1
|
Cui X, Dong Y, Zhan Q, Huang Y, Zhu Q, Zhang Z, Yang G, Wang L, Shen S, Zhao J, Lin Z, Sun J, Su Z, Xiao Y, Zhang C, Liang Y, Shen L, Ji L, Zhang X, Yin J, Wang H, Chen Z, Ju Z, Jiang C, Le R, Gao S. Altered 3D genome reorganization mediates precocious myeloid differentiation of aged hematopoietic stem cells in inflammation. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1209-1225. [PMID: 39754007 DOI: 10.1007/s11427-024-2754-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/09/2024] [Indexed: 05/23/2025]
Abstract
Inflammation is a driving force of hematopoietic stem cells (HSCs) aging, causing irreversible exhaustion of functional HSCs. However, the underlying mechanism of HSCs erosion by inflammatory insult remains poorly understood. Here, we find that transient LPS exposure primes aged HSCs to undergo accelerated differentiation at the expense of self-renewal, leading to depletion of HSCs. Meanwhile, the central regulator nuclear factor kappa B (NF-κB) mediating functional impairment by inflammation insult induces differential transcriptional response in aged HSCs compared with young HSCs, with precocious activation of myeloid lineage genes. Altered compartmentalization and chromatin loop formation are associated with aging-related differential transcriptional response in HSCs upon lipopolysaccharide (LPS) stimulation. Mechanistically, enhancer and promoter regions of myeloid lineage genes in aged HSCs are more accessible and display more rapid and prominent CTCF occupancy upon LPS stimulation. Our study provides comprehensive resources for the three-dimensional (3D) genome structure of HSCs and sheds light into the ordered genome organization and the associated transcriptome signature underlying HSCs aging.
Collapse
Affiliation(s)
- Xinyu Cui
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yu Dong
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiang Zhan
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yixin Huang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qianshu Zhu
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zihao Zhang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Guang Yang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Liping Wang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shijun Shen
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jia Zhao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhiyi Lin
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jiatong Sun
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhongqu Su
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China
| | - Yihan Xiao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Chuyu Zhang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yuwei Liang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Lu Shen
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Lichen Ji
- Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Xuguang Zhang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiqing Yin
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Hong Wang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhiyang Chen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Cizhong Jiang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Rongrong Le
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
2
|
Kapadia CD, Williams N, Dawson KJ, Watson C, Yousefzadeh MJ, Le D, Nyamondo K, Kodavali S, Cagan A, Waldvogel S, Zhang X, De La Fuente J, Leongamornlert D, Mitchell E, Florez MA, Sosnowski K, Aguilar R, Martell A, Guzman A, Harrison D, Niedernhofer LJ, King KY, Campbell PJ, Blundell J, Goodell MA, Nangalia J. Clonal dynamics and somatic evolution of haematopoiesis in mouse. Nature 2025; 641:681-689. [PMID: 40044850 PMCID: PMC12074984 DOI: 10.1038/s41586-025-08625-8] [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: 11/02/2023] [Accepted: 01/10/2025] [Indexed: 03/12/2025]
Abstract
Haematopoietic stem cells maintain blood production throughout life1. Although extensively characterized using the laboratory mouse, little is known about clonal selection and population dynamics of the haematopoietic stem cell pool during murine ageing. We isolated stem cells and progenitors from young and old mice, identifying 221,890 somatic mutations genome-wide in 1,845 single-cell-derived colonies. Mouse stem cells and progenitors accrue approximately 45 somatic mutations per year, a rate only approximately threefold greater than human progenitors despite the vastly different organismal sizes and lifespans. Phylogenetic patterns show that stem and multipotent progenitor cell pools are established during embryogenesis, after which they independently self-renew in parallel over life, evenly contributing to differentiated progenitors and peripheral blood. The stem cell pool grows steadily over the mouse lifespan to about 70,000 cells, self-renewing about every 6 weeks. Aged mice did not display the profound loss of clonal diversity characteristic of human haematopoietic ageing. However, targeted sequencing showed small, expanded clones in the context of murine ageing, which were larger and more numerous following haematological perturbations, exhibiting a selection landscape similar to humans. Our data illustrate both conserved features of population dynamics of blood and distinct patterns of age-associated somatic evolution in the short-lived mouse.
Collapse
Affiliation(s)
- Chiraag D Kapadia
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | | | - Kevin J Dawson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Caroline Watson
- Early Cancer Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Matthew J Yousefzadeh
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Columbia Center for Translational Immunology, Columbia Center for Human Longevity, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Duy Le
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Kudzai Nyamondo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Sreeya Kodavali
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Alex Cagan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Departments of Genetics, Pathology & Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Sarah Waldvogel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Xiaoyan Zhang
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Josephine De La Fuente
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | | | - Emily Mitchell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Marcus A Florez
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Krzysztof Sosnowski
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Rogelio Aguilar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Alejandra Martell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Anna Guzman
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | | | - Laura J Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Katherine Y King
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Peter J Campbell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Jamie Blundell
- Early Cancer Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Margaret A Goodell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
| | - Jyoti Nangalia
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK.
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK.
- Department of Haematology, University of Cambridge, Cambridge, UK.
| |
Collapse
|
3
|
Stomper J, Niroula A, Belizaire R, McConkey M, Bandaru TS, Ebert BL. Sex differences in DNMT3A-mutant clonal hematopoiesis and the effects of estrogen. Cell Rep 2025; 44:115494. [PMID: 40178977 DOI: 10.1016/j.celrep.2025.115494] [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: 06/27/2024] [Revised: 02/03/2025] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
Blood cancers are generally more common in males, and the prevalence of most mutations that drive clonal hematopoiesis and myeloid malignancies is higher in males. In contrast, hematopoietic DNMT3A mutations are more common in females. Among ∼450,000 participants in the UK Biobank, the prevalence of DNMT3A mutations and copy-number abnormalities is higher in females than males. In a murine model, Dnmt3a-mutant hematopoietic stem cells (HSCs) from unperturbed female mice had increased stemness gene expression compared to male and wild-type (WT) mice. Estrogen regulates HSCs, and we found that Dnmt3a mutations maintain stemness in the setting of estrogen-induced proliferative stress. Dnmt3a-mutant myeloid cells outcompeted WT cells under chronic estrogen treatment, an effect that was dependent on cell-intrinsic estrogen receptor alpha activity. Our studies indicate that estrogen might contribute to the female predominance of DNMT3A-mutant clonal hematopoiesis.
Collapse
Affiliation(s)
- Julia Stomper
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Abhishek Niroula
- Broad Institute, Cambridge, MA 02142, USA; Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, 40530 Gothenburg, Sweden; SciLifeLab, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Roger Belizaire
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Marie McConkey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Tagore Sanketh Bandaru
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, 40530 Gothenburg, Sweden
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Broad Institute, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Boston, MA 02215, USA.
| |
Collapse
|
4
|
Fu J, Deng T, Zheng T, Shi P, Zhu W, Tao M, Wen Z, Wu X. Development and Validation of a Predictive Nomogram for Myelosuppression Risk in Chronic Hepatitis B Patients Treated with Peginterferon. Infect Drug Resist 2025; 18:1793-1805. [PMID: 40225103 PMCID: PMC11994083 DOI: 10.2147/idr.s508538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
Purpose Peginterferon (Peg-IFN) is a common treatment for chronic hepatitis B (CHB); however, some patients developing myelosuppression as a side-effect. In this study, we identified risk factors associated with increased myelosuppression, and incorporated them into a predictive nomogram. Patients and Methods This study is designed as a case-control study. A total of 312 CHB patients treated with Peg-IFN from two medical centers were retrospectively enrolled between December 2019 and December 2022. Patients from the First Affiliated Hospital of Nanchang University were randomly divided into a training cohort (n=153) and a test cohort (n=55) at a 3:1 ratio. Patients from the Jiangxi Provincial People's Hospital composed the validation cohort (n= 104). In the training cohort, based on the blood routine results of patients 1 week after Peg-IFN treatment, patients were further divided into Normal (myelosuppression grades 0-I) and Myelosuppression (grades II-IV) groups. Then uni- and multivariate logistic regression analyses were carried out to identify myelosuppression risk factors, which were subsequently incorporated into a predictive nomogram. The capability of the predictive nomogram was validated using an area under the curve (AUC) of the receiver operating characteristic (ROC) curve. The Hosmer-Lemeshow test, calibration curves, and decision curve analysis (DCA) were used to evaluate the nomogram. Finally, the developed predictive nomogram was validated both internally and externally using separate test and validation cohorts. Results Body mass index (BMI; odds ratio [OR]=0.841, 95% confidence interval [CI] 0.738-0.959, P=0.010), white blood cell counts (WBC; OR=0.657, 95% CI 0.497-0.868, P=0.003), globulin (GLB; OR=0.796, 95% CI 0.713-0.889, P<0.001) and serum creatinine levels (SCR; OR=1.029, 95% CI 1.002-1.058, P=0.038) are independent risk factors for myelosuppression in Peg-IFN-treated CHB patients. A predictive nomogram was constructed by incorporating the above independent risk factors, and its performance was assessed across the training, test, and validation cohorts. The model demonstrated AUC values of 0.824 (95% CI 0.757-0.891), 0.812 (95% CI 0.701-0.923), and 0.870 (95% CI 0.802-0.940), respectively, highlighting its good predictive accuracy. As for Hosmer-Lemeshow, it was P=0.351, (χ2= 8.898) for training, P=0.514 (χ2=6.226) for the test, and P=0.442 (χ2=7.918) for the validation cohort. The results of the calibration curves and DCA demonstrated good concordance between predicted probabilities and observed outcomes, with the model showing higher clinical net benefit. Conclusion Lower BMI, WBC counts, GLB, and higher SCR levels are independent risk factors for myelosuppression among Peg-IFN-treated CHB patients. The predictive nomogram, based on those factors, is able to identify high-risk individuals for myelosuppression, thereby aiding in early alleviation of this side-effect.
Collapse
Affiliation(s)
- Jiwei Fu
- Department of Infectious Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, People’s Republic of China
| | - Ting Deng
- Second Department of Cardiovascular Medicine, Jiangxi Provincial People’s Hospital, The First Affiliated Hospital of Nanchang Medical College, Nanchang, Jiangxi Province, 330006, People’s Republic of China
| | - Ting Zheng
- Department of Infectious Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, People’s Republic of China
| | - Pei Shi
- Department of Infectious Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, People’s Republic of China
| | - Wentao Zhu
- Department of Infectious Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, People’s Republic of China
| | - Mengyu Tao
- Department of Infectious Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, People’s Republic of China
| | - Zhilong Wen
- Department of Infectious Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, People’s Republic of China
| | - Xiaoping Wu
- Department of Infectious Diseases, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, People’s Republic of China
| |
Collapse
|
5
|
Molteni R, Fiumara M, Campochiaro C, Alfieri R, Pacini G, Licari E, Tomelleri A, Diral E, Varesi A, Weber A, Quaranta P, Albano L, Gaddoni C, Basso-Ricci L, Stefanoni D, Alessandrini L, Degl'Innocenti S, Sanvito F, Bergonzi GM, Annoni A, Panigada M, Cantoni E, Canarutto D, Xie SZ, D'Alessandro A, Di Micco R, Aiuti A, Ciceri F, De Luca G, Dagna L, Matucci-Cerinic M, Merelli I, Cenci S, Scala S, Cavalli G, Naldini L, Ferrari S. Mechanisms of hematopoietic clonal dominance in VEXAS syndrome. Nat Med 2025:10.1038/s41591-025-03623-9. [PMID: 40195449 DOI: 10.1038/s41591-025-03623-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 02/28/2025] [Indexed: 04/09/2025]
Abstract
Clonal dominance characterizes hematopoiesis during aging and increases susceptibility to blood cancers and common nonmalignant disorders. VEXAS syndrome is a recently discovered, adult-onset, autoinflammatory disease burdened by a high mortality rate and caused by dominant hematopoietic clones bearing somatic mutations in the UBA1 gene. However, pathogenic mechanisms driving clonal dominance are unknown. Moreover, the lack of disease models hampers the development of disease-modifying therapies. In the present study, we performed immunophenotype characterization of hematopoiesis and single-cell transcriptomics in a cohort of nine male patients with VEXAS syndrome, revealing pervasive inflammation across all lineages. Hematopoietic stem and progenitor cells (HSPCs) in patients are skewed toward myelopoiesis and acquire senescence-like programs. Humanized models of VEXAS syndrome, generated by inserting the causative mutation in healthy HSPCs through base editing, recapitulated proteostatic defects, cytological alterations and senescence signatures of patients' cells, as well as hematological and inflammatory disease hallmarks. Competitive transplantations of human UBA1-mutant and wild-type HSPCs showed that, although mutant cells are more resilient to the inflammatory milieu, probably through the acquisition of the senescence-like state, wild-type ones are progressively exhausted and overwhelmed by VEXAS clones, overall impairing functional hematopoiesis and leading to bone marrow failure. Our study unveils the mechanism of clonal dominance and provides models for preclinical studies and preliminary insights that could inform therapeutic strategies.
Collapse
Affiliation(s)
- Raffaella Molteni
- Vita-Salute San Raffaele University, Milan, Italy.
- Inflammation Fibrosis and Ageing Initiative (INFLAGE), Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| | - Martina Fiumara
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Corrado Campochiaro
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Roberta Alfieri
- National Research Council, Institute for Biomedical Technologies, Segrate, Italy
| | - Guido Pacini
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eugenia Licari
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Tomelleri
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Elisa Diral
- Unit of Hematology and Stem Cell Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angelica Varesi
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, Canada
| | - Alessandra Weber
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Pamela Quaranta
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luisa Albano
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Chiara Gaddoni
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luca Basso-Ricci
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Davide Stefanoni
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Laura Alessandrini
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sara Degl'Innocenti
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Sanvito
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pathology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gregorio Maria Bergonzi
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Hematology and Stem Cell Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Andrea Annoni
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maddalena Panigada
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Eleonora Cantoni
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Daniele Canarutto
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stephanie Z Xie
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Denver, CO, USA
| | - Raffaella Di Micco
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- University School of Advanced Studies IUSS, Pavia, Italy
| | - Alessandro Aiuti
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology Unit and BMT Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Fabio Ciceri
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Hematology and Stem Cell Transplantation, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giacomo De Luca
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Dagna
- Vita-Salute San Raffaele University, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Matucci-Cerinic
- Vita-Salute San Raffaele University, Milan, Italy
- Inflammation Fibrosis and Ageing Initiative (INFLAGE), Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ivan Merelli
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
- National Research Council, Institute for Biomedical Technologies, Segrate, Italy
| | - Simone Cenci
- Vita-Salute San Raffaele University, Milan, Italy
- Inflammation Fibrosis and Ageing Initiative (INFLAGE), Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Serena Scala
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giulio Cavalli
- Unit of Immunology, Rheumatology, Allergy and Rare diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Division of Genetics and Cell Biology, IRCCS San Raffaele Scientific Institute, Milan, Italy
- Translational Medicine, Novartis Pharma, Basel, Switzerland
| | - Luigi Naldini
- Vita-Salute San Raffaele University, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Samuele Ferrari
- Vita-Salute San Raffaele University, Milan, Italy.
