1
|
Sun S, Sproviero D, Payán-Gómez C, Hoeijmakers JHJ, Maslov AY, Mastroberardino PG, Vijg J. RNA sequence analysis of somatic mutations in aging and Parkinson's Disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.26.645360. [PMID: 40196509 PMCID: PMC11974798 DOI: 10.1101/2025.03.26.645360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Parkinson's Disease (PD) is an age-related neurodegenerative disorder that has been associated with increased DNA damage. To test if PD is associated with increased somatic mutations, we analyzed RNA-seq data in whole blood from 5 visits of the Parkinson's Progression Markers Initiative for clonally amplified somatic variants. Comprehensive analysis of RNA-sequencing data revealed a total of 5,927 somatic variants (2.4 variants per sample on average). Mutation frequencies were significantly elevated in PD subjects as compared to age-matched controls at the time of the last visit. This was confirmed by RNA analysis of substantia nigra. By contrast, the fraction of carriers with clonal hematopoiesis, was significantly reduced in old PD patients as compared to old healthy controls. These results indicate that while the overall mutation rate is higher in PD, specific clonally amplified mutations are protective against PD, as has been found for Alzheimer's Disease.
Collapse
|
2
|
Yu W, Jie S, Su G, Zhuangwei N, Yiqing Z, Suhong C, Guiyuan L. The ultrafine powder of atractylodis macrocephalae rhizoma improves immune function in naturally aging rats by regulating the PI3K/Akt/NF-κB signaling pathway. Front Pharmacol 2025; 16:1550357. [PMID: 40255567 PMCID: PMC12006087 DOI: 10.3389/fphar.2025.1550357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 03/21/2025] [Indexed: 04/22/2025] Open
Abstract
Background The phenomenon of population aging presents a significant global challenge, with the aging population in China steadily increasing. As individuals progress in age, there is a gradual deterioration of human organs and systems, as well as a decline in the immune system, referred to as immunosenescence. Atractylodis macrocephalae rhizoma (BZ) has been historically used in China for its medicinal properties, including gastrointestinal improvement, immunomodulation, anti-aging, antioxidant effects, and anti-tumor effects. Nevertheless, there remains a gap in understanding the pharmacological and molecular mechanisms underlying its anti-immunosenescence effects. Methods This study employed UPLC-ESI-MS and network pharmacology to create a network map of BZ ultrafine powder (BZU) and its aging targets. Enrichment analysis was then used to identify the primary mechanistic pathways underlying BZU's anti-immunosenescence effects. The primary components of BZU were quantitatively analyzed using high-performance liquid chromatography (HPLC). Naturally aging rats were used to examine the effects of different oral doses (0.25, 0.5, and 1 g/kg) of BZU over 5 weeks on aging performance, peripheral blood immunophenotyping and cell count, and splenic lymphocyte proliferation rate. To validate the findings of network pharmacology, quantitative RT-PCR, Western blotting, and immunofluorescence analyses were conducted. Results Our analyses demonstrated that BZU improved various indicators of aging in naturally aging rats, such as increasing the number of voluntary activities, enhance grip strength and fatigue resistance, increasing the microcirculatory blood flow and improving hematological levels. The BZU administration enhanced T and B lymphocyte proliferation and significantly improved the lymphocyte-to-T cell subpopulation ratio. It can elevate serum IL-2 and IL-4 levels while reducing IL-6, IFN-γ and TNF-α levels in naturally aging rats. Finally, it increased CD3 protein expression in the spleen while decreasing protein levels of PI3K, p-AKT, IKKα/β, and NF-κB. It also decreased the mRNA expression of Pik3cg, Akt1, Pdk1 and Nfκb1. Conclusion These findings suggest that BZU may enhance lymphocyte proliferation by inhibiting the PI3K/Akt/NF-κB signaling pathway, correcting immune cell imbalances, reducing inflammatory responses, and ultimately enhancing immune function and potentially delaying aging.
Collapse
Affiliation(s)
| | | | | | | | | | - Chen Suhong
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lv Guiyuan
- College of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
3
|
Li C, Zhang C, Li X. Clonal hematopoiesis of indeterminate potential: contribution to disease and promising interventions. Mol Cell Biochem 2025:10.1007/s11010-025-05261-8. [PMID: 40140229 DOI: 10.1007/s11010-025-05261-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 03/16/2025] [Indexed: 03/28/2025]
Abstract
In clonal hematopoiesis of indeterminate potential (CHIP), subpopulations of blood cells carrying somatic mutations expand as the individual ages, and this expansion may elevate risk of blood cancers as well as cardiovascular disease. Individuals at higher risk of CHIP and therefore of CHIP-associated disease can be identified through mutational profiling, and the apparently central role of inflammation in CHIP-associated disease has emerged as a potential therapeutic target. While CHIP is often associated with negative health outcomes, emerging evidence suggests that some CHIP-related mutations may also exert beneficial effects, indicating a more complex role in human health. This review examines current understanding of the epidemiology and clinical significance of CHIP and the role of inflammation in driving its association with disease risk. It explores the mechanisms linking CHIP to inflammation and risk of cardiovascular and other diseases, as well as the potential of personalizing therapies against those diseases for individuals with CHIP.
Collapse
Affiliation(s)
- Chongjie Li
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China
- School of Pharmacy, Southwest Medical University, LuZhou, 646000, Sichuan, People's Republic of China
| | - Chunxiang Zhang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
- School of Pharmacy, Southwest Medical University, LuZhou, 646000, Sichuan, People's Republic of China.
| | - Xiuying Li
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, People's Republic of China.
- School of Pharmacy, Southwest Medical University, LuZhou, 646000, Sichuan, People's Republic of China.
| |
Collapse
|
4
|
Ryu G, Koh Y, Jaiswal S, Yoon SS. Clonal hematopoiesis: elements associated with clonal expansion and diseases. Blood Res 2025; 60:17. [PMID: 40080235 PMCID: PMC11906933 DOI: 10.1007/s44313-025-00065-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2025] [Accepted: 02/24/2025] [Indexed: 03/15/2025] Open
Abstract
Clonal hematopoiesis (CH), characterized by the expansion of hematopoietic stem and progenitor cells harboring somatic mutations, has emerged as a significant age-related phenomenon with profound implications for human health. While initially recognized in the 1960s, recent technological advances have revealed its complex nature and widespread prevalence, affecting up to 84% of individuals aged ≥ 70 years. The clinical significance of CH extends beyond its well-established role as a precursor to hematological malignancies, encompassing its association with cardiovascular diseases, chronic kidney disease, and other non-malignant disorders. This comprehensive review synthesizes the current understanding of CH, focusing on recent advances in genetic and molecular mechanisms, particularly the roles of commonly mutated genes such as DNMT3A, TET2, and ASXL1. We address the emerging distinction between myeloid and lymphoid CH, their differential impacts on disease progression, and the complex interplay between CH and inflammation. Special attention is given to newly identified genetic determinants of clonal expansion rates and their implications for disease progression. The review also examines the revolutionary concept of passenger-approximated clonal expansion rate and its utility in understanding CH dynamics. Furthermore, we discuss therapeutic strategies targeting inflammatory pathways and their potential in mitigating CH-associated complications. By integrating recent findings from genetic, molecular, and clinical studies, this review provides a framework for understanding CH as a systemic condition and highlights promising directions for therapeutic interventions.
Collapse
Affiliation(s)
- Gangpyo Ryu
- Cancer Research Institute, Seoul National University, Seoul, Korea
- The Interdisciplinary Program of Cancer Biology, Seoul National University, Seoul, Korea
- Department of Pathology, Stanford University, Stanford, CA, USA
| | - Youngil Koh
- Cancer Research Institute, Seoul National University, Seoul, Korea.
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.
| | - Siddhartha Jaiswal
- Department of Pathology, Stanford University, Stanford, CA, USA.
- Stanford Cardiovascular Institute, Stanford University, Stanford, CA, USA.
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University, Stanford, CA, USA.
| | - Sung-Soo Yoon
- Cancer Research Institute, Seoul National University, Seoul, Korea.
- The Interdisciplinary Program of Cancer Biology, Seoul National University, Seoul, Korea.
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Korea.
- College of Medicine, Seoul National University, Seoul, Korea.
| |
Collapse
|
5
|
Yang Y, Cathelin S, Liu ACH, Subedi A, Maher A, Hosseini M, Manikoth Ayyathan D, Vanner R, Chan SM. TET2 deficiency increases the competitive advantage of hematopoietic stem and progenitor cells through upregulation of thrombopoietin receptor signaling. Nat Commun 2025; 16:2384. [PMID: 40064887 PMCID: PMC11894142 DOI: 10.1038/s41467-025-57614-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
Ten-Eleven Translocation-2 (TET2) mutations drive the expansion of mutant hematopoietic stem cells (HSCs) in clonal hematopoiesis (CH). However, the precise mechanisms by which TET2 mutations confer a competitive advantage to HSCs remain unclear. Here, through an epigenetic drug screen, we discover that inhibition of disruptor of telomeric silencing 1-like (DOT1L), a H3K79 methyltransferase, selectively reduces the fitness of Tet2 knockout (Tet2KO) hematopoietic stem and progenitor cells (HSPCs). Mechanistically, we find that TET2 deficiency increases H3K79 dimethylation and expression of Mpl, which encodes the thrombopoietin receptor (TPO-R). Correspondingly, TET2 deficiency is associated with a higher proportion of primitive Mpl-expressing (Mpl+) cells in the HSC compartment. Importantly, inhibition of Mpl expression or the signaling downstream of TPO-R is sufficient to reduce the competitive advantage of murine and human TET2-deficient HSPCs. Our findings demonstrate a critical role for aberrant TPO-R signaling in TET2 mutation-driven CH and uncover potential therapeutic strategies against this condition.
Collapse
Affiliation(s)
- Yitong Yang
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | - Severine Cathelin
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | - Alex C H Liu
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, M5G 1L7, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | - Amit Subedi
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | - Abdula Maher
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
- Institute of Medical Science, Temerty Faculty of Medicine, University of Toronto, Toronto, M5S 1A8, Canada
| | - Mohsen Hosseini
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | | | - Robert Vanner
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada
| | - Steven M Chan
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, M5G 1L7, Canada.
- Princess Margaret Cancer Centre, University Health Network, Toronto, M5G 1L7, Canada.
| |
Collapse
|
6
|
Tan Z, Zhang X, Feng J, Zhao Y, Hu H, Wu D, Yu Q, Zhang Y, Wu L, Hu T, Yan Z, Ye B, Liu W. Hematopoietic stem cell transplantation and immunosuppressive therapy: implications of clonal haematopoiesis. Ann Hematol 2025; 104:1877-1886. [PMID: 39873798 PMCID: PMC12031881 DOI: 10.1007/s00277-024-06152-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025]
Abstract
Aplastic anemia (AA) is a life-threatening bone marrow failure syndrome. The advent of next-generation sequencing (NGS) has shed light on the link between somatic mutations (SM) and the efficacy of immunosuppressive therapy (IST) in AA patients. However, the relationship between SM and hematopoietic stem cell transplantation (HSCT) has not been extensively explored. In this retrospective analysis, we examined 166 AA patients who received HSCT or IST at our institution between May 2019 and December 2023. NGS was conducted on 66 genes within bone marrow cells to investigate the correlation between SM and the prognosis and therapeutic response in AA patients, as well as to assess the impact of mutation types on HSCT outcomes. Clinical data were gathered from 166 AA patients, comprising 84 males and 82 females, with a median age of 32 years (ranging from 9 to 75 years). In our study, a total of 151 somatic mutations were identified across 84 patients (50.6%), with 42 patients (25.3%) presenting a single mutation and 26 patients (15.7%) harboring two mutations. The top five genes with the highest mutation frequency were BCOR/BCORL1 (12.6%), ASXL1 (8.6%), TET2 (6.6%), CEBPA (5.3%), and GATA2 (4.6%). We stratified patients into SM and No-SM groups based on the presence of mutations and further divided them into HSCT and IST groups to assess the influence of mutation types on treatment response and survival within and between these groups. The findings were as follows: 1.Patients in the HSCT group exhibited a higher treatment response (OR 85.9% vs. 68.4%, p < 0.05), although there was no significant difference in survival. 2.Patients with favorable mutations, such as PIGA and BCOR/BCORL1, experienced significantly improved response and survival compared to those with unfavorable mutations like ASXL1, DNMT3A, and TET2 (OR 93.7% vs. 72%, p < 0.05) (3-year OS 93.7% vs. 80%, p > 0.05). 3.The HSCT-Favorable group demonstrated superior response rates (OR 100% vs. 67.7%, p < 0.05) and longer survival (3-year OS 100% vs. 67.7%, p < 0.05) compared to the IST-Favorable group. This study underscores that AA patients carrying favorable mutations, particularly BCOR/BCORL1, tend to have a more robust response and better prognosis than those without mutations or those with unfavorable mutations, such as ASXL1/DNMT3A. These findings are especially pertinent to HSCT, highlighting the importance of NGS prior to initiating treatment.
Collapse
Affiliation(s)
- Zhengwei Tan
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinhe Zhang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jia Feng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuechao Zhao
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Huijin Hu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Dijiong Wu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Qinghong Yu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Yu Zhang
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Liqiang Wu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Tonglin Hu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Zhengsong Yan
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Baodong Ye
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Wenbin Liu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China.
| |
Collapse
|
7
|
Kallai A, Ungvari Z, Fekete M, Maier AB, Mikala G, Andrikovics H, Lehoczki A. Genomic instability and genetic heterogeneity in aging: insights from clonal hematopoiesis (CHIP), monoclonal gammopathy (MGUS), and monoclonal B-cell lymphocytosis (MBL). GeroScience 2025; 47:703-720. [PMID: 39405013 PMCID: PMC11872960 DOI: 10.1007/s11357-024-01374-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 10/01/2024] [Indexed: 03/04/2025] Open
Abstract
Aging is a multifaceted process characterized by a gradual decline in physiological function and increased susceptibility to a range of chronic diseases. Among the molecular and cellular mechanisms driving aging, genomic instability is a fundamental hallmark, contributing to increased mutation load and genetic heterogeneity within cellular populations. This review explores the role of genomic instability and genetic heterogeneity in aging in the hematopoietic system, with a particular focus on clonal hematopoiesis of indeterminate potential (CHIP), monoclonal gammopathy of undetermined significance (MGUS), and monoclonal B-cell lymphocytosis (MBL) as biomarkers. CHIP involves the clonal expansion of hematopoietic stem cells with somatic mutations. In contrast, MGUS is characterized by the presence of clonal plasma cells producing monoclonal immunoglobulins, while MBL is characterized by clonal proliferation of B cells. These conditions are prevalent in the aging population and serve as measurable indicators of underlying genomic instability. Studying these entities offers valuable insights into the mechanisms by which somatic mutations accumulate and drive clonal evolution in the hematopoietic system, providing a deeper understanding of how aging impacts cellular and tissue homeostasis. In summary, the hematopoietic system serves as a powerful model for investigating the interplay between genomic instability and aging. Incorporating age-related hematological conditions into aging research, alongside other biomarkers such as epigenetic clocks, can enhance the precision and predictive power of biological age assessments. These biomarkers provide a comprehensive view of the aging process, facilitating the early detection of age-related diseases and hopefully enabling personalized healthcare strategies.
Collapse
Affiliation(s)
- Attila Kallai
- Healthy Aging Program, Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
| | - Zoltan Ungvari
- Vascular Cognitive Impairment, Neurodegeneration and Healthy Brain Aging Program, Department of Neurosurgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Stephenson Cancer Center, University of Oklahoma, Oklahoma City, OK, USA
- Oklahoma Center for Geroscience and Healthy Brain Aging, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Department of Health Promotion Sciences, College of Public Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
- Doctoral College/Institute of Preventive Medicine and Public Health, International Training Program in Geroscience, Semmelweis University, Budapest, Hungary
| | - Mónika Fekete
- Healthy Aging Program, Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
| | - Andrea B Maier
- Department of Medicine and Aged Care, @AgeMelbourne, The Royal Melbourne Hospital, The University of Melbourne, Parkville, VIC, Australia
- Department of Human Movement Sciences, @AgeAmsterdam, Faculty of Behavioural and Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, the Netherlands
- Centre for Healthy Longevity, @AgeSingapore, National University Health System, Singapore, Republic of Singapore
- Healthy Longevity Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, @AgeSingapore, National University Health System, Singapore, Republic of Singapore
| | - Gabor Mikala
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary
- Department of Hematology and Stem Cell Transplantation, Central Hospital of Southern Pest, National Institute for Hematology and Infectious Diseases, Budapest, Hungary
| | - Hajnalka Andrikovics
- Healthy Aging Program, Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary
- Laboratory of Molecular Genetics, Central Hospital of Southern Pest, National Institute for Hematology and Infectious Diseases, Budapest, Hungary
| | - Andrea Lehoczki
- Healthy Aging Program, Institute of Preventive Medicine and Public Health, Semmelweis University, Budapest, Hungary.
