1
|
Tsourveloudis I, Georgiadi EC, Vatalis G, Kotsi P. Case report of a patient with VEXAS syndrome. Medicine (Baltimore) 2023; 102:e36738. [PMID: 38206689 PMCID: PMC10754568 DOI: 10.1097/md.0000000000036738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 11/30/2023] [Indexed: 01/13/2024] Open
Abstract
RATIONALE Hematological malignancies have always been a challenge for scientists because there is a constant need to better define these entities. Myelodysplastic syndromes (MDS) are clonal hematopoietic disorders characterized by ineffective hematopoiesis. Cytogenetics and molecular findings are a prerequisite for these syndromes as they confirm the clonal nature of the disease. However, MDS is often linked to autoimmunity and inflammation as part of its pathogenesis. Recently, VEXAS syndrome (vacuoles, E1 enzyme, X-linked, autoinflammatory, somatic) linked these two in a single mutation, suggesting that the heterogeneity among hematological malignancies often demands a more personalized medicine by tailoring medical treatment to the individual characteristics of each patient. PATIENT CONCERNS We present a case of VEXAS syndrome regarding a 63-year-old male patient who initially presented with episodes of low fever, polyarthritis of the knees and ankles, polymyalgia, and fatigue. His laboratory examinations revealed increased levels of serum inflammatory markers. DIAGNOSES Diagnosis was based on high clinical suspicion, laboratory findings, and vacuolization of the erythroid and myeloid precursors in the bone marrow evaluation. Mutational status of ubiquitin-like modifier activating enzyme 1 gene was positive with a 68.8% allelomorph frequency (rs782416867). INTERVENTIONS Therapy was based on controlling inflammation with the use of glucocorticoids and treating MDS-related anemia with the use of erythropoietin. OUTCOMES Currently, the patient visits our department regularly. He is still receiving the aforementioned treatment. He did not mention any new incidents for the time being. LESSONS VEXAS syndrome as a newly identified entity might be often underestimated since its clinical presentation is notably diverse.
Collapse
Affiliation(s)
| | - Eleni C. Georgiadi
- Transfusion Department, University General Hospital of Larissa, Larissa, Greece
| | | | - Paraskevi Kotsi
- Transfusion Department, University General Hospital of Larissa, Larissa, Greece
| |
Collapse
|
2
|
Yu D, Yu X, Ye A, Xu C, Li X, Geng W, Zhu L. Profiling of gut microbial dysbiosis in adults with myeloid leukemia. FEBS Open Bio 2021; 11:2050-2059. [PMID: 33993646 PMCID: PMC8406483 DOI: 10.1002/2211-5463.13193] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 04/25/2021] [Accepted: 05/12/2021] [Indexed: 11/24/2022] Open
Abstract
Dysregulation of gut microbiota is implicated in the pathogenesis of various diseases, including metabolic diseases, inflammatory diseases, and cancer. To date, the link between gut microbiota and myeloid leukemia (ML) remains largely unelucidated. Herein, a total of 29 patients with acute myeloid leukemia (AML), 17 patients with chronic myeloid leukemia (CML), and 33 healthy subjects were enrolled, and gut microbiota were profiled via Illumina sequencing of the 16S rRNA. We evaluated the correlation between ML and gut microbiota. The microbial α‐diversity and β‐diversity exhibited significant differences between ML patients and healthy controls (HCs). Compared to healthy subjects, we found that at the phylum level, the relative abundance of Actinobacteria, Acidobacteria, and Chloroflexi was increased, while that of Tenericutes was decreased. Correspondingly, at the genus level in ML, Streptococcus were increased, especially in AML patients, while Megamonas (P = 0.02), Lachnospiraceae NC2004 group, and Prevotella 9 (P = 0.007) were decreased. Moreover, ML‐enriched species, including Sphingomonas, Lysobacyer, Helicobacter, Lactobacillus, Enterococcus, and Clostridium sensu stricto 1, were identified. Our results indicate that the gut microbiota was altered in ML patients compared to that of healthy subjects, which could contribute to the elucidation of microbiota‐related pathogenesis of ML, and the development of novel therapeutic strategies in the treatment of ML.
Collapse
Affiliation(s)
- Dandan Yu
- Department of Clinical Laboratory Sciences, The First Affiliated Hospital of Wenzhou Medical University, China
| | - Xiaomin Yu
- Department of Clinical Laboratory Sciences, The First Affiliated Hospital of Wenzhou Medical University, China
| | - Aifang Ye
- Department of Translational Medicine Center, The First Affiliated Hospital of Wenzhou Medical University, China
| | - Chunquan Xu
- Department of Clinical Laboratory Sciences, The First Affiliated Hospital of Wenzhou Medical University, China
| | - Xiaolong Li
- Department of Clinical Laboratory Sciences, The First Affiliated Hospital of Wenzhou Medical University, China
| | - Wujun Geng
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, China
| | - Liqing Zhu
- Department of Clinical Laboratory Sciences, The First Affiliated Hospital of Wenzhou Medical University, China
| |
Collapse
|
3
|
Sant'Antonio E, Camerini C, Rizzo V, Musolino C, Allegra A. Genetic Heterogeneity in Chronic Myeloid Leukemia: How Clonal Hematopoiesis and Clonal Evolution May Influence Prognosis, Treatment Outcome, and Risk of Cardiovascular Events. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:573-579. [PMID: 34078586 DOI: 10.1016/j.clml.2021.04.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2021] [Revised: 04/18/2021] [Accepted: 04/23/2021] [Indexed: 12/17/2022]
Abstract
Chronic myeloid leukemia (CML) has long been considered as a model of cancer caused by a single-driver genetic lesion (BCR/ABL1 rearrangement) that codes for a unique, gain-of-function, deregulated protein. However, in the last decade, high-throughput sequencing technologies have shed light on a more complex genetic landscape, in which additional mutations may be found in different disease phases, including diagnosis. These genetic lesions may even precede the occurrence of the Philadelphia (Ph) chromosome, pointing to an antecedent premalignant state of clonal hematopoiesis (CH) at least in some patients. Preliminary data support the hypothesis that the most frequent CH-associated mutations (DNMT3A, TET2, and ASXL1) may be associated with a risk of vascular event, but a definitive answer for this topic is still lacking. Moreover, several recent studies have linked a much more complex genetic background in chronic-phase CML, including signs of clonal evolution over time, with depth of treatment responses or with patient survival. In the present review, we address the current state of the art on age-related CH, its association with cardiovascular risk, and its pathophysiology; review the current knowledge on CH that precedes the acquisition of the Ph chromosome in CML patients; and discuss available evidence on the prognostic and predictive value of additional mutations in chronic-phase CML, either as a sign of clonal dynamics under treatment or as markers of an antecedent CH.