- San Raffaele Telethon Institute for Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
6
|
Jenkins MT, Chu YE, Franceski AM, Potts CR, Dubin R, Dickerson KM, Lee SC, Lu R, Welner RS, Ferrell PB. TET2-loss enhances immediate and time-resolved interferon-γ signaling responses across myeloid differentiation. Exp Hematol 2025; 144:104727. [PMID: 39855457 DOI: 10.1016/j.exphem.2025.104727] [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: 11/12/2024] [Revised: 01/11/2025] [Accepted: 01/13/2025] [Indexed: 01/27/2025]
Abstract
Signaling responses to cytokines are disrupted in clonal hematopoiesis and myeloid malignancies. To better identify specific signaling response alterations in the presence or absence of TET2, we developed a 36-parameter cytometry by time-of-flight (CyTOF) panel of both surface marker and phosphoprotein antigens in murine bone marrow (BM). We show diverse, cell-type specific inflammatory cytokine responses in healthy hematopoietic cells. We next investigated changes associated with BM cells from Tet2KO mice. High-dimensional surface marker phenotyping revealed expansion of hematopoietic stem and progenitor cells (HSPCs), committed cKIT+Ly6C+ myeloid progenitors, and monocytes. Loss of TET2 function increased the magnitude of response to extracellular perturbations, including interferon (IFN)γ and H2O2. Response time courses revealed that IFNγ-mediated pSTAT1 remains elevated over time in Tet2KO. Further, IFNγ resulted in a more significant increase in major histocompatibility complex class II (MHCII) expression in Tet2KO immortalized progenitor cells than in Tet2WT. Inhibition of Janus kinase 1 and 2 (JAK1/2) with ruxolitinib significantly reduced STAT1 phosphorylation and MHCII expression in Tet2KO cells. Our results identify targetable disrupted signaling responses in Tet2KO cells.
Collapse
Affiliation(s)
- Matthew T Jenkins
- Department of Pharmacology, Vanderbilt University, Nashville, TN; Department of Medicine, Division of Hematology/Oncology & Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Yunli E Chu
- Department of Medicine, Division of Hematology/Oncology & Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN
| | - Alana M Franceski
- Division of Hematology/Oncology and O'Neal Comprehensive Cancer Center at the University of Alabama at Birmingham, Birmingham, AL
| | - Chad R Potts
- Department of Medicine, Division of Hematology/Oncology & Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Rebecca Dubin
- Department of Medicine, Division of Hematology/Oncology & Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Kirsten M Dickerson
- Department of Medicine, Division of Hematology/Oncology & Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN
| | - Stanley C Lee
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Rui Lu
- Division of Hematology/Oncology and O'Neal Comprehensive Cancer Center at the University of Alabama at Birmingham, Birmingham, AL
| | - Robert S Welner
- Division of Hematology/Oncology and O'Neal Comprehensive Cancer Center at the University of Alabama at Birmingham, Birmingham, AL
| | - P Brent Ferrell
- Department of Medicine, Division of Hematology/Oncology & Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN; Nashville Veterans Affairs Hospital, Tennessee Valley Health Care, Department of Veterans Affairs, Nashville, TN.
| |
Collapse
|
7
|
Sobrino S, Joseph L, Magrin E, Chalumeau A, Hebert N, Corsia A, Denis A, Roudaut C, Aussel C, Leblanc O, Brusson M, Felix T, Diana JS, Petrichenko A, El Etri J, Godard A, Tibi E, Manceau S, Treluyer JM, Mavilio F, Bushman FD, Marcais A, Castelle M, Neven B, Hermine O, Renolleau S, Magnani A, Asnafi V, El Nemer W, Bartolucci P, Six E, Semeraro M, Miccio A, Cavazzana M. Severe inflammation and lineage skewing are associated with poor engraftment of engineered hematopoietic stem cells in patients with sickle cell disease. Nat Commun 2025; 16:3137. [PMID: 40169559 PMCID: PMC11961595 DOI: 10.1038/s41467-025-58321-4] [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: 08/14/2024] [Accepted: 03/04/2025] [Indexed: 04/03/2025] Open
Abstract
In sickle cell disease (SCD), the β6Glu→Val substitution in the β-globin leads to red blood cell sickling. The transplantation of autologous, genetically modified hematopoietic stem and progenitor cells (HSPCs) is a promising treatment option for patients with SCD. We completed a Phase I/II open-label clinical trial (NCT03964792) for patients with SCD using a lentiviral vector (DREPAGLOBE) expressing a potent anti-sickling β-globin. The primary endpoint was to evaluate the short-term safety and secondary endpoints included the efficacy and the long-term safety. We report on the results after 18 to 36 months of follow-up. No drug-related adverse events or signs of clonal hematopoiesis were observed. Despite similar vector copy numbers in the drug product, gene-marking in peripheral blood mononuclear cells and correction of the clinical phenotype varied from one patient to another. Single-cell transcriptome analyses show that in the patients with poor engraftment, the most immature HSCs display an exacerbated inflammatory signature (via IL-1 or TNF-α and interferon signaling pathways). This signature is accompanied by a lineage bias in the HSCs. Our clinical data indicates that the DREPAGLOBE-based gene therapy (GT) is safe. However, its efficacy is variable and probably depends on the number of infused HSCs and intrinsic, engraftment-impairing inflammatory alterations in HSCs. Trial: NCT03964792.
Collapse
Affiliation(s)
- Steicy Sobrino
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Laure Joseph
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
| | - Elisa Magrin
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Anne Chalumeau
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
| | - Nicolas Hebert
- Univ Paris Est Créteil, IMRB, Laboratory of Excellence LABEX GRex, Créteil, France
- Etablissement Français du Sang, Créteil, France
| | - Alice Corsia
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Department of Adult Hematology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Laboratory of Excellence LABEX GRex, Paris, France
| | - Adeline Denis
- Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Cécile Roudaut
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Clotilde Aussel
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Olivia Leblanc
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Mégane Brusson
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
| | - Tristan Felix
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
| | - Jean-Sebastien Diana
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Angelina Petrichenko
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jana El Etri
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
- Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Auria Godard
- Aix Marseille Univ, CNRS, EFS, ADES, Labex GR-Ex, Marseille, France
| | - Eden Tibi
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
| | - Sandra Manceau
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Department of Adult Hematology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Laboratory of Excellence LABEX GRex, Paris, France
| | - Jean Marc Treluyer
- Centre d'Investigation Clinique-Unité de Recherche Clinique, Hôpital Universitaire Necker Enfants-Malades, GH Paris Centre, Paris, France
- Université Paris Cité, Paris, France
| | - Fulvio Mavilio
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Frederic D Bushman
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ambroise Marcais
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Department of Adult Hematology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Laboratory of Excellence LABEX GRex, Paris, France
| | - Martin Castelle
- Pediatric Immunology and Hematology Department, Hôpital Necker Enfants-Malades, Paris, France
| | - Benedicte Neven
- Pediatric Immunology and Hematology Department, Hôpital Necker Enfants-Malades, Paris, France
| | - Olivier Hermine
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Department of Adult Hematology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Laboratory of Excellence LABEX GRex, Paris, France
| | - Sylvain Renolleau
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Department of Adult Hematology, Necker Hospital, Assistance Publique-Hôpitaux de Paris, Laboratory of Excellence LABEX GRex, Paris, France
| | - Alessandra Magnani
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France
| | - Vahid Asnafi
- Laboratory of Onco-Hematology, Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Wassim El Nemer
- Aix Marseille Univ, CNRS, EFS, ADES, Labex GR-Ex, Marseille, France
| | - Pablo Bartolucci
- Univ Paris Est Créteil, IMRB, Laboratory of Excellence LABEX GRex, Créteil, France
- Paris-East Créteil University, Henri Mondor University Hospitals, APHP, Sickle Cell Referral Center-UMGGR, Créteil, France
| | - Emmanuelle Six
- Laboratory of Human Lymphohematopoiesis, INSERM, Imagine Institute, Paris, France
| | - Michaela Semeraro
- Centre d'Investigation Clinique-Unité de Recherche Clinique, Hôpital Universitaire Necker Enfants-Malades, GH Paris Centre, Paris, France
- Université Paris Cité, Inserm, Pharmacologie et évaluations des thérapeutiques chez l'enfant et la femme enceinte, Paris, France
| | - Annarita Miccio
- Laboratory of Chromatin and Gene Regulation During Development, University Paris Cite, UMR1163 INSERM, Imagine Institute, Paris, France
| | - Marina Cavazzana
- Departement of Biotherapy Necker Children's Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP) Groupe Hospitalier Universitaire Centre, Université Paris Cité, Paris, France.
- Biotherapy Clinical Investigation Center, AP-HP, INSERM, Institut Imagine, Paris, France.
| |
Collapse
|
8
|
Mhlanga MM, Fanucchi S, Ozturk M, Divangahi M. Cellular and Molecular Mechanisms of Innate Memory Responses. Annu Rev Immunol 2025; 43:615-640. [PMID: 40279311 DOI: 10.1146/annurev-immunol-101721-035114] [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] [Indexed: 04/27/2025]
Abstract
There has been an increasing effort to understand the memory responses of a complex interplay among innate, adaptive, and structural cells in peripheral organs and bone marrow. Trained immunity is coined as the de facto memory of innate immune cells and their progenitors. These cells acquire epigenetic modifications and shift their metabolism to equip an imprinted signature to a persistent fast-responsive functional state. Recent studies highlight the contribution of noncoding RNAs and modulation of chromatin structures in establishing this epigenetic readiness for potential immune perturbations. In this review, we discuss recent studies that highlight trained immunity-mediated memory responses emerging intrinsically in innate immune cells and as a complex interplay with other cells at the organ level. Lastly, we survey epigenetic contributors to trained immunity phenotypes-specifically, a recently discovered regulatory circuit coordinating the regulation of a key driver of trained immunity.
Collapse
Affiliation(s)
- Musa M Mhlanga
- Epigenomics & Single Cell Biophysics Group, Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, Nijmegen, The Netherlands;
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Human Genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - Mumin Ozturk
- Epigenomics & Single Cell Biophysics Group, Department of Cell Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University, Nijmegen, The Netherlands;
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maziar Divangahi
- Departments of Medicine, Pathology, and Microbiology & Immunology, McGill University, Montreal, Quebec, Canada
- McGill University Health Centre, McGill International TB Centre, and Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada;
| |
Collapse
|
9
|
Bruno F, Georgiou C, Cunningham D, Bett L, Secchi MA, Atkinson S, González Antón S, Birch F, Langhorne J, Lo Celso C. Differential Response and Recovery Dynamics of HSPC Populations Following Plasmodium chabaudi Infection. Int J Mol Sci 2025; 26:2816. [PMID: 40141458 PMCID: PMC11943058 DOI: 10.3390/ijms26062816] [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: 11/28/2024] [Revised: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 03/28/2025] Open
Abstract
Severe infections such as malaria are on the rise worldwide, driven by both climate change and increasing drug resistance. It is therefore paramount that we better understand how the host responds to severe infection. Hematopoiesis is particularly of interest in this context because hematopoietic stem and progenitor cells (HSPCs) maintain the turnover of all blood cells, including all immune cells. Severe infections have been widely acknowledged to affect HSPCs; however, this disruption has been mainly studied during the acute phase, and the process and level of HSPC recovery remain understudied. Using a self-resolving model of natural rodent malaria, infection by Plasmodium chabaudi, here we systematically assess phenotypically defined HSPCs' acute response and recovery upon pathogen clearance. We demonstrate that during the acute phase of infection the most quiescent and functional stem cells are depleted, multipotent progenitor compartments are drastically enlarged, and oligopotent progenitors virtually disappear, underpinned by dramatic, population-specific and sometimes unexpected changes in proliferation rates. HSPC populations return to homeostatic size and proliferation rate again through specific patterns of recovery. Overall, our data demonstrate that HSPC populations adopt different responses to cope with severe infection and suggest that the ability to adjust proliferative capacity becomes more restricted as differentiation progresses.
Collapse
Affiliation(s)
- Federica Bruno
- Department of Life Sciences, Imperial College London, London SW10 9NH, UK; (F.B.)
- Sir Francis Crick Institute, London NW1 1AT, UK
| | - Christiana Georgiou
- Department of Life Sciences, Imperial College London, London SW10 9NH, UK; (F.B.)
- Sir Francis Crick Institute, London NW1 1AT, UK
| | | | - Lucy Bett
- Department of Life Sciences, Imperial College London, London SW10 9NH, UK; (F.B.)
| | - Marine A. Secchi
- Department of Life Sciences, Imperial College London, London SW10 9NH, UK; (F.B.)
- Sir Francis Crick Institute, London NW1 1AT, UK
| | - Samantha Atkinson
- Department of Life Sciences, Imperial College London, London SW10 9NH, UK; (F.B.)
- Sir Francis Crick Institute, London NW1 1AT, UK
| | - Sara González Antón
- Department of Life Sciences, Imperial College London, London SW10 9NH, UK; (F.B.)
- Sir Francis Crick Institute, London NW1 1AT, UK
| | - Flora Birch
- Department of Life Sciences, Imperial College London, London SW10 9NH, UK; (F.B.)
- Sir Francis Crick Institute, London NW1 1AT, UK
| | | | - Cristina Lo Celso
- Department of Life Sciences, Imperial College London, London SW10 9NH, UK; (F.B.)
- Sir Francis Crick Institute, London NW1 1AT, UK
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London W12 0NN, UK
| |
Collapse
|
10
|
Du L, Freitas-Cortez MA, Zhang J, Xue Y, Veettil RT, Zhao Z, Morrison SJ. Periarteriolar niches become inflamed in aging bone marrow, remodeling the stromal microenvironment and depleting lymphoid progenitors. Proc Natl Acad Sci U S A 2025; 122:e2412317122. [PMID: 40063797 PMCID: PMC11929388 DOI: 10.1073/pnas.2412317122] [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: 06/19/2024] [Accepted: 02/05/2025] [Indexed: 03/25/2025] Open
Abstract
In early postnatal and young adult bone marrow, Leptin receptor-expressing (LepR+) stromal cells and endothelial cells synthesize factors required for hematopoietic stem cell (HSC) maintenance, including Stem Cell Factor (SCF) and Cxcl12. However, little is known about how these stromal cells change during aging. We performed single-cell RNA sequencing of mouse bone marrow stromal cells at 2, 12, and 24 mo of age. We identified five transcriptionally distinct subsets of LepR+ cells, all of which expressed the highest levels of Scf and Cxcl12 in bone marrow throughout adult life. In aging bone marrow, SCF from LepR+ cells, but not endothelial cells, continued to be necessary for the maintenance of HSCs and early restricted progenitors. However, arteriolar endothelial cells and other periarteriolar cells expressed increasing levels of interferon during aging. This increased the numbers of periarteriolar Sca1+Cxcl9+LepR+ cells with an inflammatory gene signature and depleted lymphoid progenitors, at least some of which are also periarteriolar. The periarteriolar environment thus became particularly inflamed during aging, remodeling the stromal microenvironment and depleting lymphoid progenitors in an interferon-dependent manner.