- Doctoral College, Health Sciences Program, Semmelweis University, Budapest, Hungary.
| |
Collapse
|
8
|
Xie Y, Kazakova V, Weeks LD, Gerber JM, Tai J, Zhang TY, Lowsky R, Wu X, Yang C, Patel SA. Effects of donor-engrafted clonal hematopoiesis in allogeneic and autologous stem cell transplantation: a systematic review and meta-analysis. Bone Marrow Transplant 2024; 59:1585-1593. [PMID: 39183321 PMCID: PMC11531373 DOI: 10.1038/s41409-024-02403-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/13/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
Donor stem cell health may be critically important to the success of hematopoietic stem cell transplantation (HSCT). Herein, we performed this systematic review and meta-analysis including meta-regression to assess the impact of donor-engrafted clonal hematopoiesis (CH) in allogeneic HSCT (allo-HSCT) and impact of pre-transplant CH in autologous HSCT (auto-HSCT). We applied random-effects models to analyze 5 allo-HSCT studies with 3192 donor-recipient pairs and 9 auto-HSCT studies with 2854 patients. We found that donor-engrafted CH after allo-HSCT decreased the risk of disease relapse [Hazard Ratio (HR) = 0.79, 95% Confidence Interval (CI): (0.67, 0.93)], but did not affect overall survival (OS) [HR = 0.91, 95% CI: (0.75, 1.11)], progression-free survival (PFS) [HR = 0.94, 95% CI: (0.63, 1.41)], or non-relapse mortality [HR = 1.06, 95% CI: (0.81, 1.39)]. In contrast, pre-transplant CH in auto-HSCT recipients resulted in inferior OS [HR = 1.30, 95% CI: (1.16, 1.46)], inferior PFS [HR = 1.35, 95% CI: (1.18, 1.54)], and higher risk for therapy-related myeloid neoplasm [HR = 4.85, 95% CI: (2.39, 9.82)] when compared to auto-HSCT recipients without CH. This study sheds light onto the debate about prospective "CHIP screening" for stem cell donors and addresses the impact of CH as a transmissible phenomenon.
Collapse
Affiliation(s)
- Yiyu Xie
- Dept. of Medicine - Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA
| | - Vera Kazakova
- Dept. of Medicine - Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA
| | - Lachelle D Weeks
- Center for Early Detection and Interception of Blood Cancers, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jonathan M Gerber
- Dept. of Medicine - Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA
- Center for Clinical and Translational Science, UMass Chan Medical School, Worcester, MA, USA
| | - Jesse Tai
- Dept. of Medicine - Division of Hematology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Tian Y Zhang
- Dept. of Medicine - Division of Hematology, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Robert Lowsky
- Dept. of Medicine - Division of Blood & Marrow Transplantation & Cellular Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Xiaojin Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Chengwu Yang
- Measurement, Outcome, and Design Section, Division of Biostatistics and Health Service Research, Department of Population and Quantitative Health Sciences, T.H. Chan School of Medicine, UMass Chan Medical School, Worcester, MA, USA.
- Department of Obstetrics & Gynecology, T.H. Chan School of Medicine, UMass Chan Medical School, Worcester, MA, USA.
| | - Shyam A Patel
- Dept. of Medicine - Division of Hematology/Oncology, UMass Memorial Medical Center, UMass Chan Medical School, Worcester, MA, USA.
- Center for Clinical and Translational Science, UMass Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
9
|
Woo KA, Kim HJ, Lee CY, Shin JH, Sun C, Im H, An H, Lim J, Choi SY, Koh Y, Jeon B. Parkinson's disease is associated with clonal hematopoiesis with TET2 mutation. NPJ Parkinsons Dis 2024; 10:168. [PMID: 39242596 PMCID: PMC11379878 DOI: 10.1038/s41531-024-00784-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 08/27/2024] [Indexed: 09/09/2024] Open
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP), a premalignant expansion of mutated hematopoietic stem cells, is linked to immune alterations. Given the role of neuroinflammation and immune dysfunction in Parkinson's disease (PD), we hypothesized a connection between CHIP and PD. We analyzed peripheral blood DNA from 341 PD, 92 isolated REM sleep behavior disorder (iRBD) patients, and 5003 controls using targeted sequencing of 24 genes associated with hematologic neoplasms. PD cases were classified by clinical progression mode: fast, slow, and typical. Using multivariable logistic regression models, CHIP prevalence was assessed against controls with a 1.0% variant allele fraction threshold. CHIP with TET2 mutations was more prevalent in PD than controls (aOR 1.75, 95% CI 1.11-2.77, p = 0.017), particularly in the fast motor progression subgroup (aOR 3.19, p = 0.004). No distinct associations were observed with iRBD. PD is linked to increased odds of CHIP with TET2 mutations, suggesting immune dysregulation in PD pathophysiology.
Collapse
Affiliation(s)
- Kyung Ah Woo
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Han-Joon Kim
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea.
| | - Chan Young Lee
- Department of Neurology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | - Jung Hwan Shin
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | | | - Hogune Im
- NOBO Medicine Inc, Seoul, Republic of Korea
| | - Hongyul An
- NOBO Medicine Inc, Seoul, Republic of Korea
| | - Jiwoo Lim
- NOBO Medicine Inc, Seoul, Republic of Korea
| | - Su-Yeon Choi
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul National University Hospital Healthcare System Gangnam Center, Seoul, Republic of Korea
| | - Youngil Koh
- NOBO Medicine Inc, Seoul, Republic of Korea
- Department of Internal Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Beomseok Jeon
- Department of Neurology, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
10
|
Burren OS, Dhindsa RS, Deevi SVV, Wen S, Nag A, Mitchell J, Hu F, Loesch DP, Smith KR, Razdan N, Olsson H, Platt A, Vitsios D, Wu Q, Codd V, Nelson CP, Samani NJ, March RE, Wasilewski S, Carss K, Fabre M, Wang Q, Pangalos MN, Petrovski S. Genetic architecture of telomere length in 462,666 UK Biobank whole-genome sequences. Nat Genet 2024; 56:1832-1840. [PMID: 39192095 PMCID: PMC11387196 DOI: 10.1038/s41588-024-01884-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Accepted: 07/25/2024] [Indexed: 08/29/2024]
Abstract
Telomeres protect chromosome ends from damage and their length is linked with human disease and aging. We developed a joint telomere length metric, combining quantitative PCR and whole-genome sequencing measurements from 462,666 UK Biobank participants. This metric increased SNP heritability, suggesting that it better captures genetic regulation of telomere length. Exome-wide rare-variant and gene-level collapsing association studies identified 64 variants and 30 genes significantly associated with telomere length, including allelic series in ACD and RTEL1. Notably, 16% of these genes are known drivers of clonal hematopoiesis-an age-related somatic mosaicism associated with myeloid cancers and several nonmalignant diseases. Somatic variant analyses revealed gene-specific associations with telomere length, including lengthened telomeres in individuals with large SRSF2-mutant clones, compared with shortened telomeres in individuals with clonal expansions driven by other genes. Collectively, our findings demonstrate the impact of rare variants on telomere length, with larger effects observed among genes also associated with clonal hematopoiesis.
Collapse
Affiliation(s)
- Oliver S Burren
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Ryan S Dhindsa
- Center for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
| | - Sri V V Deevi
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Sean Wen
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Abhishek Nag
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Jonathan Mitchell
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Fengyuan Hu
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Douglas P Loesch
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Katherine R Smith
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Neetu Razdan
- Biosciences COPD & IPF, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gaithersburg, MD, USA
| | - Henric Olsson
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden
| | - Adam Platt
- Translational Science and Experimental Medicine, Research and Early Development, Respiratory & Immunology, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Dimitrios Vitsios
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Qiang Wu
- Center for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
- Department of Mathematical Sciences, Middle Tennessee State University, Murfreesboro, TN, USA
| | - Veryan Codd
- Department of Cardiovascular Sciences, University of Leicester and Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Christopher P Nelson
- Department of Cardiovascular Sciences, University of Leicester and Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester and Leicester NIHR Biomedical Research Centre, Leicester, UK
| | - Ruth E March
- Precision Medicine & Biosamples, Oncology R&D, AstraZeneca, Dublin, Ireland
| | - Sebastian Wasilewski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Keren Carss
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - Margarete Fabre
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK
- Department of Haematology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Quanli Wang
- Center for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Waltham, MA, USA
| | | | - Slavé Petrovski
- Centre for Genomics Research, Discovery Sciences, BioPharmaceuticals R&D, AstraZeneca, Cambridge, UK.
- Department of Medicine, University of Melbourne, Austin Health, Melbourne, Victoria, Australia.
| |
Collapse
|
11
|
Wilcox NS, Amit U, Reibel JB, Berlin E, Howell K, Ky B. Cardiovascular disease and cancer: shared risk factors and mechanisms. Nat Rev Cardiol 2024; 21:617-631. [PMID: 38600368 PMCID: PMC11324377 DOI: 10.1038/s41569-024-01017-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/20/2024] [Indexed: 04/12/2024]
Abstract
Cardiovascular disease (CVD) and cancer are among the leading causes of morbidity and mortality globally, and these conditions are increasingly recognized to be fundamentally interconnected. In this Review, we present the current epidemiological data for each of the modifiable risk factors shared by the two diseases, including hypertension, hyperlipidaemia, diabetes mellitus, obesity, smoking, diet, physical activity and the social determinants of health. We then review the epidemiological data demonstrating the increased risk of CVD in patients with cancer, as well as the increased risk of cancer in patients with CVD. We also discuss the shared mechanisms implicated in the development of these conditions, highlighting their inherent bidirectional relationship. We conclude with a perspective on future research directions for the field of cardio-oncology to advance the care of patients with CVD and cancer.
Collapse
Affiliation(s)
- Nicholas S Wilcox
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Uri Amit
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jacob B Reibel
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Division of Hematology Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Eva Berlin
- Department of Radiation Oncology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kendyl Howell
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bonnie Ky
- Division of Cardiology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Biostatistics, Epidemiology and Informatics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
12
|
Jurgens SJ, Wang X, Choi SH, Weng LC, Koyama S, Pirruccello JP, Nguyen T, Smadbeck P, Jang D, Chaffin M, Walsh R, Roselli C, Elliott AL, Wijdeveld LFJM, Biddinger KJ, Kany S, Rämö JT, Natarajan P, Aragam KG, Flannick J, Burtt NP, Bezzina CR, Lubitz SA, Lunetta KL, Ellinor PT. Rare coding variant analysis for human diseases across biobanks and ancestries. Nat Genet 2024; 56:1811-1820. [PMID: 39210047 DOI: 10.1038/s41588-024-01894-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 08/01/2024] [Indexed: 09/04/2024]
Abstract
Large-scale sequencing has enabled unparalleled opportunities to investigate the role of rare coding variation in human phenotypic variability. Here, we present a pan-ancestry analysis of sequencing data from three large biobanks, including the All of Us research program. Using mixed-effects models, we performed gene-based rare variant testing for 601 diseases across 748,879 individuals, including 155,236 with ancestry dissimilar to European. We identified 363 significant associations, which highlighted core genes for the human disease phenome and identified potential novel associations, including UBR3 for cardiometabolic disease and YLPM1 for psychiatric disease. Pan-ancestry burden testing represented an inclusive and useful approach for discovery in diverse datasets, although we also highlight the importance of ancestry-specific sensitivity analyses in this setting. Finally, we found that effect sizes for rare protein-disrupting variants were concordant between samples similar to European ancestry and other genetic ancestries (βDeming = 0.7-1.0). Our results have implications for multi-ancestry and cross-biobank approaches in sequencing association studies for human disease.
Collapse
Affiliation(s)
- Sean J Jurgens
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Xin Wang
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Seung Hoan Choi
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
| | - Lu-Chen Weng
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Satoshi Koyama
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - James P Pirruccello
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Cardiology, University of California, San Francisco, CA, USA
| | - Trang Nguyen
- Metabolism Program, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Patrick Smadbeck
- Metabolism Program, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Dongkeun Jang
- Metabolism Program, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Mark Chaffin
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Roddy Walsh
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Carolina Roselli
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Amanda L Elliott
- Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Psychiatry and Center for Genomic Medicine, Psychiatric and Neurodevelopmental Genetics Unit, Massachusetts General Hospital,Harvard Medical School, Boston, MA, USA
- Center for Psychiatric Research, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Leonoor F J M Wijdeveld
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Physiology, Amsterdam UMC location VU, Amsterdam, The Netherlands
| | - Kiran J Biddinger
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shinwan Kany
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Cardiology, University Heart and Vascular Center Hamburg-Eppendorf, Hamburg, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Hamburg/Kiel/Lübeck, Hamburg, Germany
| | - Joel T Rämö
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Institute for Molecular Medicine Finland (FIMM), Helsinki Institute of Life Science (HiLIFE), University of Helsinki, Helsinki, Finland
| | - Pradeep Natarajan
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Center for Genomic Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Krishna G Aragam
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Jason Flannick
- Metabolism Program, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Division of Genetics and Genomics, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Noël P Burtt
- Metabolism Program, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Program in Medical and Population Genetics, The Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Connie R Bezzina
- Department of Experimental Cardiology, Heart Center, Amsterdam Cardiovascular Sciences, Heart Failure and Arrhythmias, Amsterdam UMC location University of Amsterdam, Amsterdam, The Netherlands
| | - Steven A Lubitz
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA
| | - Kathryn L Lunetta
- Department of Biostatistics, Boston University School of Public Health, Boston, MA, USA
- NHLBI and Boston University's Framingham Heart Study, Framingham, MA, USA
| | - Patrick T Ellinor
- Cardiovascular Disease Initiative, The Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Cardiovascular Research Center, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
- Demoulas Center for Cardiac Arrhythmias, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
13
|
Muhsen IN, Zubair AC, Niederwieser T, Hashmi SK. Space exploration and cancer: the risks of deeper space adventures. Leukemia 2024; 38:1872-1875. [PMID: 38969730 DOI: 10.1038/s41375-024-02298-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 05/06/2024] [Accepted: 05/29/2024] [Indexed: 07/07/2024]
Affiliation(s)
- Ibrahim N Muhsen
- Section of Hematology and Oncology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Abba C Zubair
- Department of Laboratory Medicine and Pathology, Jacksonville, FL, USA
- Sheikh Shakhbout Medical City-Mayo Clinic Joint Venture, Abu Dhabi, UAE
| | - Tobias Niederwieser
- University of Colorado Boulder, Ann and H.J. Smead Department of Aerospace Engineering Sciences, BioServe Space Technologies, 429 UCB, Boulder, CO, 80309, USA
| | - Shahrukh K Hashmi
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN, USA.
- Department of Medicine, Sheikh Shakbout Medical City, Abu Dhabi, UAE.
- Medical and Clinical Affairs, Khalifa University, Abu Dhabi, UAE.
| |
Collapse
|
14
|
Werneth CM, Patel ZS, Thompson MS, Blattnig SR, Huff JL. Considering clonal hematopoiesis of indeterminate potential in space radiation risk analysis for hematologic cancers and cardiovascular disease. COMMUNICATIONS MEDICINE 2024; 4:105. [PMID: 38862635 PMCID: PMC11166645 DOI: 10.1038/s43856-023-00408-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 11/16/2023] [Indexed: 06/13/2024] Open
Abstract
BACKGROUND Expanding human presence in space through long-duration exploration missions and commercial space operations warrants improvements in approaches for quantifying crew space radiation health risks. Currently, risk assessment models for radiogenic cancer and cardiovascular disease consider age, sex, and tobacco use, but do not incorporate other modifiable (e.g., body weight, physical activity, diet, environment) and non-modifiable individual risk factors (e.g., genetics, medical history, race/ethnicity, family history) that may greatly influence crew health both in-mission and long-term. For example, clonal hematopoiesis of indeterminate potential (CHIP) is a relatively common age-related condition that is an emerging risk factor for a variety of diseases including cardiovascular disease and cancer. CHIP carrier status may therefore exacerbate health risks associated with space radiation exposure. METHODS In the present study, published CHIP hazard ratios were used to modify background hazard rates for coronary heart disease, stroke, and hematologic cancers in the National Aeronautics and Space Administration space radiation risk assessment model. The risk of radiation exposure-induced death for these endpoints was projected for a future Mars exploration mission scenario. RESULTS Here we show appreciable increases in the lifetime risk of exposure-induced death for hematologic malignancies, coronary heart disease, and stroke, which are observed as a function of age after radiation exposure for male and female crew members that are directly attributable to the elevated health risks for CHIP carriers. CONCLUSIONS We discuss the importance of evaluating individual risk factors such as CHIP as part of a comprehensive space radiation risk assessment strategy aimed at effective risk communication and disease surveillance for astronauts embarking on future exploration missions.