Collapse
Affiliation(s)
- Emanuela Sant'Antonio
- Division of Hematology, Azienda USL Toscana Nord Ovest, Ospedale San Luca, Lucca, Italy.
| | - Chiara Camerini
- Division of Hematology, Azienda USL Toscana Nord Ovest, Ospedale San Luca, Lucca, Italy.
| | - Vincenzo Rizzo
- Department of Clinical and Experimental Medicine, University of Messina, Messina, Italy.
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Messina, Italy.
| | - Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood "Gaetano Barresi", University of Messina, Messina, Italy.
| |
Collapse
|
4
|
Papa V, Marracino L, Fortini F, Rizzo P, Campo G, Vaccarezza M, Vieceli Dalla Sega F. Translating Evidence from Clonal Hematopoiesis to Cardiovascular Disease: A Systematic Review. J Clin Med 2020; 9:jcm9082480. [PMID: 32748835 PMCID: PMC7465104 DOI: 10.3390/jcm9082480] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2020] [Revised: 07/21/2020] [Accepted: 07/28/2020] [Indexed: 02/07/2023] Open
Abstract
Some random mutations can confer a selective advantage to a hematopoietic stem cell. As a result, mutated hematopoietic stem cells can give rise to a significant proportion of mutated clones of blood cells. This event is known as "clonal hematopoiesis." Clonal hematopoiesis is closely associated with age, and carriers show an increased risk of developing blood cancers. Clonal hematopoiesis of indeterminate potential is defined by the presence of clones carrying a mutation associated with a blood neoplasm without obvious hematological malignancies. Unexpectedly, in recent years, it has emerged that clonal hematopoiesis of indeterminate potential carriers also have an increased risk of developing cardiovascular disease. Mechanisms linking clonal hematopoiesis of indeterminate potential to cardiovascular disease are only partially known. Findings in animal models indicate that clonal hematopoiesis of indeterminate potential-related mutations amplify inflammatory responses. Consistently, clinical studies have revealed that clonal hematopoiesis of indeterminate potential carriers display increased levels of inflammatory markers. In this review, we describe progress in our understanding of clonal hematopoiesis in the context of cancer, and we discuss the most recent findings linking clonal hematopoiesis of indeterminate potential and cardiovascular diseases.
Collapse
Affiliation(s)
- Veronica Papa
- Department of Motor Sciences and Wellness (DiSMeB), Università Degli Studi di Napoli “Parthenope,” 80133 Napoli, Italy;
- FAPAB Research Center, 96012 Avola (SR), Italy
| | - Luisa Marracino
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Via Fossato di Mortara 64/B, 44121 Ferrara, Italy;
| | - Francesca Fortini
- Translational Research Center, Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; (F.F.); (G.C.); (F.V.D.S.)
| | - Paola Rizzo
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Via Fossato di Mortara 64/B, 44121 Ferrara, Italy;
- Translational Research Center, Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; (F.F.); (G.C.); (F.V.D.S.)
- Correspondence: (P.R.); (M.V.)
| | - Gianluca Campo
- Translational Research Center, Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; (F.F.); (G.C.); (F.V.D.S.)
- Department of Medical Sciences, Cardiovascular Institute, Azienda Ospedaliero-Universitaria of Ferrara, University of Ferrara, 44124 Cona, Italy
| | - Mauro Vaccarezza
- Department of Morphology, Surgery and Experimental Medicine and Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, Via Fossato di Mortara 64/B, 44121 Ferrara, Italy;
- School of Pharmacy and Biomedical Sciences, Faculty of Health Sciences, Curtin University, Curtin Perth Campus, Bentley, Perth, WA 6102, Australia
- Correspondence: (P.R.); (M.V.)
| | - Francesco Vieceli Dalla Sega
- Translational Research Center, Maria Cecilia Hospital, GVM Care & Research, Cotignola, 48033 Ravenna, Italy; (F.F.); (G.C.); (F.V.D.S.)