Collapse
Affiliation(s)
- Liming Du
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Maria Angelica Freitas-Cortez
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Jingzhu Zhang
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Yuanyuan Xue
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Reshma T. Veettil
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Zhiyu Zhao
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX75390
| | - Sean J. Morrison
- Children’s Research Institute and the Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX75390
- HHMI, University of Texas Southwestern Medical Center, Dallas, TX75390
| |
Collapse
|
11
|
van der Geest R, Lee JS. Role of the basic leucine zipper transcription factor BATF2 in modulating immune responses and inflammation in health and disease. J Leukoc Biol 2025; 117:qiae245. [PMID: 39504573 PMCID: PMC11953073 DOI: 10.1093/jleuko/qiae245] [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: 07/20/2024] [Accepted: 11/06/2024] [Indexed: 11/08/2024] Open
Abstract
BATF2 is a transcription factor known to exhibit tumor-suppressive activity in cancer cells. Within recent years, however, BATF2 has also emerged as an important transcriptional regulator of the immune system. Through its immunomodulatory function, BATF2 has been implicated in a variety of (patho)physiological processes, including host defense against infection, antitumor immunity, and maintenance of tissue inflammatory homeostasis. Below, we discuss recent literature that has provided insight into the role of BATF2 as a transcriptional regulator of immune responses in health and disease, including the cell types that express BATF2, the different diseases in which the immunomodulatory effects of BATF2 have been shown to play a role, and the molecular mechanisms through which BATF2 is thought to exert those effects. In doing so, we highlight that the immunological effects of BATF2 are highly context dependent, and we point out the overlap between the mechanisms of action of BATF2 in infectious and noninfectious diseases. We also discuss areas of interest for future research, the clinical relevance of better understanding BATF2 function, and potential strategies for therapeutic modulation of BATF2.
Collapse
Affiliation(s)
- Rick van der Geest
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, 3459 Fifth Avenue, Pittsburgh, PA 15213, United States
| | - Janet S Lee
- Acute Lung Injury Center of Excellence, Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, University of Pittsburgh, 3459 Fifth Avenue, Pittsburgh, PA 15213, United States
- Vascular Medicine Institute, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, United States
- Division of Pulmonary and Critical Care Medicine, Washington University in St. Louis, 660 South Euclid Avenue, St. Louis, MO 63110, United States
| |
Collapse
|
12
|
Ramalingam P, Gutkin MC, Poulos MG, Winiarski A, Smith A, Carter C, Doughty C, Tillery T, Redmond D, Freire AG, Butler JM. Suppression of thrombospondin-1-mediated inflammaging prolongs hematopoietic health span. Sci Immunol 2025; 10:eads1556. [PMID: 39752538 PMCID: PMC12068530 DOI: 10.1126/sciimmunol.ads1556] [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: 07/31/2024] [Accepted: 12/02/2024] [Indexed: 04/04/2025]
Abstract
Chronic low-grade inflammation observed in older adults, termed inflammaging, is a common feature underlying a multitude of aging-associated maladies including a decline in hematopoietic activity. However, whether suppression of inflammaging can preserve hematopoietic health span remains unclear, in part because of a lack of tools to measure inflammaging within hematopoietic stem cells (HSCs). Here, we identify thrombospondin-1 (Thbs1) as an essential regulator of inflammaging within HSCs. We describe a transcriptomics-based approach for measuring inflammaging within stem cells and demonstrate that deletion of Thbs1 is sufficient to prevent HSC inflammaging. Our results demonstrate that suppression of HSC inflammaging prevents aging-associated defects in hematopoietic activity including loss of HSC self-renewal, myeloid-biased HSC differentiation, and anemia. Our findings indicate that suppression of HSC inflammaging may also prolong overall systemic health span.
Collapse
Affiliation(s)
- Pradeep Ramalingam
- Division of Hematology and Oncology, Department of Medicine, University of Florida; Gainesville, FL, 32610, USA
| | - Michael C. Gutkin
- Center for Discovery and Innovation, Hackensack University Medical Center; Nutley, NJ, 07110, USA
| | - Michael G. Poulos
- Division of Hematology and Oncology, Department of Medicine, University of Florida; Gainesville, FL, 32610, USA
| | - Agatha Winiarski
- Division of Hematology and Oncology, Department of Medicine, University of Florida; Gainesville, FL, 32610, USA
| | - Arianna Smith
- Division of Hematology and Oncology, Department of Medicine, University of Florida; Gainesville, FL, 32610, USA
| | - Cody Carter
- Division of Hematology and Oncology, Department of Medicine, University of Florida; Gainesville, FL, 32610, USA
| | - Chelsea Doughty
- Center for Discovery and Innovation, Hackensack University Medical Center; Nutley, NJ, 07110, USA
| | - Taylor Tillery
- Center for Discovery and Innovation, Hackensack University Medical Center; Nutley, NJ, 07110, USA
| | - David Redmond
- Department of Medicine, Weill Cornell Medicine; New York, NY, 10065, USA
| | - Ana G. Freire
- Center for Discovery and Innovation, Hackensack University Medical Center; Nutley, NJ, 07110, USA
| | - Jason M. Butler
- Division of Hematology and Oncology, Department of Medicine, University of Florida; Gainesville, FL, 32610, USA
| |
Collapse
|
13
|
Zhang C, Hao T, Bortoluzzi A, Chen MH, Wu X, Wang J, Ermel R, Kim Y, Chen S, Chen W. Sex-dependent differences in hematopoietic stem cell aging and leukemogenic potential. Oncogene 2025; 44:64-78. [PMID: 39487323 PMCID: PMC11706783 DOI: 10.1038/s41388-024-03197-9] [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: 07/15/2024] [Revised: 10/09/2024] [Accepted: 10/11/2024] [Indexed: 11/04/2024]
Abstract
Sex influences many biological outcomes, but how sex affects hematopoietic stem cell (HSC) aging and hematological disorders is poorly understood. The widespread use of young animal models to study age-related diseases further complicates these matters. Using aged and long-lived BALB/c mouse models, we discovered that aging mice exhibit sex-dependent disparities, mirroring aging humans, in developing myeloid skewing, anemia, and leukemia. These disparities are underlined by sex-differentiated HSC aging characteristics across the population, single-cell, and molecular levels. The HSC population expanded significantly with aging and longevity in males, but this occurred to a much lesser degree in aging females that instead expanded committed progenitors. Aging male HSCs are more susceptible to BCR-ABL1 transformation with faster development of chronic myeloid leukemia (CML) than female HSCs. Additionally, the loss of the aging regulator Sirt1 inhibited CML development in aging male but not female mice. Our results showed for the first time that sex-differentiated HSC aging impacts hematopoiesis, leukemogenesis, and certain gene functions. This discovery provides insights into understanding age-dependent hematological diseases and sex-targeted strategies for the treatment and prevention of certain blood disorders and cancer.
Collapse
MESH Headings
- Animals
- Hematopoietic Stem Cells/metabolism
- Hematopoietic Stem Cells/pathology
- Female
- Male
- Mice
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics
- Cellular Senescence
- Aging/pathology
- Aging/physiology
- Mice, Inbred BALB C
- Sirtuin 1/metabolism
- Sirtuin 1/genetics
- Hematopoiesis
- Sex Characteristics
- Humans
- Cell Transformation, Neoplastic/pathology
- Cell Transformation, Neoplastic/metabolism
- Cell Transformation, Neoplastic/genetics
- Sex Factors
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
Collapse
Affiliation(s)
- Chunxiao Zhang
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
- Amgen, Thousand Oaks, CA, USA
| | - Taisen Hao
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
- Bristol Myers Squibb, Seattle, WA, USA
| | - Alessia Bortoluzzi
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Min-Hsuan Chen
- Integrative Genomics Core, Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Xiwei Wu
- Integrative Genomics Core, Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Jinhui Wang
- Integrative Genomics Core, Department of Molecular and Cellular Biology, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Richard Ermel
- Center for Comparative Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - Young Kim
- Department of Pathology, City of Hope National Medical Center, Duarte, CA, 91010, USA
| | - Shiuan Chen
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA
| | - WenYong Chen
- Department of Cancer Biology and Molecular Medicine, Beckman Research Institute, City of Hope, Duarte, CA, 91010, USA.
| |
Collapse
|
14
|
Stevens CS, Carmichael JC, Watkinson R, Kowdle S, Reis RA, Hamane K, Jang J, Park A, Pernet O, Khamaikawin W, Hong P, Thibault P, Gowlikar A, An DS, Lee B. A temperature-sensitive and less immunogenic Sendai virus for efficient gene editing. J Virol 2024; 98:e0083224. [PMID: 39494910 PMCID: PMC11650993 DOI: 10.1128/jvi.00832-24] [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: 05/09/2024] [Accepted: 10/10/2024] [Indexed: 11/05/2024] Open
Abstract
The therapeutic potential of gene editing technologies hinges on the development of safe and effective delivery methods. In this study, we developed a temperature-sensitive and less immunogenic Sendai virus (ts SeV) as a novel delivery vector for CRISPR-Cas9 and for efficient gene editing in sensitive human cell types with limited induction of an innate immune response. ts SeV demonstrates high transduction efficiency in human CD34+ hematopoietic stem and progenitor cells (HSPCs) including transduction of the CD34+/CD38-/CD45RA-/CD90+(Thy1+)/CD49fhigh stem cell enriched subpopulation. The frequency of CCR5 editing exceeded 90% and bi-allelic CCR5 editing exceeded 70% resulting in significant inhibition of HIV-1 infection in primary human CD14+ monocytes. These results demonstrate the potential of the ts SeV platform as a safe, efficient, and flexible addition to the current gene-editing tool delivery methods, which may help further expand the possibilities in personalized medicine and the treatment of genetic disorders. IMPORTANCE Gene editing has the potential to be a powerful tool for the treatment of human diseases including HIV, β-thalassemias, and sickle cell disease. Recent advances have begun to overcome one of the major limiting factors of this technology, namely delivery of the CRISPR-Cas9 gene editing machinery, by utilizing viral vectors. However, gene editing therapies have yet to be implemented due to inherent risks associated with the DNA viral vectors typically used for delivery. As an alternative strategy, we have developed an RNA-based Sendai virus CRISPR-Cas9 delivery vector that does not integrate into the genome, is temperature sensitive, and does not induce a significant host interferon response. This recombinant SeV successfully delivered CRISPR-Cas9 in primary human CD14+ monocytes ex vivo resulting in a high level of CCR5 editing and inhibition of HIV infection.
Collapse
Affiliation(s)
- Christian S. Stevens
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jillian C. Carmichael
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Ruth Watkinson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shreyas Kowdle
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Rebecca A. Reis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Kory Hamane
- UCLA School of Nursing, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Jason Jang
- UCLA School of Nursing, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Arnold Park
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Olivier Pernet
- UCLA School of Nursing, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Wannisa Khamaikawin
- UCLA School of Nursing, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Patrick Hong
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Patricia Thibault
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Aditya Gowlikar
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Dong Sung An
- UCLA School of Nursing, Los Angeles, California, USA
- UCLA AIDS Institute, Los Angeles, California, USA
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| |
Collapse
|
15
|
Kim YS, Lupatov AY, Burunova VV, Bagmet NN, Chardarov NK, Malov SL, Kholodenko RV, Shatverian GA, Manukyan GV, Yarygin KN, Kholodenko IV. Human Liver MSCs Retain Their Basic Cellular Properties in Chronically Inflamed Liver Tissue. Int J Mol Sci 2024; 25:13374. [PMID: 39769138 PMCID: PMC11676302 DOI: 10.3390/ijms252413374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/04/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Every 25th death worldwide is associated with liver pathology. The development of novel approaches to liver diseases therapy and protocols for maintaining the vital functions of patients on the liver transplant waiting list are urgently needed. Resident mesenchymal stem cells (MSCs) play a significant role in supporting liver tissue integrity and improve the liver condition after infusion. However, it remains unclear whether MSCs isolated from chronically inflamed livers are similar in their basic cellular properties to MSCs obtained from healthy livers. We applied a large array of tests to compare resident MSCs isolated from apparently normal liver tissue and from chronically inflamed livers of patients with fibrosis, cirrhosis, and viral hepatitis. Chronic inflammatory environment did not alter the major cellular characteristics of MSCs, including the expression of MSC markers, stem cell markers, adhesion molecules, and the hallmarks of senescence, as well as cell proliferation, migration, and secretome. Only the expression of some immune checkpoints and toll-like receptors was different. Evidently, MSCs with unchanged cellular properties are present in human liver even at late stages of inflammatory diseases. These cells can be isolated and used as starting material in the development of cell therapies of liver diseases.
Collapse
Affiliation(s)
- Yan S. Kim
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| | - Alexey Yu. Lupatov
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| | - Veronika V. Burunova
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| | - Nikolay N. Bagmet
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Nikita K. Chardarov
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Svyatoslav L. Malov
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Roman V. Kholodenko
- Laboratory of Molecular Immunology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, 117997 Moscow, Russia
| | - Garnik A. Shatverian
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Garik V. Manukyan
- Department of Abdominal Surgery and Oncology, Laboratory of Emergency Surgery and Portal Hypertension, Petrovsky National Research Centre of Surgery, 119435 Moscow, Russia
| | - Konstantin N. Yarygin
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
- Department of General Pathology and Pathophysiology, Russian Medical Academy of Continuous Professional Education, 125284 Moscow, Russia
| | - Irina V. Kholodenko
- Laboratory of Cell Biology, V.N. Orekhovich Institute of Biomedical Chemistry, 119121 Moscow, Russia (K.N.Y.)
| |
Collapse
|
16
|
Ruffinatto L, Groult Y, Iacono J, Sarrazin S, de Laval B. Hematopoietic stem cell a reservoir of innate immune memory. Front Immunol 2024; 15:1491729. [PMID: 39720722 PMCID: PMC11666435 DOI: 10.3389/fimmu.2024.1491729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/30/2024] [Indexed: 12/26/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are a rare, long-lived and multipotent population that give rise to majority of blood cells and some tissue-resident immune cells. There is growing evidence that inflammatory stimuli can trigger persistent reprogramming in HSCs that enhances or inhibits the cellular functions of these HSCs and their progeny in response to subsequent infections. This newly discovered property makes HSCs a reservoir for innate immune memory. The molecular mechanisms underlying innate immune memory in HSCs are similar to those observed in innate immune cells, although their full elucidation is still pending. In this review, we examine the current state of knowledge on how an inflammatory response leads to reprogramming of HSCs. Understanding the full spectrum of consequences of reshaping early hematopoiesis is critical for assessing the potential benefits and risks under physiological and pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | - Bérengère de Laval
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut
National de la Santé et de la Recherche Médicale (INSERM), Centre d’Immunologie de Marseille-Luminy (CIML), Marseille, France
| |
Collapse
|
17
|
Frati G, Brusson M, Sartre G, Mlayah B, Felix T, Chalumeau A, Antoniou P, Hardouin G, Concordet JP, Romano O, Turchiano G, Miccio A. Safety and efficacy studies of CRISPR-Cas9 treatment of sickle cell disease highlights disease-specific responses. Mol Ther 2024; 32:4337-4352. [PMID: 39044427 PMCID: PMC11638826 DOI: 10.1016/j.ymthe.2024.07.015] [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: 01/26/2024] [Revised: 05/15/2024] [Accepted: 07/18/2024] [Indexed: 07/25/2024] Open
Abstract
Fetal hemoglobin (HbF) reactivation expression through CRISPR-Cas9 is a promising strategy for the treatment of sickle cell disease (SCD). Here, we describe a genome editing strategy leading to reactivation of HbF expression by targeting the binding sites (BSs) for the lymphoma-related factor (LRF) repressor in the γ-globin promoters. CRISPR-Cas9 treatment in healthy donor (HD) and patient-derived HSPCs resulted in a high frequency of LRF BS disruption and potent HbF synthesis in their erythroid progeny. LRF BS disruption did not impair HSPC engraftment and differentiation but was more efficient in SCD than in HD cells. However, SCD HSPCs showed a reduced engraftment and a myeloid bias compared with HD cells. We detected off-target activity and chromosomal rearrangements, particularly in SCD samples (likely because of the higher overall editing efficiency) but did not impact the target gene expression and HSPC engraftment and differentiation. Transcriptomic analyses showed that the editing procedure results in the up-regulation of genes involved in DNA damage and inflammatory responses, which was more evident in SCD HSPCs. This study provides evidence of efficacy and safety for an editing strategy based on HbF reactivation and highlights the need of performing safety studies in clinically relevant conditions, i.e., in patient-derived HSPCs.