Collapse
Affiliation(s)
| | - Zarana S Patel
- Center for Scientific Review, National Institutes of Health, Bethesda, MD, USA
| | | | | | | |
Collapse
|
15
|
Graham JH, Schlachetzki JCM, Yang X, Breuss MW. Genomic Mosaicism of the Brain: Origin, Impact, and Utility. Neurosci Bull 2024; 40:759-776. [PMID: 37898991 PMCID: PMC11178748 DOI: 10.1007/s12264-023-01124-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/16/2023] [Indexed: 10/31/2023] Open
Abstract
Genomic mosaicism describes the phenomenon where some but not all cells within a tissue harbor unique genetic mutations. Traditionally, research focused on the impact of genomic mosaicism on clinical phenotype-motivated by its involvement in cancers and overgrowth syndromes. More recently, we increasingly shifted towards the plethora of neutral mosaic variants that can act as recorders of cellular lineage and environmental exposures. Here, we summarize the current state of the field of genomic mosaicism research with a special emphasis on our current understanding of this phenomenon in brain development and homeostasis. Although the field of genomic mosaicism has a rich history, technological advances in the last decade have changed our approaches and greatly improved our knowledge. We will provide current definitions and an overview of contemporary detection approaches for genomic mosaicism. Finally, we will discuss the impact and utility of genomic mosaicism.
Collapse
Affiliation(s)
- Jared H Graham
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado School of Medicine, Aurora, 80045-2581, CO, USA
| | - Johannes C M Schlachetzki
- Department of Cellular and Molecular Medicine, University of California San Diego, La Jolla, 92093-0021, San Diego, CA, USA
| | - Xiaoxu Yang
- Department of Neurosciences, University of California San Diego, La Jolla, 92093-0021, San Diego, CA, USA
- Rady Children's Institute for Genomic Medicine, San Diego, 92123, CA, USA
| | - Martin W Breuss
- Department of Pediatrics, Section of Clinical Genetics and Metabolism, University of Colorado School of Medicine, Aurora, 80045-2581, CO, USA.
| |
Collapse
|
16
|
Desai A, Pasquina LW, Nulsen C, Keller-Evans RB, Mata DA, Tukachinsky H, Oxnard GR. Putting comprehensive genomic profiling of ctDNA to work: 10 proposed use cases. THE JOURNAL OF LIQUID BIOPSY 2024; 4:100140. [PMID: 40027147 PMCID: PMC11863816 DOI: 10.1016/j.jlb.2024.100140] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 03/05/2025]
Abstract
Liquid biopsy profiling of circulating tumor DNA (ctDNA) has become established as a compelling, pragmatic diagnostic in the care of cancer patients and is now endorsed by multiple cancer care guidelines. Moreover, ctDNA profiling technologies have advanced significantly and offer increasingly comprehensive and reliable insights into cancer. In this review, we focus on applications of ctDNA and propose that a critical untapped opportunity is in considering how we utilize these accessible, scalable technologies across diverse potential applications. With a specific focus on clinical applications, rather than research uses, we describe 10 use cases for ctDNA profiling across four categories: (1) established and (2) emerging applications of ctDNA profiling for therapy selection, (3) incidental detection of secondary genomic findings, and (4) quantification of plasma DNA tumor content.
Collapse
Affiliation(s)
- Aakash Desai
- Division of Hematology and Oncology, Department of Medicine, University of Alabama at Birmingham, AL, USA
| | | | | | | | | | | | | |
Collapse
|
17
|
Lorenzi L, Haferlach T, Mori L, Simbeni M, Walter W, Balzarini P, Meggendorfer M, Döring C, Lonardi S, Bugatti M, Agostinelli C, Mehta J, Borges A, Agaimy A, Simonitsch-Klupp I, Cabeçadas J, Campo E, Pileri SA, Facchetti F, Leo Hansmann M, Hartmann S. Massive parallel sequencing unveils homologous recombination deficiency in follicular dendritic cell sarcoma. Haematologica 2024; 109:1815-1824. [PMID: 37994105 PMCID: PMC11141687 DOI: 10.3324/haematol.2023.283669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 11/13/2023] [Indexed: 11/24/2023] Open
Abstract
Standardized treatment options are lacking for patients with unresectable or multifocal follicular dendritic cell sarcoma (FDCS) and disease-related mortality is as high as 20%. Applying whole-genome sequencing (WGS) in one case and whole-exome sequencing (WES) in additional twelve cases, this study adds information on the molecular landscape of FDCS, expanding knowledge on pathobiological mechanisms and identifying novel markers of potential theragnostic significance. Massive parallel sequencing showed high frequency of mutations on oncosuppressor genes, particularly in RB1, CARS and BRCA2 and unveiled alterations on homologous recombination DNA damage repair-related genes in 70% (9/13) of cases. This indicates that patients with high-stage FDCS may be eligible for poly ADP ribose polymerase inhibition protocols. Low tumor mutational burden was confirmed in this study despite common PDL1 expression in FDCS arguing on the efficacy of immune checkpoint inhibitors. CDKN2A deletion, detected by WGS and confirmed by fluorescence in situ hybridization in 41% of cases (9/22) indicates that impairment of cell cycle regulation may sustain oncogenesis in FDCS. Absence of mutations in the RAS/RAF/MAPK pathway and lack of clonal hematopoiesis-related mutations in FDCS sanction its differences from dendritic cell-derived neoplasms of hematopoietic derivation. WGS and WES in FDCS provides additional information on the molecular landscape of this rare tumor, proposing novel candidate genes for innovative therapeutical approaches to improve survival of patients with multifocal disease.
Collapse
Affiliation(s)
- Luisa Lorenzi
- Pathology Unit, ASST Spedali Civili di Brescia, Department of Molecular and Translational Medicine, University of Brescia, Brescia.
| | | | - Luigi Mori
- Laboratory of Molecular Medicine, Department of Clinical and Experimental Science, University of Brescia, Brescia
| | - Matteo Simbeni
- Pathology Unit, ASST Spedali Civili di Brescia, Department of Molecular and Translational Medicine, University of Brescia, Brescia
| | | | - Piera Balzarini
- Pathology Unit, ASST Spedali Civili di Brescia, Department of Molecular and Translational Medicine, University of Brescia, Brescia
| | | | - Claudia Döring
- Dr Senckenberg Institute of Pathology, Goethe University, Frankfurt
| | - Silvia Lonardi
- Pathology Unit, ASST Spedali Civili di Brescia, Department of Molecular and Translational Medicine, University of Brescia, Brescia
| | - Mattia Bugatti
- Pathology Unit, ASST Spedali Civili di Brescia, Department of Molecular and Translational Medicine, University of Brescia, Brescia
| | - Claudio Agostinelli
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy"
| | | | | | - Abbas Agaimy
- Institute of Pathology, University Hospital, Erlangen
| | | | - José Cabeçadas
- Department of Pathology, Portuguese Institute of Oncology, Lisbon, Portugal
| | - Elias Campo
- Hematopathology Section, Hospital Clinic, IDIBAPS, University of Barcelona, Barcelona, Spain
| | - Stefano Aldo Pileri
- Division of Haematopathology, European Institute of Oncology (IEO) IRCCS, Milan
| | - Fabio Facchetti
- Pathology Unit, ASST Spedali Civili di Brescia, Department of Molecular and Translational Medicine, University of Brescia, Brescia
| | - Martin Leo Hansmann
- Institute for General Pharmacology and Toxicology, Goethe University, Frankfurt
| | - Sylvia Hartmann
- Dr Senckenberg Institute of Pathology, Goethe University, Frankfurt
| |
Collapse
|
18
|
Ogurchenok NE, Khalin KD, Bryukhovetskiy IS. Chemoprophylaxis of precancerous lesions in patients who are at a high risk of developing colorectal cancer (Review). MEDICINE INTERNATIONAL 2024; 4:25. [PMID: 38628384 PMCID: PMC11019464 DOI: 10.3892/mi.2024.149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Accepted: 03/07/2024] [Indexed: 04/19/2024]
Abstract
The diagnostics of colorectal cancer (CRC) and precancerous lesions in the colon is one of the most urgent matters to be considered for the modern protocols of complex examination, recommended for use from the age of 45 years, and including both instrumental and laboratory methods of research: Colonoscopy, CT colonography, flexible sigmoidoscopy, fecal occult blood test, fecal immunohistochemistry test and stool DNA test Nevertheless, the removal of those precancerous lesions does not solve the issue, and, apart from the regular endoscopic monitoring of patients who are at a high risk of developing CRC, the pharmacological treatment of certain key pathogenic mechanisms leading to the development of CRC is required. The present review to discusses the function of β-catenin in the transformation of precancerous colorectal lesions into CRC, when collaborating with PI3K/AKT/mTOR signaling pathway and other mechanisms. The existing methods for the early diagnostics and prevention of discovered anomalies are described and categorized. The analysis of the approaches to chemoprophylaxis of CRC, depending on the results of endoscopic, morphological and molecular-genetic tests, is presented.
Collapse
Affiliation(s)
- Nonna E. Ogurchenok
- Far Eastern Federal University, School of Medicine and Life Sciences, FEFU Medical Center, Russky Island, 690091 Vladivostok, Russian Federation
- Primorskiy Regional Clinical Hospital N1, Medical Center, Russky Island, 690091 Vladivostok, Russian Federation
| | - Konstantin D. Khalin
- Far Eastern Federal University, School of Medicine and Life Sciences, FEFU Medical Center, Russky Island, 690091 Vladivostok, Russian Federation
- Far Eastern Federal University, Medical Center, Russky Island, 690091 Vladivostok, Russian Federation
| | - Igor S. Bryukhovetskiy
- Far Eastern Federal University, Medical Center, Russky Island, 690091 Vladivostok, Russian Federation
| |
Collapse
|
19
|
Daman AW, Cheong JG, Berneking L, Josefowicz SZ. The potency of hematopoietic stem cell reprogramming for changing immune tone. Immunol Rev 2024; 323:197-208. [PMID: 38632868 DOI: 10.1111/imr.13335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/19/2024]
Abstract
Innate immune memory endows innate immune cells with antigen independent heightened responsiveness to subsequent challenges. The durability of this response can be mediated by inflammation induced epigenetic and metabolic reprogramming in hematopoietic stem and progenitor cells (HSPCs) that are maintained through differentiation to mature immune progeny. Understanding the mechanisms and extent of trained immunity induction by pathogens and vaccines, such as BCG, in HSPC remains a critical area of exploration with important implications for health and disease. Here we review these concepts and present new analysis to highlight how inflammatory reprogramming of HSPC can potently alter immune tone, including to enhance specific anti-tumor responses. New findings in the field pave the way for novel HSPC targeting therapeutic strategies in cancer and other contexts of immune modulation. Future studies are expected to unravel diverse and extensive effects of infections, vaccines, microbiota, and sterile inflammation on hematopoietic progenitor cells and begin to illuminate the broad spectrum of immunologic tuning that can be established through altering HSPC phenotypes. The purpose of this review is to draw attention to emerging and speculative topics in this field where we posit that focused study of HSPC in the framework of trained immunity holds significant promise.
Collapse
Affiliation(s)
- Andrew W Daman
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jin Gyu Cheong
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Laura Berneking
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Steven Z Josefowicz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
20
|
Lin AE, Bapat AC, Xiao L, Niroula A, Ye J, Wong WJ, Agrawal M, Farady CJ, Boettcher A, Hergott CB, McConkey M, Flores-Bringas P, Shkolnik V, Bick AG, Milan D, Natarajan P, Libby P, Ellinor PT, Ebert BL. Clonal Hematopoiesis of Indeterminate Potential With Loss of Tet2 Enhances Risk for Atrial Fibrillation Through Nlrp3 Inflammasome Activation. Circulation 2024; 149:1419-1434. [PMID: 38357791 PMCID: PMC11058018 DOI: 10.1161/circulationaha.123.065597] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 01/10/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP), a common age-associated phenomenon, associates with increased risk of both hematological malignancy and cardiovascular disease. Although CHIP is known to increase the risk of myocardial infarction and heart failure, the influence of CHIP in cardiac arrhythmias, such as atrial fibrillation (AF), is less explored. METHODS CHIP prevalence was determined in the UK Biobank, and incident AF analysis was stratified by CHIP status and clone size using Cox proportional hazard models. Lethally irradiated mice were transplanted with hematopoietic-specific loss of Tet2, hematopoietic-specific loss of Tet2 and Nlrp3, or wild-type control and fed a Western diet, compounded with or without NLRP3 (NLR [NACHT, LRR {leucine rich repeat}] family pyrin domain containing protein 3) inhibitor, NP3-361, for 6 to 9 weeks. Mice underwent in vivo invasive electrophysiology studies and ex vivo optical mapping. Cardiomyocytes from Ldlr-/- mice with hematopoietic-specific loss of Tet2 or wild-type control and fed a Western diet were isolated to evaluate calcium signaling dynamics and analysis. Cocultures of pluripotent stem cell-derived atrial cardiomyocytes were incubated with Tet2-deficient bone marrow-derived macrophages, wild-type control, or cytokines IL-1β (interleukin 1β) or IL-6 (interleukin 6). RESULTS Analysis of the UK Biobank showed individuals with CHIP, in particular TET2 CHIP, have increased incident AF. Hematopoietic-specific inactivation of Tet2 increases AF propensity in atherogenic and nonatherogenic mouse models and is associated with increased Nlrp3 expression and CaMKII (Ca2+/calmodulin-dependent protein kinase II) activation, with AF susceptibility prevented by inactivation of Nlrp3. Cardiomyocytes isolated from Ldlr-/- mice with hematopoietic inactivation of Tet2 and fed a Western diet have impaired calcium release from the sarcoplasmic reticulum into the cytosol, contributing to atrial arrhythmogenesis. Abnormal sarcoplasmic reticulum calcium release was recapitulated in cocultures of cardiomyocytes with the addition of Tet2-deficient macrophages or cytokines IL-1β or IL-6. CONCLUSIONS We identified a modest association between CHIP, particularly TET2 CHIP, and incident AF in the UK Biobank population. In a mouse model of AF resulting from hematopoietic-specific inactivation of Tet2, we propose altered calcium handling as an arrhythmogenic mechanism, dependent on Nlrp3 inflammasome activation. Our data are in keeping with previous studies of CHIP in cardiovascular disease, and further studies into the therapeutic potential of NLRP3 inhibition for individuals with TET2 CHIP may be warranted.
Collapse
Affiliation(s)
- Amy Erica Lin
- Division of Cardiovascular Medicine, Department of Medicine (A.E.L., P.L.), Brigham and Women’s Hospital, Boston, MA
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Aneesh C. Bapat
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Demoulas Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Department of Medicine (A.C.B., P.T.E.), Massachusetts General Hospital, Boston
| | - Ling Xiao
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Abhishek Niroula
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
- Department of Laboratory Medicine, Lund University, Sweden (A.N.)
- Institute of Biomedicine, SciLifeLab, University of Gothenburg, Sweden (A.N.)
| | - Jiangchuan Ye
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Waihay J. Wong
- Department of Pathology (W.J.W., C.B.H.), Brigham and Women’s Hospital, Boston, MA
| | - Mridul Agrawal
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Christopher J. Farady
- Novartis Institutes for BioMedical Research Forum 1, Basel, Switzerland (C.J.F., A.B.)
| | - Andreas Boettcher
- Novartis Institutes for BioMedical Research Forum 1, Basel, Switzerland (C.J.F., A.B.)
| | - Christopher B. Hergott
- Department of Pathology (W.J.W., C.B.H.), Brigham and Women’s Hospital, Boston, MA
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Marie McConkey
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Patricio Flores-Bringas
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Veronica Shkolnik
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
| | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, Nashville, TN (A.G.B.)
| | - David Milan
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Leducq Foundation, Boston, MA (D.M.)
| | - Pradeep Natarajan
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Peter Libby
- Division of Cardiovascular Medicine, Department of Medicine (A.E.L., P.L.), Brigham and Women’s Hospital, Boston, MA
| | - Patrick T. Ellinor
- Cardiovascular Research Center (A.C.B., L.X., J.Y., D.M., P.N., P.T.E.), Massachusetts General Hospital, Boston
- Demoulas Cardiac Arrhythmia Service, Division of Cardiovascular Medicine, Department of Medicine (A.C.B., P.T.E.), Massachusetts General Hospital, Boston
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana Farber Cancer Institute, Harvard Medical School, Boston, MA (A.E.L., A.N., M.A., C.B.H., M.M.C., V.S., B.L.E.)
