1
|
Liu M, Yang L, Liu Y, Niu H, Zhang M, Shao Z, Xing L, Wang H. Accelerated senescence of bone marrow erythrocyte precursors in myelodysplastic syndrome. Ann Med 2025; 57:2494676. [PMID: 40277030 PMCID: PMC12035936 DOI: 10.1080/07853890.2025.2494676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 06/04/2024] [Accepted: 03/24/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND Myelodysplastic syndromes (MDS) represent a heterogeneous group of clonal hematopoietic disorders characterized by ineffective haematopoiesis, refractory cytopenia, and an increased risk of progression to acute myeloid leukaemia. This study investigates the presence of cellular senescence in bone marrow (BM) CD235a+ erythrocyte precursors of MDS patients and explores its correlation with anaemia. METHODS We assessed senescence-related markers and cell cycle distribution in BM CD235a+ erythrocyte precursors of MDS patients. Correlation analyses were conducted between the relative mRNA expression of p16INK4A, a key senescence regulator, and peripheral blood parameters. RESULTS MDS patients exhibited heightened cellular senescence characterized by increased SA-β-gal positivity, elevated p16INK4A and p21CIP1 expression, reduced CyclinD1 levels, and elevated IL-6. Cell cycle analysis revealed G0/G1 phase arrest. Correlation analysis established a negative association between p16INK4A expression and reticulocyte count, RBC count, haemoglobin concentration, indicating a direct link between BM erythrocyte precursors senescence and anaemia severity. CONCLUSION MDS patients have accelerated senescence of bone marrow erythrocyte precursors, which is related to their anaemia. The observed correlation underscores the potential significance of senescence-targeted interventions in managing anaemia in MDS.Key MessagesBone marrow CD235a⁺ erythroid precursors in MDS patients exhibit accelerated senescence, characterized by cell cycle arrest and increased inflammatory markers. p16INK4A expression negatively correlates with anaemia severity, suggesting senescence as a key contributor to MDS-related anaemia.
Collapse
Affiliation(s)
- Mengyuan Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Liyan Yang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yumei Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Haiyue Niu
- Department of Hematology, Capital Medical University Affiliated Beijing Friendship Hospital, Beijing, China
| | - Mengying Zhang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zonghong Shao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Limin Xing
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Huaquan Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
2
|
Sato Y. Immune Aging and Its Implication for Age-Related Disease Progression. Physiology (Bethesda) 2025; 40:0. [PMID: 39887318 DOI: 10.1152/physiol.00051.2024] [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: 11/11/2024] [Revised: 12/18/2024] [Accepted: 01/25/2025] [Indexed: 02/01/2025] Open
Abstract
As life expectancy increases globally, the prevalence and severity of age-related diseases have risen, significantly impacting patients' quality of life and increasing dependency on the healthcare system. Age-related diseases share several pathological commonalities, and emerging evidence suggests that targeting these biological processes ameliorates multiple age-related diseases. Immune aging plays a critical role in the pathogenesis of age-related diseases, given its involvement not only in controlling infection and cancer but also in facilitating tissue homeostasis and repair. Aging causes compositional and functional changes in both innate and adaptive immune cells, thereby significantly contributing to the pathogenesis of age-related disease and systemic low-grade inflammation, termed "inflammaging." This review article aims to describe the current understanding of immune aging and its impact on age-related diseases with particular emphasis on kidney and autoimmune diseases. In addition, this review highlights tertiary lymphoid structures (TLS) as a hallmark of immune aging, exploring their roles in inflammation, tissue damage, and potential therapeutic targeting.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
3
|
Chiang YH, Emmrich S, Vannini N. Metabolic Alterations in HSCs during Aging and Leukemogenesis. Physiology (Bethesda) 2025; 40:0. [PMID: 40019828 DOI: 10.1152/physiol.00054.2024] [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: 11/20/2024] [Revised: 12/15/2024] [Accepted: 02/23/2025] [Indexed: 04/26/2025] Open
Abstract
Aging is a multifaceted process associated with a functional decline in cellular function over time, affecting all lifeforms. During the aging process, metabolism, a fundamental hallmark of life (1), is profoundly altered. In the context of hematopoiesis, the proper function of hematopoietic stem cells, at the apex of the blood system, is tightly linked to their energy metabolism, which in turn shapes hematopoietic output. Here, we review the latest developments in our understanding of the metabolic states and changes in aged hematopoietic stem cells, molecular players and pathways involved in aged hematopoietic stem cell metabolism, the consequences of perturbed metabolism on clonal hematopoiesis and leukemogenesis, and pharmacologic/genetic strategies to reverse or rejuvenate altered metabolic phenotypes.
Collapse
Affiliation(s)
- Yi-Hsuan Chiang
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Stephan Emmrich
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Nicola Vannini
- Department of Oncology, Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| |
Collapse
|
4
|
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
|
5
|
Arjmand B, Mehran P, Badamchizadeh S, Alavi-Moghadam S, Arjmand R, Rezaei-Tavirani M, Aghayan HR, Larijani B, Vaezi M, Janbabaei G, Hajifathali A. The Role of Aging and Senescence in Bone Marrow Transplantation Outcome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40259169 DOI: 10.1007/5584_2025_861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/23/2025]
Abstract
Bone marrow transplantation is considered a cornerstone in the treatment of hematologic malignancies and blood disorders. While it may offer the possibility of a cure through the use of high-dose chemotherapy and radiation, outcomes are significantly impacted by biological and medical factors. Herein, aging is associated with reduced hematopoiesis, immune function, and overall regenerative capacity of tissues. Growth arrest, a crucial property of cellular senescence, inhibits bone marrow function, lowers immune surveillance in aged adults, and reduces the efficiency of bone marrow transplantation. The clinical course for older recipients is further complicated by the presence of prolonged immunosuppression, slower recovery, and higher complication rates, including life-threatening graft-versus-host disease. Accordingly, there is increasing interest in explaining how aging, cellular senescence, and transplant outcomes are interrelated. The current chapter outlines the mechanisms whereby aging and senescence contribute to the immunological dysregulation and poor bone marrow transplantation outcomes observed in elderly cancer patients. The authors' goal is to suggest therapeutic approaches that will enhance the quality of life and survival rates of elderly bone marrow transplant recipients.