| |
Collapse
|
5
|
Calvillo-Argüelles O, Jaiswal S, Shlush LI, Moslehi JJ, Schimmer A, Barac A, Thavendiranathan P. Connections Between Clonal Hematopoiesis, Cardiovascular Disease, and Cancer: A Review. JAMA Cardiol 2020; 4:380-387. [PMID: 30865214 DOI: 10.1001/jamacardio.2019.0302] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Importance Clonal hematopoiesis (CH) has been recently described as a novel driver for cancer and cardiovascular disease (CVD). Clonal hematopoiesis is a common, age-associated disorder marked by expansion of hematopoietic clones carrying recurrent somatic mutations. Current literature suggests that patients with CH have a higher risk of subsequent hematological malignant conditions and mortality attributable to excess CVD. This review discusses the association of cancer with CVD with CH as a potential unifying factor. Observations The prevalence of CH varies based on the sequencing depth, diagnostic criteria, and patient age and ranges from less than 1% in those younger than 40 years to more than 15% to 20% in those 90 years and older. Clonal hematopoiesis is associated with a 0.5% to 1.0% absolute annual risk of hematological malignant condition and a 2-fold to 4-fold higher risk of coronary artery disease, stroke, and CVD deaths, independent of traditional cardiovascular risk factors. In fact, CH appears to have a relative risk similar to that of traditional cardiovascular risk factors for CVD. Experimental studies suggest that the link between CVD and CH is causal, with inflammation as 1 potential mechanism. There may be also a link between CH and CVD in survivors of cancer; however, data to support this association are currently limited. Conclusions and Relevance Clonal hematopoiesis represents a premalignant state, with carriers having an increased risk of hematological malignant conditions. Although most carriers will not develop a malignant condition, CH confers an increased risk of CVD, possibly via inflammation. Clonal hematopoiesis may also contribute to CVD in survivors of cancer, although this hypothesis requires validation. Clinically, as advanced sequencing techniques become available, CH may pave the way for precision medicine in the field of cardio-oncology.
Collapse
Affiliation(s)
- Oscar Calvillo-Argüelles
- Ted Rogers Program in Cardiotoxicity Prevention, Toronto General Hospital, Toronto, Ontario, Canada.,Instituto Nacional de Cancerología, Mexico City, Mexico
| | - Siddhartha Jaiswal
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| | - Liran I Shlush
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Javid J Moslehi
- Division of Cardiovascular Medicine, Cardio-oncology Program, Vanderbilt University Medical Center, Nashville, Tennessee.,Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Aaron Schimmer
- Princess Margaret Cancer Center, Toronto, Ontario, Canada
| | - Ana Barac
- MedStar Heart and Vascular Institute, Georgetown University, Washington, DC
| | | |
Collapse
|
6
|
Mutations associated with age-related clonal hematopoiesis in PMF patients with rapid progression to myelofibrosis. Leukemia 2019; 34:1364-1372. [PMID: 31776465 DOI: 10.1038/s41375-019-0668-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 10/29/2019] [Accepted: 11/17/2019] [Indexed: 01/07/2023]
Abstract
Besides histopathological findings there are no indicators of increased risk for fibrotic progression in myeloproliferative neoplasms (MPN). Age-related clonal hematopoiesis (ARCH/CHIP) is a frequent finding in the elderly and combinations with MPN driver mutations (JAK2, MPL, and CALR) have been described. To determine the impact of ARCH/CHIP-related mutations for development of fibrosis in primary myelofibrosis (PMF), the mutational status of cases with fibrotic progression from grade 0 to grade 2/3 (n = 77) as evidenced by follow-up bone marrow biopsies (median 6.2 years) was compared with prefibrotic PMF samples without development of fibrosis (n = 27; median follow-up 7.3 years). Frequent ARCH/CHIP-associated mutations (TET2, ASXL1, and DNMT3A) demonstrable at presentation were not connected with fibrotic progression. However, mutations which are rarely found in ARCH/CHIP (SRSF2, U2AF1, SF3B1, IDH1/2, and EZH2) were present in 24.7% of cases with later development of fibrosis and not detectable in cases staying free from fibrosis (P = 0.0028). Determination of the tumor mutational burden (TMB) in a subgroup of cases (n = 32) did not show significant differences (7.68 mutations/MB vs. 6.85 mutations/MB). We conclude that mutations rarely found in ARCH/CHIP provide an independent risk factor for rapid fibrotic progression (median 2.0 years) when manifest already at first presentation.
Collapse
|
7
|
Mazzarella L, Botteri E, Matthews A, Gatti E, Di Salvatore D, Bagnardi V, Breccia M, Montesinos P, Bernal T, Gil C, Ley TJ, Sanz M, Bhaskaran K, Coco FL, Pelicci PG. Obesity is a risk factor for acute promyelocytic leukemia: evidence from population and cross-sectional studies and correlation with FLT3 mutations and polyunsaturated fatty acid metabolism. Haematologica 2019; 105:1559-1566. [PMID: 31515354 PMCID: PMC7271575 DOI: 10.3324/haematol.2019.223925] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/11/2019] [Indexed: 12/12/2022] Open
Abstract
Obesity correlates with hematologic malignancies including leukemias, but risk of specific leukemia subtypes like acute promyelocytic leukemia and underlying molecular mechanisms are poorly understood. We explored multiple datasets for correlation between leukemia, body mass index (BMI) and molecular features. In a population-based study (n=5.2 million), we correlated BMI with promyelocytic leukemia, and other acute myeloid, lymphoid or other leukemias. In cross-sectional studies, we tested BMI deviation in promyelocytic leukemia trial cohorts from that expected based on national surveys. We explored The Cancer Genome Atlas for transcriptional signatures and mutations enriched in promyelocytic leukemia and/or obesity, and confirmed a correlation between body mass and FLT3 mutations in promyelocytic leukemia cohorts by logistic regression. In the population-based study, hazard ratio per 5 kg/m2 increase was: promyelocytic leukemia 1.44 (95%CI: 1.0-2.08), non-promyelocytic acute myeloid leukemias 1.17 (95%CI: 1.10-1.26), lymphoid leukemias 1.04 (95%CI: 1.0-1.09), other 1.10 (95%CI: 1.04-1.15). In cross-sectional studies, body mass deviated significantly from that expected (Italy: P<0.001; Spain: P=0.011; USA: P<0.001). Promyelocytic leukemia showed upregulation of polyunsaturated fatty acid metabolism genes. Odds of FLT3 mutations were higher in obese acute myeloid leukemias (odds ratio=2.4, P=0.007), whether promyelocytic or not, a correlation confirmed in the pooled promyelocytic leukemia cohorts (OR=1.22, 1.05-1.43 per 5 kg/m2). These results strengthen the evidence for obesity as a bona fide risk factor for myeloid leukemias, and in particular APL. FLT3 mutations and polyunsaturated fatty acid metabolism may play a previously under-appreciated role in obesity-associated leukemogenesis.