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge (L.X., A.N., J.Y., P.F.-B., P.N., P.T.E., B.L.E.)
- Howard Hughes Medical Institute, Boston, MA (B.L.E.)
| |
Collapse
|
21
|
Buchheit SF, Collins JM, Anthony KM, Love SAM, Stewart JD, Gondalia R, Huang DY, Manson JE, Reiner AP, Schwartz GG, Vitolins MZ, Schumann RR, Smith RL, Whitsel EA. Radon Exposure and Incident Stroke Risk in the Women's Health Initiative. Neurology 2024; 102:e209143. [PMID: 38546022 PMCID: PMC11770674 DOI: 10.1212/wnl.0000000000209143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 11/15/2023] [Indexed: 09/14/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Little is known about the role of radon in the epidemiology of stroke among women. We therefore examined the association between home radon exposure and risk of stroke among middle-aged and older women in the United States. METHODS We conducted a prospective cohort study of postmenopausal women aged 50-79 years at baseline (1993-1998) in the Women's Health Initiative. We measured exposures as 2-day, indoor, lowest living-level average radon concentrations in picocuries per liter (pCi/L) as estimated in 1993 by the US Geological Survey and reviewed by the Association of American State Geologists under the Indoor Radon Abatement Act. We used Cox proportional hazards models to estimate risk of incident, neurologist-adjudicated stroke during follow-up through 2020 as a hazard ratio and 95% CI, adjusting for study design and participant demographic, social, behavioral, and clinical characteristics. RESULTS Among 158,910 women without stroke at baseline (mean age 63.2 years; 83% white), 6,979 incident strokes were identified over follow-up (mean 13.4 years). Incidence rates were 333, 343, and 349 strokes per 100,000 woman-years at radon concentrations of <2, 2-4, and >4 pCi/L, respectively. Compared with women living at concentrations <2 pCi/L, those at 2-4 and >4 pCi/L had higher covariate-adjusted risks of incident stroke: hazard ratio (95% CI) 1.06 (0.99-1.13) and 1.14 (1.05-1.22). Using nonlinear spline functions to model radon, stroke risk was significantly elevated at concentrations ranging from 2 to 4 pCi/L (p = 0.0004), that is, below the United States Environmental Protection Agency Radon Action Level for mitigation (4 pCi/L). Associations were slightly stronger for ischemic (especially cardioembolic, small vessel occlusive, and large artery atherosclerotic) than hemorrhagic stroke, but otherwise robust in sensitivity analyses. DISCUSSION Radon exposure is associated with moderately increased stroke risk among middle-aged and older women in the United States, suggesting that promulgation of a lower Radon Action Level may help reduce the domestic impact of cerebrovascular disease on public health.
Collapse
Affiliation(s)
- Sophie F Buchheit
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Jason M Collins
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Kurtis M Anthony
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Shelly-Ann M Love
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - James D Stewart
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Rahul Gondalia
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - David Y Huang
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - JoAnn E Manson
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Alexander P Reiner
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Gary G Schwartz
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Mara Z Vitolins
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - R Randall Schumann
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Richard L Smith
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Eric A Whitsel
- From the Brown University (S.F.B.), Providence, RI; Family Medicine (S.F.B.), Mountain Area Health Education Center, Asheville, NC; Department of Epidemiology (J.M.C., K.M.A., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, and Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Public Health Sciences (A.P.R.), Fred Hutchinson Cancer Research Center and Department of Epidemiology, University of Washington, Seattle; Department of Population Health (G.G.S.), School of Medicine & Health Sciences, University of North Dakota, Grand Forks; Department of Epidemiology and Prevention (M.Z.V.), Wake Forest School of Medicine, Winston-Salem, NC; Geosciences and Environmental Change Science Center (R.R.S.), U.S. Geological Survey, U.S. Department of the Interior, Denver, CO; Department of Statistics & Operations Research and Department of Biostatistics (R.L.S.), Gillings School of Global Public Health, and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| |
Collapse
|
22
|
Mitroulis I, Hajishengallis G, Chavakis T. Bone marrow inflammatory memory in cardiometabolic disease and inflammatory comorbidities. Cardiovasc Res 2024; 119:2801-2812. [PMID: 36655373 PMCID: PMC10874275 DOI: 10.1093/cvr/cvad003] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/08/2022] [Accepted: 08/01/2022] [Indexed: 01/20/2023] Open
Abstract
Cardiometabolic disorders are chief causes of morbidity and mortality, with chronic inflammation playing a crucial role in their pathogenesis. The release of differentiated myeloid cells with elevated pro-inflammatory potential, as a result of maladaptively trained myelopoiesis may be a crucial factor for the perpetuation of inflammation. Several cardiovascular risk factors, including sedentary lifestyle, unhealthy diet, hypercholesterolemia, and hyperglycemia, may modulate bone marrow hematopoietic progenitors, causing sustained functional changes that favour chronic metabolic and vascular inflammation. In the present review, we summarize recent studies that support the function of long-term inflammatory memory in progenitors of the bone marrow for the development and progression of cardiometabolic disease and related inflammatory comorbidities, including periodontitis and arthritis. We also discuss how maladaptive myelopoiesis associated with the presence of mutated hematopoietic clones, as present in clonal hematopoiesis, may accelerate atherosclerosis via increased inflammation.
Collapse
Affiliation(s)
- Ioannis Mitroulis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- First Department of Internal Medicine and Department of Haematology, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, 01307 Dresden, Germany
- Centre for Cardiovascular Science, QMRI, University of Edinburgh, Edinburgh EH16 4TJ, UK
| |
Collapse
|
23
|
Cai Y, Wang Y, Yang J, Zhu Z. Concerns regarding myelofibrosis-type megakaryocyte dysplasia. Leukemia 2024; 38:467-468. [PMID: 38263434 PMCID: PMC10844077 DOI: 10.1038/s41375-024-02160-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2023] [Revised: 01/15/2024] [Accepted: 01/16/2024] [Indexed: 01/25/2024]
Affiliation(s)
- Yanan Cai
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Yuebo Wang
- Clinical Research Service Center, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Jing Yang
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China
| | - Zunmin Zhu
- Department of Hematology, Henan Provincial People's Hospital, Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
24
|
Kosianova А, Pak O, Bryukhovetskiy I. Regulation of cancer stem cells and immunotherapy of glioblastoma (Review). Biomed Rep 2024; 20:24. [PMID: 38170016 PMCID: PMC10758921 DOI: 10.3892/br.2023.1712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 11/24/2023] [Indexed: 01/05/2024] Open
Abstract
Glioblastoma (GB) is one of the most adverse diagnoses in oncology. Complex current treatment results in a median survival of 15 months. Resistance to treatment is associated with the presence of cancer stem cells (CSCs). The present review aimed to analyze the mechanisms of CSC plasticity, showing the particular role of β-catenin in regulating vital functions of CSCs, and to describe the molecular mechanisms of Wnt-independent increase of β-catenin levels, which is influenced by the local microenvironment of CSCs. The present review also analyzed the reasons for the low effectiveness of using medication in the regulation of CSCs, and proposed the development of immunotherapy scenarios with tumor cell vaccines, containing heterogenous cancer cells able of producing a multidirectional antineoplastic immune response. Additionally, the possibility of managing lymphopenia by transplanting hematopoietic stem cells from a healthy sibling and using clofazimine or other repurposed drugs that reduce β-catenin concentration in CSCs was discussed in the present study.
Collapse
Affiliation(s)
- Аleksandra Kosianova
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| | - Oleg Pak
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| | - Igor Bryukhovetskiy
- Medical Center, School of Medicine and Life Science, Far Eastern Federal University, Vladivostok 690091, Russian Federation
| |
Collapse
|
25
|
Pasquer H, Daltro de Oliveira R, Vasseur L, Soret-Dulphy J, Maslah N, Zhao LP, Marcault C, Cazaux M, Gauthier N, Verger E, Parquet N, Vainchenker W, Raffoux E, Ugo V, Luque Paz D, Roy L, Lambert WC, Ianotto JC, Lippert E, Giraudier S, Cassinat B, Kiladjian JJ, Benajiba L. Distinct clinico-molecular arterial and venous thrombosis scores for myeloproliferative neoplasms risk stratification. Leukemia 2024; 38:326-339. [PMID: 38148396 DOI: 10.1038/s41375-023-02114-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/30/2023] [Accepted: 12/06/2023] [Indexed: 12/28/2023]
Abstract
Current recommended risk scores to predict thrombotic events associated with myeloproliferative neoplasms (MPN) do not discriminate between arterial and venous thrombosis despite their different physiopathology. To define novel stratification systems, we delineated a comprehensive landscape of MPN associated thrombosis across a large long-term follow-up MPN cohort. Prior arterial thrombosis, age >60 years, cardiovascular risk factors and presence of TET2 or DNMT3A mutations were independently associated with arterial thrombosis in multivariable analysis. ARTS, an ARterial Thrombosis Score, based on these four factors, defined low- (0.37% patients-year) and high-risk (1.19% patients-year) patients. ARTS performance was superior to the two-tiered conventional risk stratification in our training cohort, across all MPN subtypes, as well as in two external validation cohorts. Prior venous thrombosis and presence of a JAK2V617F mutation with a variant allelic frequency ≥50% were independently associated with venous thrombosis. The discrimination potential of VETS, a VEnous Thrombosis Score based on these two factors, was poor, similar to the two-tiered conventional risk stratification. Our study pinpoints arterial and venous thrombosis clinico-molecular differences and proposes an arterial risk score for more accurate patients' stratification. Further improvement of venous risk scores, accounting for additional factors and considering venous thrombosis as a heterogeneous entity is warranted.
Collapse
Affiliation(s)
- Hélène Pasquer
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
- INSERM UMR 944, Institut de Recherche Saint-Louis, Paris, France
| | - Rafael Daltro de Oliveira
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Loic Vasseur
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Juliette Soret-Dulphy
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Nabih Maslah
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Lin-Pierre Zhao
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Clémence Marcault
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Marine Cazaux
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Nicolas Gauthier
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
| | - Emmanuelle Verger
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Nathalie Parquet
- Université Paris Cité, APHP, Hôpital Saint-Louis, Département d'hématologie et d'Immunologie, Paris, France
| | - William Vainchenker
- APHP, Hôpital Saint-Louis, Département d'hématologie et d'Immunologie, Paris, France
| | - Emmanuel Raffoux
- Université Paris Cité, APHP, Hôpital Saint-Louis, Département d'hématologie et d'Immunologie, Paris, France
| | - Valérie Ugo
- Univ Angers, Nantes Université, CHU Angers, Inserm, CNRS, CRCI2NA, Angers, France
| | - Damien Luque Paz
- Univ Angers, Nantes Université, CHU Angers, Inserm, CNRS, CRCI2NA, Angers, France
| | - Lydia Roy
- Université Paris Est Créteil, APHP, Hôpital Henri Mondor, Service d'hématologie, Créteil, France
| | - Wayne-Corentin Lambert
- Université de Bretagne Occidentale, CHU de Brest, Service d'Hématologie Biologique, Brest, France
| | - Jean-Christophe Ianotto
- Université de Bretagne Occidentale, CHU de Brest, Service d'Hématologie et d'Hémostase Clinique, Brest, France
| | - Eric Lippert
- Université de Bretagne Occidentale, CHU de Brest, Service d'Hématologie Biologique, Brest, France
| | - Stéphane Giraudier
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Bruno Cassinat
- Université Paris Cité, APHP, Hôpital Saint-Louis, Laboratoire de Biologie Cellulaire, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Jean-Jacques Kiladjian
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France
- INSERM UMR 1131, Institut de Recherche Saint-Louis, Paris, France
| | - Lina Benajiba
- Université Paris Cité, APHP, Hôpital Saint-Louis, Centre d'Investigations Cliniques, INSERM CIC 1427, Paris, France.
- INSERM UMR 944, Institut de Recherche Saint-Louis, Paris, France.
| |
Collapse
|
26
|
Anthony KM, Collins JM, Love SAM, Stewart JD, Buchheit SF, Gondalia R, Schwartz GG, Huang DY, Meliker JR, Zhang Z, Barac A, Desai P, Hayden KM, Honigberg MC, Jaiswal S, Natarajan P, Bick AG, Kooperberg C, Manson JE, Reiner AP, Whitsel EA. Radon Exposure, Clonal Hematopoiesis, and Stroke Susceptibility in the Women's Health Initiative. Neurology 2024; 102:e208055. [PMID: 38170948 PMCID: PMC10870742 DOI: 10.1212/wnl.0000000000208055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 10/30/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND AND OBJECTIVES Studies suggest that clonal hematopoiesis of indeterminate potential (CHIP) may increase risk of hematologic malignancy and cardiovascular disease, including stroke. However, few studies have investigated plausible environmental risk factors for CHIP such as radon, despite the climate-related increases in and documented infrequency of testing for this common indoor air pollutant.The purpose of this study was to estimate the risk of CHIP related to radon, an established environmental mutagen. METHODS We linked geocoded addresses of 10,799 Women's Health Initiative Trans-Omics for Precision Medicine (WHI TOPMed) participants to US Environmental Protection Agency-predicted, county-level, indoor average screening radon concentrations, categorized as follows: Zone 1 (>4 pCi/L), Zone 2 (2-4 pCi/L), and Zone 3 (<2 pCi/L). We defined CHIP as the presence of one or more leukemogenic driver mutations with variant allele frequency >0.02. We identified prevalent and incident ischemic and hemorrhagic strokes; subtyped ischemic stroke using Trial of ORG 10172 in Acute Stroke Treatment (TOAST) criteria; and then estimated radon-related risk of CHIP as an odds ratio (OR) and 95% CI using multivariable-adjusted, design-weighted logistic regression stratified by age, race/ethnicity, smoking status, and stroke type/subtype. RESULTS The percentages of participants with CHIP in Zones 1, 2, and 3 were 9.0%, 8.4%, and 7.7%, respectively (ptrend = 0.06). Among participants with ischemic stroke, Zones 2 and 1 were associated with higher estimated risks of CHIP relative to Zone 3: 1.39 (1.15-1.68) and 1.46 (1.15-1.87), but not among participants with hemorrhagic stroke: 0.98 (0.68-1.40) and 1.03 (0.70-1.52), or without stroke: 1.04 (0.74-1.46) and 0.95 (0.63-1.42), respectively (pinteraction = 0.03). Corresponding estimates were particularly high among TOAST-subtyped cardioembolism: 1.78 (1.30-2.47) and 1.88 (1.31-2.72), or other ischemic etiologies: 1.37 (1.06-1.78) and 1.50 (1.11-2.04), but not small vessel occlusion: 1.05 (0.74-1.49) and 1.00 (0.68-1.47), respectively (pinteraction = 0.10). Observed patterns of association among strata were insensitive to attrition weighting, ancestry adjustment, prevalent stroke exclusion, separate analysis of DNMT3A driver mutations, and substitution with 3 alternative estimates of radon exposure. DISCUSSION The robust elevation of radon-related risk of CHIP among postmenopausal women who develop incident cardioembolic stroke is consistent with a potential role of somatic genomic mutation in this societally burdensome form of cerebrovascular disease, although the mechanism has yet to be confirmed.