Collapse
Affiliation(s)
- Giacomo Frati
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Megane Brusson
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Gilles Sartre
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Bochra Mlayah
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Tristan Felix
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Anne Chalumeau
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Panagiotis Antoniou
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Giulia Hardouin
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France
| | - Jean-Paul Concordet
- INSERM U1154, CNRS UMR7196, Museum National d'Histoire Naturelle, Paris, France
| | - Oriana Romano
- University of Padova, Department of Molecular Medicine, Padova, Italy
| | | | - Annarita Miccio
- Université Paris Cité, Imagine Institute, Laboratory of Chromatin and Gene Regulation During Development, INSERM UMR 1163, Paris, France.
| |
Collapse
|
18
|
Florez MA, Thatavarty A, Le DT, Hill HA, Jeong Y, Ho BM, Kus P, Wathan TK, Kain BN, Huang S, Park D, King KY. BST2 facilitates activation of hematopoietic stem cells through ERK signaling. Exp Hematol 2024; 140:104653. [PMID: 39362577 DOI: 10.1016/j.exphem.2024.104653] [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: 06/21/2024] [Revised: 09/20/2024] [Accepted: 09/22/2024] [Indexed: 10/05/2024]
Abstract
The proinflammatory cytokine interferon gamma (IFNγ) is upregulated in a variety of infections and contributes to bone marrow failure through hematopoietic stem cell (HSC) activation and subsequent exhaustion. The cell-surface protein, bone marrow stromal antigen 2 (BST2), is a key mediator of this process, because it is induced upon IFN stimulation and required for IFN-dependent HSC activation. To identify the mechanism by which BST2 promotes IFN-dependent HSC activation, we evaluated its role in niche localization, immune cell function, lipid raft formation, and intracellular signaling. Our studies indicated that knockout (KO) of BST2 in a murine model does not disrupt immune cell responses to IFN-inducing mycobacterial infection. Furthermore, intravital imaging studies indicate that BST2 KO does not disrupt localization of HSCs relative to endothelial or osteoblastic niches in the bone marrow. However, using imaging-based flow cytometry, we found that IFNγ treatment shifts the lipid raft polarity of wild-type (WT) but not Bst2-/- hematopoietic stem and progenitor cells (HSPCs). Furthermore, RNAseq analysis, reverse-phase protein array and western blot analysis of HSPCs indicate that BST2 promotes ERK1/2 phosphorylation during IFNγ-mediated stress. Overall, we find that BST2 facilitates HSC division by promoting cell polarization and ERK activation, thus elucidating a key mechanism of IFN-dependent HSPC activation. These findings inform future approaches in the treatment of cancer and bone marrow failure.
Collapse
Affiliation(s)
- Marcus A Florez
- Program in Translational Biology and Molecular Medicine, Graduate School of Biomedical Sciences (GSBS) and Medical Scientist Training Program, Baylor College of Medicine, Houston, TX; Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Apoorva Thatavarty
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Program in Genetics and Genomics, GSBS, and Medical Scientist Training Program, Baylor College of Medicine, Houston, TX
| | - Duy T Le
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Program in Immunology, GSBS, Baylor College of Medicine, Houston, TX
| | - Holly A Hill
- Department of Statistics, School of Engineering, Rice University, Houston, TX
| | - Youngjae Jeong
- Program in Genetics and Genomics, GSBS, and Medical Scientist Training Program, Baylor College of Medicine, Houston, TX
| | - Brian M Ho
- Program in Quantitative and Computational Biosciences, Baylor College of Medicine, Houston, TX
| | - Pawel Kus
- Department of Systems Biology and Engineering, Silesian University of Technology, Gliwice, Poland
| | - Trisha K Wathan
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine and Texas Children's Hospital, Houston, TX
| | - Bailee N Kain
- Program in Translational Biology and Molecular Medicine, Graduate School of Biomedical Sciences (GSBS) and Medical Scientist Training Program, Baylor College of Medicine, Houston, TX
| | - Shixia Huang
- Advanced Technology Cores, Department of Molecular and Cellular Biology, Department of Education, Innovation & Technology, Houston, TX; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX
| | - Dongsu Park
- Program in Genetics and Genomics, GSBS, and Medical Scientist Training Program, Baylor College of Medicine, Houston, TX
| | - Katherine Y King
- Program in Translational Biology and Molecular Medicine, Graduate School of Biomedical Sciences (GSBS) and Medical Scientist Training Program, Baylor College of Medicine, Houston, TX; Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine and Texas Children's Hospital, Houston, TX; Program in Immunology, GSBS, Baylor College of Medicine, Houston, TX; Dan L. Duncan Cancer Center, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
19
|
Patel SB, Moskop DR, Jordan CT, Pietras EM. Understanding MDS stem cells: Advances and limitations. Semin Hematol 2024; 61:409-419. [PMID: 39472255 DOI: 10.1053/j.seminhematol.2024.09.007] [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: 06/25/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 11/10/2024]
Abstract
In work spanning several decades, extensive studies have focused on the properties of malignant stem cells that drive the pathogenesis of acute myeloid leukemia (AML). However, relatively little attention has been devoted to several serious myeloid malignancies that occur prior to the onset of frank leukemia, including myelodysplastic syndrome (MDS). Like leukemia, MDS is hypothesized to arise from a pool of immature malignant stem and progenitor cells (MDS-SCs) that serve as a reservoir for disease evolution and progression1. While multiple studies have sought to identify and characterize the biology and vulnerabilities of MDS-SCs, yet translation of scientific concepts to therapeutically impactful regimens has been limited. Here, we evaluate the currently known properties of MDS-SCs as well as the post-transcriptional mechanisms that drive MDS pathogenesis at a stem and progenitor level. We highlight limits and gaps in our characterization and understanding of MDS-SCs and address the extent to which the properties of MDS-SC are (and can be) inferred from the characterization of LSCs.
Collapse
Affiliation(s)
- Sweta B Patel
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Daniel R Moskop
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Craig T Jordan
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| | - Eric M Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| |
Collapse
|
20
|
Rodriguez-Sevilla JJ, Colla S. Inflammation in myelodysplastic syndrome pathogenesis. Semin Hematol 2024; 61:385-396. [PMID: 39424469 DOI: 10.1053/j.seminhematol.2024.09.005] [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: 09/07/2024] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
Inflammation is a key driver of the progression of preleukemic myeloid conditions, such as clonal hematopoiesis of indeterminate potential (CHIP) and clonal cytopenia of undetermined significance (CCUS), to myelodysplastic syndromes (MDS). Inflammation is a critical mediator in the complex interplay of the genetic, epigenetic, and microenvironmental factors contributing to clonal evolution. Under inflammatory conditions, somatic mutations in TET2, DNMT3A, and ASXL1, the most frequently mutated genes in CHIP and CCUS, induce a competitive advantage to hematopoietic stem and progenitor cells, which leads to their clonal expansion in the bone marrow. Chronic inflammation also drives metabolic reprogramming and immune system deregulation, further promoting the expansion of malignant clones. This review underscores the urgent need to fully elucidate the role of inflammation in MDS initiation and highlights the potential of the therapeutical targeting of inflammatory pathways as an early intervention in MDS.
Collapse
Affiliation(s)
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX.
| |
Collapse
|
21
|
Mills TS, Kain B, Burchill MA, Danis E, Lucas ED, Culp-Hill R, Cowan CM, Schleicher WE, Patel SB, Tran BT, Cao R, Goodspeed A, Ferrara S, Bevers S, Jirón Tamburini BA, Roede JR, D'Alessandro A, King KY, Pietras EM. A distinct metabolic and epigenetic state drives trained immunity in HSC-derived macrophages from autoimmune mice. Cell Stem Cell 2024; 31:1630-1649.e8. [PMID: 39413777 PMCID: PMC11560650 DOI: 10.1016/j.stem.2024.09.010] [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: 03/06/2024] [Revised: 06/18/2024] [Accepted: 09/11/2024] [Indexed: 10/18/2024]
Abstract
Here, we investigate the contribution of long-term hematopoietic stem cells (HSCsLT) to trained immunity (TI) in the setting of chronic autoimmune disease. Using a mouse model of systemic lupus erythematosus (SLE), we show that bone marrow-derived macrophages (BMDMs) from autoimmune mice exhibit hallmark features of TI, including increased Mycobacterium avium killing and inflammatory cytokine production, which are mechanistically linked to increased glycolytic metabolism. We show that HSCs from autoimmune mice constitute a transplantable, long-term reservoir for macrophages that exhibit the functional properties of TI. However, these BMDMs exhibit reduced glycolytic activity and chromatin accessibility at metabolic genes while retaining elevated expression of TI-associated transcriptional regulators. Hence, HSC exposed to autoimmune inflammation can give rise to macrophages in which the functional and metabolic properties of TI are decoupled. Our data support a model in which TI is characterized by a spectrum of molecular and metabolic states driving augmented immune function.
Collapse
Affiliation(s)
- Taylor S Mills
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Immunology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Bailee Kain
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Matt A Burchill
- Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Etienne Danis
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Erin D Lucas
- Immunology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Rachel Culp-Hill
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Courtney M Cowan
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Immunology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Wolfgang E Schleicher
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sweta B Patel
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Brandon T Tran
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Ruoqiong Cao
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Andrew Goodspeed
- Department of Biomedical Informatics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Sarah Ferrara
- University of Colorado Comprehensive Cancer Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Shaun Bevers
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Beth A Jirón Tamburini
- Immunology Graduate Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Division of Gastroenterology and Hepatology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - James R Roede
- Department of Pharmaceutical Sciences, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of Colorado, Aurora, CO 80045, USA
| | - Angelo D'Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Katherine Y King
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX 77030, USA
| | - Eric M Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA; Department of Immunology and Microbiology, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA.
| |
Collapse
|
22
|
Kapadia CD, Williams N, Dawson KJ, Watson C, Yousefzadeh MJ, Le D, Nyamondo K, Cagan A, Waldvogel S, De La Fuente J, Leongamornlert D, Mitchell E, Florez MA, Aguilar R, Martell A, Guzman A, Harrison D, Niedernhofer LJ, King KY, Campbell PJ, Blundell J, Goodell MA, Nangalia J. Clonal dynamics and somatic evolution of haematopoiesis in mouse. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.17.613129. [PMID: 39345649 PMCID: PMC11429886 DOI: 10.1101/2024.09.17.613129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Haematopoietic stem cells maintain blood production throughout life. While extensively characterised using the laboratory mouse, little is known about how the population is sustained and evolves with age. We isolated stem cells and progenitors from young and old mice, identifying 221,890 somatic mutations genome-wide in 1845 single cell-derived colonies, and used phylogenetic analysis to infer the ontogeny and population dynamics of the stem cell pool. Mouse stem cells and progenitors accrue ~45 somatic mutations per year, a rate only about 2-fold greater than human progenitors despite the vastly different organismal sizes and lifespans. Phylogenetic patterns reveal that stem and multipotent progenitor cell pools are both established during embryogenesis, after which they independently self-renew in parallel over life. The stem cell pool grows steadily over the mouse lifespan to approximately 70,000 cells, self-renewing about every six weeks. Aged mice did not display the profound loss of stem cell clonal diversity characteristic of human haematopoietic ageing. However, targeted sequencing revealed small, expanded clones in the context of murine ageing, which were larger and more numerous following haematological perturbations and exhibited a selection landscape similar to humans. Our data illustrate both conserved features of population dynamics of blood and distinct patterns of age-associated somatic evolution in the short-lived mouse.
Collapse
Affiliation(s)
- Chiraag D. Kapadia
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | | | - Kevin J. Dawson
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
| | - Caroline Watson
- Early Cancer Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Matthew J. Yousefzadeh
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
- Columbia Center for Translational Immunology, Columbia Center for Human Longevity, Department of Medicine, Columbia University Medical Center, New York, NY, USA
| | - Duy Le
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Kudzai Nyamondo
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
| | - Alex Cagan
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Departments of Genetics, Pathology & Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - Sarah Waldvogel
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Josephine De La Fuente
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | | | - Emily Mitchell
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| | - Marcus A. Florez
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | - Rogelio Aguilar
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Alejandra Martell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Anna Guzman
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | | | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, MN, USA
| | - Katherine Y. King
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX, USA
| | | | - Jamie Blundell
- Early Cancer Institute, University of Cambridge, Cambridge Biomedical Campus, Cambridge, UK
| | - Margaret A. Goodell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Jyoti Nangalia
- Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, UK
- Wellcome-MRC Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge, UK
- Department of Haematology, University of Cambridge, Cambridge, UK
| |
Collapse
|
23
|
Aliazis K, Yenyuwadee S, Phikulsod P, Boussiotis VA. Emergency myelopoiesis in solid cancers. Br J Haematol 2024; 205:798-811. [PMID: 39044285 DOI: 10.1111/bjh.19656] [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: 05/13/2024] [Accepted: 07/09/2024] [Indexed: 07/25/2024]
Abstract
Cells of the innate and adaptive immune systems are the progeny of haematopoietic stem and progenitor cells (HSPCs). During steady-state myelopoiesis, HSPC undergo differentiation and proliferation but are called to respond directly and acutely to various signals that lead to emergency myelopoiesis, including bone marrow ablation, infections, and sterile inflammation. There is extensive evidence that many solid tumours have the potential to secrete classical myelopoiesis-promoting growth factors and other products able to mimic emergency haematopoiesis, and to aberrantly re-direct myeloid cell development into immunosuppressive cells with tumour promoting properties. Here, we summarize the current literature regarding the effects of solid cancers on HSPCs function and discuss how these effects might shape antitumour responses via a mechanism initiated at a site distal from the tumour microenvironment.
Collapse
Affiliation(s)
- Konstantinos Aliazis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
| | - Sasitorn Yenyuwadee
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Department of Dermatology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Ployploen Phikulsod
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Vassiliki A Boussiotis
- Department of Hematology-Oncology, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA
- Cancer Center, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
24
|
Thompson Z, Anderson GA, Hernandez M, Alfaro Quinde C, Marchione A, Rodriguez M, Gabriel S, Binder V, Taylor AM, Kathrein KL. Ing4-deficiency promotes a quiescent yet transcriptionally poised state in hematopoietic stem cells. iScience 2024; 27:110521. [PMID: 39175773 PMCID: PMC11340613 DOI: 10.1016/j.isci.2024.110521] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 05/14/2024] [Accepted: 07/12/2024] [Indexed: 08/24/2024] Open
Abstract
Defining the mechanisms that regulate stem cell maintenance, proliferation, and differentiation is critical for identifying therapies for improving stem cell function under stress. Here, we have identified the tumor suppressor, inhibitor of growth 4 (Ing4), as a critical regulator of hematopoietic stem cell (HSC) homeostasis. Cancer cell line models with Ing4 deficiency have shown that Ing4 functions as a tumor suppressor, in part, due to Ing4-mediated regulation of several major signaling pathways, including c-Myc. In HSCs, we show Ing4 deficiency promotes gene expression signatures associated with activation, yet HSCs are arrested in G0, expressing several markers of quiescence. Functionally, Ing4-deficient HSCs demonstrate robust regenerative capacity following transplantation. Our findings suggest Ing4 deficiency promotes a poised state in HSCs, where they appear transcriptionally primed for activation but remain in a resting state. Our model provides key tools for further identification and characterization of pathways that control quiescence and self-renewal in HSCs.