Collapse
Affiliation(s)
- Luca Mazzarella
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy .,Division of Early Drug Development for Innovative Therapies, European Institute of Oncology IRCCS, Milan, Italy
| | | | | | - Elena Gatti
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy
| | | | | | - Massimo Breccia
- Division of Hematology, Department of Cellular Biotechnologies and Hematology, Sapienza University, Rome, Italy
| | - Pau Montesinos
- Hematology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | | | - Timothy J Ley
- Department of Medicine, Division of Oncology, Washington University in St. Louis, Saint Louis, MO, USA
| | - Miguel Sanz
- Hematology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
| | | | | | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, European Institute of Oncology IRCCS, Milan, Italy .,Department of Oncology and Hemato-Oncology, University of Milan
| |
Collapse
|
8
|
Sano S, Oshima K, Wang Y, MacLauchlan S, Katanasaka Y, Sano M, Zuriaga MA, Yoshiyama M, Goukassian D, Cooper MA, Fuster JJ, Walsh K. Tet2-Mediated Clonal Hematopoiesis Accelerates Heart Failure Through a Mechanism Involving the IL-1β/NLRP3 Inflammasome. J Am Coll Cardiol 2019; 71:875-886. [PMID: 29471939 DOI: 10.1016/j.jacc.2017.12.037] [Citation(s) in RCA: 482] [Impact Index Per Article: 80.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Revised: 12/08/2017] [Accepted: 12/12/2017] [Indexed: 12/17/2022]
Abstract
BACKGROUND Recent studies have shown that hematopoietic stem cells can undergo clonal expansion secondary to somatic mutations in leukemia-related genes, thus leading to an age-dependent accumulation of mutant leukocytes in the blood. This somatic mutation-related clonal hematopoiesis is common in healthy older individuals, but it has been associated with an increased incidence of future cardiovascular disease. The epigenetic regulator TET2 is frequently mutated in blood cells of individuals exhibiting clonal hematopoiesis. OBJECTIVES This study investigated whether Tet2 mutations within hematopoietic cells can contribute to heart failure in 2 models of cardiac injury. METHODS Heart failure was induced in mice by pressure overload, achieved by transverse aortic constriction or chronic ischemia induced by the permanent ligation of the left anterior descending artery. Competitive bone marrow transplantation strategies with Tet2-deficient cells were used to mimic TET2 mutation-driven clonal hematopoiesis. Alternatively, Tet2 was specifically ablated in myeloid cells using Cre recombinase expressed from the LysM promoter. RESULTS In both experimental heart failure models, hematopoietic or myeloid Tet2 deficiency worsened cardiac remodeling and function, in parallel with increased interleukin-1beta (IL-1β) expression. Treatment with a selective NLRP3 inflammasome inhibitor protected against the development of heart failure and eliminated the differences in cardiac parameters between Tet2-deficient and wild-type mice. CONCLUSIONS Tet2 deficiency in hematopoietic cells is associated with greater cardiac dysfunction in murine models of heart failure as a result of elevated IL-1β signaling. These data suggest that individuals with TET2-mediated clonal hematopoiesis may be at greater risk of developing heart failure and respond better to IL-1β-NLRP3 inflammasome inhibition.
Collapse
Affiliation(s)
- Soichi Sano
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts; Department of Cardiovascular Medicine, Osaka City University of Medicine, Osaka, Japan
| | - Kosei Oshima
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Ying Wang
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Susan MacLauchlan
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Yasufumi Katanasaka
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts; Division of Molecular Medicine, Graduate School of Pharmaceutical Sciences, University of Shizuoka, Yada, Japan
| | - Miho Sano
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - María A Zuriaga
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Minoru Yoshiyama
- Department of Cardiovascular Medicine, Osaka City University of Medicine, Osaka, Japan
| | - David Goukassian
- Center for Transnational Medicine, Temple University School of Medicine, Philadelphia, Pennsylvania
| | - Matthew A Cooper
- Institute for Molecular Bioscience, University of Queensland, Brisbane, Queensland, Australia
| | - José J Fuster
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts
| | - Kenneth Walsh
- Molecular Cardiology/Whitaker Cardiovascular Institute, Boston University School of Medicine, Boston, Massachusetts.