Collapse
Affiliation(s)
- Kurtis M Anthony
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Jason M Collins
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Shelly-Ann M Love
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - James D Stewart
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Sophie F Buchheit
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Rahul Gondalia
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Gary G Schwartz
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - David Y Huang
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Jaymie R Meliker
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Zhenzhen Zhang
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Ana Barac
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Pinkal Desai
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Kathleen M Hayden
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Michael C Honigberg
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Siddhartha Jaiswal
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Pradeep Natarajan
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Alexander G Bick
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Charles Kooperberg
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - JoAnn E Manson
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Alexander P Reiner
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| | - Eric A Whitsel
- From the Department of Epidemiology (K.M.A., J.M.C., S.-A.M.L., J.D.S., R.G., E.A.W.), Gillings School of Global Public Health, University of North Carolina, Chapel Hill; Brown University (S.F.B.), Providence, RI; Department of Population Health (G.G.S.), University of North Dakota School of Medicine & Health Sciences, Grand Forks; Department of Neurology (D.Y.H.), School of Medicine, University of North Carolina, Chapel Hill; Program in Public Health (J.R.M.), Stony Brook University, Stony Brook, NY; Division of Oncological Sciences (Z.Z.), Knight Cancer Institute, Oregon Health & Science University, Portland; Department of Cardiology (A.B.), Medstar Washington Hospital Center, Washington, DC; Department of Medicine (A.B.), Georgetown University, Washington, DC; Division of Hematology and Oncology (P.D.), Weill Cornell Medicine, New York; Department of Social Sciences and Health Policy (K.M.H.), Wake Forest University School of Medicine, Winston-Salem, NC; Cardiology Division (M.C.H.), Massachusetts General Hospital, Boston; Program in Medical and Population Genetics (M.C.H., P.N.), Broad Institute of Harvard and MIT, Cambridge, MA; Department of Pathology (S.J.), Stanford University School of Medicine, CA; Cardiovascular Research Center and Center for Genomic Medicine (P.N.), Massachusetts General Hospital, Boston; Department of Medicine (P.N.), Harvard Medical School, Boston; Division of Genetic Medicine (A.G.B.), Department of Medicine, Vanderbilt University Medical Center, Nashville, TN; Division of Public Health Sciences (C.K., A.P.R.), Fred Hutchinson Cancer Center, Seattle, WA; Department of Medicine (J.E.M.), Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Department of Epidemiology (A.P.R.), University of Washington, Seattle; and Department of Medicine (E.A.W.), School of Medicine, University of North Carolina, Chapel Hill
| |
Collapse
|
27
|
Yang S, Xu J, Dai Y, Jin S, Sun Y, Li J, Liu C, Ma X, Chen Z, Chen L, Hou J, Mi JQ, Chen SJ. Neutrophil activation and clonal CAR-T re-expansion underpinning cytokine release syndrome during ciltacabtagene autoleucel therapy in multiple myeloma. Nat Commun 2024; 15:360. [PMID: 38191582 PMCID: PMC10774397 DOI: 10.1038/s41467-023-44648-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024] Open
Abstract
Cytokine release syndrome (CRS) is the most common complication of chimeric antigen receptor redirected T cells (CAR-T) therapy. CAR-T toxicity management has been greatly improved, but CRS remains a prime safety concern. Here we follow serum cytokine levels and circulating immune cell transcriptomes longitudinally in 26 relapsed/refractory multiple myeloma patients receiving the CAR-T product, ciltacabtagene autoleucel, to understand the immunological kinetics of CRS. We find that although T lymphocytes and monocytes/macrophages are the major overall cytokine source in manifest CRS, neutrophil activation peaks earlier, before the onset of severe symptoms. Intracellularly, signaling activation dominated by JAK/STAT pathway occurred prior to cytokine cascade and displayed regular kinetic changes. CRS severity is accurately described and potentially predicted by temporal cytokine secretion signatures. Notably, CAR-T re-expansion is found in three patients, including a fatal case characterized by somatic TET2-mutation, clonal expanded cytotoxic CAR-T, broadened cytokine profiles and irreversible hepatic toxicity. Together, our findings show that a latent phase with distinct immunological changes precedes manifest CRS, providing an optimal window and potential targets for CRS therapeutic intervention and that CAR-T re-expansion warrants close clinical attention and laboratory investigation to mitigate the lethal risk.
Collapse
Affiliation(s)
- Shuangshuang Yang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jie Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuting Dai
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shiwei Jin
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yan Sun
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jianfeng Li
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chenglin Liu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaolin Ma
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zhu Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Lijuan Chen
- Department of Hematology, First affiliated Hospital of Nanjing Medical University, Jiangsu Province Hospital, Nanjing, 210029, China
| | - Jian Hou
- Department of Hematology, Ren Ji Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China
| | - Jian-Qing Mi
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Sai-Juan Chen
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
28
|
Sato Y, Fukatsu M, Suzuki T, Sasajima T, Gunji N, Yoshida S, Asano N, Fukuchi K, Mori H, Takano M, Hayashi K, Takahashi H, Shirado-Harada K, Kimura S, Koyama D, Migita K, Ikezoe T. Successful allogeneic hematopoietic stem cell transplantation for myelodysplastic neoplasms complicated with secondary pulmonary alveolar proteinosis and Behçet's disease harboring GATA2 mutation. Int J Hematol 2023; 118:642-646. [PMID: 37084069 DOI: 10.1007/s12185-023-03603-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/13/2023] [Accepted: 04/14/2023] [Indexed: 04/22/2023]
Abstract
Myelodysplastic neoplasms (MDS) are defined by cytopenia and morphologic dysplasia originating from clonal hematopoiesis. They are also frequently complicated with diseases caused by immune dysfunction, such as Behçet's disease (BD) and secondary pulmonary alveolar proteinosis (sPAP). MDS with both BD and sPAP is extremely rare, and their prognosis is poor. In addition, haploinsufficiency of the hematopoietic transcription factor gene GATA2 is recognized as a cause of familial MDS and is frequently complicated by sPAP. Herein, we report a case of MDS combined with both BD and sPAP in association with GATA2 deficiency in a Japanese woman. Because she developed progressive leukopenia and macrocytic anemia during BD treatment at the age of 61, she underwent a bone-marrow examination and was diagnosed with MDS. She subsequently developed sPAP. At the age of 63, she underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT). Since allo-HSCT, she has maintained complete remission of MDS as well as the symptoms of BD and sPAP. Furthermore, we performed whole exome sequencing and identified the GATA2 Ala164Thr germline mutation. These findings suggest that patients with MDS, BD and sPAP should be considered for early allo-HSCT.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Masahiko Fukatsu
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Tomohiro Suzuki
- Department of Gastroenterology, Fukushima Rosai Hospital, Iwaki, Fukushima, 973-8403, Japan
| | - Tomomi Sasajima
- Department of Rheumatology, Fukushima Rosai Hospital, Iwaki, Fukushima, 973-8403, Japan
| | - Naohiko Gunji
- Department of Gastroenterology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Shuhei Yoshida
- Department of Rheumatology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Naomi Asano
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Koichiro Fukuchi
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Hirotaka Mori
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Motoki Takano
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Kiyohito Hayashi
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Hiroshi Takahashi
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Kayo Shirado-Harada
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Satoshi Kimura
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Daisuke Koyama
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan.
| | - Kiyoshi Migita
- Department of Rheumatology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| | - Takayuki Ikezoe
- Department of Hematology, Fukushima Medical University, Fukushima, Fukushima, 960-1295, Japan
| |
Collapse
|
29
|
Lassalle F, Duployez N, Vincent F, Rauch A, Denimal T, Rosa M, Labreuche J, Dombrowicz D, Staels B, Preudhomme C, Susen S, Van Belle E, Dupont A. Negative Impact of TET2 Mutations on Long-Term Survival After Transcatheter Aortic Valve Replacement. JACC Basic Transl Sci 2023; 8:1424-1435. [PMID: 38093739 PMCID: PMC10714177 DOI: 10.1016/j.jacbts.2023.04.010] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Revised: 04/18/2023] [Accepted: 04/18/2023] [Indexed: 07/01/2024]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is considered as being a novel age-related risk factor for cardiovascular diseases. By capture-sequencing of a 67-gene panel, we established a large spectrum of CHIP in 258 patients with aortic valve stenosis undergoing transcatheter aortic valve replacement (TAVR) and assessed their association with long-term survival after TAVR. One or several CHIP variants in 35 genes were identified in 68% of the cohort, DNMT3A and TET2 being the 2 most frequently mutated genes. Patients carrying a TET2-CHIP-driver variant with low variant allele frequency (2%-10%) had a significant decrease in overall survival 5 years after TAVR.
Collapse
Affiliation(s)
- Fanny Lassalle
- University of Lille, Inserm, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Nicolas Duployez
- Unite Mixte de Recherche (UMR) 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche contre le Cancer de Lille, University of Lille, CNRS, Inserm, Centre Hospitalier Universitaire Lille, Lille, France
| | - Flavien Vincent
- University of Lille, Inserm, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Antoine Rauch
- University of Lille, Inserm, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Tom Denimal
- University of Lille, Inserm, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Mickael Rosa
- University of Lille, Inserm, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Julien Labreuche
- Department of Biostatistics, Centre Hospitalier Universitaire Lille, Lille, France
| | - David Dombrowicz
- University of Lille, Inserm, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Bart Staels
- University of Lille, Inserm, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Claude Preudhomme
- Unite Mixte de Recherche (UMR) 9020–UMR-S 1277–Canther–Cancer Heterogeneity, Plasticity and Resistance to Therapies, Institut de Recherche contre le Cancer de Lille, University of Lille, CNRS, Inserm, Centre Hospitalier Universitaire Lille, Lille, France
| | - Sophie Susen
- University of Lille, Inserm, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Eric Van Belle
- University of Lille, Inserm, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| | - Annabelle Dupont
- University of Lille, Inserm, Centre Hospitalier Universitaire Lille, Institut Pasteur de Lille, U1011-EGID, Lille, France
| |
Collapse
|
30
|
Ogura Y, Mimura I. Epigenetic roles in clonal hematopoiesis and aging kidney-related chronic kidney disease. Front Cell Dev Biol 2023; 11:1281850. [PMID: 37928907 PMCID: PMC10623128 DOI: 10.3389/fcell.2023.1281850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 10/11/2023] [Indexed: 11/07/2023] Open
Abstract
Accumulation of somatic hematopoietic stem cell mutations with aging has been revealed by the recent genome-wide analysis. Clonal expansion, known as clonal hematopoiesis of indeterminate potential (CHIP), is a premalignant condition of hematological cancers. It is defined as the absence of definitive morphological evidence of a hematological neoplasm and occurrence of ≥2% of mutant allele fraction in the peripheral blood. In CHIP, the most frequently mutated genes are epigenetic regulators such as DNMT3A, TET2, and ASXL1. CHIP induces inflammation. CHIP is shown to be associated with not only hematological malignancy but also non-malignant disorders such as atherosclerosis, cardiovascular diseases and chronic liver disease. In addition, recent several large clinical trials have shown that CHIP is also the risk factor for developing chronic kidney disease (CKD). In this review article, we proposed novel findings about CHIP and CHIP related kidney disease based on the recent basic and clinical research. The possible mechanism of the kidney injury in CHIP is supposed to be due to the clonal expansion in both myeloid and lymphoid cell lines. In myeloid cell lines, the mutated macrophages increase the inflammatory cytokine level and induce chronic inflammation. It leads to epigenetic downregulation of kidney and macrophage klotho level. In lymphoid cell lines, CHIP might be related to monoclonal gammopathy of renal significance (MGRS). It describes any B cell or plasma cell clonal disorder that does not fulfill the criteria for cancer yet produces a nephrotoxic monoclonal immunoglobulin that leads to kidney injury or disease. MGRS causes M-protein related nephropathy frequently observed among aged CKD patients. It is important to consider the CHIP-related complications such as hematological malignancy, cardiovascular diseases and metabolic disorders in managing the elderly CKD patients. There are no established therapies for CHIP and CHIP-related CKD yet. However, recent studies have supported the development of effective CHIP therapies, such as blocking the expansion of aberrant HSCs and inhibiting chronic inflammation. In addition, drugs targeting the epigenetic regulation of Klotho in the kidney and macrophages might be therapeutic targets of CHIP in the kidney.
Collapse
Affiliation(s)
| | - Imari Mimura
- Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, Tokyo, Japan
| |
Collapse
|
31
|
Schneider M, Rolfs C, Trumpp M, Winter S, Fischer L, Richter M, Menger V, Nenoff K, Grieb N, Metzeler KH, Kubasch AS, Sockel K, Thiede C, Wu J, Woo J, Brüderle A, Hofbauer LC, Lützner J, Roth A, Cross M, Platzbecker U. Activation of distinct inflammatory pathways in subgroups of LR-MDS. Leukemia 2023; 37:1709-1718. [PMID: 37420006 PMCID: PMC10400420 DOI: 10.1038/s41375-023-01949-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 05/17/2023] [Accepted: 06/19/2023] [Indexed: 07/09/2023]
Abstract
Aberrant innate immune signaling has been identified as a potential key driver of the complex pathophysiology of myelodysplastic neoplasms (MDS). This study of a large, clinically and genetically well-characterized cohort of treatment-naïve MDS patients confirms intrinsic activation of inflammatory pathways in general mediated by caspase-1, interleukin (IL)-1β and IL-18 in low-risk (LR)-MDS bone marrow and reveals a previously unrecognized heterogeneity of inflammation between genetically defined LR-MDS subgroups. Principal component analysis resolved two LR-MDS phenotypes with low (cluster 1) and high (cluster 2) levels of IL1B gene expression, respectively. Cluster 1 contained 14/17 SF3B1-mutated cases, while cluster 2 contained 8/8 del(5q) cases. Targeted gene expression analysis of sorted cell populations showed that the majority of the inflammasome-related genes, including IL1B, were primarily expressed in the monocyte compartment, consistent with a dominant role in determining the inflammatory bone marrow environment. However, the highest levels of IL18 expression were found in hematopoietic stem and progenitor cells (HSPCs). The colony forming activity of healthy donor HSPCs exposed to monocytes from LR-MDS was increased by the IL-1β-neutralizing antibody canakinumab. This work reveals distinct inflammatory profiles in LR-MDS that are of likely relevance to the personalization of emerging anti-inflammatory therapies.
Collapse
Affiliation(s)
- Marie Schneider
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Medical Center Leipzig, Leipzig, Germany
| | - Clara Rolfs
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Medical Center Leipzig, Leipzig, Germany
| | - Matthias Trumpp
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Medical Center Leipzig, Leipzig, Germany
| | - Susann Winter
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Luise Fischer
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Mandy Richter
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Medical Center Leipzig, Leipzig, Germany
| | - Victoria Menger
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Medical Center Leipzig, Leipzig, Germany
| | - Kolja Nenoff
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Medical Center Leipzig, Leipzig, Germany
| | - Nora Grieb
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Medical Center Leipzig, Leipzig, Germany
| | - Klaus H Metzeler
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Medical Center Leipzig, Leipzig, Germany
| | - Anne Sophie Kubasch
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Medical Center Leipzig, Leipzig, Germany
| | - Katja Sockel
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Christian Thiede
- Department of Medicine I, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Jincheng Wu
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | - Janghee Woo
- Novartis Institutes for BioMedical Research, Cambridge, MA, USA
| | | | - Lorenz C Hofbauer
- UniversityCenter for Healthy Aging & Department of Medicine III, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Jörg Lützner
- Department of Orthopedic Surgery, University Hospital Carl Gustav Carus, Dresden, Germany
| | - Andreas Roth
- Department of Orthopedic Surgery, University Medical Center Leipzig, Leipzig, Germany
| | - Michael Cross
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Medical Center Leipzig, Leipzig, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University Medical Center Leipzig, Leipzig, Germany.
| |
Collapse
|
32
|
Sikking MA, Stroeks SLVM, Waring OJ, Henkens MTHM, Riksen NP, Hoischen A, Heymans SRB, Verdonschot JAJ. Clonal Hematopoiesis of Indeterminate Potential From a Heart Failure Specialist's Point of View. J Am Heart Assoc 2023; 12:e030603. [PMID: 37489738 PMCID: PMC10492961 DOI: 10.1161/jaha.123.030603] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2023] [Accepted: 06/06/2023] [Indexed: 07/26/2023]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is a common bone marrow abnormality induced by age-related DNA mutations, which give rise to proinflammatory immune cells. These immune cells exacerbate atherosclerotic cardiovascular disease and may induce or accelerate heart failure. The mechanisms involved are complex but point toward a central role for proinflammatory macrophages and an inflammasome-dependent immune response (IL-1 [interleukin-1] and IL-6 [interleukin-6]) in the atherosclerotic plaque or directly in the myocardium. Intracardiac inflammation may decrease cardiac function and induce cardiac fibrosis, even in the absence of atherosclerotic cardiovascular disease. The pathophysiology and consequences of CHIP may differ among implicated genes as well as subgroups of patients with heart failure, based on cause (ischemic versus nonischemic) and ejection fraction (reduced ejection fraction versus preserved ejection fraction). Evidence is accumulating that CHIP is associated with cardiovascular mortality in ischemic and nonischemic heart failure with reduced ejection fraction and involved in the development of heart failure with preserved ejection fraction. CHIP and corresponding inflammatory pathways provide a highly potent therapeutic target. Randomized controlled trials in patients with well-phenotyped heart failure, where readily available anti-inflammatory therapies are used to intervene with clonal hematopoiesis, may pave the way for a new area of heart failure treatment. The first clinical trials that target CHIP are already registered.