Collapse
Affiliation(s)
- Zanshé Thompson
- University of South Carolina, Department of Biomedical Engineering, Columbia, SC, USA
| | - Georgina A. Anderson
- University of South Carolina, Department of Biological Sciences, Columbia, SC, USA
| | - Marco Hernandez
- University of South Carolina, Department of Biological Sciences, Columbia, SC, USA
| | - Carlos Alfaro Quinde
- University of South Carolina, Department of Biological Sciences, Columbia, SC, USA
| | - Alissa Marchione
- University of South Carolina, Department of Biological Sciences, Columbia, SC, USA
| | - Melanie Rodriguez
- University of South Carolina, Department of Biological Sciences, Columbia, SC, USA
| | - Seth Gabriel
- University of South Carolina, Department of Biological Sciences, Columbia, SC, USA
| | - Vera Binder
- Department of Hematology and Oncology, Dr. von Hauner Children’s Hospital, Ludwig-Maximilians University, 80539 Munich, Germany
| | - Alison M. Taylor
- Columbia University Medical Center, Department of Pathology and Cell Biology, Herbert Irving Comprehensive Cancer Center, New York, NY 10032, USA
| | - Katie L. Kathrein
- University of South Carolina, Department of Biological Sciences, Columbia, SC, USA
| |
Collapse
|
25
|
Navarro-Bailón A, López-Parra M, Veiga-Vaz Á, Villarón EM, Díez-Campelo M, Martín AÁ, Pérez-López E, Cabrero M, Vázquez L, López-Corral L, Sánchez-Guijo F. Treatment of post-allogeneic hematopoietic stem cell transplant cytopenias with sequential doses of multipotent mesenchymal stromal/stem cells. Cytotherapy 2024; 26:806-812. [PMID: 38727653 DOI: 10.1016/j.jcyt.2024.04.006] [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: 11/29/2023] [Revised: 04/08/2024] [Accepted: 04/15/2024] [Indexed: 06/01/2024]
Abstract
BACKGROUND AIMS Cytopenias after allogeneic stem cell transplantation (allo-SCT) are a common complication, the underlying pathogenic mechanisms of which remain incompletely understood. Multipotent mesenchymal stromal/stem cell (MSC) therapy has been successfully employed in the treatment of immune-related disorders and can aid in the restoration of the hematopoietic niche. METHODS A phase II clinical trial to assess the efficacy and safety of administering four sequential doses of ex-vivo expanded bone marrow MSCs from a third-party donor to patients with persistent severe cytopenias after allo-SCT was performed. RESULTS The overall response rate on day 90 was 75% among the 27 evaluable patients (comprising 12 complete responses, 8 partial responses, and 7 with no response). The median time to respond was 14.5 days. Responses were observed across different profiles, including single or multiple affected lineages, primary or secondary timing, and potential immune-mediated or post-infectious pathophysiology versus idiopathic origin. With a median follow-up for surviving patients of 85 months after MSC infusion, 53% of patients are alive. Notably, no adverse events related to MSC therapy were reported. CONCLUSIONS In summary, the sequential infusion of third-party MSCs emerges as a viable and safe therapeutic option, exhibiting potential benefits for patients experiencing cytopenias following allo-SCT.
Collapse
Affiliation(s)
- Almudena Navarro-Bailón
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Faculty of Medicine, University of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain.
| | - Miriam López-Parra
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain
| | - Álvaro Veiga-Vaz
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain
| | - Eva María Villarón
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain
| | - María Díez-Campelo
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Faculty of Medicine, University of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain; Biomedical Research Networking Center for Cancer (CIBERONC), ISCIII, Spain
| | - Ana África Martín
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain
| | - Estefanía Pérez-López
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain
| | - Mónica Cabrero
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain
| | - Lourdes Vázquez
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Faculty of Medicine, University of Salamanca, Salamanca, Spain
| | - Lucía López-Corral
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Faculty of Medicine, University of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain; Biomedical Research Networking Center for Cancer (CIBERONC), ISCIII, Spain
| | - Fermín Sánchez-Guijo
- Hematology Department, Cell Therapy Area, IBSAL-University Hospital of Salamanca, Salamanca, Spain; Faculty of Medicine, University of Salamanca, Salamanca, Spain; Network Center for Regenerative Medicine and Cellular Therapy of Castilla y León, Spain; RICORS of Advanced Therapies (TERAV), ISCIII, Spain; Biomedical Research Networking Center for Cancer (CIBERONC), ISCIII, Spain
| |
Collapse
|
26
|
Mincham KT, Lauzon-Joset JF, Read JF, Holt PG, Stumbles PA, Strickland DH. Mapping Lung Hematopoietic Progenitors: Developmental Kinetics and Response to Influenza A Infection. Am J Respir Cell Mol Biol 2024; 71:219-228. [PMID: 38669465 DOI: 10.1165/rcmb.2023-0395oc] [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: 11/13/2023] [Accepted: 04/26/2024] [Indexed: 04/28/2024] Open
Abstract
The bone marrow is a specialized niche responsible for the maintenance of hematopoietic stem and progenitor cells during homeostasis and inflammation. Recent studies, however, have extended this essential role to the extramedullary and extravascular lung microenvironment. Here, we provide further evidence for a reservoir of hematopoietic stem and progenitor cells within the lung from Embryonic Day 18.5 until adulthood. These lung progenitors display distinct microenvironment-specific developmental kinetics compared with their bone marrow counterparts, exemplified by a rapid shift from a common myeloid to a megakaryocyte-erythrocyte progenitor-dominated niche with increasing age. In adult mice, influenza A viral infection results in a transient reduction in multipotent progenitors within the lungs, with a parallel increase in downstream granulocyte-monocyte progenitors and dendritic cell populations associated with acute viral infections. Our findings suggest that lung hematopoietic progenitors play a role in reestablishing immunological homeostasis in the respiratory mucosa, which may have significant clinical implications for maintaining pulmonary health after inflammatory perturbation.
Collapse
Affiliation(s)
- Kyle T Mincham
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Jean-Francois Lauzon-Joset
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
- Centre de Recherche de l'Institut de Cardiologie et de Pneumologie de Québec, Université Laval, Québec, Québec, Canada
| | - James F Read
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
- Asthma and Airway Disease Research Center, University of Arizona, Tucson, Arizona
| | - Patrick G Holt
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
- Child Health Research Centre, University of Queensland, Brisbane, Queensland, Australia; and
| | - Philip A Stumbles
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
- Medical, Molecular and Forensic Sciences, Murdoch University, Perth, Western Australia, Australia
| | - Deborah H Strickland
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, Nedlands, Western Australia, Australia
| |
Collapse
|
27
|
Jakobsen NA, Turkalj S, Zeng AGX, Stoilova B, Metzner M, Rahmig S, Nagree MS, Shah S, Moore R, Usukhbayar B, Angulo Salazar M, Gafencu GA, Kennedy A, Newman S, Kendrick BJL, Taylor AH, Afinowi-Luitz R, Gundle R, Watkins B, Wheway K, Beazley D, Murison A, Aguilar-Navarro AG, Flores-Figueroa E, Dakin SG, Carr AJ, Nerlov C, Dick JE, Xie SZ, Vyas P. Selective advantage of mutant stem cells in human clonal hematopoiesis is associated with attenuated response to inflammation and aging. Cell Stem Cell 2024; 31:1127-1144.e17. [PMID: 38917807 PMCID: PMC11512683 DOI: 10.1016/j.stem.2024.05.010] [Citation(s) in RCA: 26] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 01/29/2024] [Accepted: 05/30/2024] [Indexed: 06/27/2024]
Abstract
Clonal hematopoiesis (CH) arises when hematopoietic stem cells (HSCs) acquire mutations, most frequently in the DNMT3A and TET2 genes, conferring a competitive advantage through mechanisms that remain unclear. To gain insight into how CH mutations enable gradual clonal expansion, we used single-cell multi-omics with high-fidelity genotyping on human CH bone marrow (BM) samples. Most of the selective advantage of mutant cells occurs within HSCs. DNMT3A- and TET2-mutant clones expand further in early progenitors, while TET2 mutations accelerate myeloid maturation in a dose-dependent manner. Unexpectedly, both mutant and non-mutant HSCs from CH samples are enriched for inflammatory and aging transcriptomic signatures, compared with HSCs from non-CH samples, revealing a non-cell-autonomous effect. However, DNMT3A- and TET2-mutant HSCs have an attenuated inflammatory response relative to wild-type HSCs within the same sample. Our data support a model whereby CH clones are gradually selected because they are resistant to the deleterious impact of inflammation and aging.
Collapse
Affiliation(s)
- Niels Asger Jakobsen
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK; Oxford Centre for Haematology, NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Sven Turkalj
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK; Oxford Centre for Haematology, NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Andy G X Zeng
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Bilyana Stoilova
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Marlen Metzner
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Susann Rahmig
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Murtaza S Nagree
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Sayyam Shah
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Rachel Moore
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Batchimeg Usukhbayar
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Mirian Angulo Salazar
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Grigore-Aristide Gafencu
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Alison Kennedy
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK; Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Simon Newman
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK; Nuffield Orthopaedic Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Benjamin J L Kendrick
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK; Nuffield Orthopaedic Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Adrian H Taylor
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK; Nuffield Orthopaedic Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Rasheed Afinowi-Luitz
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK; Nuffield Orthopaedic Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Roger Gundle
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK; Nuffield Orthopaedic Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Bridget Watkins
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Kim Wheway
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Debra Beazley
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Alex Murison
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Alicia G Aguilar-Navarro
- Unidad de Investigación Médica en Enfermedades Oncológicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Eugenia Flores-Figueroa
- Unidad de Investigación Médica en Enfermedades Oncológicas, Centro Médico Nacional Siglo XXI, Instituto Mexicano del Seguro Social, Mexico City, Mexico
| | - Stephanie G Dakin
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK
| | - Andrew J Carr
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Botnar Research Centre, University of Oxford, Oxford, UK; Nuffield Orthopaedic Centre, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Claus Nerlov
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - John E Dick
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada
| | - Stephanie Z Xie
- Princess Margaret Cancer Centre, University Health Network, Toronto, ON, Canada
| | - Paresh Vyas
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK; Oxford Centre for Haematology, NIHR Oxford Biomedical Research Centre, Oxford, UK; Department of Haematology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| |
Collapse
|
28
|
Schleicher WE, Hoag B, De Dominici M, DeGregori J, Pietras EM. CHIP: a clonal odyssey of the bone marrow niche. J Clin Invest 2024; 134:e180068. [PMID: 39087468 PMCID: PMC11290965 DOI: 10.1172/jci180068] [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] [Indexed: 08/02/2024] Open
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is characterized by the selective expansion of hematopoietic stem and progenitor cells (HSPCs) carrying somatic mutations. While CHIP is typically asymptomatic, it has garnered substantial attention due to its association with the pathogenesis of multiple disease conditions, including cardiovascular disease (CVD) and hematological malignancies. In this Review, we will discuss seminal and recent studies that have advanced our understanding of mechanisms that drive selection for mutant HSPCs in the BM niche. Next, we will address recent studies evaluating potential relationships between the clonal dynamics of CHIP and hematopoietic development across the lifespan. Next, we will examine the roles of systemic factors that can influence hematopoietic stem cell (HSC) fitness, including inflammation, and exposures to cytotoxic agents in driving selection for CHIP clones. Furthermore, we will consider how - through their impact on the BM niche - lifestyle factors, including diet, exercise, and psychosocial stressors, might contribute to the process of somatic evolution in the BM that culminates in CHIP. Finally, we will review the role of old age as a major driver of selection in CHIP.
Collapse
Affiliation(s)
| | - Bridget Hoag
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - Marco De Dominici
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | - James DeGregori
- Division of Hematology, Department of Medicine, and
- Department of Biochemistry and Molecular Genetics, University of Colorado Anschutz Medical Campus, Aurora, Colorado, USA
| | | |
Collapse
|
29
|
Pendse S, Loeffler D. Decoding Clonal Hematopoiesis: Emerging Themes and Novel Mechanistic Insights. Cancers (Basel) 2024; 16:2634. [PMID: 39123361 PMCID: PMC11311828 DOI: 10.3390/cancers16152634] [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: 06/27/2024] [Revised: 07/22/2024] [Accepted: 07/22/2024] [Indexed: 08/12/2024] Open
Abstract
Clonal hematopoiesis (CH), the relative expansion of mutant clones, is derived from hematopoietic stem cells (HSCs) with acquired somatic or cytogenetic alterations that improve cellular fitness. Individuals with CH have a higher risk for hematological and non-hematological diseases, such as cardiovascular disease, and have an overall higher mortality rate. Originally thought to be restricted to a small fraction of elderly people, recent advances in single-cell sequencing and bioinformatics have revealed that CH with multiple expanded mutant clones is universal in the elderly population. Just a few years ago, phylogenetic reconstruction across the human lifespan and novel sensitive sequencing techniques showed that CH can start earlier in life, decades before it was thought possible. These studies also suggest that environmental factors acting through aberrant inflammation might be a common theme promoting clonal expansion and disease progression. However, numerous aspects of this phenomenon remain to be elucidated and the precise mechanisms, context-specific drivers, and pathways of clonal expansion remain to be established. Here, we review our current understanding of the cellular mechanisms driving CH and specifically focus on how pro-inflammatory factors affect normal and mutant HSC fates to promote clonal selection.
Collapse
Affiliation(s)
- Shalmali Pendse
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Comprehensive Cancer Center, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Pathology & Laboratory Medicine, The University of Tennessee, Memphis, TN 37996, USA
| | - Dirk Loeffler
- Department of Hematology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Comprehensive Cancer Center, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
- Department of Pathology & Laboratory Medicine, The University of Tennessee, Memphis, TN 37996, USA
| |
Collapse
|
30
|
Cao R, Thatavarty A, King KY. Forged in the fire: Lasting impacts of inflammation on hematopoietic progenitors. Exp Hematol 2024; 134:104215. [PMID: 38580008 DOI: 10.1016/j.exphem.2024.104215] [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: 01/11/2024] [Revised: 03/26/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
Quiescence and differentiation of hematopoietic stem and progenitor cells (HSPC) can be modified by systemic inflammatory cues. Such cues can not only yield short-term changes in HSPCs such as in supporting emergency granulopoiesis but can also promote lasting influences on the HSPC compartment. First, inflammation can be a driver for clonal expansion, promoting clonal hematopoiesis for certain mutant clones, reducing overall clonal diversity, and reshaping the composition of the HSPC pool with significant health consequences. Second, inflammation can generate lasting cell-autonomous changes in HSPCs themselves, leading to changes in the epigenetic state, metabolism, and function of downstream innate immune cells. This concept, termed "trained immunity," suggests that inflammatory stimuli can alter subsequent immune responses leading to improved innate immunity or, conversely, autoimmunity. Both of these concepts have major implications in human health. Here we reviewed current literature about the lasting effects of inflammation on the HSPC compartment and opportunities for future advancement in this fast-developing field.