Collapse
Affiliation(s)
- Babak Arjmand
- Hematology-Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Pouya Mehran
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Sepideh Alavi-Moghadam
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasta Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Hamid Reza Aghayan
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Larijani
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohamad Vaezi
- Hematology-Oncology and Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghasem Janbabaei
- Hematologic Malignancies Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Hajifathali
- Hematopoietic Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
6
|
Vicenzi S, Gao F, Côté P, Hartman JD, Avsharian LC, Vora AA, Rowe RG, Li H, Skowronska-Krawczyk D, Crews LA. Systemic deficits in lipid homeostasis promote aging-associated impairments in B cell progenitor development. GeroScience 2025:10.1007/s11357-025-01594-w. [PMID: 40232347 DOI: 10.1007/s11357-025-01594-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 02/28/2025] [Indexed: 04/16/2025] Open
Abstract
Organismal aging has been associated with diverse metabolic and functional changes across tissues. Within the immune system, key features of physiological hematopoietic cell aging include increased fat deposition in the bone marrow, impaired hematopoietic stem and progenitor cell (HSPC) function, and a propensity towards myeloid differentiation. This shift in lineage bias can lead to pre-malignant bone marrow conditions such as clonal hematopoiesis of indeterminate potential (CHIP) or clonal cytopenias of undetermined significance (CCUS), frequently setting the stage for subsequent development of age-related cancers in myeloid or lymphoid lineages. Human aging has also been associated with diverse lipid alterations across tissues, such as decreased phospholipid membrane fluidity that arises as a result of increased saturated fatty acid (FA) accumulation and a decay in n-3 polyunsaturated fatty acid (PUFA) species by the age of 80 years, however the extent to which impaired FA metabolism contributes to hematopoietic aging is less clear. Here, comprehensive multi-omics analyses uncovered a role for a key PUFA biosynthesis gene, ELOVL2, in mouse and human immune cell aging. Whole transcriptome RNA-sequencing studies and complementary flow cytometric analyses of bone marrow from aged Elovl2 mutant (enzyme-deficient) mice compared with age-matched controls revealed global downregulation in lymphoid cell markers and expression of genes involved specifically in B cell development. These studies unveiled CD79B, a vital molecular regulator of lymphoid progenitor development from the pro-B to pre-B cell stage, as a putative surface biomarker whose loss is associated with accelerated immune aging. The lipidome of mutant versus wild-type mice also displayed significant changes in the biophysical properties of cellular membranes. To investigate the relevance of these finding to human bone marrow aging, analyses of a single cell RNA-seq dataset of human HSPCs across the spectrum of human development and aging uncovered a rare subpopulation (< 7%) of CD34+ HSPCs that expresses ELOVL2 in healthy adult bone marrow. This HSPC subset, along with CD79B-expressing lymphoid-committed cells, were almost completely absent in CD34+ cells isolated from elderly bone marrow samples. Together, these findings uncover new roles for lipid metabolism enzymes in the molecular regulation of cellular aging and immune cell function in mouse and human hematopoiesis. In addition, because systemic loss of ELOVL2 enzymatic activity resulted in downregulation of B cell genes that are also associated with lymphoproliferative neoplasms, this study sheds light on an intriguing metabolic pathway that could be leveraged in future studies as a novel therapeutic modality to target blood cancers or other age-related conditions involving the B cell lineage.
Collapse
Affiliation(s)
- Silvia Vicenzi
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, 92037, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92037, USA
| | - Fangyuan Gao
- Center for Translational Vision Research, Departments of Physiology and Biophysics and Department of Ophthalmology, University of California, Irvine, CA, 92617, USA
| | - Parker Côté
- Department of Pediatrics, University of California San Diego, La Jolla, CA, 92037, USA
| | - Joshua D Hartman
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, 92037, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92037, USA
| | - Lara C Avsharian
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, 92037, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92037, USA
| | - Ashni A Vora
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, 92037, USA
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92037, USA
| | - R Grant Rowe
- Boston Children's Hospital, Dana-Farber Cancer Institute, Harvard Stem Cell Institute, and Harvard Medical School, Boston, MA, 02115, USA
| | - Hojun Li
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92037, USA
- Department of Pediatrics, University of California San Diego, La Jolla, CA, 92037, USA
- Division of Hematology/Oncology, Rady Children's Hospital, San Diego, CA, 92123, USA
| | - Dorota Skowronska-Krawczyk
- Center for Translational Vision Research, Departments of Physiology and Biophysics and Department of Ophthalmology, University of California, Irvine, CA, 92617, USA.
| | - Leslie A Crews
- Division of Regenerative Medicine, Department of Medicine, University of California San Diego, La Jolla, CA, 92037, USA.
- Moores Cancer Center, University of California San Diego, La Jolla, CA, 92037, USA.
- Sanford Stem Cell Institute, University of California San Diego, La Jolla, CA, 92037, USA.
| |
Collapse
|
7
|
Skamagki M, Zhang C, Hacisuleyman E, Galleti G, Wu C, Vinagolu RK, Cha H, Ata D, Kim J, Weiskittel T, Diop M, Aung T, Del Latto M, Kim AS, Li Z, Miele M, Zhao R, Tang LH, Hendrickson RC, Romesser PB, Smith JJ, Giannakakou P, Darnell RB, Bott MJ, Li H, Kim K. Aging-dependent dysregulation of EXOSC2 is maintained in cancer as a dependency. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.04.647279. [PMID: 40236131 PMCID: PMC11996493 DOI: 10.1101/2025.04.04.647279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Reprogramming of aged donor tissue cells into induced pluripotent stem cells (A-iPSC) preserved the epigenetic memory of aged-donor tissue, defined as genomic instability and poor tissue differentiation in our previous study. The unbalanced expression of RNA exosome subunits affects the RNA degradation complex function and is associated with geriatric diseases including premature aging and cancer progression. We hypothesized that the age-dependent progressive subtle dysregulation of EXOSC2 (exosome component 2) causes the aging traits (abnormal cell cycle and poor tissue differentiation). We used embryonic stem cells as a tool to study EXOSC2 function as the aging trait epigenetic memory determined in A-iPSC because these aging traits could not be studied in senesced aged cells or immortalized cancer cells. We found that the regulatory subunit of PP2A phosphatase, PPP2R5E, is a key target of EXOSC2 and this regulation is preserved in stem cells and cancer.