| |
Collapse
|
9
|
Rožman P. How Could We Slow or Reverse the Human Aging Process and Extend the Healthy Life Span with Heterochronous Autologous Hematopoietic Stem Cell Transplantation. Rejuvenation Res 2019; 23:159-170. [PMID: 31203790 DOI: 10.1089/rej.2018.2164] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The senescence of the immune system contributes considerably to the age-related diseases that are the main causes of death after the age of 65. In this study, we present an appealing option for the prevention of immune senescence and slowing or reversing the aging process, which can be achieved by heterochronous autologous hematopoietic stem cell transplantation (haHSCT), where healthy autologous bone marrow stem cells are collected from donors while young, cryopreserved and stored for a long period, and reinfused at a later time when indicated. After reinfusion and homing, these young HSCs could participate in normal hemato- and immunopoiesis and improve several immune functions by expanding the immune- as well as hematopoietic cell repertoire. Several animal studies have already confirmed the feasibility of this procedure, which extended the longevity of the treated animals. If translated to human medicine, haHSCT could prevent or mitigate age-related immune defects and extend the healthy life span. In this review, we describe the concept of haHSCT, recent studies that confirm its feasibility, and discuss the further research needed to translate this heterochronous methodology.
Collapse
Affiliation(s)
- Primož Rožman
- Immunohaematology Department, Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia
| |
Collapse
|
10
|
Clonal Hematopoiesis with Oncogenic Potential (CHOP): Separation from CHIP and Roads to AML. Int J Mol Sci 2019; 20:ijms20030789. [PMID: 30759825 PMCID: PMC6387423 DOI: 10.3390/ijms20030789] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2019] [Revised: 02/10/2019] [Accepted: 02/11/2019] [Indexed: 12/21/2022] Open
Abstract
The development of leukemia is a step-wise process that is associated with molecular diversification and clonal selection of neoplastic stem cells. Depending on the number and combinations of lesions, one or more sub-clones expand/s after a variable latency period. Initial stages may develop early in life or later in adulthood and include premalignant (indolent) stages and the malignant phase, defined by an acute leukemia. We recently proposed a cancer model in which the earliest somatic lesions are often age-related early mutations detectable in apparently healthy individuals and where additional oncogenic mutations will lead to the development of an overt neoplasm that is usually a preleukemic condition such as a myelodysplastic syndrome. These neoplasms may or may not transform to overt acute leukemia over time. Thus, depending on the type and number of somatic mutations, clonal hematopoiesis (CH) can be divided into CH with indeterminate potential (CHIP) and CH with oncogenic potential (CHOP). Whereas CHIP mutations per se usually create the molecular background of a neoplastic process, CHOP mutations are disease-related or even disease-specific lesions that trigger differentiation and/or proliferation of neoplastic cells. Over time, the acquisition of additional oncogenic events converts preleukemic neoplasms into secondary acute myeloid leukemia (sAML). In the present article, recent developments in the field are discussed with a focus on CHOP mutations that lead to distinct myeloid neoplasms, their role in disease evolution, and the impact of additional lesions that can drive a preleukemic neoplasm into sAML.
Collapse
|
11
|
Konieczny J, Arranz L. Updates on Old and Weary Haematopoiesis. Int J Mol Sci 2018; 19:ijms19092567. [PMID: 30158459 PMCID: PMC6163425 DOI: 10.3390/ijms19092567] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 08/20/2018] [Accepted: 08/26/2018] [Indexed: 12/13/2022] Open
Abstract
Blood formation, or haematopoiesis, originates from haematopoietic stem cells (HSCs), whose functions and maintenance are regulated in both cell- and cell non-autonomous ways. The surroundings of HSCs in the bone marrow create a specific niche or microenvironment where HSCs nest that allows them to retain their unique characteristics and respond rapidly to external stimuli. Ageing is accompanied by reduced regenerative capacity of the organism affecting all systems, due to the progressive decline of stem cell functions. This includes blood and HSCs, which contributes to age-related haematological disorders, anaemia, and immunosenescence, among others. Furthermore, chronological ageing is characterised by myeloid and platelet HSC skewing, inflammageing, and expanded clonal haematopoiesis, which may be the result of the accumulation of preleukaemic lesions in HSCs. Intriguingly, haematological malignancies such as acute myeloid leukaemia have a high incidence among elderly patients, yet not all individuals with clonal haematopoiesis develop leukaemias. Here, we discuss recent work on these aspects, their potential underlying molecular mechanisms, and the first cues linking age-related changes in the HSC niche to poor HSC maintenance. Future work is needed for a better understanding of haematopoiesis during ageing. This field may open new avenues for HSC rejuvenation and therapeutic strategies in the elderly.
Collapse
Affiliation(s)
- Joanna Konieczny
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9019 Tromsø, Norway.
| | - Lorena Arranz
- Stem Cell Aging and Cancer Research Group, Department of Medical Biology, Faculty of Health Sciences, UiT, The Arctic University of Norway, 9019 Tromsø, Norway.
- Department of Hematology, University Hospital of North Norway, 9019 Tromsø, Norway.
- Young Associate Investigator, Norwegian Center for Molecular Medicine (NCMM), 0349 Oslo, Norway.