Collapse
Affiliation(s)
- Maurits A. Sikking
- Department of CardiologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
| | - Sophie L. V. M. Stroeks
- Department of CardiologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
| | - Olivia J. Waring
- Department of PathologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
| | - Michiel T. H. M. Henkens
- Department of PathologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
- Netherlands Heart Institute (NLHI)Utrechtthe Netherlands
| | - Niels P. Riksen
- Department of Internal MedicineRadboud University Medical CenterNijmegenthe Netherlands
| | - Alexander Hoischen
- Department of Human GeneticsRadboud University Medical CenterNijmegenthe Netherlands
| | - Stephane R. B. Heymans
- Department of CardiologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
- Department of Cardiovascular ResearchUniversity of LeuvenBelgium
| | - Job A. J. Verdonschot
- Department of CardiologyCardiovascular Research Institute Maastricht (CARIM), Maastricht University Medical Center (MUMC)Maastrichtthe Netherlands
- Department of Clinical GeneticsMaastricht University Medical Center (MUMC)Maastrichtthe Netherlands
| |
Collapse
|
33
|
Bilton EJ, Mollan SP. Giant cell arteritis: reviewing the advancing diagnostics and management. Eye (Lond) 2023; 37:2365-2373. [PMID: 36788362 PMCID: PMC9927059 DOI: 10.1038/s41433-023-02433-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/16/2023] [Accepted: 01/30/2023] [Indexed: 02/16/2023] Open
Abstract
Giant Cell Arteritis (GCA) is well known to be a critical ischaemic disease that requires immediate medical recognition to initiate treatment and where one in five people still suffer visual loss. The immunopathophysiology has continued to be characterised, and the influencing of ageing in the development of GCA is beginning to be understood. Recent national and international guidelines have supported the directed use of cranial ultrasound to reduce diagnostic delay and improve clinical outcomes. Immediate high dose glucocorticoids remain the standard emergency treatment for GCA, with a number of targeted agents that have been shown in clinical trials to have superior clinical efficacy and steroid sparing effects. The aim of this review was to present the latest advances in GCA that have the potential to influence routine clinical practice.
Collapse
Affiliation(s)
- Edward J Bilton
- Ophthalmology Department, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TH, UK
- INSIGHT Health Data Research hub for eye health, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TH, UK
| | - Susan P Mollan
- Ophthalmology Department, Queen Elizabeth Hospital, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TH, UK.
- INSIGHT Health Data Research hub for eye health, University Hospitals Birmingham NHS Foundation Trust, Birmingham, B15 2TH, UK.
- Transitional Brain Science, Institute of Metabolism and Systems Research, College of Medical and Dental Sciences, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
34
|
Chavez JS, Rabe JL, Niño KE, Wells HH, Gessner RL, Mills TS, Hernandez G, Pietras EM. PU.1 is required to restrain myelopoiesis during chronic inflammatory stress. Front Cell Dev Biol 2023; 11:1204160. [PMID: 37497478 PMCID: PMC10368259 DOI: 10.3389/fcell.2023.1204160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Accepted: 06/19/2023] [Indexed: 07/28/2023] Open
Abstract
Chronic inflammation is a common feature of aging and numerous diseases such as diabetes, obesity, and autoimmune syndromes and has been linked to the development of hematological malignancy. Blood-forming hematopoietic stem cells (HSC) can contribute to these diseases via the production of tissue-damaging myeloid cells and/or the acquisition of mutations in epigenetic and transcriptional regulators that initiate evolution toward leukemogenesis. We previously showed that the myeloid "master regulator" transcription factor PU.1 is robustly induced in HSC by pro-inflammatory cytokines such as interleukin (IL)-1β and limits their proliferative activity. Here, we used a PU.1-deficient mouse model to investigate the broader role of PU.1 in regulating hematopoietic activity in response to chronic inflammatory challenges. We found that PU.1 is critical in restraining inflammatory myelopoiesis via suppression of cell cycle and self-renewal gene programs in myeloid-biased multipotent progenitor (MPP) cells. Our data show that while PU.1 functions as a key driver of myeloid differentiation, it plays an equally critical role in tailoring hematopoietic responses to inflammatory stimuli while limiting expansion and self-renewal gene expression in MPPs. These data identify PU.1 as a key regulator of "emergency" myelopoiesis relevant to inflammatory disease and leukemogenesis.
Collapse
Affiliation(s)
- James S. Chavez
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Jennifer L. Rabe
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Katia E. Niño
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Harrison H. Wells
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rachel L. Gessner
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Taylor S. Mills
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Giovanny Hernandez
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Eric M. Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
- Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| |
Collapse
|
35
|
Ktena YP, Dionysiou M, Gondek LP, Cooke KR. The impact of epigenetic modifications on allogeneic hematopoietic stem cell transplantation. Front Immunol 2023; 14:1188853. [PMID: 37325668 PMCID: PMC10264773 DOI: 10.3389/fimmu.2023.1188853] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 05/18/2023] [Indexed: 06/17/2023] Open
Abstract
The field of epigenetics studies the complex processes that regulate gene expression without altering the DNA sequence itself. It is well established that epigenetic modifications are crucial to cellular homeostasis and differentiation and play a vital role in hematopoiesis and immunity. Epigenetic marks can be mitotically and/or meiotically heritable upon cell division, forming the basis of cellular memory, and have the potential to be reversed between cellular fate transitions. Hence, over the past decade, there has been increasing interest in the role that epigenetic modifications may have on the outcomes of allogeneic hematopoietic transplantation and growing enthusiasm in the therapeutic potential these pathways may hold. In this brief review, we provide a basic overview of the types of epigenetic modifications and their biological functions, summarizing the current literature with a focus on hematopoiesis and immunity specifically in the context of allogeneic hematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Yiouli P. Ktena
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | | | | | | |
Collapse
|
36
|
Bao H, Cao J, Chen M, Chen M, Chen W, Chen X, Chen Y, Chen Y, Chen Y, Chen Z, Chhetri JK, Ding Y, Feng J, Guo J, Guo M, He C, Jia Y, Jiang H, Jing Y, Li D, Li J, Li J, Liang Q, Liang R, Liu F, Liu X, Liu Z, Luo OJ, Lv J, Ma J, Mao K, Nie J, Qiao X, Sun X, Tang X, Wang J, Wang Q, Wang S, Wang X, Wang Y, Wang Y, Wu R, Xia K, Xiao FH, Xu L, Xu Y, Yan H, Yang L, Yang R, Yang Y, Ying Y, Zhang L, Zhang W, Zhang W, Zhang X, Zhang Z, Zhou M, Zhou R, Zhu Q, Zhu Z, Cao F, Cao Z, Chan P, Chen C, Chen G, Chen HZ, Chen J, Ci W, Ding BS, Ding Q, Gao F, Han JDJ, Huang K, Ju Z, Kong QP, Li J, Li J, Li X, Liu B, Liu F, Liu L, Liu Q, Liu Q, Liu X, Liu Y, Luo X, Ma S, Ma X, Mao Z, Nie J, Peng Y, Qu J, Ren J, Ren R, Song M, Songyang Z, Sun YE, Sun Y, Tian M, Wang S, et alBao H, Cao J, Chen M, Chen M, Chen W, Chen X, Chen Y, Chen Y, Chen Y, Chen Z, Chhetri JK, Ding Y, Feng J, Guo J, Guo M, He C, Jia Y, Jiang H, Jing Y, Li D, Li J, Li J, Liang Q, Liang R, Liu F, Liu X, Liu Z, Luo OJ, Lv J, Ma J, Mao K, Nie J, Qiao X, Sun X, Tang X, Wang J, Wang Q, Wang S, Wang X, Wang Y, Wang Y, Wu R, Xia K, Xiao FH, Xu L, Xu Y, Yan H, Yang L, Yang R, Yang Y, Ying Y, Zhang L, Zhang W, Zhang W, Zhang X, Zhang Z, Zhou M, Zhou R, Zhu Q, Zhu Z, Cao F, Cao Z, Chan P, Chen C, Chen G, Chen HZ, Chen J, Ci W, Ding BS, Ding Q, Gao F, Han JDJ, Huang K, Ju Z, Kong QP, Li J, Li J, Li X, Liu B, Liu F, Liu L, Liu Q, Liu Q, Liu X, Liu Y, Luo X, Ma S, Ma X, Mao Z, Nie J, Peng Y, Qu J, Ren J, Ren R, Song M, Songyang Z, Sun YE, Sun Y, Tian M, Wang S, Wang S, Wang X, Wang X, Wang YJ, Wang Y, Wong CCL, Xiang AP, Xiao Y, Xie Z, Xu D, Ye J, Yue R, Zhang C, Zhang H, Zhang L, Zhang W, Zhang Y, Zhang YW, Zhang Z, Zhao T, Zhao Y, Zhu D, Zou W, Pei G, Liu GH. Biomarkers of aging. SCIENCE CHINA. LIFE SCIENCES 2023; 66:893-1066. [PMID: 37076725 PMCID: PMC10115486 DOI: 10.1007/s11427-023-2305-0] [Show More Authors] [Citation(s) in RCA: 154] [Impact Index Per Article: 77.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 02/27/2023] [Indexed: 04/21/2023]
Abstract
Aging biomarkers are a combination of biological parameters to (i) assess age-related changes, (ii) track the physiological aging process, and (iii) predict the transition into a pathological status. Although a broad spectrum of aging biomarkers has been developed, their potential uses and limitations remain poorly characterized. An immediate goal of biomarkers is to help us answer the following three fundamental questions in aging research: How old are we? Why do we get old? And how can we age slower? This review aims to address this need. Here, we summarize our current knowledge of biomarkers developed for cellular, organ, and organismal levels of aging, comprising six pillars: physiological characteristics, medical imaging, histological features, cellular alterations, molecular changes, and secretory factors. To fulfill all these requisites, we propose that aging biomarkers should qualify for being specific, systemic, and clinically relevant.
Collapse
Affiliation(s)
- Hainan Bao
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Jiani Cao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Mengting Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Min Chen
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Wei Chen
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Xiao Chen
- Department of Nuclear Medicine, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Yanhao Chen
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yu Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yutian Chen
- The Department of Endovascular Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Zhiyang Chen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, 510632, China
| | - Jagadish K Chhetri
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
| | - Yingjie Ding
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Junlin Feng
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jun Guo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China
| | - Mengmeng Guo
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China
| | - Chuting He
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Yujuan Jia
- Department of Neurology, First Affiliated Hospital, Shanxi Medical University, Taiyuan, 030001, China
| | - Haiping Jiang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Ying Jing
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China
| | - Jiaming Li
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jingyi Li
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Qinhao Liang
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China
| | - Rui Liang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300384, China
| | - Feng Liu
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, 510275, China
| | - Xiaoqian Liu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Zuojun Liu
- School of Life Sciences, Hainan University, Haikou, 570228, China
| | - Oscar Junhong Luo
- Department of Systems Biomedical Sciences, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Jianwei Lv
- School of Life Sciences, Xiamen University, Xiamen, 361102, China
| | - Jingyi Ma
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Kehang Mao
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China
| | - Jiawei Nie
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine (Shanghai), International Center for Aging and Cancer, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xinpei Sun
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, 100101, China
| | - Xiaoqiang Tang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianfang Wang
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiaoran Wang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Siyuan Wang
- Clinical Research Institute, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China
| | - Xuan Wang
- Hepatobiliary and Pancreatic Center, Medical Research Center, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China
| | - Yaning Wang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Yuhan Wang
- University of Chinese Academy of Sciences, Beijing, 100049, China
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Rimo Wu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China
| | - Kai Xia
- Center for Stem Cell Biologyand Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China
| | - Fu-Hui Xiao
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China
| | - Lingyan Xu
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China
| | - Yingying Xu
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China
| | - Haoteng Yan
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China
| | - Liang Yang
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China
| | - Ruici Yang
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Yuanxin Yang
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China
| | - Yilin Ying
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
- International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China
| | - Le Zhang
- Gerontology Center of Hubei Province, Wuhan, 430000, China
- Institute of Gerontology, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Weiwei Zhang
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, 100853, China
| | - Wenwan Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Xing Zhang
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Zhuo Zhang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Min Zhou
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China
| | - Rui Zhou
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Qingchen Zhu
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Zhengmao Zhu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, 300071, China
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
| | - Feng Cao
- Department of Cardiology, The Second Medical Centre, Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, 100853, China.
| | - Zhongwei Cao
- State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Piu Chan
- National Clinical Research Center for Geriatric Diseases, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Guobing Chen
- Department of Microbiology and Immunology, School of Medicine, Jinan University, Guangzhou, 510632, China.
- Guangdong-Hong Kong-Macau Great Bay Area Geroscience Joint Laboratory, Guangzhou, 510000, China.
| | - Hou-Zao Chen
- Department of Biochemistryand Molecular Biology, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100005, China.
| | - Jun Chen
- Peking University Research Center on Aging, Beijing Key Laboratory of Protein Posttranslational Modifications and Cell Function, Department of Biochemistry and Molecular Biology, Department of Integration of Chinese and Western Medicine, School of Basic Medical Science, Peking University, Beijing, 100191, China.
| | - Weimin Ci
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
| | - Bi-Sen Ding
- State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| | - Qiurong Ding
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Feng Gao
- Key Laboratory of Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jing-Dong J Han
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Center for Quantitative Biology (CQB), Peking University, Beijing, 100871, China.
| | - Kai Huang
- Clinic Center of Human Gene Research, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Clinical Research Center of Metabolic and Cardiovascular Disease, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Metabolic Abnormalities and Vascular Aging, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Ageing and Regenerative Medicine, Jinan University, Guangzhou, 510632, China.
| | - Qing-Peng Kong
- CAS Center for Excellence in Animal Evolution and Genetics, Chinese Academy of Sciences, Kunming, 650223, China.
- State Key Laboratory of Genetic Resources and Evolution, Key Laboratory of Healthy Aging Research of Yunnan Province, Kunming Key Laboratory of Healthy Aging Study, KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650223, China.
| | - Ji Li
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China.
| | - Jian Li
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, 100730, China.
| | - Xin Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Baohua Liu
- School of Basic Medical Sciences, Shenzhen University Medical School, Shenzhen, 518060, China.
| | - Feng Liu
- Metabolic Syndrome Research Center, The Second Xiangya Hospital, Central South Unversity, Changsha, 410011, China.
| | - Lin Liu
- Department of Genetics and Cell Biology, College of Life Science, Nankai University, Tianjin, 300071, China.
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Institute of Translational Medicine, Tianjin Union Medical Center, Nankai University, Tianjin, 300000, China.
- State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin, 300350, China.
| | - Qiang Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230036, China.
| | - Qiang Liu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, 300052, China.
- Tianjin Institute of Immunology, Tianjin Medical University, Tianjin, 300070, China.
| | - Xingguo Liu
- CAS Key Laboratory of Regenerative Biology, Joint School of Life Sciences, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou Medical University, Guangzhou, 510530, China.
| | - Yong Liu
- College of Life Sciences, TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430072, China.
| | - Xianghang Luo
- Department of Endocrinology, Endocrinology Research Center, Xiangya Hospital of Central South University, Changsha, 410008, China.
| | - Shuai Ma
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Xinran Ma
- Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, 200241, China.
| | - Zhiyong Mao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Jing Nie
- The State Key Laboratory of Organ Failure Research, National Clinical Research Center of Kidney Disease, Division of Nephrology, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| | - Yaojin Peng
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Jing Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Jie Ren
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Ruibao Ren
- Shanghai Institute of Hematology, State Key Laboratory for Medical Genomics, National Research Center for Translational Medicine (Shanghai), International Center for Aging and Cancer, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- International Center for Aging and Cancer, Hainan Medical University, Haikou, 571199, China.
| | - Moshi Song
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Zhou Songyang
- MOE Key Laboratory of Gene Function and Regulation, Guangzhou Key Laboratory of Healthy Aging Research, School of Life Sciences, Institute of Healthy Aging Research, Sun Yat-sen University, Guangzhou, 510275, China.
- Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, China.
| | - Yi Eve Sun
- Stem Cell Translational Research Center, Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China.
| | - Yu Sun
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Department of Medicine and VAPSHCS, University of Washington, Seattle, WA, 98195, USA.
| | - Mei Tian
- Human Phenome Institute, Fudan University, Shanghai, 201203, China.
| | - Shusen Wang
- Research Institute of Transplant Medicine, Organ Transplant Center, NHC Key Laboratory for Critical Care Medicine, Tianjin First Central Hospital, Nankai University, Tianjin, 300384, China.
| | - Si Wang
- Beijing Municipal Geriatric Medical Research Center, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Aging Translational Medicine Center, International Center for Aging and Cancer, Xuanwu Hospital, Capital Medical University, Beijing, 100053, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| | - Xia Wang
- School of Pharmaceutical Sciences, Tsinghua University, Beijing, 100084, China.
| | - Xiaoning Wang
- Institute of Geriatrics, The second Medical Center, Beijing Key Laboratory of Aging and Geriatrics, National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, China.
| | - Yan-Jiang Wang
- Department of Neurology and Center for Clinical Neuroscience, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| | - Yunfang Wang
- Hepatobiliary and Pancreatic Center, Medical Research Center, Beijing Tsinghua Changgung Hospital, Beijing, 102218, China.
| | - Catherine C L Wong
- Clinical Research Institute, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Beijing, 100730, China.
| | - Andy Peng Xiang
- Center for Stem Cell Biologyand Tissue Engineering, Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Sun Yat-sen University, Guangzhou, 510080, China.