Collapse
|
8
|
Brown G. Cell Lineage Affiliation During Hematopoiesis. Int J Mol Sci 2025; 26:3346. [PMID: 40244205 PMCID: PMC11989489 DOI: 10.3390/ijms26073346] [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: 02/24/2025] [Revised: 03/30/2025] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
By the mid-1960s, hematopoietic stem cells (HSCs) were well described. They generate perhaps the most complex array of functionally mature cells in an adult organism. HSCs and their descendants have been studied extensively, and findings have provided principles that have been applied to the development of many cell systems. However, there are uncertainties about the process of HSC development. They center around when and how HSCs become affiliated with a single-cell lineage. A longstanding view is that this occurs late in development and stepwise via a series of committed oligopotent progenitor cells, which eventually give rise to unipotent progenitors. A very different view is that lineage affiliation can occur as early as within HSCs, and the development of these cells to a mature end cell is then a continuous process. A key consideration is the extent to which lineage-affiliated HSCs self-renew to make a major contribution to hematopoiesis. This review examines the above aspects in relation to our understanding of hematopoiesis.
Collapse
Affiliation(s)
- Geoffrey Brown
- Department of Biomedical Sciences, School of Infection, Inflammation, and Immunology, College of Medicine and Health, University of Birmingham, Edgbaston, Birmingham B15 2TT, UK
| |
Collapse
|
9
|
Toghani D, Gupte S, Zeng S, Mahammadov E, Crosse EI, Seyedhassantehrani N, Burns C, Gravano D, Radtke S, Kiem HP, Rodriguez S, Carlesso N, Pradeep A, Georgiades A, Lucas F, Wilson NK, Kinston SJ, Göttgens B, Zong L, Beerman I, Park B, Janssens DH, Jones D, Toghani A, Nerlov C, Pietras EM, Mesnieres M, Maes C, Kumanogoh A, Worzfeld T, Cheong JG, Josefowicz SZ, Kharchenko P, Scadden DT, Scialdone A, Spencer JA, Silberstein L. Niche-derived Semaphorin 4A safeguards functional identity of myeloid-biased hematopoietic stem cells. NATURE AGING 2025; 5:558-575. [PMID: 39881190 PMCID: PMC12025894 DOI: 10.1038/s43587-024-00798-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/17/2024] [Indexed: 01/31/2025]
Abstract
Somatic stem cell pools comprise diverse, highly specialized subsets whose individual contribution is critical for the overall regenerative function. In the bone marrow, myeloid-biased hematopoietic stem cells (myHSCs) are indispensable for replenishment of myeloid cells and platelets during inflammatory response but, at the same time, become irreversibly damaged during inflammation and aging. Here we identify an extrinsic factor, Semaphorin 4A (Sema4A), which non-cell-autonomously confers myHSC resilience to inflammatory stress. We show that, in the absence of Sema4A, myHSC inflammatory hyper-responsiveness in young mice drives excessive myHSC expansion, myeloid bias and profound loss of regenerative function with age. Mechanistically, Sema4A is mainly produced by neutrophils, signals via a cell surface receptor, Plexin D1, and safeguards the myHSC epigenetic state. Our study shows that, by selectively protecting a distinct stem cell subset, an extrinsic factor preserves functional diversity of somatic stem cell pool throughout organismal lifespan.
Collapse
Affiliation(s)
- Dorsa Toghani
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sanika Gupte
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sharon Zeng
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Elmir Mahammadov
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum Muenchen, Munich, Germany
| | - Edie I Crosse
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | | | - Christian Burns
- Department of Bioengineering, University of California, Merced, Merced, CA, USA
| | - David Gravano
- Department of Bioengineering, University of California, Merced, Merced, CA, USA
| | - Stefan Radtke
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Hans-Peter Kiem
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Sonia Rodriguez
- Department of Stem Cell Biology & Regenerative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Nadia Carlesso
- Department of Stem Cell Biology & Regenerative Medicine, Beckman Research Institute of City of Hope, Duarte, CA, USA
| | - Amogh Pradeep
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Alexis Georgiades
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Fabienne Lucas
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Nicola K Wilson
- Department of Haematology, Jeffrey Cheah Biomedical Centre, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Sarah J Kinston
- Department of Haematology, Jeffrey Cheah Biomedical Centre, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Berthold Göttgens
- Department of Haematology, Jeffrey Cheah Biomedical Centre, Wellcome - MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Le Zong
- Epigenetics and Stem Cell Aging Unit, National Institute of Aging, Baltimore, MD, USA
| | - Isabel Beerman
- Epigenetics and Stem Cell Aging Unit, National Institute of Aging, Baltimore, MD, USA
| | - Bongsoo Park
- Epigenetics and Stem Cell Aging Unit, National Institute of Aging, Baltimore, MD, USA
| | - Derek H Janssens
- Department of Epigenetics, Van Del Institute, Grand Rapids, MI, USA
| | - Daniel Jones
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Ali Toghani
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Claus Nerlov
- Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Eric M Pietras
- Department of Medicine-Hematology, University of Colorado School of Medicine, Aurora, CO, USA
| | - Marion Mesnieres
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Christa Maes
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine, Allergy and Rheumatic Diseases, University of Osaka, Osaka, Japan
| | - Thomas Worzfeld
- Faculty of Medicine, Institute of Pharmacology, University of Marburg, Marburg, Germany
| | - Jin-Gyu Cheong
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Steven Z Josefowicz
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY, USA
| | - Peter Kharchenko
- Department of Stem Cell and Regenerative Biology, Harvard University, Boston, MA, USA
| | - David T Scadden
- Department of Stem Cell and Regenerative Biology, Harvard University, Boston, MA, USA
| | - Antonio Scialdone
- Institute of Epigenetics and Stem Cells (IES), Helmholtz Zentrum Muenchen, Munich, Germany
| | - Joel A Spencer
- Department of Bioengineering, University of California, Merced, Merced, CA, USA
| | - Lev Silberstein
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA.