| |
Collapse
|
12
|
Darwiche W, Gubler B, Marolleau JP, Ghamlouch H. Chronic Lymphocytic Leukemia B-Cell Normal Cellular Counterpart: Clues From a Functional Perspective. Front Immunol 2018; 9:683. [PMID: 29670635 PMCID: PMC5893869 DOI: 10.3389/fimmu.2018.00683] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 03/20/2018] [Indexed: 12/20/2022] Open
Abstract
Chronic lymphocytic leukemia (CLL) is characterized by the clonal expansion of small mature-looking CD19+ CD23+ CD5+ B-cells that accumulate in the blood, bone marrow, and lymphoid organs. To date, no consensus has been reached concerning the normal cellular counterpart of CLL B-cells and several B-cell types have been proposed. CLL B-cells have remarkable phenotypic and gene expression profile homogeneity. In recent years, the molecular and cellular biology of CLL has been enriched by seminal insights that are leading to a better understanding of the natural history of the disease. Immunophenotypic and molecular approaches (including immunoglobulin heavy-chain variable gene mutational status, transcriptional and epigenetic profiling) comparing the normal B-cell subset and CLL B-cells provide some new insights into the normal cellular counterpart. Functional characteristics (including activation requirements and propensity for plasma cell differentiation) of CLL B-cells have now been investigated for 50 years. B-cell subsets differ substantially in terms of their functional features. Analysis of shared functional characteristics may reveal similarities between normal B-cell subsets and CLL B-cells, allowing speculative assignment of a normal cellular counterpart for CLL B-cells. In this review, we summarize current data regarding peripheral B-cell differentiation and human B-cell subsets and suggest possibilities for a normal cellular counterpart based on the functional characteristics of CLL B-cells. However, a definitive normal cellular counterpart cannot be attributed on the basis of the available data. We discuss the functional characteristics required for a cell to be logically considered to be the normal counterpart of CLL B-cells.
Collapse
Affiliation(s)
- Walaa Darwiche
- EA 4666 Lymphocyte Normal - Pathologique et Cancers, HEMATIM, Université de Picardie Jules Verne, Amiens, France.,Laboratoire d'Hématologie, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
| | - Brigitte Gubler
- EA 4666 Lymphocyte Normal - Pathologique et Cancers, HEMATIM, Université de Picardie Jules Verne, Amiens, France.,Laboratoire d'Oncobiologie Moléculaire, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
| | - Jean-Pierre Marolleau
- EA 4666 Lymphocyte Normal - Pathologique et Cancers, HEMATIM, Université de Picardie Jules Verne, Amiens, France.,Service d'Hématologie Clinique et Thérapie cellulaire, Centre Hospitalier Universitaire Amiens-Picardie, Amiens, France
| | - Hussein Ghamlouch
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1170, Gustave Roussy, Villejuif, France.,Institut Gustave Roussy, Villejuif, France.,Université Paris-Sud, Faculté de Médecine, Le Kremlin-Bicêtre, France
| |
Collapse
|
13
|
Abegunde SO, Buckstein R, Wells RA, Rauh MJ. An inflammatory environment containing TNFα favors Tet2-mutant clonal hematopoiesis. Exp Hematol 2017; 59:60-65. [PMID: 29195897 DOI: 10.1016/j.exphem.2017.11.002] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 11/15/2017] [Accepted: 11/21/2017] [Indexed: 01/16/2023]
Abstract
Clonal hematopoiesis of aging and indeterminate potential (ARCH or CHIP), driven mainly by mutations in DNMT3A and TET2, is an emerging public health issue, affecting at least 10-15% of adults older than 65 years. CHIP is associated with increased risks of de novo and therapy-related hematological neoplasms and serves as a reservoir for leukemic relapse. CHIP is also associated with increased all-cause mortality and risk of cardio-metabolic disease. The latter association may be explained, at least in part, by the effects of inactivating mutations in TET2 on progeny macrophages. We and others have shown recently that TET2-deficient macrophages are hyperinflammatory and this may exacerbate processes such as atherosclerosis. We postulated an inflammatory state associated with TET2 inactivation and/or unhealthy aging may also favor TET2-mutant hematopoietic stem and progenitor cell (HSPC) expansion. Herein, we demonstrate a clonogenic advantage for Tet2-knockout murine and TET2-mutant human HSPCs in an in vitro environment that contains the proinflammatory cytokine tumor necrosis factor-alpha (TNFα). This phenotype emerges on chronic TNFα exposure and is associated with myeloid skewing and resistance to apoptosis. To our knowledge, this is the first evidence to suggest that TET2 mutations promote clonal dominance with aging by conferring TNFα resistance to sensitive bone marrow progenitors while also propagating such an inflammatory environment. Normalizing the immune environment may present a novel strategy to control or eradicate mutant CHIP clones.
Collapse
Affiliation(s)
- Samuel O Abegunde
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada
| | - Rena Buckstein
- Division of Hematology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Richard A Wells
- Division of Hematology, Odette Cancer Centre, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Michael J Rauh
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, Ontario, Canada.
| |
Collapse
|
14
|
Corces MR, Chang HY, Majeti R. Preleukemic Hematopoietic Stem Cells in Human Acute Myeloid Leukemia. Front Oncol 2017; 7:263. [PMID: 29164062 PMCID: PMC5681525 DOI: 10.3389/fonc.2017.00263] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 10/19/2017] [Indexed: 12/16/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive malignancy of the bone marrow characterized by an uncontrolled proliferation of undifferentiated myeloid lineage cells. Decades of research have demonstrated that AML evolves from the sequential acquisition of genetic alterations within a single lineage of hematopoietic cells. More recently, the advent of high-throughput sequencing has enabled the identification of a premalignant phase of AML termed preleukemia. Multiple studies have demonstrated that AML can arise from the accumulation of mutations within hematopoietic stem cells (HSCs). These HSCs have been termed "preleukemic HSCs" as they represent the evolutionary ancestors of the leukemia. Through examination of the biological and clinical characteristics of these preleukemic HSCs, this review aims to shed light on some of the unexplored questions in the field. We note that some of the material discussed is speculative in nature and is presented in order to motivate future work.