- National-Local Joint Engineering Research Center for Stem Cells and Regenerative Medicine, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Yichuan Xiao
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Zhengwei Xie
- Peking University International Cancer Institute, Health Science Center, Peking University, Beijing, 100101, China.
- Beijing & Qingdao Langu Pharmaceutical R&D Platform, Beijing Gigaceuticals Tech. Co. Ltd., Beijing, 100101, China.
| | - Daichao Xu
- Interdisciplinary Research Center on Biology and Chemistry, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai, 201210, China.
| | - Jing Ye
- Department of Geriatrics, Medical Center on Aging of Shanghai Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- International Laboratory in Hematology and Cancer, Shanghai Jiao Tong University School of Medicine/Ruijin Hospital, Shanghai, 200025, China.
| | - Rui Yue
- Institute for Regenerative Medicine, Shanghai East Hospital, Frontier Science Center for Stem Cell Research, Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Cuntai Zhang
- Gerontology Center of Hubei Province, Wuhan, 430000, China.
- Institute of Gerontology, Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| | - Hongbo Zhang
- Key Laboratory for Stem Cells and Tissue Engineering, Ministry of Education, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
- Advanced Medical Technology Center, The First Affiliated Hospital, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Liang Zhang
- CAS Key Laboratory of Tissue Microenvironment and Tumor, Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences and China National Center for Bioinformation, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
| | - Yong Zhang
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Yun-Wu Zhang
- Fujian Provincial Key Laboratory of Neurodegenerative Disease and Aging Research, Institute of Neuroscience, School of Medicine, Xiamen University, Xiamen, 361102, China.
| | - Zhuohua Zhang
- Key Laboratory of Molecular Precision Medicine of Hunan Province and Center for Medical Genetics, Institute of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China.
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421001, China.
| | - Tongbiao Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
| | - Yuzheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China.
- Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| | - Dahai Zhu
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou, 510005, China.
- The State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing, 100005, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, Shanghai Institute of Biochemistry and Cell Biology, Center for Excellence in Molecular Cell Science, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
| | - Gang Pei
- Shanghai Key Laboratory of Signaling and Disease Research, Laboratory of Receptor-Based Biomedicine, The Collaborative Innovation Center for Brain Science, School of Life Sciences and Technology, Tongji University, Shanghai, 200070, China.
| | - Guang-Hui Liu
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China.
- Institute for Stem Cell and Regeneration, Chinese Academy of Sciences, Beijing, 100101, China.
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China.
- Advanced Innovation Center for Human Brain Protection, and National Clinical Research Center for Geriatric Disorders, Xuanwu Hospital Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
37
|
Reed SC, Croessmann S, Park BH. CHIP Happens: Clonal Hematopoiesis of Indeterminate Potential and Its Relationship to Solid Tumors. Clin Cancer Res 2023; 29:1403-1411. [PMID: 36454121 PMCID: PMC10106364 DOI: 10.1158/1078-0432.ccr-22-2598] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 10/21/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is characterized by the expansion of hematopoietic cells harboring leukemia-associated somatic mutations in otherwise healthy people and occurs in at least 10% of adults over 70. It is well established that people with CHIP have increased rates of hematologic malignancy, increased risk of cardiovascular disease, and worse all-cause mortality compared with those without CHIP. Despite recent advancements in understanding CHIP as it relates to these known outcomes, much remains to be learned about the development and role of CHIP in other disease states. Emerging research has identified high rates of CHIP in patients with solid tumors, driven in part by oncologic therapy, and revealed associations between CHIP and differential outcomes in both solid tumors and other diseases. Recent studies have demonstrated that CHIP can contribute to dysregulated inflammatory signaling in multiple contexts, underscoring the importance of interrogating how CHIP might alter tumor immunology. Here, we review the role of CHIP mutations in clonal expansion of hematopoietic cells, explore the relationship between CHIP and solid tumors, and discuss the potential roles of CHIP in inflammation and solid tumor biology.
Collapse
Affiliation(s)
- Sarah C. Reed
- The Vanderbilt-Ingram Cancer Center, Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Sarah Croessmann
- The Vanderbilt-Ingram Cancer Center, Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Ben Ho Park
- The Vanderbilt-Ingram Cancer Center, Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
38
|
Goldman EA, Spellman PT, Agarwal A. Defining clonal hematopoiesis of indeterminate potential: evolutionary dynamics and detection under aging and inflammation. Cold Spring Harb Mol Case Stud 2023; 9:a006251. [PMID: 36889927 PMCID: PMC10240836 DOI: 10.1101/mcs.a006251] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 02/24/2023] [Indexed: 03/10/2023] Open
Abstract
Clonal hematopoiesis (CH), in which hematopoietic stem and progenitor cell (HSPC) clones and their progeny expand in the circulating blood cell population, occurs following the acquisition of somatic driver mutations. Individuals diagnosed with clonal hematopoiesis of indeterminate potential (CHIP) carry somatic mutations in hematological malignancy-associated driver genes, historically at or above a variant allele frequency of 2%, but do not exhibit abnormal blood cell counts or any other symptoms of hematologic disease. However, CHIP is associated with moderately increased risk of hematological cancer and a greater likelihood of cardiovascular and pulmonary disease. Recent advances in the resolution of high-throughput sequencing experiments suggest CHIP is much more prevalent in the population than once thought, particularly among those aged 60 and over. Although CHIP does elevate the risk of eventual hematological malignancy, only one in 10 individuals with CHIP will receive such a diagnosis; the problem lies in the continued difficulty in accurately separating the 10% of CHIP patients who are most likely to be in a premalignant state from those who are not, given the heterogeneity of this condition and the etiology of the associated hematological cancers. Concerns over the risk of eventual malignancies must be balanced with growing recognition of CH as a common age-dependent occurrence, and efforts to better characterize and differentiate oncogenic clonal expansion from that which is much more benign. In this review, we discuss evolutionary dynamics of CH and CHIP, the relationship of CH to aging and inflammation, and the role of the epigenome in promoting potentially pathogenic or benign cellular trajectories. We outline molecular mechanisms that may contribute to heterogeneity in the etiology of CHIP and the incidence of malignant disease among individuals. Finally, we discuss epigenetic markers and modifications for CHIP detection and monitoring with the potential for translational applications and clinical utility in the near future.
Collapse
Affiliation(s)
- Elisabeth A Goldman
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon 97239, USA;
| | - Paul T Spellman
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Department of Molecular and Medical Genetics, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| | - Anupriya Agarwal
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Department of Molecular and Medical Genetics, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
- Division of Oncological Sciences, Knight Cancer Institute, Oregon Health & Science University, Portland, Oregon 97239, USA
| |
Collapse
|
39
|
Moon I, Kong MG, Ji YS, Kim SH, Park SK, Suh J, Jang MA. Clinical, Mutational, and Transcriptomic Characteristics in Elderly Korean Individuals With Clonal Hematopoiesis Driver Mutations. Ann Lab Med 2023; 43:145-152. [PMID: 36281508 PMCID: PMC9618905 DOI: 10.3343/alm.2023.43.2.145] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/07/2022] [Accepted: 09/29/2022] [Indexed: 11/07/2022] Open
Abstract
Background Clonal hematopoiesis of indeterminate potential (CHIP), which is defined as the presence of blood cells originating from somatically mutated hematopoietic stem cells, is common among the elderly and is associated with an increased risk of hematologic malignancies. We investigated the clinical, mutational, and transcriptomic characteristics in elderly Korean individuals with CHIP mutations. Methods We investigated CHIP in 90 elderly individuals aged ≥60 years with normal complete blood counts at a tertiary-care hospital in Korea between June 2021 and February 2022. Clinical and laboratory data were prospectively obtained. Targeted next-generation sequencing of 49 myeloid malignancy driver genes and massively parallel RNA sequencing were performed to explore the molecular spectrum and transcriptomic characteristics of CHIP mutations. Results We detected 51 mutations in 10 genes in 37 (41%) of the study individuals. CHIP prevalence increased with age. CHIP mutations were observed with high prevalence in DNMT3A (26 individuals) and TET2 (eight individuals) and were also found in various other genes, including KDM6A, SMC3, TP53, BRAF, PPM1D, SRSF2, STAG1, and ZRSR2. Baseline characteristics, including age, confounding diseases, and blood cell parameters, showed no significant differences. Using mRNA sequencing, we characterized the altered gene expression profile, implicating neutrophil degranulation and innate immune system dysregulation. Conclusions Somatic CHIP driver mutations are common among the elderly in Korea and are detected in various genes, including DNMT3A and TET2. Our study highlights that chronic dysregulation of innate immune signaling is associated with the pathogenesis of various diseases, including hematologic malignancies.
Collapse
Affiliation(s)
- Inki Moon
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Min Gyu Kong
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Young Sok Ji
- Division of Hematooncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Se Hyung Kim
- Division of Hematooncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Seong Kyu Park
- Division of Hematooncology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Jon Suh
- Division of Cardiology, Department of Internal Medicine, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| | - Mi-Ae Jang
- Department of Laboratory Medicine and Genetics, Soonchunhyang University Bucheon Hospital, Soonchunhyang University College of Medicine, Bucheon, Korea
| |
Collapse
|
40
|
Yu K, Deuitch N, Merguerian M, Cunningham L, Davis J, Bresciani E, Diemer J, Andrews E, Young A, Donovan F, Sood R, Craft K, Chong S, Chandrasekharappa S, Mullikin J, Liu PP. Genomic Landscape of Patients with Germline RUNX1 Variants and Familial Platelet Disorder with Myeloid Malignancy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.17.524290. [PMID: 36789433 PMCID: PMC9928034 DOI: 10.1101/2023.01.17.524290] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Germline RUNX1 mutations lead to familial platelet disorder with associated myeloid malignancies (FPDMM), which is characterized by thrombocytopenia and a life-long risk (35-45%) of hematological malignancies. We recently launched a longitudinal natural history study for patients with FPDMM at the NIH Clinical Center. Among 29 families with research genomic data, 28 different germline RUNX1 variants were detected. Besides missense mutations enriched in Runt homology domain and loss-of-function mutations distributed throughout the gene, splice-region mutations and large deletions were detected in 6 and 7 families, respectively. In 24 of 54 (44.4%) non-malignant patients, somatic mutations were detected in at least one of the clonal hematopoiesis of indeterminate potential (CHIP) genes or acute myeloid leukemia (AML) driver genes. BCOR was the most frequently mutated gene (in 9 patients), and multiple BCOR mutations were identified in 4 patients. Mutations in 7 other CHIP or AML driver genes ( DNMT3A, TET2, NRAS, SETBP1, SF3B1, KMT2C , and LRP1B ) were also found in more than one non-malignant patient. Moreover, three unrelated patients (one with myeloid malignancy) carried somatic mutations in NFE2 , which regulates erythroid and megakaryocytic differentiation. Sequential sequencing data from 19 patients demonstrated dynamic changes of somatic mutations over time, and stable clones were more frequently found in elderly patients. In summary, there are diverse types of germline RUNX1 mutations and high frequency of somatic mutations related to clonal hematopoiesis in patients with FPDMM. Monitoring dynamic changes of somatic mutations prospectively will benefit patients’ clinical management and reveal mechanisms for progression to myeloid malignancies. Key Points Comprehensive genomic profile of patients with FPDMM with germline RUNX1 mutations. Rising clonal hematopoiesis related secondary mutations that may lead to myeloid malignancies.
Collapse
|
41
|
Van Horebeek L, Dedoncker N, Dubois B, Goris A. Frequent somatic mosaicism in T lymphocyte subsets in individuals with and without multiple sclerosis. Front Immunol 2022; 13:993178. [PMID: 36618380 PMCID: PMC9817019 DOI: 10.3389/fimmu.2022.993178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 12/09/2022] [Indexed: 12/25/2022] Open
Abstract
Background Somatic variants are variations in an individual's genome acquired after the zygotic stadium and result from mitotic errors or not (fully) repaired DNA damage. Objectives To investigate whether somatic mosaicism in T lymphocyte subsets is enriched early in multiple sclerosis (MS). Methods We identified somatic variants with variant allele fractions ≥1% across the whole exome in CD4+ and CD8+ T lymphocytes of 21 treatment-naive MS patients with <5 years of disease duration and 16 partially age-matched healthy controls. We investigated the known somatic STAT3 variant p.Y640F in peripheral blood in a larger cohort of 446 MS patients and 259 controls. Results All subjects carried 1-142 variants in CD4+ or CD8+ T lymphocytes. Variants were more common, more abundant, and increased with age in CD8+ T lymphocytes. Somatic variants were common in the genes DNMT3A and especially STAT3. Overall, the presence or abundance of somatic variants, including the STAT3 p.Y640F variant, did not differ between MS patients and controls. Conclusions Somatic variation in T lymphocyte subsets is widespread in both control individuals and MS patients. Somatic mosaicism in T lymphocyte subsets is not enriched in early MS and thus unlikely to contribute to MS risk, but future research needs to address whether a subset of variants influences disease susceptibility.
Collapse
Affiliation(s)
- Lies Van Horebeek
- Laboratory for Neuroimmunology, Department of Neurosciences, Leuven Brain Institute, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Nina Dedoncker
- Laboratory for Neuroimmunology, Department of Neurosciences, Leuven Brain Institute, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Bénédicte Dubois
- Laboratory for Neuroimmunology, Department of Neurosciences, Leuven Brain Institute, Katholieke Universiteit (KU) Leuven, Leuven, Belgium,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| | - An Goris
- Laboratory for Neuroimmunology, Department of Neurosciences, Leuven Brain Institute, Katholieke Universiteit (KU) Leuven, Leuven, Belgium,*Correspondence: An Goris,
| |
Collapse
|
42
|
Urban VS, Cegledi A, Mikala G. Multiple myeloma, a quintessential malignant disease of aging: a geroscience perspective on pathogenesis and treatment. GeroScience 2022; 45:727-746. [PMID: 36508077 PMCID: PMC9742673 DOI: 10.1007/s11357-022-00698-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 11/18/2022] [Indexed: 12/14/2022] Open
Abstract
Multiple myeloma (MM) is an incurable plasma cell malignancy, which is predominantly a disease of older adults (the median age at diagnosis is 70 years). The slow progression from asymptomatic stages and the late-onset of MM suggest fundamental differences compared to many other hematopoietic system-related malignancies. The concept discussed in this review is that age-related changes at the level of terminally differentiated plasma cells act as the main risk factors for the development of MM. Epigenetic and genetic changes that characterize both MM development and normal aging are highlighted. The relationships between cellular aging processes, genetic mosaicism in plasma cells, and risk for MM and the stochastic processes contributing to clonal selection and expansion of mutated plasma cells are investigated. In line with the DNA damage accumulation theory of aging, in this review, the evolution of monoclonal gammopathy to symptomatic MM is considered. Therapeutic consequences of age-dependent comorbidities that lead to frailty and have fundamental influence on treatment outcome are described. The importance of considering geriatric states when planning the life-long treatment course of an elderly MM patient in order to achieve maximal therapeutic benefit is emphasized.