| |
Collapse
|
10
|
Salminen A. Cooperation between inhibitory immune checkpoints of senescent cells with immunosuppressive network to promote immunosenescence and the aging process. Ageing Res Rev 2025; 106:102694. [PMID: 39984130 DOI: 10.1016/j.arr.2025.102694] [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: 11/22/2024] [Revised: 12/30/2024] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
The accumulation of senescent cells within tissues promotes the aging process by remodelling the functions of the immune system. For many years, it has been known that senescent cells secrete pro-inflammatory cytokines and chemokines, a phenotype called the senescence-associated secretory phenotype (SASP). Chemokines and colony-stimulating factors stimulate myelopoiesis and recruit myeloid cells into aging tissues. Interestingly, recent studies have demonstrated that senescent cells are not only secretory but they also express an increased level of ligand proteins for many inhibitory immune checkpoint receptors. These ligands represent "don't eat me" markers in senescent cells and moreover, they are able to induce an exhaustion of many immune cells, such as surveying natural killer (NK) cells, cytotoxic CD8+ T cells, and macrophages. The programmed cell death protein-1 (PD-1) and its ligand PD-L1 represent the best known inhibitory immune checkpoint pathway. Importantly, the activation of inhibitory checkpoint receptors, e.g., in chronic inflammatory states, can also induce certain immune cells to differentiate toward their immunosuppressive phenotype. This can be observed in myeloid derived suppressor cells (MDSC), tissue regulatory T cells (Treg), and M2 macrophages. Conversely, these immunosuppressive cells stimulate in senescent cells the expression of many ligand proteins for inhibitory checkpoint receptors. Paradoxically, senescent cells not only promote the pro-inflammatory state but they maintain it at a low-grade level by expressing ligands for inhibitory immune checkpoint receptors. Thus, the cooperation between senescent cells and immunosuppressive cells enhances the senescence state of immune cells, i.e., immune senescence/exhaustion, and cellular senescence within tissues via bystander effects.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, Kuopio FI-70211, Finland.
| |
Collapse
|
11
|
Singh VR, O'Donnell LA. Age-Stratified Treg Responses During Viral Infections of the Central Nervous System: A Literature Review. J Med Virol 2025; 97:e70315. [PMID: 40178106 PMCID: PMC11967158 DOI: 10.1002/jmv.70315] [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: 08/20/2024] [Revised: 12/24/2024] [Accepted: 03/07/2025] [Indexed: 04/05/2025]
Abstract
Regulatory T cells (Tregs) play a vital role in limiting inflammation and resolving the immune response after a viral infection. Within the central nervous system (CNS), Tregs are especially important for the protection of neurons, which have limited regenerative capacity, and the preservation of myelin sheaths, which support neuronal function and survival. Nevertheless, viral infections of the CNS often result in enduring neurological dysfunction, especially in more vulnerable age groups such as newborns and the elderly. Although it is appreciated that Treg activity changes with age, it is unclear how these age-dependent changes impact viral CNS infections. In this review, we explore Treg development over the life of the host and discuss evidence for age-dependent Treg responses to peripheral viral infections. We also discuss the CNS-specific roles of Tregs, where both immunomodulatory and neuroprotective functions can contribute to preservation of brain cells. Finally, we examine the current evidence for Treg activity in neurotropic infections in the context of age, and highlight gaps in our understanding of Treg function in younger and older hosts. Overall, a better understanding of age-dependent Treg activity in the CNS may reveal opportunities for therapeutic interventions tailored to the most vulnerable ages.
Collapse
Affiliation(s)
- Vivek R. Singh
- School of Pharmacy and the Graduate School of Pharmaceutical SciencesDuquesne UniversityPittsburghPennsylvaniaUSA
| | - Lauren A. O'Donnell
- School of Pharmacy and the Graduate School of Pharmaceutical SciencesDuquesne UniversityPittsburghPennsylvaniaUSA
| |
Collapse
|
12
|
Yanai H, McNeely T, Ayyar S, Leone M, Zong L, Park B, Beerman I. DNA methylation drives hematopoietic stem cell aging phenotypes after proliferative stress. GeroScience 2025; 47:1873-1886. [PMID: 39390312 PMCID: PMC11978565 DOI: 10.1007/s11357-024-01360-4] [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: 07/17/2024] [Accepted: 09/17/2024] [Indexed: 10/12/2024] Open
Abstract
Aging of hematopoietic stem cells (HSCs) is implicated in various aging phenotypes, including immune dysfunction, anemia, and malignancies. The role of HSC proliferation in driving these aging phenotypes, particularly under stress conditions, remains unclear. Therefore, we induced forced replications of HSCs in vivo by a cyclical treatment with low-dose fluorouracil (5FU) and examined the impact on HSC aging. Our findings show that proliferative stress induces several aging phenotypes, including altered leukocyte counts, decreased lymphoid progenitors, accumulation of HSCs with high expression of Slamf1, and reduced reconstitution potential, without affecting stem cell self-renewal capacity. The divisional history of HSCs was imprinted in the DNA methylome, consistent with functional decline. Specifically, DNA methylation changes included global hypermethylation in non-coding regions and similar frequencies of hypo- and hyper-methylation at promoter regions, particularly affecting genes targeted by the PRC2 complex. Importantly, initial forced replication promoted DNA damage repair accumulated with age, but continuous proliferative stress led to the accumulation of double-strand breaks, independent of functional decline. Overall, our results suggest that HSC proliferation can drive some aging phenotypes primarily through epigenetic mechanisms, including DNA methylation changes.