Collapse
Affiliation(s)
- M. Ryan Corces
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, United States
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, United States
| | - Howard Y. Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, United States
- Program in Epithelial Biology, Stanford University School of Medicine, Stanford, CA, United States
| | - Ravindra Majeti
- Program in Cancer Biology, Cancer Institute, Institute for Stem Cell Biology and Regenerative Medicine, Ludwig Center, Stanford University School of Medicine, Stanford, CA, United States
| |
Collapse
|
15
|
Ultra-sensitive Sequencing Identifies High Prevalence of Clonal Hematopoiesis-Associated Mutations throughout Adult Life. Am J Hum Genet 2017; 101:50-64. [PMID: 28669404 DOI: 10.1016/j.ajhg.2017.05.013] [Citation(s) in RCA: 184] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2017] [Accepted: 05/18/2017] [Indexed: 12/13/2022] Open
Abstract
Clonal hematopoiesis results from somatic mutations in hematopoietic stem cells, which give an advantage to mutant cells, driving their clonal expansion and potentially leading to leukemia. The acquisition of clonal hematopoiesis-driver mutations (CHDMs) occurs with normal aging and these mutations have been detected in more than 10% of individuals ≥65 years. We aimed to examine the prevalence and characteristics of CHDMs throughout adult life. We developed a targeted re-sequencing assay combining high-throughput with ultra-high sensitivity based on single-molecule molecular inversion probes (smMIPs). Using smMIPs, we screened more than 100 loci for CHDMs in more than 2,000 blood DNA samples from population controls between 20 and 69 years of age. Loci screened included 40 regions known to drive clonal hematopoiesis when mutated and 64 novel candidate loci. We identified 224 somatic mutations throughout our cohort, of which 216 were coding mutations in known driver genes (DNMT3A, JAK2, GNAS, TET2, and ASXL1), including 196 point mutations and 20 indels. Our assay's improved sensitivity allowed us to detect mutations with variant allele frequencies as low as 0.001. CHDMs were identified in more than 20% of individuals 60 to 69 years of age and in 3% of individuals 20 to 29 years of age, approximately double the previously reported prevalence despite screening a limited set of loci. Our findings support the occurrence of clonal hematopoiesis-associated mutations as a widespread mechanism linked with aging, suggesting that mosaicism as a result of clonal evolution of cells harboring somatic mutations is a universal mechanism occurring at all ages in healthy humans.
Collapse
|
16
|
Inversion 3 Cytogenetic Abnormality in an Allogeneic Hematopoietic Cell Transplant Recipient Representative of a Donor-Derived Constitutional Abnormality. Biol Blood Marrow Transplant 2017; 23:1582-1587. [PMID: 28549770 DOI: 10.1016/j.bbmt.2017.05.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 05/12/2017] [Indexed: 11/23/2022]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) is an important treatment for many severe hematologic disorders; however, HCT can be associated with significant complications, including organ toxicity, graft-versus-host disease, and relapse. Another serious, but rare, complication is the transmission of hematologic and nonhematologic diseases from the donor to the recipient. With older donors, the risk of an abnormality may be increased. Here we describe the transmission of an inversion 3 constitutional cytogenetic abnormality from an unrelated donor to a recipient, and review the clinical implications of the discovery of donor-derived constitutional cytogenetic abnormalities.
Collapse
|
17
|
Alexeev I. Lorenz system in the thermodynamic modelling of leukaemia malignancy. Med Hypotheses 2017; 102:150-155. [PMID: 28478821 DOI: 10.1016/j.mehy.2017.03.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2017] [Revised: 03/19/2017] [Accepted: 03/25/2017] [Indexed: 11/24/2022]
Abstract
The core idea of the proposed thermodynamic modelling of malignancy in leukaemia is entropy arising within normal haematopoiesis. Mathematically its description is supposed to be similar to the Lorenz system of ordinary differential equations for simplified processes of heat flow in fluids. The hypothetical model provides a description of remission and relapse in leukaemia as two hierarchical and qualitatively different states of normal haematopoiesis with their own phase spaces. Phase space transition is possible through pitchfork bifurcation, which is considered the common symmetrical scenario for relapse, induced remission and the spontaneous remission of leukaemia. Cytopenia is regarded as an adaptive reaction of haematopoiesis to an increase in entropy caused by leukaemia clones. The following predictions are formulated: a) the percentage of leukaemia cells in marrow as a criterion of remission or relapse is not necessarily constant but is a variable value; b) the probability of remission depends upon normal haematopoiesis reaching bifurcation; c) the duration of remission depends upon the eradication of leukaemia cells through induction or consolidation therapies; d) excessively high doses of chemotherapy in consolidation may induce relapse.
Collapse
|
18
|
Pan Y, Meng M, Zheng N, Cao Z, Yang P, Xi X, Zhou Q. Targeting of multiple senescence-promoting genes and signaling pathways by triptonide induces complete senescence of acute myeloid leukemia cells. Biochem Pharmacol 2017; 126:34-50. [PMID: 27908660 DOI: 10.1016/j.bcp.2016.11.024] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 11/26/2016] [Indexed: 01/08/2023]
Abstract
Leukemia cells aberrantly overexpress senescence-suppression telomerase reverse transcriptase (TERT) and down-regulate key senescence-promoting genes to escape complete senescence, resulting in immortalization and malignant progression. Accordingly, induction of complete senescence is a sensible strategy for anti-leukemia therapy. However, effective senescence-based anti-leukemia drugs with low toxicity are currently lacking. In this study, we found that triptonide (chemical name diterpene triepoxide), a small molecule derived from the herb Tripterygium wilfordii Hook, strongly induced complete senescence in cultured acute myeloid leukemia (AML) cell lines, and potently inhibited growth and colony formation of U937 and HL-60 AML cell line with IC50 values of 7.5 and 12nM, respectively. Strikingly, triptonide (4mg/kg) nearly completely suppressed human leukemia cell tumorigenicity (>99%) without obvious toxicity in a mouse xenograft model. Mechanistic studies showed that triptonide induced senescence followed by apoptosis mainly by suppressing transcription of TERT and oncogenic c-Myc, while concomitantly promoting transcription of senescence-promoting genes p16 and p21 and the pro-apoptotic gene encoding DNA damage-inducible transcript 3. These effects of triptonide are mediated by selective mitogen-activated protein kinase kinase-3/p38 signaling pathway activation. Our study provides a conceptual framework for inducing complete senescence as an effective anti-leukemia therapeutic strategy through a "multiple-hits" model and supports further development of triptonide as an anti-cancer agent.