Collapse
Affiliation(s)
- Veronika S. Urban
- Department of Morphology and Physiology, Faculty of Health Sciences, Semmelweis University, Budapest, Hungary
| | - Andrea Cegledi
- Department of Hematology and Stem Cell Transplantation, South Pest Central Hospital–National Institute for Hematology and Infectious Diseases, Budapest, Hungary
| | - Gabor Mikala
- Department of Hematology and Stem Cell Transplantation, South Pest Central Hospital-National Institute for Hematology and Infectious Diseases, Budapest, Hungary.
| |
Collapse
|
43
|
Ambinder AJ, DeZern AE. Navigating the contested borders between myelodysplastic syndrome and acute myeloid leukemia. Front Oncol 2022; 12:1033534. [PMID: 36387170 PMCID: PMC9650616 DOI: 10.3389/fonc.2022.1033534] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/11/2022] [Indexed: 10/23/2023] Open
Abstract
Myelodysplastic syndrome and acute myeloid leukemia are heterogeneous myeloid neoplasms which arise from the accumulation of mutations in a myeloid stem cell or progenitor that confer survival or growth advantages. These disease processes are formally differentiated by clinical, laboratory, and morphological presentations, especially with regard to the preponderance of blasts in the peripheral blood or bone marrow (AML); however, they are closely associated through their shared lineage as well as their existence on a spectrum with some cases of MDS displaying increased blasts, a feature that reflects more AML-like behavior, and the propensity for MDS to transform into AML. It is increasingly recognized that the distinctions between these two entities result from the divergent patterns of genetic alterations that drive each of them. Mutations in genes related to chromatin-remodeling and the spliceosome are seen in both MDS and AML arising out of antecedent MDS, while mutations in genes related to signaling pathways such as RAS or FLT3 are more typically seen in AML or otherwise are a harbinger of transformation. In this review, we focus on the insights into the biological and genetic distinctions and similarities between MDS and AML that are now used to refine clinical prognostication, guide disease management, and to inform development of novel therapeutic approaches.
Collapse
Affiliation(s)
| | - Amy E. DeZern
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
44
|
Yeaton A, Cayanan G, Loghavi S, Dolgalev I, Leddin EM, Loo CE, Torabifard H, Nicolet D, Wang J, Corrigan K, Paraskevopoulou V, Starczynowski DT, Wang E, Abdel-Wahab O, Viny AD, Stone RM, Byrd JC, Guryanova OA, Kohli RM, Cisneros GA, Tsirigos A, Eisfeld AK, Aifantis I, Guillamot M. The Impact of Inflammation-Induced Tumor Plasticity during Myeloid Transformation. Cancer Discov 2022; 12:2392-2413. [PMID: 35924979 PMCID: PMC9547930 DOI: 10.1158/2159-8290.cd-21-1146] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 05/26/2022] [Accepted: 08/01/2022] [Indexed: 11/16/2022]
Abstract
Clonal hematopoiesis (CH) is an aging-associated condition characterized by the clonal outgrowth of mutated preleukemic cells. Individuals with CH are at an increased risk of developing hematopoietic malignancies. Here, we describe a novel animal model carrying a recurrent TET2 missense mutation frequently found in patients with CH and leukemia. In a fashion similar to CH, animals show signs of disease late in life when they develop a wide range of myeloid neoplasms, including acute myeloid leukemia (AML). Using single-cell transcriptomic profiling of the bone marrow, we show that disease progression in aged animals correlates with an enhanced inflammatory response and the emergence of an aberrant inflammatory monocytic cell population. The gene signature characteristic of this inflammatory population is associated with poor prognosis in patients with AML. Our study illustrates an example of collaboration between a genetic lesion found in CH and inflammation, leading to transformation and the establishment of blood neoplasms. SIGNIFICANCE Progression from a preleukemic state to transformation, in the presence of TET2 mutations, is coupled with the emergence of inflammation and a novel population of inflammatory monocytes. Genes characteristic of this inflammatory population are associated with the worst prognosis in patients with AML. These studies connect inflammation to progression to leukemia. See related commentary by Pietras and DeGregori, p. 2234 . This article is highlighted in the In This Issue feature, p. 2221.
Collapse
Affiliation(s)
- Anna Yeaton
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Geraldine Cayanan
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas, MD Anderson Cancer Center, Houston, TX, USA
| | - Igor Dolgalev
- Applied Bioinformatics Laboratories, Office of Science & Research, NYU School of Medicine, New York, NY, USA
| | - Emmett M. Leddin
- Department of Physics, University of Texas at Dallas, Richardson, TX, USA; Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA
| | - Christian E. Loo
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Hedieh Torabifard
- Department of Physics, University of Texas at Dallas, Richardson, TX, USA; Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA
| | - Deedra Nicolet
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingjing Wang
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Kate Corrigan
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Varvara Paraskevopoulou
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Daniel T Starczynowski
- Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA; Department of Cancer Biology, University of Cincinnati, Cincinnati, OH, USA; Division of Experimental Hematology and Cancer Biology, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
| | - Eric Wang
- MSK Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Omar Abdel-Wahab
- MSK Center for Hematologic Malignancies, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Aaron D Viny
- Department of Genetics & Development, Columbia University, New York, NY, USA; Columbia Stem Cell Initiative, Columbia University, New York, NY, USA; Cancer Genomics and Epigenomics Program, Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Richard M. Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - John C. Byrd
- Department of Internal Medicine, University of Cincinnati, Cincinnati, OH, USA
| | - Olga A. Guryanova
- Department of Pharmacology and Therapeutics, University of Florida College of Medicine, Gainesville, FL, USA
| | - Rahul M. Kohli
- Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - G. Andrés Cisneros
- Department of Physics, University of Texas at Dallas, Richardson, TX, USA; Department of Chemistry and Biochemistry, University of Texas at Dallas, Richardson, TX, USA
| | - Aristotelis Tsirigos
- Applied Bioinformatics Laboratories, Office of Science & Research, NYU School of Medicine, New York, NY, USA
| | - Ann-Kathrin Eisfeld
- Clara D. Bloomfield Center for Leukemia Outcomes Research; The Ohio State University, Comprehensive Cancer Center, Columbus, OH, USA
- Division of Hematology, The Ohio State University, Comprehensive Cancer Center, Columbus/OH, USA
| | - Iannis Aifantis
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| | - Maria Guillamot
- Department of Pathology, NYU Grossman School of Medicine, New York, NY, USA
- Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, NY, USA
| |
Collapse
|
45
|
Kanai M. Current Clinical Practice of Precision Medicine Using Comprehensive Genomic Profiling Tests in Biliary Tract Cancer in Japan. Curr Oncol 2022; 29:7272-7284. [PMID: 36290850 PMCID: PMC9599999 DOI: 10.3390/curroncol29100573] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/06/2022] Open
Abstract
With the recent advances of next generation sequencing technologies, comprehensive genomic profiling (CGP) tests, which are designed to measure more than hundreds of cancer-related genes at a time, have now been widely introduced into daily clinical practice. For the patients whose tumor samples are not fit for tissue-based CGP tests, a blood-based CGP test (liquid biopsy) is available as an alternative option. Three CGP tests, "OncoGuide NCC™Oncopanel System (124 genes)", "FoundationOne®CDx (324 genes)", and "Founda-tionOne®CDx Liquid (324 genes)", are now reimbursed by public insurance in 233 hospitals designated for cancer genomic medicine in Japan. In biliary tract cancer, the prevalence of druggable variants is relatively higher compared to other cancer types and the European Society for Medical Oncology recommends routine use of CGP tests for advanced biliary tract cancer to guide treatment options. The latest National Cancer Center Network guideline lists eight druggable markers (NTRK fusion, MSI-H, TMB-H, BRAF V600E, FGFR2 fusions/rearrangement, IDH1 mutations, RET fusion, and HER2 overexpression) and matched therapies. In Japan, matched therapies for four markers (NTRK, MSI-H, TMB-H, and FGFR2) are reimbursed by public insurance (as of September 2022). The progress of genomic profiling technology will contribute to the improvement of the dismal clinical outcomes of this disease in the future.
Collapse
Affiliation(s)
- Masashi Kanai
- Department of Therapeutic Oncology, Graduate School of Medicine, Kyoto University, Kyoto 606-8507, Japan
| |
Collapse
|
46
|
Stolzenbach V, Woods DC, Tilly JL. Non-neutral clonal selection and its potential role in mammalian germline stem cell dysfunction with advancing age. Front Cell Dev Biol 2022; 10:942652. [PMID: 36081905 PMCID: PMC9445274 DOI: 10.3389/fcell.2022.942652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
The concept of natural selection, or "survival of the fittest", refers to an evolutionary process in nature whereby traits emerge in individuals of a population through random gene alterations that enable those individuals to better adapt to changing environmental conditions. This genetic variance allows certain members of the population to gain an advantage over others in the same population to survive and reproduce in greater numbers under new environmental pressures, with the perpetuation of those advantageous traits in future progeny. Here we present that the behavior of adult stem cells in a tissue over time can, in many respects, be viewed in the same manner as evolution, with each stem cell clone being representative of an individual within a population. As stem cells divide or are subjected to cumulative oxidative damage over the lifespan of the organism, random genetic alterations are introduced into each clone that create variance in the population. These changes may occur in parallel to, or in response to, aging-associated changes in microenvironmental cues perceived by the stem cell population. While many of these alterations will be neutral or silent in terms of affecting cell function, a small fraction of these changes will enable certain clones to respond differently to shifts in microenvironmental conditions that arise with advancing age. In some cases, the same advantageous genetic changes that support survival and expansion of certain clones over others in the population (viz. non-neutral competition) could be detrimental to the downstream function of the differentiated stem cell descendants. In the context of the germline, such a situation would be devastating to successful propagation of the species across generations. However, even within a single generation, the “evolution” of stem cell lineages in the body over time can manifest into aging-related organ dysfunction and failure, as well as lead to chronic inflammation, hyperplasia, and cancer. Increased research efforts to evaluate stem cells within a population as individual entities will improve our understanding of how organisms age and how certain diseases develop, which in turn may open new opportunities for clinical detection and management of diverse pathologies.
Collapse
|
47
|
Gaulin C, Kelemen K, Arana Yi C. Molecular Pathways in Clonal Hematopoiesis: From the Acquisition of Somatic Mutations to Transformation into Hematologic Neoplasm. Life (Basel) 2022; 12:1135. [PMID: 36013314 PMCID: PMC9410004 DOI: 10.3390/life12081135] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 07/23/2022] [Accepted: 07/26/2022] [Indexed: 11/16/2022] Open
Abstract
Hematopoietic stem cell aging, through the acquisition of somatic mutations, gives rise to clonal hematopoiesis (CH). While a high prevalence of CH has been described in otherwise healthy older adults, CH confers an increased risk of both hematologic and non-hematologic diseases. Classification of CH into clonal hematopoiesis of indeterminate potential (CHIP) and clonal cytopenia of undetermined significance (CCUS) further describes this neoplastic myeloid precursor state and stratifies individuals at risk of developing clinically significant complications. The sequential acquisition of driver mutations, such as DNMT3A, TET2, and ASXL1, provide a selective advantage and lead to clonal expansion. Inflammation, microbiome signatures, and external selective pressures also contribute to clonal evolution. Despite significant progress in recent years, the precise molecular mechanisms driving CH transformation to hematologic neoplasms are not well defined. Further understanding of these complex mechanisms may improve risk stratification and introduce therapeutic interventions in CH. Here we discuss the genetic drivers underpinning CH, mechanisms for clonal evolution, and transformation to hematologic neoplasm.
Collapse
Affiliation(s)
- Charles Gaulin
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ 85054, USA;
| | - Katalin Kelemen
- Division of Hematopathology, Department of Laboratory Medicine and Pathology, Mayo Clinic, Phoenix, AZ 85054, USA;
| | - Cecilia Arana Yi
- Division of Hematology and Medical Oncology, Department of Medicine, Mayo Clinic, Phoenix, AZ 85054, USA;
| |
Collapse
|
48
|
Hoermann G. Clinical Significance of Clonal Hematopoiesis of Indeterminate Potential in Hematology and Cardiovascular Disease. Diagnostics (Basel) 2022; 12:1613. [PMID: 35885518 PMCID: PMC9317488 DOI: 10.3390/diagnostics12071613] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/26/2022] [Accepted: 06/29/2022] [Indexed: 01/15/2023] Open
Abstract
Liquid profiling uses circulating tumor DNA (ctDNA) for minimal invasive tumor mutational profiling from peripheral blood. The presence of somatic mutations in peripheral blood cells without further evidence of a hematologic neoplasm defines clonal hematopoiesis of indeterminate potential (CHIP). CHIP-mutations can be found in the cell-free DNA (cfDNA) of plasma, are a potential cause of false positive results in liquid profiling, and thus limit its usage in screening settings. Various strategies are in place to mitigate the effect of CHIP on the performance of ctDNA assays, but the detection of CHIP also represents a clinically significant incidental finding. The sequelae of CHIP comprise the risk of progression to a hematologic neoplasm including therapy-related myeloid neoplasms. While the hematological risk increases with the co-occurrence of unexplained blood count abnormalities, a number of non-hematologic diseases have independently been associated with CHIP. In particular, CHIP represents a major risk factor for cardiovascular disease such as atherosclerosis or heart failure. The management of CHIP requires an interdisciplinary setting and represents a new topic in the field of cardio-oncology. In the future, the information on CHIP may be taken into account for personalized therapy of cancer patients.
Collapse
|
49
|
Law SM, Akizuki S, Morinobu A, Ohmura K. A case of refractory systemic lupus erythematosus with monocytosis exhibiting somatic KRAS mutation. Inflamm Regen 2022; 42:10. [PMID: 35361277 PMCID: PMC8973904 DOI: 10.1186/s41232-022-00195-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/20/2022] [Indexed: 11/11/2022] Open
Abstract
Background Systemic lupus erythematosus (SLE), an autoimmune disorder that damages various organ systems, is caused by a combination of genetic and environmental factors. Although germline mutations of several genes are known to cause juvenile SLE, most of the susceptibility genetic variants of adult SLE are common variants of the population, somatic mutations that cause or exacerbate SLE have not been reported. We hereby report a refractory SLE case with monocytosis accompanying somatic KRAS mutation that have been shown to cause lupus-like symptoms. Case presentation A 60-year-old female patient who had been diagnosed with SLE was admitted to our hospital. Although prednisolone and tacrolimus treatments had kept her thrombocytopenia and anti-DNA Ab level at bay for more than 4 years, a diagnosis of transverse myelitis was made when she became acutely ill with pleocytosis. Elevated cells (predominately monocytes), protein, IgG, and IL-6 levels were also found in the cerebrospinal fluid (CSF) of the patient. Standard pulse treatments of methylprednisolone, high-dose of prednisolone, and intravenous cyclophosphamide in combination with plasma exchange could not alleviate the refractory neural and autoimmune manifestation. Monocytosis of peripheral blood was also noted. Flow cytometric analysis revealed elevated ratio of CD14+CD16+ atypical monocytes, which excluded the possibility of chronic myelomonocytic leukemia. Lupus-like symptoms with monocytosis reminded us of Ras-associated autoimmune leukoproliferative disorder, and Sanger sequencing of KRAS and NRAS genes from the patients’ peripheral blood mononuclear cells (PBMC), sorted CD3+ lymphocytes and CD14+ monocytes, and cerebrospinal fluid were performed. An activating KRAS somatic mutation was found in the patients’ DNA at the time of encephalomyelitis diagnosis. Conclusion Somatic mutations of some genes including KRAS may cause the refractoriness of SLE.
Collapse
Affiliation(s)
- Sze-Ming Law
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, 54 kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Shuji Akizuki
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, 54 kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Akio Morinobu
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, 54 kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Koichiro Ohmura
- Department of Rheumatology and Clinical Immunology, Graduate School of Medicine, Kyoto University, 54 kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan. .,Department of Rheumatology, Kobe City Medical Center General Hospital, Kobe, Japan.
| |
Collapse
|
50
|
Wang Y, Yu T, Dong Q, Liu S, Yu Y, Zhao HY, Ma J, Dong L, Wang L, Ma D, Zhao Y, Hou Y, Liu X, Peng J, Hou M. Clonal hematopoiesis in primary immune thrombocytopenia. Blood Cancer J 2022; 12:40. [PMID: 35293382 PMCID: PMC8924250 DOI: 10.1038/s41408-022-00641-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 11/15/2022] Open
Affiliation(s)
- Yanming Wang
- Department of Hematology, Yantai Yuhuangding Hospital Affiliated to Qingdao University, Yantai, China.,Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Tianshu Yu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Qiaofeng Dong
- Department of Hematology, Heze Municipal Hospital, Heze, China
| | - Shuang Liu
- Department of Hematology, Taian Central Hospital, Taian, China
| | - Yafei Yu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Hong Yu Zhao
- Department of Hematology, Jinan Central Hospital Affiliated to Shandong University, Jinan, China
| | - Ji Ma
- Department of Medical Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Lin Dong
- Department of Hematology, The First Affiliated Hospital of Shandong First Medical University, Taian, China
| | - Liang Wang
- Department of Hematology, Shengli Oilfield Central Hospital, Dongying, China
| | - Daoxin Ma
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yajing Zhao
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Yu Hou
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Xinguang Liu
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.
| | - Jun Peng
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Ming Hou
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Shandong Provincial Key Laboratory of Immunohaematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Leading Research Group of Scientific Innovation, Department of Science and Technology of Shandong Province, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China.,Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|