Collapse
Affiliation(s)
- Hagai Yanai
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, NIH, 251 Bayview Blvd, Suite 100/10C220, Baltimore, MD, 21224, USA
| | - Taylor McNeely
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, NIH, 251 Bayview Blvd, Suite 100/10C220, Baltimore, MD, 21224, USA
| | - Saipriya Ayyar
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, NIH, 251 Bayview Blvd, Suite 100/10C220, Baltimore, MD, 21224, USA
| | - Michael Leone
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, NIH, 251 Bayview Blvd, Suite 100/10C220, Baltimore, MD, 21224, USA
| | - Le Zong
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, NIH, 251 Bayview Blvd, Suite 100/10C220, Baltimore, MD, 21224, USA
| | - Bongsoo Park
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, NIH, 251 Bayview Blvd, Suite 100/10C220, Baltimore, MD, 21224, USA
| | - Isabel Beerman
- Epigenetics and Stem Cell Unit, Translational Gerontology Branch, National Institute On Aging, NIH, 251 Bayview Blvd, Suite 100/10C220, Baltimore, MD, 21224, USA.
| |
Collapse
|
13
|
Mistry P, Potgieter JJC, Pepper MS, Durandt C. Phenotypic Characterisation of Bone Marrow-Derived Haematopoietic Stem/Progenitor Cells from HIV-Infected Individuals. Stem Cell Rev Rep 2025; 21:900-903. [PMID: 39836357 DOI: 10.1007/s12015-024-10834-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/11/2024] [Indexed: 01/22/2025]
Affiliation(s)
- Priyal Mistry
- Institute for Cellular and Molecular Medicine, Department of Immunology, SAMRC Extramural Unit for Stem Cell Research and Therapy, University of Pretoria, Pretoria, 0084, South Africa
| | - Joachim J C Potgieter
- Department of Haematology, University of Pretoria, and National Health Laboratory Service (NHLS) Tshwane Academic Division (TAD), Pretoria, 0084, South Africa
| | - Michael S Pepper
- Institute for Cellular and Molecular Medicine, Department of Immunology, SAMRC Extramural Unit for Stem Cell Research and Therapy, University of Pretoria, Pretoria, 0084, South Africa
| | - Chrisna Durandt
- Institute for Cellular and Molecular Medicine, Department of Immunology, SAMRC Extramural Unit for Stem Cell Research and Therapy, University of Pretoria, Pretoria, 0084, South Africa.
| |
Collapse
|
14
|
Jia Y, Yan L, Fan C, Sun H, Zhou X, Shi Z. Progress of immune senescence in multiple myeloma treatment resistance. Discov Oncol 2025; 16:402. [PMID: 40138127 PMCID: PMC11947401 DOI: 10.1007/s12672-025-02136-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/12/2025] [Indexed: 03/29/2025] Open
Abstract
Multiple myeloma has become the second most common hematologic malignancy threatening human health with the increasing incidence in the population, and the emergence of drug resistance in its treatment has become a problem that needs to be solved urgently. Recent studies have shown that the immune system is closely related to the development of multiple myeloma, and immune senescence plays an extremely critical role in MM treatment resistance. In this paper, we review the connection between immune senescence and the development of MM and its possible role in the drug resistance of MM treatment, to provide new research ideas for the in-depth study of the mechanism of immune senescence and the search for new immunotherapeutic targets to overcome the phenomenon of drug resistance in the immunotherapy of MM patients.
Collapse
Affiliation(s)
- Yanan Jia
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Lixiang Yan
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Chenyang Fan
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Hui Sun
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Xinli Zhou
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China
| | - Zhexin Shi
- Department of Hematology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, 300381, China.
| |
Collapse
|
15
|
Niazi V, Parseh B, Ghafouri-Fard S. The role of genetic/epigenetic factors and microenvironment in hematopoietic stem cell ageing. Biogerontology 2025; 26:76. [PMID: 40119993 DOI: 10.1007/s10522-025-10218-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/09/2025] [Indexed: 03/25/2025]
Abstract
Hematopoietic stem cells (HSCs) ageing is a phenomenon described by reduction in self-renewal capacity, compromised homing, a bias towards myeloid differentiation, and defective reconstitution function. The molecular mechanisms of HSCs ageing have been investigated by several groups. In a broad classification, the underlying causes can be grouped into the intrinsic factors and those related to the microenvironment. Determination of the exact mechanism of HSCs ageing and detailed molecular events during its initiation and progression will help in the establishment of novel therapies for the treatment or prevention of ageing-related hematopoietic disorders. This review offers an overview of genetic and epigenetic causes of HSCs ageing. The findings of these investigations paved the way for design of novel strategies for rejuvenation of HSCs.