Collapse
Affiliation(s)
- Yanyan Pan
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Mei Meng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Nana Zheng
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Zhifei Cao
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Ping Yang
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China
| | - Xiaodong Xi
- State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Quansheng Zhou
- Cyrus Tang Hematology Center, Jiangsu Institute of Hematology, 2011 Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
19
|
Nguyen TB, Sakata-Yanagimoto M, Asabe Y, Matsubara D, Kano J, Yoshida K, Shiraishi Y, Chiba K, Tanaka H, Miyano S, Izutsu K, Nakamura N, Takeuchi K, Miyoshi H, Ohshima K, Minowa T, Ogawa S, Noguchi M, Chiba S. Identification of cell-type-specific mutations in nodal T-cell lymphomas. Blood Cancer J 2017; 7:e516. [PMID: 28157189 PMCID: PMC5301031 DOI: 10.1038/bcj.2016.122] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/07/2016] [Indexed: 12/26/2022] Open
Abstract
Recent genetic analysis has identified frequent mutations in ten-eleven translocation 2 (TET2), DNA methyltransferase 3A (DNMT3A), isocitrate dehydrogenase 2 (IDH2) and ras homolog family member A (RHOA) in nodal T-cell lymphomas, including angioimmunoblastic T-cell lymphoma and peripheral T-cell lymphoma, not otherwise specified. We examined the distribution of mutations in these subtypes of mature T-/natural killer cell neoplasms to determine their clonal architecture. Targeted sequencing was performed for 71 genes in tumor-derived DNA of 87 cases. The mutations were then analyzed in a programmed death-1 (PD1)-positive population enriched with tumor cells and CD20-positive B cells purified by laser microdissection from 19 cases. TET2 and DNMT3A mutations were identified in both the PD1+ cells and the CD20+ cells in 15/16 and 4/7 cases, respectively. All the RHOA and IDH2 mutations were confined to the PD1+ cells, indicating that some, including RHOA and IDH2 mutations, being specific events in tumor cells. Notably, we found that all NOTCH1 mutations were detected only in the CD20+ cells. In conclusion, we identified both B- as well as T-cell-specific mutations, and mutations common to both T and B cells. These findings indicate the expansion of a clone after multistep and multilineal acquisition of gene mutations.
Collapse
Affiliation(s)
- T B Nguyen
- Department of Hematology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Hematology, Faculty of Medicine, University of Medicine and Pharmacy, Ho Chi Minh City, Vietnam.,Stem Cell Transplantation Zone, Blood Transfusion Hematology Hospital, Ho Chi Minh City, Vietnam
| | - M Sakata-Yanagimoto
- Department of Hematology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Hematology, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
| | - Y Asabe
- Department of Hematology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - D Matsubara
- Department of Integrative Pathology, Jichii Medical University, Shimotsuke, Tochigi, Japan
| | - J Kano
- Department of Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - K Yoshida
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Y Shiraishi
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - K Chiba
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - H Tanaka
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - S Miyano
- Human Genome Center, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - K Izutsu
- Department of Hematology, Toranomon Hospital, Tokyo, Japan.,Okinaka Memorial Institute for Medical Research, Tokyo, Japan
| | - N Nakamura
- Department of Pathology, Tokai University School of Medicine, Isehara, Kanagawa, Japan
| | - K Takeuchi
- Pathology Project for Molecular Targets, The Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - H Miyoshi
- Department of Pathology, Kurume University, Kurume, Fukuoka, Japan
| | - K Ohshima
- Department of Pathology, Kurume University, Kurume, Fukuoka, Japan
| | - T Minowa
- Nanotechnology Innovation Station, National Institute for Materials Science, Tsukuba, Ibaraki, Japan
| | - S Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - M Noguchi
- Department of Pathology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - S Chiba
- Department of Hematology, Graduate School of Comprehensive Human Sciences, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Hematology, Faculty of Medicine, University of Tsukuba, Tsukuba, Ibaraki, Japan.,Department of Hematology, University of Tsukuba Hospital, Tsukuba, Ibaraki, Japan
| |
Collapse
|
20
|
Iron-induced epigenetic abnormalities of mouse bone marrow through aberrant activation of aconitase and isocitrate dehydrogenase. Int J Hematol 2016; 104:491-501. [DOI: 10.1007/s12185-016-2054-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2016] [Revised: 06/24/2016] [Accepted: 06/24/2016] [Indexed: 12/19/2022]
|
21
|
Marsh JCW, Mufti GJ. Clinical significance of acquired somatic mutations in aplastic anaemia. Int J Hematol 2016; 104:159-67. [DOI: 10.1007/s12185-016-1972-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 02/19/2016] [Accepted: 03/02/2016] [Indexed: 11/29/2022]
|