Collapse
Affiliation(s)
- Ruoqiong Cao
- Department of Pediatrics - Division of Infectious Disease, Texas Children's Hospital, Baylor College of Medicine, Houston, TX; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX
| | - Apoorva Thatavarty
- Department of Pediatrics - Division of Infectious Disease, Texas Children's Hospital, Baylor College of Medicine, Houston, TX; Graduate Program in Genetics and Genomics, Baylor College of Medicine, Houston, Texas; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX
| | - Katherine Y King
- Department of Pediatrics - Division of Infectious Disease, Texas Children's Hospital, Baylor College of Medicine, Houston, TX; Graduate Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX; Medical Scientist Training Program, Baylor College of Medicine, Houston, TX; Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX.
| |
Collapse
|
31
|
Brix A, Belleri L, Pezzotta A, Pettinato E, Mazzola M, Zoccolillo M, Marozzi A, Monteiro R, Del Bene F, Mortellaro A, Pistocchi A. ADA2 regulates inflammation and hematopoietic stem cell emergence via the A 2bR pathway in zebrafish. Commun Biol 2024; 7:615. [PMID: 38777862 PMCID: PMC11111730 DOI: 10.1038/s42003-024-06286-3] [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: 09/28/2023] [Accepted: 05/03/2024] [Indexed: 05/25/2024] Open
Abstract
Deficiency of adenosine deaminase 2 (DADA2) is an inborn error of immunity caused by loss-of-function mutations in the adenosine deaminase 2 (ADA2) gene. Clinical manifestations of DADA2 include vasculopathy and immuno-hematological abnormalities, culminating in bone marrow failure. A major gap exists in our knowledge of the regulatory functions of ADA2 during inflammation and hematopoiesis, mainly due to the absence of an ADA2 orthologue in rodents. Exploring these mechanisms is essential for understanding disease pathology and developing new treatments. Zebrafish possess two ADA2 orthologues, cecr1a and cecr1b, with the latter showing functional conservation with human ADA2. We establish a cecr1b-loss-of-function zebrafish model that recapitulates the immuno-hematological and vascular manifestations observed in humans. Loss of Cecr1b disrupts hematopoietic stem cell specification, resulting in defective hematopoiesis. This defect is caused by induced inflammation in the vascular endothelium. Blocking inflammation, pharmacological modulation of the A2r pathway, or the administration of the recombinant human ADA2 corrects these defects, providing insights into the mechanistic link between ADA2 deficiency, inflammation and immuno-hematological abnormalities. Our findings open up potential therapeutic avenues for DADA2 patients.
Collapse
Affiliation(s)
- Alessia Brix
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Laura Belleri
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
- Department of Development, Institut de la Vision, 17 Rue Moreau, 75012, Paris, France
| | - Alex Pezzotta
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Emanuela Pettinato
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
| | - Mara Mazzola
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Matteo Zoccolillo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy
| | - Anna Marozzi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy
| | - Rui Monteiro
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, Edgbaston, B15 2TTB, UK
| | - Filippo Del Bene
- Department of Development, Institut de la Vision, 17 Rue Moreau, 75012, Paris, France
| | - Alessandra Mortellaro
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, via Olgettina 60, 20132, Milan, Italy.
| | - Anna Pistocchi
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, L.I.T.A., via Fratelli Cervi 93, Segrate, 20054, Milan, Italy.
| |
Collapse
|
32
|
Stevens CS, Carmichael J, Watkinson R, Kowdle S, Reis RA, Hamane K, Jang J, Park A, Pernet O, Khamaikawin W, Hong P, Thibault P, Gowlikar A, An DS, Lee B. A temperature-sensitive and interferon-silent Sendai virus vector for CRISPR-Cas9 delivery and gene editing in primary human cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.03.592383. [PMID: 38746439 PMCID: PMC11092779 DOI: 10.1101/2024.05.03.592383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
The transformative potential of gene editing technologies hinges on the development of safe and effective delivery methods. In this study, we developed a temperature-sensitive and interferon-silent Sendai virus (ts SeV) as a novel delivery vector for CRISPR-Cas9 and for efficient gene editing in sensitive human cell types without inducing IFN responses. ts SeV demonstrates unprecedented transduction efficiency in human CD34+ hematopoietic stem and progenitor cells (HSPCs) including transduction of the CD34+/CD38-/CD45RA-/CD90+(Thy1+)/CD49fhigh stem cell enriched subpopulation. The frequency of CCR5 editing exceeded 90% and bi-allelic CCR5 editing exceeded 70% resulting in significant inhibition of HIV-1 infection in primary human CD14+ monocytes. These results demonstrate the potential of the ts SeV platform as a safe, efficient, and flexible addition to the current gene-editing tool delivery methods, which may help to further expand the possibilities in personalized medicine and the treatment of genetic disorders.
Collapse
Affiliation(s)
- Christian S Stevens
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Jillian Carmichael
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Ruth Watkinson
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Shreyas Kowdle
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Rebecca A Reis
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Kory Hamane
- UCLA School of Nursing, Los Angeles, California, 90095
- UCLA AIDS Institute, Los Angeles, California, 90095
| | - Jason Jang
- UCLA School of Nursing, Los Angeles, California, 90095
- UCLA AIDS Institute, Los Angeles, California, 90095
| | - Arnold Park
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Olivier Pernet
- UCLA School of Nursing, Los Angeles, California, 90095
- UCLA AIDS Institute, Los Angeles, California, 90095
| | - Wannisa Khamaikawin
- UCLA School of Nursing, Los Angeles, California, 90095
- UCLA AIDS Institute, Los Angeles, California, 90095
| | - Patrick Hong
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Patricia Thibault
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Aditya Gowlikar
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| | - Dong Sung An
- UCLA School of Nursing, Los Angeles, California, 90095
- UCLA AIDS Institute, Los Angeles, California, 90095
| | - Benhur Lee
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029
| |
Collapse
|
33
|
Sadeghi M, Divangahi M. Discovering adaptive features of innate immune memory. Immunol Rev 2024; 323:186-196. [PMID: 38563500 DOI: 10.1111/imr.13328] [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: 03/04/2024] [Accepted: 03/11/2024] [Indexed: 04/04/2024]
Abstract
Conventionally, it was thought that innate immunity operated through a simple system of nonspecific responses to an insult. However, this perspective now seems overly simplistic. It has become evident that intricate cooperation and networking among various cells, receptors, signaling pathways, and protein complexes are essential for regulating and defining the overall activation status of the immune response, where the distinction between innate and adaptive immunity becomes ambiguous. Given the evolutionary timeline of vertebrates and the success of plants and invertebrates which depend solely on innate immunity, immune memory cannot be considered an innovation of only the lymphoid lineage. Indeed, the evolutionary innate immune memory program is a conserved mechanism whereby innate immune cells can induce a heightened response to a secondary stimulus due to metabolic and epigenetic reprogramming. Importantly, the longevity of this memory phenotype can be attributed to the reprogramming of self-renewing hematopoietic stem cells (HSCs) in the bone marrow, which is subsequently transmitted to lineage-committed innate immune cells. HSCs reside within a complex regulated network of immune and stromal cells that govern their two primary functions: self-renewal and differentiation. In this review, we delve into the emerging cellular and molecular mechanisms as well as metabolic pathways of innate memory in HSCs, which harbor substantial therapeutic promise.
Collapse
Affiliation(s)
- Mina Sadeghi
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| | - Maziar Divangahi
- Department of Medicine, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Microbiology & Immunology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
- Department of Pathology, McGill University Health Centre, McGill International TB Centre, Meakins-Christie Laboratories, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
34
|
Cook CV, Lighty AM, Smith BJ, Ford Versypt AN. A review of mathematical modeling of bone remodeling from a systems biology perspective. FRONTIERS IN SYSTEMS BIOLOGY 2024; 4:1368555. [PMID: 40012834 PMCID: PMC11864782 DOI: 10.3389/fsysb.2024.1368555] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Bone remodeling is an essential, delicately balanced physiological process of coordinated activity of bone cells that remove and deposit new bone tissue in the adult skeleton. Due to the complex nature of this process, many mathematical models of bone remodeling have been developed. Each of these models has unique features, but they have underlying patterns. In this review, the authors highlight the important aspects frequently found in mathematical models for bone remodeling and discuss how and why these aspects are included when considering the physiology of the bone basic multicellular unit, which is the term used for the collection of cells responsible for bone remodeling. The review also emphasizes the view of bone remodeling from a systems biology perspective. Understanding the systemic mechanisms involved in remodeling will help provide information on bone pathology associated with aging, endocrine disorders, cancers, and inflammatory conditions and enhance systems pharmacology. Furthermore, some features of the bone remodeling cycle and interactions with other organ systems that have not yet been modeled mathematically are discussed as promising future directions in the field.
Collapse
Affiliation(s)
- Carley V. Cook
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Ariel M. Lighty
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
| | - Brenda J. Smith
- Indiana Center for Musculoskeletal Health, School of Medicine, Indiana University, Indianapolis, IN, United States
- Department of Obstetrics and Gynecology, School of Medicine, Indiana University, Indianapolis, IN, United States
| | - Ashlee N. Ford Versypt
- Department of Chemical and Biological Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
- Department of Biomedical Engineering, University at Buffalo, The State University of New York, Buffalo, NY, United States
- Institute for Artificial Intelligence and Data Science, University at Buffalo, The State University of New York, Buffalo, NY, United States
| |
Collapse
|
35
|
Chen DW, Fan JM, Schrey JM, Mitchell DV, Jung SK, Hurwitz SN, Perez EB, Muraro MJ, Carroll M, Taylor DM, Kurre P. Inflammatory recruitment of healthy hematopoietic stem and progenitor cells in the acute myeloid leukemia niche. Leukemia 2024; 38:741-750. [PMID: 38228679 PMCID: PMC10997516 DOI: 10.1038/s41375-024-02136-7] [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: 08/03/2023] [Revised: 12/21/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Abstract
Inflammation in the bone marrow (BM) microenvironment is a constitutive component of leukemogenesis in acute myeloid leukemia (AML). Current evidence suggests that both leukemic blasts and stroma secrete proinflammatory factors that actively suppress the function of healthy hematopoietic stem and progenitor cells (HSPCs). HSPCs are also cellular components of the innate immune system, and we reasoned that they may actively propagate the inflammation in the leukemic niche. In two separate congenic models of AML we confirm by evaluation of the BM plasma secretome and HSPC-selective single-cell RNA sequencing (scRNA-Seq) that multipotent progenitors and long-lived stem cells adopt inflammatory gene expression programs, even at low leukemic infiltration of the BM. In particular, we observe interferon gamma (IFN-γ) pathway activation, along with secretion of its chemokine target, CXCL10. We show that AML-derived nanometer-sized extracellular vesicles (EVAML) are sufficient to trigger this inflammatory HSPC response, both in vitro and in vivo. Altogether, our studies indicate that HSPCs are an unrecognized component of the inflammatory adaptation of the BM by leukemic cells. The pro-inflammatory conversion and long-lived presence of HSPCs in the BM along with their regenerative re-expansion during remission may impact clonal selection and disease evolution.
Collapse
Affiliation(s)
- Ding-Wen Chen
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Jian-Meng Fan
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Julie M Schrey
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Dana V Mitchell
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Seul K Jung
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Stephanie N Hurwitz
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | | | | | - Martin Carroll
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Deanne M Taylor
- Department of Biomedical and Health Informatics, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Peter Kurre
- Comprehensive Bone Marrow Failure Center, Division of Hematology, Department of Pediatrics, Children's Hospital of Philadelphia, Philadelphia, PA, USA.
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
36
|
Wang B, Reville PK, Yassouf MY, Jelloul FZ, Ly C, Desai PN, Wang Z, Borges P, Veletic I, Dasdemir E, Burks JK, Tang G, Guo S, Garza AI, Nasnas C, Vaughn NR, Baran N, Deng Q, Matthews J, Gunaratne PH, Antunes DA, Ekmekcioglu S, Sasaki K, Garcia MB, Cuglievan B, Hao D, Daver N, Green MR, Konopleva M, Futreal A, Post SM, Abbas HA. Comprehensive characterization of IFNγ signaling in acute myeloid leukemia reveals prognostic and therapeutic strategies. Nat Commun 2024; 15:1821. [PMID: 38418901 PMCID: PMC10902356 DOI: 10.1038/s41467-024-45916-6] [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: 07/07/2023] [Accepted: 02/05/2024] [Indexed: 03/02/2024] Open
Abstract
Interferon gamma (IFNγ) is a critical cytokine known for its diverse roles in immune regulation, inflammation, and tumor surveillance. However, while IFNγ levels were elevated in sera of most newly diagnosed acute myeloid leukemia (AML) patients, its complex interplay in AML remains insufficiently understood. We aim to characterize these complex interactions through comprehensive bulk and single-cell approaches in bone marrow of newly diagnosed AML patients. We identify monocytic AML as having a unique microenvironment characterized by IFNγ producing T and NK cells, high IFNγ signaling, and immunosuppressive features. IFNγ signaling score strongly correlates with venetoclax resistance in primary AML patient cells. Additionally, IFNγ treatment of primary AML patient cells increased venetoclax resistance. Lastly, a parsimonious 47-gene IFNγ score demonstrates robust prognostic value. In summary, our findings suggest that inhibiting IFNγ is a potential treatment strategy to overcoming venetoclax resistance and immune evasion in AML patients.