Collapse
Affiliation(s)
- Vahid Niazi
- Stem Cell Research Center, Golestan University of Medical Science, Gorgan, Iran
- School of Advanced Technologies in Medicine, Golestan University of Medical Science, Gorgan, Iran
| | - Benyamin Parseh
- Stem Cell Research Center, Golestan University of Medical Science, Gorgan, Iran
- School of Advanced Technologies in Medicine, Golestan University of Medical Science, Gorgan, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Singh A, Chia JJ, Rao DS, Hoffmann A. Population dynamics modeling reveals that myeloid bias involves both HSC differentiation and progenitor proliferation biases. Blood 2025; 145:1293-1308. [PMID: 39791596 PMCID: PMC11952015 DOI: 10.1182/blood.2024025598] [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: 06/10/2024] [Revised: 11/01/2024] [Accepted: 11/20/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Aging and chronic inflammation are associated with overabundant myeloid-primed multipotent progenitors (MPPs) among hematopoietic stem and progenitor cells (HSPCs). Although hematopoietic stem cell (HSC) differentiation bias has been considered a primary cause of myeloid bias, whether it is sufficient has not been quantitatively evaluated. Here, we analyzed bone marrow data from the IκB- (Nfkbia+/-Nfkbib-/-Nfkbie-/-) mouse model of inflammation with elevated NFκB activity, which reveals increased myeloid-biased MPPs. We interpreted these data with differential equation models of population dynamics to identify alterations of HSPC proliferation and differentiation rates. This analysis revealed that short-term HSC differentiation bias alone is likely insufficient to account for the increase in myeloid-biased MPPs. To explore additional mechanisms, we used single-cell RNA sequencing (scRNA-seq) measurements of IκB- and wild-type HSPCs to track the continuous differentiation trajectories from HSCs to erythrocyte/megakaryocyte, myeloid, and lymphoid primed progenitors. Fitting a partial differential equations model of population dynamics to these data revealed not only less lymphoid-fate specification among HSCs but also increased expansion of early myeloid-primed progenitors. Differentially expressed genes along the differentiation trajectories supported increased proliferation among these progenitors. These findings were conserved when wild-type HSPCs were transplanted into IκB- recipients, indicating that an inflamed bone marrow microenvironment is a sufficient driver. We then applied our analysis pipeline to scRNA-seq measurements of HSPCs isolated from aged mice and human patients with myeloid neoplasms. These analyses identified the same myeloid-primed progenitor expansion as in the IκB- models, suggesting that it is a common feature across different settings of myeloid bias.
Collapse
Affiliation(s)
- Apeksha Singh
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA
| | - Jennifer J. Chia
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA
| | - Dinesh S. Rao
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine, University of California, Los Angeles, CA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA
| | - Alexander Hoffmann
- Signaling Systems Laboratory, Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, CA
- Institute for Quantitative and Computational Biosciences, University of California, Los Angeles, CA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA
- Broad Stem Cell Research Center, University of California, Los Angeles, CA
| |
Collapse
|
17
|
Fassoni AC, Glauche I. Math models expose myeloid bias mechanisms in hematopoiesis. Blood 2025; 145:1231-1232. [PMID: 40111337 DOI: 10.1182/blood.2024027777] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025] Open
|
18
|
Komic H, Schmachtel T, Simoes C, Külp M, Yu W, Jolly A, Nilsson MS, Gonzalez C, Prosper F, Bonig H, Paiva B, Thorén FB, Rieger MA. Continuous map of early hematopoietic stem cell differentiation across human lifetime. Nat Commun 2025; 16:2287. [PMID: 40055319 PMCID: PMC11889232 DOI: 10.1038/s41467-025-57096-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 02/11/2025] [Indexed: 05/13/2025] Open
Abstract
Uncovering early gene network changes of human hematopoietic stem cells (HSCs) leading to differentiation induction is of utmost importance for therapeutic manipulation. We employed single cell proteo-transcriptomic sequencing to FACS-enriched bone marrow hematopoietic stem and progenitor cells (HSPCs) from 15 healthy donors. Pseudotime analysis reveals four major differentiation trajectories, which remain consistent upon aging, with an early branching point into megakaryocyte-erythroid progenitors. However, young donors suggest a more productive differentiation from HSPCs to committed progenitors of all lineages. tradeSeq analysis depicts continuous changes in gene expression of HSPC-related genes (DLK1, ADGRG6), and provides a roadmap of gene expression at the earliest branching points. We identify CD273/PD-L2 to be highly expressed in a subfraction of immature multipotent HSPCs with enhanced quiescence. Functional experiments confirm the immune-modulatory function of CD273/PD-L2 on HSPCs in regulating T-cell activation and cytokine release. Here, we present a molecular map of early HSPC differentiation across human life.
Collapse
Affiliation(s)
- Hana Komic
- TIMM Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tessa Schmachtel
- Department of Medicine 2, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Catia Simoes
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Marius Külp
- Department of Medicine 2, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany
- Cardio-Pulmonary-Institute, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Weijia Yu
- Department of Medicine 2, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Adrien Jolly
- Department of Medicine 2, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Malin S Nilsson
- TIMM Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Microbiology and Immunology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Carmen Gonzalez
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Felipe Prosper
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Bruno Paiva
- Cancer Center Clínica Universidad de Navarra, Centro de Investigación Médica Aplicada (CIMA), IDISNA, CIBER-ONC number CB16/12/00369 and CB16/12/00489, Pamplona, Spain
| | - Fredrik B Thorén
- TIMM Laboratory at Sahlgrenska Center for Cancer Research, University of Gothenburg, Gothenburg, Sweden
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Michael A Rieger
- Department of Medicine 2, Hematology/Oncology, Goethe University Frankfurt, Frankfurt am Main, Germany.
- Cardio-Pulmonary-Institute, Frankfurt am Main, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Frankfurt Cancer Institute, Frankfurt am Main, Germany.
| |
Collapse
|
19
|
Joly P, Labsy R, Silvin A. Aging and neurodegeneration: when systemic dysregulations affect brain macrophage heterogeneity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkae034. [PMID: 40073104 DOI: 10.1093/jimmun/vkae034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/14/2024] [Indexed: 03/14/2025]
Abstract
Microglia, the major population of brain resident macrophages, differentiate from yolk sac progenitors in the embryo and play multiple nonimmune roles in brain organization throughout development and life. Various microglia subtypes have been described by transcriptomic and proteomic signatures, involved metabolic pathways, morphology, intracellular complexity, time of residency, and ontogeny, both in development and in disease settings. Such macrophage heterogeneity increases with aging or neurodegeneration. Monocytes' infiltration and differentiation into monocyte-derived macrophages (MDMs) in the brain contribute to this diversity. Microbiota's role in brain diseases has been recently highlighted, revealing how microbial signals, such as metabolites, influence microglia and MDMs. In this brief review, we describe how these signals can influence microglia through their sensome and shape MDMs from their development in the bone marrow to their differentiation in the brain. Monocytes could then be a crucial player in the constitution of a dysbiotic gut-brain axis in neurodegenerative diseases and aging.
Collapse
Affiliation(s)
- Paul Joly
- INSERM U1015, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, Villejuif, 94805, France
| | - Reyhane Labsy
- INSERM U1015, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, Villejuif, 94805, France
| | - Aymeric Silvin
- INSERM U1015, Gustave Roussy Cancer Campus, 114 rue Edouard Vaillant, Villejuif, 94805, France
| |
Collapse
|
20
|
Prescott J, Keyser AJ, Litwin P, Dunbar MD, McClelland R, Ruple A, Ernst H, Butler BL, Kauffman M, Avery A, Harrison BR, Partida-Aguilar M, McCoy BM, Slikas E, Greenier AK, Muller E, Algavi YM, Bamberger T, Creevy KE, Borenstein E, Snyder-Mackler N, Promislow DEL. Rationale and design of the Dog Aging Project precision cohort: a multi-omic resource for longitudinal research in geroscience. GeroScience 2025:10.1007/s11357-025-01571-3. [PMID: 40038157 DOI: 10.1007/s11357-025-01571-3] [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: 10/25/2024] [Accepted: 02/17/2025] [Indexed: 03/06/2025] Open
Abstract
A significant challenge in multi-omic geroscience research is the collection of high quality, fit-for-purpose biospecimens from a diverse and well-characterized study population with sufficient sample size to detect age-related changes in physiological biomarkers. The Dog Aging Project designed the precision cohort to study the mechanisms underlying age-related change in the metabolome, microbiome, and epigenome in companion dogs, an emerging model system for translational geroscience research. One thousand dog-owner pairs were recruited into cohort strata based on life stage, sex, size, and geography. We designed and built a novel implementation of the REDCap electronic data capture system to manage study participants, logistics, and biospecimen and survey data collection in a secure online platform. In collaboration with primary care veterinarians, we collected and processed blood, urine, fecal, and hair samples from 976 dogs. The resulting data include complete blood count, chemistry profile, immunophenotyping by flow cytometry, metabolite quantification, fecal microbiome characterization, epigenomic profile, urinalysis, and associated metadata characterizing sample conditions at collection and during lab processing. The project, which has already begun collecting second- and third-year samples from precision cohort dogs, demonstrates that scientifically useful biospecimens can be collected from a geographically dispersed population through collaboration with private veterinary clinics and downstream labs. The data collection infrastructure developed for the precision cohort can be leveraged for future studies. Most important, the Dog Aging Project is an open data project. We encourage researchers around the world to apply for data access and utilize this rich, constantly growing dataset in their own work.
Collapse
Affiliation(s)
- Jena Prescott
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Amber J Keyser
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Paul Litwin
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Matthew D Dunbar
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Robyn McClelland
- Biostatistics and Collaborative Health Studies Coordinating Center, University of Washington, Seattle, WA, USA
| | - Audrey Ruple
- Department of Population Health Science, Virginia Tech, Blacksburg, VA, USA
| | - Holley Ernst
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Brianna L Butler
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Mandy Kauffman
- Center for Studies in Demography and Ecology, University of Washington, Seattle, WA, USA
| | - Anne Avery
- College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, CO, USA
| | - Benjamin R Harrison
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Maria Partida-Aguilar
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Brianah M McCoy
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Elizabeth Slikas
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | | | - Efrat Muller
- The Blavatnik School of Computer Science, Tel Aviv University, Tel Aviv, Israel
| | - Yadid M Algavi
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tal Bamberger
- Faculty of Medical & Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Kate E Creevy
- Department of Small Animal Clinical Sciences, Texas a&M University, College Station, TX, USA
| | - Elhanan Borenstein
- Blavatnik School of Computer Science and Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv, Israel
| | | | - Daniel E L Promislow
- Jean Mayer USDA Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA.
| |
Collapse
|
21
|
Tchouto MN, Bucher CH, Mess AK, Haas S, Schmidt-Bleek K, Duda GN, Beule D, Milek M. Pronounced impairment of B cell differentiation during bone regeneration in adult immune experienced mice. Front Immunol 2025; 16:1511902. [PMID: 40098964 PMCID: PMC11911212 DOI: 10.3389/fimmu.2025.1511902] [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: 10/16/2024] [Accepted: 02/13/2025] [Indexed: 03/19/2025] Open
Abstract
Introduction Alterations of the adaptive immune system have been shown to impact bone healing and may result in impaired healing in some patients. Apart from T cells, B cells are the key drivers of adaptive immunity. Therefore, their role in age-associated impairments of bone healing might be essential to understand delays during the healing process. B cells are essential for bone formation, and their dysfunction has been associated with aging or autoimmune diseases. But whether age-associated changes in B cell phenotypes are involved in bone regeneration is unknown. Methods Here, we aimed to characterize the role of immune aging in B cell phenotypes during the early inflammatory phase of bone healing. By comparing non-immune experienced with young and immune experienced mice we aimed to analyze the effect of gained immune experience on B cells. Our single cell proteo-genomics analysis quantified thousands of transcriptomes of cells that were isolated from post osteotomy hematoma and the proximal and distal bone marrow cavities, and enabled us to evaluate cell proportion, differential gene expression and cell trajectories. Results While the B cell proportion in young and non-immune experienced animals did not significantly change from 2 to 5 days post osteotomy in the hematoma, we found a significant decrease of the B cell proportion in the immune experienced mice, which was accompanied by the decreased expression of B cell specific genes, suggesting a specific response in immune experienced animals. Furthermore, we detected the most extensive B cell differentiation block in immune-experienced mice compared to non-immune experienced and young animals, predominantly in the transition from immature to mature B cells. Discussion Our results suggest that the pronounced impairment of B cell production found in immune experienced animals plays an important role in the initial phase leading to delayed bone healing. Therefore, novel therapeutic approaches may be able target the B cell differentiation defect to retain B cell functionality even in the immune experienced setting, which is prone to delayed healing.