Collapse
Affiliation(s)
- Bofei Wang
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick K Reville
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mhd Yousuf Yassouf
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Fatima Z Jelloul
- Department of Hematopathology, Division of Pathology & Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Christopher Ly
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Poonam N Desai
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- School of Biomedical Informatics, The University of Texas Health Science Center, Houston, TX, USA
| | - Zhe Wang
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pamella Borges
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Ivo Veletic
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Enes Dasdemir
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Jared K Burks
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guilin Tang
- Department of Hematopathology, Division of Pathology & Laboratory Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shengnan Guo
- School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Araceli Isabella Garza
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cedric Nasnas
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicole R Vaughn
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalia Baran
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Qing Deng
- Department of Lymphoma & Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jairo Matthews
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Preethi H Gunaratne
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Dinler A Antunes
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Suhendan Ekmekcioglu
- Department of Melanoma Medical Oncology, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Miriam B Garcia
- Department of Pediatrics, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Branko Cuglievan
- Department of Pediatrics, Division of Pediatrics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dapeng Hao
- School of Basic Medical Sciences, Harbin Medical University, Harbin, Heilongjiang, China
| | - Naval Daver
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael R Green
- Department of Lymphoma & Myeloma, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Oncology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Andrew Futreal
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sean M Post
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hussein A Abbas
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
37
|
Zhao XC, Ju B, Xiu NN, Sun XY, Meng FJ. When inflammatory stressors dramatically change, disease phenotypes may transform between autoimmune hematopoietic failure and myeloid neoplasms. Front Immunol 2024; 15:1339971. [PMID: 38426096 PMCID: PMC10902444 DOI: 10.3389/fimmu.2024.1339971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 01/24/2024] [Indexed: 03/02/2024] Open
Abstract
Aplastic anemia (AA) and hypoplastic myelodysplastic syndrome are paradigms of autoimmune hematopoietic failure (AHF). Myelodysplastic syndrome and acute myeloid leukemia are unequivocal myeloid neoplasms (MNs). Currently, AA is also known to be a clonal hematological disease. Genetic aberrations typically observed in MNs are detected in approximately one-third of AA patients. In AA patients harboring MN-related genetic aberrations, a poor response to immunosuppressive therapy (IST) and an increased risk of transformation to MNs occurring either naturally or after IST are predicted. Approximately 10%-15% of patients with severe AA transform the disease phenotype to MNs following IST, and in some patients, leukemic transformation emerges during or shortly after IST. Phenotypic transformations between AHF and MNs can occur reciprocally. A fraction of advanced MN patients experience an aplastic crisis during which leukemic blasts are repressed. The switch that shapes the disease phenotype is a change in the strength of extramedullary inflammation. Both AHF and MNs have an immune-active bone marrow (BM) environment (BME). In AHF patients, an inflamed BME can be evoked by infiltrated immune cells targeting neoplastic molecules, which contributes to the BM-specific autoimmune impairment. Autoimmune responses in AHF may represent an antileukemic mechanism, and inflammatory stressors strengthen antileukemic immunity, at least in a significant proportion of patients who have MN-related genetic aberrations. During active inflammatory episodes, normal and leukemic hematopoieses are suppressed, which leads to the occurrence of aplastic cytopenia and leukemic cell regression. The successful treatment of underlying infections mitigates inflammatory stress-related antileukemic activities and promotes the penetration of leukemic hematopoiesis. The effect of IST is similar to that of treating underlying infections. Investigating inflammatory stress-powered antileukemic immunity is highly important in theoretical studies and clinical practice, especially given the wide application of immune-activating agents and immune checkpoint inhibitors in the treatment of hematological neoplasms.
Collapse
Affiliation(s)
- Xi-Chen Zhao
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Bo Ju
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Nuan-Nuan Xiu
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Xiao-Yun Sun
- Department of Hematology, The Central Hospital of Qingdao West Coast New Area, Qingdao, Shandong, China
| | - Fan-Jun Meng
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| |
Collapse
|
38
|
Wijshake T, Rose J, Wang J, Zielke J, Marlar-Pavey M, Chen W, Collins JJ, Agathocleous M. Schistosome Infection Impacts Hematopoiesis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:607-616. [PMID: 38169327 PMCID: PMC10872488 DOI: 10.4049/jimmunol.2300195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 12/06/2023] [Indexed: 01/05/2024]
Abstract
Helminth infections are common in animals. However, the impact of a helminth infection on the function of hematopoietic stem cells (HSCs) and other hematopoietic cells has not been comprehensively defined. In this article, we describe the hematopoietic response to infection of mice with Schistosoma mansoni, a parasitic flatworm that causes schistosomiasis. We analyzed the frequency or number of hematopoietic cell types in the bone marrow, spleen, liver, thymus, and blood and observed multiple hematopoietic changes caused by infection. Schistosome infection impaired bone marrow HSC function after serial transplantation. Functional HSCs were present in the infected liver. Infection blocked bone marrow erythropoiesis and augmented spleen erythropoiesis, observations consistent with the anemia and splenomegaly prevalent in schistosomiasis patients. This work defines the hematopoietic response to schistosomiasis, a debilitating disease afflicting more than 200 million people, and identifies impairments in HSC function and erythropoiesis.
Collapse
Affiliation(s)
- Tobias Wijshake
- Children’s Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Joseph Rose
- Children’s Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jipeng Wang
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
- Current address: State Key Laboratory of Genetic Engineering, School of Life Sciences at Fudan University, Shanghai, China
| | - Jacob Zielke
- Children’s Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX, USA
| | - Madeleine Marlar-Pavey
- Children’s Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Weina Chen
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - James J. Collins
- Department of Pharmacology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Michalis Agathocleous
- Children’s Medical Center Research Institute, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Pediatrics, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
39
|
Long H, Steimle JD, Grisanti Canozo FJ, Kim JH, Li X, Morikawa Y, Park M, Turaga D, Adachi I, Wythe JD, Samee MAH, Martin JF. Endothelial cells adopt a pro-reparative immune responsive signature during cardiac injury. Life Sci Alliance 2024; 7:e202201870. [PMID: 38012001 PMCID: PMC10681909 DOI: 10.26508/lsa.202201870] [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: 12/09/2022] [Revised: 11/11/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023] Open
Abstract
Modulation of the heart's immune microenvironment is crucial for recovery after ischemic events such as myocardial infarction (MI). Endothelial cells (ECs) can have immune regulatory functions; however, interactions between ECs and the immune environment in the heart after MI remain poorly understood. We identified an EC-specific IFN responsive and immune regulatory gene signature in adult and pediatric heart failure (HF) tissues. Single-cell transcriptomic analysis of murine hearts subjected to MI uncovered an EC population (IFN-ECs) with immunologic gene signatures similar to those in human HF. IFN-ECs were enriched in regenerative-stage mouse hearts and expressed genes encoding immune responsive transcription factors (Irf7, Batf2, and Stat1). Single-cell chromatin accessibility studies revealed an enrichment of these TF motifs at IFN-EC signature genes. Expression of immune regulatory ligand genes by IFN-ECs suggests bidirectional signaling between IFN-ECs and macrophages in regenerative-stage hearts. Our data suggest that ECs may adopt immune regulatory signatures after cardiac injury to accompany the reparative response. The presence of these signatures in human HF and murine MI models suggests a potential role for EC-mediated immune regulation in responding to stress induced by acute injury in MI and chronic adverse remodeling in HF.
Collapse
Affiliation(s)
- Hali Long
- Interdepartmental Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Jeffrey D Steimle
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | | | - Jong Hwan Kim
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Cardiomyocyte Renewal Laboratory, The Texas Heart Institute, Houston, TX, USA
| | - Xiao Li
- Cardiomyocyte Renewal Laboratory, The Texas Heart Institute, Houston, TX, USA
| | - Yuka Morikawa
- Cardiomyocyte Renewal Laboratory, The Texas Heart Institute, Houston, TX, USA
| | - Minjun Park
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - Diwakar Turaga
- Section of Critical Care Medicine, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Iki Adachi
- Section of Cardiothoracic Surgery, Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Joshua D Wythe
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
| | - Md Abul Hassan Samee
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
| | - James F Martin
- Interdepartmental Program in Integrative Molecular and Biomedical Sciences, Baylor College of Medicine, Houston, TX, USA
- Department of Integrative Physiology, Baylor College of Medicine, Houston, TX, USA
- Cardiomyocyte Renewal Laboratory, The Texas Heart Institute, Houston, TX, USA
- Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX, USA
- Center for Organ Repair and Renewal, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
40
|
Li J, Malouf C, Miles LA, Willis MB, Pietras EM, King KY. Chronic inflammation can transform the fate of normal and mutant hematopoietic stem cells. Exp Hematol 2023; 127:8-13. [PMID: 37647982 DOI: 10.1016/j.exphem.2023.08.008] [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: 08/02/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/01/2023]
Abstract
Chronic inflammation, although subtle, puts the body in a constant state of alertness and is associated with many diseases, including cancer and cardiovascular diseases. It leads hematopoietic cells to produce and release proinflammatory cytokines, which trigger specific signaling pathways in hematopoietic stem cells (HSCs) that cause changes in proliferation, differentiation, and migration. This response is essential when HSCs are needed to produce specific blood cells to eliminate an intruder, such as a pathogenic virus, but mutant HSCs can use these proinflammatory signals to their advantage and accelerate the development of hematologic disease or malignancy. Understanding this complex process is vital for monitoring and controlling disease progression in patients. In the 2023 International Society for Experimental Hematology winter webinar, Dr. Eric Pietras (University of Colorado Anschutz Medical Campus, United States) and Dr. Katherine Y. King (Baylor College of Medicine, United States) gave a presentation on this topic, which is summarized in this review article.
Collapse
Affiliation(s)
- Jingjing Li
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW, Australia.
| | | | - Linde A Miles
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH; Division of Experimental Hematology & Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Mara B Willis
- Center for Cell and Gene Therapy and Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX
| | - Eric M Pietras
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Katherine Y King
- Center for Cell and Gene Therapy and Department of Pediatrics, Division of Infectious Diseases, Baylor College of Medicine, Houston, TX
| |
Collapse
|
41
|
Fontenay M, Boussaid I, Chapuis N. [Pathophysiology of myelodysplastic syndromes]. Bull Cancer 2023; 110:1097-1105. [PMID: 37423830 DOI: 10.1016/j.bulcan.2023.02.026] [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: 01/09/2023] [Accepted: 02/15/2023] [Indexed: 07/11/2023]
Abstract
During aging, the onset of mutations at low frequency in hematopoietic cells or clonal hematopoiesis of indeterminate significance favors the evolution towards hemopathies such as myelodysplastic syndromes or acute leukemias, but also cardiovascular diseases and other pathologies. Acute or chronic inflammation related to age influences the clonal evolution and the immune response. Conversely, mutated hematopoietic cells create an inflammatory bone marrow environment facilitating their expansion. Various pathophysiological mechanisms depending on the type of mutation produce the diversity of phenotypes. Identifying factors affecting clonal selection is mandatory to improve patient care.
Collapse
Affiliation(s)
- Michaela Fontenay
- Assistance publique-Hôpitaux de Paris, université Paris Cité, hôpital Cochin, laboratoire d'hématologie, Inserm, Institut Cochin, Paris, France.
| | - Ismael Boussaid
- Assistance publique-Hôpitaux de Paris, université Paris Cité, hôpital Cochin, laboratoire d'hématologie, Inserm, Institut Cochin, Paris, France
| | - Nicolas Chapuis
- Assistance publique-Hôpitaux de Paris, université Paris Cité, hôpital Cochin, laboratoire d'hématologie, Inserm, Institut Cochin, Paris, France
| |
Collapse
|
42
|
Xu T, Dai J, Tang L, Sun L, Si L, Guo J. Systemic administration of STING agonist promotes myeloid cells maturation and antitumor immunity through regulating hematopoietic stem and progenitor cell fate. Cancer Immunol Immunother 2023; 72:3491-3505. [PMID: 37550427 PMCID: PMC10991199 DOI: 10.1007/s00262-023-03502-7] [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: 02/15/2023] [Accepted: 07/18/2023] [Indexed: 08/09/2023]
Abstract
STING is a pivotal mediator of effective innate and adaptive anti-tumor immunity; however, intratumoral administration of STING agonists have shown limited therapeutic benefit in clinical trials. The systemic effect of the intravenous delivery of STING agonists in cancer is not well-defined. Here, we demonstrated that systemic administration of STING agonist inhibited melanoma growth, improved inflammatory effector cell infiltration, and induced bone marrow mobilization and extramedullary hematopoiesis, causing widespread changes in immune components in the peripheral blood. The systemically administered STING agonist promoted HSC expansion and influenced lineage fate commitment, which was manifested as the differentiation of HSPCs was skewed toward myeloid cells at the expense of B-cell lymphopoiesis and erythropoiesis. Transcriptome analysis revealed upregulation of myeloid lineage differentiation-related and type I interferon-related genes. This myeloid-biased differentiation promoted the production and maturation of myeloid cells toward an activated phenotype. Furthermore, depletion of Gr-1+ myeloid cells attenuated the anti-tumor immunity of STING agonist. Our findings reveal the anti-tumor mechanism of systemic administration of STING agonist that involves modulating HSPC differentiation and promoting myeloid cells maturation. Our study may help explain the limited clinical activity of STING agonists administered intratumorally.
Collapse
Affiliation(s)
- Tianxiao Xu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, 52# Fucheng Road, Haidian District, Beijing, 100142, China
| | - Jie Dai
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, 52# Fucheng Road, Haidian District, Beijing, 100142, China
| | - Lirui Tang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, 52# Fucheng Road, Haidian District, Beijing, 100142, China
| | - Linzi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, 52# Fucheng Road, Haidian District, Beijing, 100142, China
| | - Lu Si
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, 52# Fucheng Road, Haidian District, Beijing, 100142, China.
| | - Jun Guo
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Melanoma and Sarcoma, Peking University Cancer Hospital and Institute, 52# Fucheng Road, Haidian District, Beijing, 100142, China.
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Genitourinary Oncology, Peking University Cancer Hospital and Institute, 52# Fucheng Road, Haidian District, Beijing, 100142, China.
| |
Collapse
|
43
|
Li F, Chen D, Zeng Q, Du Y. Possible Mechanisms of Lymphopenia in Severe Tuberculosis. Microorganisms 2023; 11:2640. [PMID: 38004652 PMCID: PMC10672989 DOI: 10.3390/microorganisms11112640] [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: 09/14/2023] [Revised: 10/20/2023] [Accepted: 10/24/2023] [Indexed: 11/26/2023] Open
Abstract
Tuberculosis (TB) is a chronic infectious disease caused by Mycobacterium tuberculosis (M. tuberculosis). In lymphopenia, T cells are typically characterized by progressive loss and a decrease in their count results. Lymphopenia can hinder immune responses and lead to systemic immunosuppression, which is strongly associated with mortality. Lymphopenia is a significant immunological abnormality in the majority of patients with severe and advanced TB, and its severity is linked to disease outcomes. However, the underlying mechanism remains unclear. Currently, the research on the pathogenesis of lymphopenia during M. tuberculosis infection mainly focuses on how it affects lymphocyte production, survival, or tissue redistribution. This includes impairing hematopoiesis, inhibiting T-cell proliferation, and inducing lymphocyte apoptosis. In this study, we have compiled the latest research on the possible mechanisms that may cause lymphopenia during M. tuberculosis infection. Lymphopenia may have serious consequences in severe TB patients. Additionally, we discuss in detail potential intervention strategies to prevent lymphopenia, which could help understand TB immunopathogenesis and achieve the goal of preventing and treating severe TB.
Collapse
Affiliation(s)
- Fei Li
- Institute of Pathogen Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China; (D.C.); (Q.Z.); (Y.D.)
| | | | | | | |
Collapse
|
44
|
Bassani B, Cucchiara M, Butera A, Kayali O, Chiesa A, Palano MT, Olmeo F, Gallazzi M, Dellavia CPB, Mortara L, Parisi L, Bruno A. Neutrophils' Contribution to Periodontitis and Periodontitis-Associated Cardiovascular Diseases. Int J Mol Sci 2023; 24:15370. [PMID: 37895050 PMCID: PMC10607037 DOI: 10.3390/ijms242015370] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 10/29/2023] Open
Abstract
Neutrophils represent the primary defense against microbial threats playing a pivotal role in maintaining tissue homeostasis. This review examines the multifaceted involvement of neutrophils in periodontitis, a chronic inflammatory condition affecting the supporting structures of teeth summarizing the contribution of neutrophil dysfunction in periodontitis and periodontal-related comorbidities. Periodontitis, a pathological condition promoted by dysbiosis of the oral microbiota, is characterized by the chronic inflammation of the gingiva and subsequent tissue destruction. Neutrophils are among the first immune cells recruited to the site of infection, releasing antimicrobial peptides, enzymes, and reactive oxygen species to eliminate pathogens. The persistent inflammatory state in periodontitis can lead to aberrant neutrophil activation and a sustained release of proinflammatory mediators, finally resulting in tissue damage, bone resorption, and disease progression. Growing evidence now points to the correlation between periodontitis and systemic comorbidities. Indeed, the release of inflammatory mediators, immune complexes, and oxidative stress by neutrophils, bridge the gap between local and systemic immunity, thus highlighting neutrophils as key players in linking periodontal inflammation to chronic conditions, including cardiovascular diseases, diabetes mellitus, and rheumatoid arthritis. This review underscores the crucial role of neutrophils in the pathogenesis of periodontitis and the complex link between neutrophil dysfunction, local inflammation, and systemic comorbidities. A comprehensive understanding of neutrophil contribution to periodontitis development and their impact on periodontal comorbidities holds significant implications for the management of oral health. Furthermore, it highlights the need for the development of novel approaches aimed at limiting the persistent recruitment and activation of neutrophils, also reducing the impact of periodontal inflammation on broader health contexts, offering promising avenues for improved disease management and patient care.
Collapse
Affiliation(s)
- Barbara Bassani
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
| | - Martina Cucchiara
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| | - Andrea Butera
- Unit of Dental Hygiene, Section of Dentistry, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (A.B.); (A.C.)
| | - Omar Kayali
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
| | - Alessandro Chiesa
- Unit of Dental Hygiene, Section of Dentistry, Department of Clinical, Surgical, Diagnostic and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy; (A.B.); (A.C.)
| | - Maria Teresa Palano
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
| | - Francesca Olmeo
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
| | - Matteo Gallazzi
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
| | | | - Lorenzo Mortara
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| | - Luca Parisi
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy;
| | - Antonino Bruno
- Laboratory of Innate Immunity, Unit of Molecular Pathology, Biochemistry and Immunology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) MultiMedica, 20138 Milan, Italy; (B.B.); (M.C.); (O.K.); (M.T.P.); (F.O.); (M.G.)
- Immunology and General Pathology Laboratory, Department of Biotechnology and Life Sciences, University of Insubria, 21100 Varese, Italy;
| |
Collapse
|
45
|
Glinin TS, Petrova MV, Shcherbinina V, Shubina AN, Dukelskaya AV, Starshova PV, Mamontova V, Burnusuz A, Godunova AO, Romashchenko AV, Moshkin MP, Khaitovich P, Daev EV. Pheromone of grouped female mice impairs genome stability in male mice through stress-mediated pathways. Sci Rep 2023; 13:17622. [PMID: 37848549 PMCID: PMC10582102 DOI: 10.1038/s41598-023-44647-w] [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: 07/22/2023] [Accepted: 10/11/2023] [Indexed: 10/19/2023] Open
Abstract
Population density is known to affect the health and survival of many species, and is especially important for social animals. In mice, living in crowded conditions results in the disruption of social interactions, chronic stress, and immune and reproductive suppression; however, the underlying mechanisms remain unclear. Here, we investigated the role of chemosignals in the regulation of mouse physiology and behavior in response to social crowding. The pheromone 2,5-dimethylpyrazine (2,5-DMP), which is released by female mice in crowded conditions, induced aversion, glucocorticoid elevation and, when chronic, resulted in reproductive and immune suppression. 2,5-DMP olfaction induced genome destabilization in bone marrow cells in a stress-dependent manner, providing a plausible mechanism for crowding-induced immune dysfunction. Interestingly, the genome-destabilizing effect of 2,5-DMP was comparable to a potent mouse stressor (immobilization), and both stressors led to correlated expression changes in genes regulating cellular stress response. Thus, our findings demonstrate that, in mice, the health effects of crowding may be explained at least in part by chemosignals and also propose a significant role of stress and genome destabilization in the emergence of crowding effects.
Collapse
Affiliation(s)
- Timofey S Glinin
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Universitetskaya Emb., 7-9, Saint Petersburg, Russia, 199034.
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA, 91403, USA.
- Endocrine Neoplasia Laboratory, Department of Surgery, University of California, San Francisco, CA, 94143, USA.
| | - Marina V Petrova
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Universitetskaya Emb., 7-9, Saint Petersburg, Russia, 199034
- Center of Molecular and Cellular Biology, Skolkovo Institute of Science and Technology, Bolshoy Blv. 30, Moscow, Russia, 121205
| | - Veronika Shcherbinina
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Universitetskaya Emb., 7-9, Saint Petersburg, Russia, 199034
- Laboratory of Higher Nervous Activity Genetics, Pavlov Institute of Physiology, Russian Academy of Sciences, Makarova Emb. 6, Saint Petersburg, Russia, 199034
| | - Anastasia N Shubina
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Universitetskaya Emb., 7-9, Saint Petersburg, Russia, 199034
- Open Longevity, 15260 Ventura Blvd, STE 2230, Sherman Oaks, CA, 91403, USA
| | - Anna V Dukelskaya
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Universitetskaya Emb., 7-9, Saint Petersburg, Russia, 199034
| | - Polina V Starshova
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Universitetskaya Emb., 7-9, Saint Petersburg, Russia, 199034
| | - Victoria Mamontova
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Universitetskaya Emb., 7-9, Saint Petersburg, Russia, 199034
- Mildred Scheel Early Career Center for Cancer Research (Mildred-Scheel-Nachwuchszentrum, MSNZ), University Hospital Würzburg, Josef-Schneider Str. 2, 97080, Würzburg, Germany
- Department of Biochemistry and Molecular Biology, Biocenter of the University of Würzburg, Am Hubland, 97074, Würzburg, Germany
| | - Alexandra Burnusuz
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Universitetskaya Emb., 7-9, Saint Petersburg, Russia, 199034
| | - Alena O Godunova
- The Federal Research Center Institute of Cytology and Genetics, SB RAS, Academician Lavrentiev Avenue, 10, Novosibirsk, Russia, 630090
| | - Alexander V Romashchenko
- The Federal Research Center Institute of Cytology and Genetics, SB RAS, Academician Lavrentiev Avenue, 10, Novosibirsk, Russia, 630090
- International Tomography Center, Institutskaya St., 3A, Novosibirsk, Russia, 630090
- Federal Research Centre of Biological Systems and Agrotechnologies, RAS, St. January 9, 29, Orenburg, Russia, 460000
| | - Mikhail P Moshkin
- The Federal Research Center Institute of Cytology and Genetics, SB RAS, Academician Lavrentiev Avenue, 10, Novosibirsk, Russia, 630090
| | - Philipp Khaitovich
- Center for Neurobiology and Brain Restoration, Skolkovo Institute of Science and Technology, 3 Nobelya St., Moscow, Russia, 121205
| | - Eugene V Daev
- Department of Genetics and Biotechnology, Saint-Petersburg State University, Universitetskaya Emb., 7-9, Saint Petersburg, Russia, 199034
- Laboratory of Higher Nervous Activity Genetics, Pavlov Institute of Physiology, Russian Academy of Sciences, Makarova Emb. 6, Saint Petersburg, Russia, 199034
| |
Collapse
|
46
|
Tu WB, Christofk HR, Plath K. Nutrient regulation of development and cell fate decisions. Development 2023; 150:dev199961. [PMID: 37260407 PMCID: PMC10281554 DOI: 10.1242/dev.199961] [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: 06/02/2023]
Abstract
Diet contributes to health at all stages of life, from embryonic development to old age. Nutrients, including vitamins, amino acids, lipids and sugars, have instructive roles in directing cell fate and function, maintaining stem cell populations, tissue homeostasis and alleviating the consequences of aging. This Review highlights recent findings that illuminate how common diets and specific nutrients impact cell fate decisions in healthy and disease contexts. We also draw attention to new models, technologies and resources that help to address outstanding questions in this emerging field and may lead to dietary approaches that promote healthy development and improve disease treatments.
Collapse
Affiliation(s)
- William B. Tu
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Heather R. Christofk
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Kathrin Plath
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA 90095, USA
- Jonsson Comprehensive Cancer Center; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
47
|
Ottesen JT, Andersen M. Aging, Inflammation, and Comorbidity in Cancers-A General In Silico Study Exemplified by Myeloproliferative Malignancies. Cancers (Basel) 2023; 15:4806. [PMID: 37835500 PMCID: PMC10572046 DOI: 10.3390/cancers15194806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/23/2023] [Accepted: 09/27/2023] [Indexed: 10/15/2023] Open
Abstract
(1) Background: We consider dormant, pre-cancerous states prevented from developing into cancer by the immune system. Inflammatory morbidity may compromise the immune system and cause the pre-cancer to escape into an actual cancerous development. The immune deficiency described is general, but the results may vary across specific cancers due to different variances (2) Methods: We formulate a general conceptual model to perform rigorous in silico consequence analysis. Relevant existing data for myeloproliferative malignancies from the literature are used to calibrate the in silico computations. (3) Results and conclusions: The hypothesis suggests a common physiological origin for many clinical and epidemiological observations in relation to cancers in general. Examples are the observed age-dependent prevalence for hematopoietic cancers, a general mechanism-based explanation for why the risk of cancer increases with age, and how somatic mutations in general, and specifically seen in screenings of citizens, sometimes are non-increased or even decrease when followed over time. The conceptual model is used to characterize different groups of citizens and patients, describing different treatment responses and development scenarios.
Collapse
Affiliation(s)
- Johnny T. Ottesen
- Mathematical Modeling—Human Health and Disease, IMFUFA, Department of Science and Environment, Roskilde University, 4000 Roskilde, Denmark;
| | | |
Collapse
|
48
|
Kain BN, Tran BT, Luna PN, Cao R, Le DT, Florez MA, Maneix L, Toups JD, Morales-Mantilla DE, Koh S, Han H, Jaksik R, Huang Y, Catic A, Shaw CA, King KY. Hematopoietic stem and progenitor cells confer cross-protective trained immunity in mouse models. iScience 2023; 26:107596. [PMID: 37664586 PMCID: PMC10470378 DOI: 10.1016/j.isci.2023.107596] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 07/24/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Recent studies suggest that infection reprograms hematopoietic stem and progenitor cells (HSPCs) to enhance innate immune responses upon secondary infectious challenge, a process called "trained immunity." However, the specificity and cell types responsible for this response remain poorly defined. We established a model of trained immunity in mice in response to Mycobacterium avium infection. scRNA-seq analysis revealed that HSPCs activate interferon gamma-response genes heterogeneously upon primary challenge, while rare cell populations expand. Macrophages derived from trained HSPCs demonstrated enhanced bacterial killing and metabolism, and a single dose of recombinant interferon gamma exposure was sufficient to induce similar training. Mice transplanted with influenza-trained HSPCs displayed enhanced immunity against M. avium challenge and vice versa, demonstrating cross protection against antigenically distinct pathogens. Together, these results indicate that heterogeneous responses to infection by HSPCs can lead to long-term production of bone marrow derived macrophages with enhanced function and confer cross-protection against alternative pathogens.
Collapse
Affiliation(s)
- Bailee N. Kain
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics – Division of Infectious Disease, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Brandon T. Tran
- Department of Pediatrics – Division of Infectious Disease, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Graduate Program in Cancer and Cell Biology, Baylor College of Medicine, Houston, TX, USA
| | - Pamela N. Luna
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Ruoqiong Cao
- Department of Pediatrics – Division of Infectious Disease, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Graduate Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Duy T. Le
- Department of Pediatrics – Division of Infectious Disease, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Graduate Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Marcus A. Florez
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics – Division of Infectious Disease, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Laure Maneix
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Jack D. Toups
- Department of Pediatrics – Division of Infectious Disease, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Daniel E. Morales-Mantilla
- Department of Pediatrics – Division of Infectious Disease, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Graduate Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| | - Scott Koh
- Department of Pediatrics – Division of Infectious Disease, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
| | - Hyojeong Han
- Department of Pediatrics – Division of Hematology Oncology, Baylor College of Medicine, Houston, TX, USA
| | - Roman Jaksik
- Department of Systems Biology and Engineering and Biotechnology Centre, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Yun Huang
- Center for Epigenetics and Disease Prevention, Institute of Biosciences and Technology, Texas A&M Health, Houston, TX, USA
| | - Andre Catic
- Huffington Center on Aging, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Chad A. Shaw
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
| | - Katherine Y. King
- Graduate Program in Translational Biology and Molecular Medicine, Baylor College of Medicine, Houston, TX, USA
- Department of Pediatrics – Division of Infectious Disease, Texas Children’s Hospital, Baylor College of Medicine, Houston, TX, USA
- Graduate Program in Immunology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Stem Cells and Regenerative Medicine Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
49
|
Mansell E, Lin DS, Loughran SJ, Milsom MD, Trowbridge JJ. New insight into the causes, consequences, and correction of hematopoietic stem cell aging. Exp Hematol 2023; 125-126:1-5. [PMID: 37433369 DOI: 10.1016/j.exphem.2023.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/13/2023]
Abstract
Aging of hematopoietic stem cells (HSCs) is characterized by lineage bias, increased clonal expansion, and functional decrease. At the molecular level, aged HSCs typically display metabolic dysregulation, upregulation of inflammatory pathways, and downregulation of DNA repair pathways. Cellular aging of HSCs, driven by cell-intrinsic and cell-extrinsic factors, causes a predisposition to anemia, adaptive immune compromise, myelodys, plasia, and malignancy. Most hematologic diseases are strongly associated with age. But what is the biological foundation for decreased fitness with age? And are there therapeutic windows to resolve age-related hematopoietic decline? These questions were the focus of the International Society for Experimental Hematology (ISEH) New Investigator Committee Fall 2022 Webinar. This review touches on the latest insights from two leading laboratories into inflammatory- and niche-driven stem cell aging and includes speculation on strategies to prevent or correct age-related decline in HSC function.
Collapse
Affiliation(s)
- Els Mansell
- Erasmus MC Hematology, Rotterdam, The Netherlands; Division of Molecular Medicine and Gene Therapy, Lund University, Lund, Sweden.
| | - Dawn S Lin
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany; Division of Stem Cells and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Stephen J Loughran
- Wellcome Trust/MRC Stem Cell Institute, University of Cambridge, Cambridge, England, UK
| | - Michael D Milsom
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany; Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | | |
Collapse
|
50
|
Avagyan S, Zon LI. Clonal hematopoiesis and inflammation - the perpetual cycle. Trends Cell Biol 2023; 33:695-707. [PMID: 36593155 PMCID: PMC10310890 DOI: 10.1016/j.tcb.2022.12.001] [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: 10/03/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 01/01/2023]
Abstract
Acquired genetic or cytogenetic alterations in a blood stem cell that confer clonal fitness promote its relative expansion leading to clonal hematopoiesis (CH). Despite a largely intact hematopoietic output, CH is associated with a heightened risk of progression to hematologic malignancies and with non-hematologic health manifestations, including cardiovascular disease and overall mortality. We focus on the evidence for the role of inflammation in establishing, maintaining and reciprocally being affected by CH. We describe the known pro-inflammatory signals associated with CH and preclinical studies that elucidated the cellular mechanisms involved. We review the evolving literature on early-onset CH in germline predisposition conditions and the possible role of immune dysregulation in this context.
Collapse
Affiliation(s)
- Serine Avagyan
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorders Center, Boston, MA, USA.
| | - Leonard I Zon
- Boston Children's Hospital, Boston, MA 02215, USA; Howard Hughes Medical Institute, USA
| |
Collapse
|