Collapse
Affiliation(s)
- Mireille Ngokingha Tchouto
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christian H. Bucher
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Ann-Kathrin Mess
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin-Brandenburg School for Regenerative Therapies, Charité – Universitätsmedizin Berlin, Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Simon Haas
- Systems Hematology, Stem Cells & Precision Medicine, Berlin Institute of Health at Charité-Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
| | - Katharina Schmidt-Bleek
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute of Biomechanics and Musculoskeletal Regeneration, Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute of Health (BIH) Center for Regenerative Therapies, Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Dieter Beule
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Miha Milek
- Core Unit Bioinformatics, Berlin Institute of Health at Charité–Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
22
|
Meng Y, Nerlov C. Epigenetic regulation of hematopoietic stem cell fate. Trends Cell Biol 2025; 35:217-229. [PMID: 39271425 DOI: 10.1016/j.tcb.2024.08.005] [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: 03/16/2024] [Revised: 08/12/2024] [Accepted: 08/14/2024] [Indexed: 09/15/2024]
Abstract
Hematopoietic stem cells (HSCs) sustain blood cell production throughout the mammalian life span. However, it has become clear that at the single cell level a subset of HSCs is stably biased in their lineage output, and that such heterogeneity may play a key role in physiological processes including aging and adaptive immunity. Analysis of chromatin accessibility, DNA methylation, and histone modifications has revealed that HSCs with different lineage bias exhibit distinct epigenetic traits inscribed at poised, lineage-specific enhancers. This allows for lineage priming without initiating lineage-specific gene expression in HSCs, controlling lineage bias while preserving self-renewal and multipotency. Here, we review our current understanding of epigenetic regulation in the establishment and maintenance of HSC fate decisions under different physiological conditions.
Collapse
Affiliation(s)
- Yiran Meng
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK
| | - Claus Nerlov
- MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford, UK.
| |
Collapse
|
23
|
Cheminant JR, Deering-Rice CE, Massa CB, Adhikari U, Noll J, Reilly CA, Venosa A. Parenchymal and inflammatory responses to ozone exposure in the aging healthy and surfactant protein C mutant lung. Am J Physiol Lung Cell Mol Physiol 2025; 328:L334-L349. [PMID: 39832482 DOI: 10.1152/ajplung.00261.2024] [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: 08/08/2024] [Revised: 09/09/2024] [Accepted: 01/02/2025] [Indexed: 01/22/2025] Open
Abstract
Ozone (O3) is a ubiquitous pollutant known to produce acute, transient inflammation through oxidative injury and inflammation. These effects are exacerbated in susceptible populations, such as the elderly and those exhibiting genetic mutations in central nodes of pulmonary function. To comprehend the impact of these predisposing factors, the present study examines structural, mechanical, and immunological responses to single acute O3 exposure (0.8 ppm, 3 h) in young (8-14-wk old), middle-aged (44-52-wk old), and old (>80-wk old) mice. Furthermore, this work compares the impact of a clinically relevant mutation in the gene encoding for the alveolar epithelial type 2 specific surfactant protein C. Aging was associated with reduced lung resistance and increases in respiratory elastic properties, the latter of which was exacerbated in SP-C mutant mice. Ozone exposure produced focal injury localized at the terminal bronchiole-to-alveolar junctions and enlarged alveoli in aged SP-C mutant lungs. Flow cytometric analysis revealed increases in mononuclear myeloid abundance in aged SP-C mutant lungs, paired with a contraction in CD8+ expressing cells. Expansion of tertiary lymphoid tissues was also noted in aged groups, more evident in the mutant mice. Spatial transcriptomics of CD68+ macrophages and CD45- nonimmune parenchymal cells highlighted age-dependent shifts in inflammatory and extracellular matrix organization signaling, and enrichment in senescence and chromatin remodeling pathways. These results illustrate the structural and immunological impact of O3 in the aging wild-type and mutant lung and emphasize the significance of modeling environmental exposure in at-risk populations.NEW & NOTEWORTHY Environmental stress and genetic mutations in key functional nodes are linked to the pathogenesis and exacerbation of respiratory pathologies. These responses are exacerbated by aging, though the impact of these factors in combination is not clearly defined. Using a surfactant protein-C mutant line, our studies describe structural changes and phenotypic responses triggered by acute ozone exposure in the young/middle-aged/old lung. Spatial transcriptomics also found regionally distinct and enhanced activation in the aged lung.
Collapse
Affiliation(s)
- Jenna R Cheminant
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah, United States
| | - Cassandra E Deering-Rice
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah, United States
| | - Christopher B Massa
- Department of Anesthesiology and Critical Care Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
- Department of Anesthesiology and Critical Care Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, United States
| | - Ujjwal Adhikari
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah, United States
| | - Jessica Noll
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah, United States
| | - Christopher A Reilly
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah, United States
| | - Alessandro Venosa
- Department of Pharmacology and Toxicology, University of Utah College of Pharmacy, Salt Lake City, Utah, United States
| |
Collapse
|
24
|
Li X, Wang J, Hu L, Cheng T. How age affects human hematopoietic stem and progenitor cells and the strategies to mitigate aging. Exp Hematol 2025; 143:104711. [PMID: 39788412 DOI: 10.1016/j.exphem.2025.104711] [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: 09/25/2024] [Revised: 12/29/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025]
Abstract
Hematopoietic stem cells (HSCs) are central to blood formation and play a pivotal role in hematopoietic and systemic aging. With aging, HSCs undergo significant functional changes, such as an increased stem cell pool, declined homing and reconstitution capacity, and skewed differentiation toward myeloid and megakaryocyte/platelet progenitors. These phenotypic alterations are likely due to the expansion of certain clones, known as clonal hematopoiesis (CH), which leads to disrupted hematopoietic homeostasis, including anemia, impaired immunity, higher risks of hematological malignancies, and even associations with cardiovascular disease, highlighting the broader impact of HSC aging on overall health. HSC aging is driven by a range of mechanisms involving both intrinsic and extrinsic factors, such as DNA damage accumulation, epigenetic remodeling, inflammaging and metabolic regulation. In this review, we summarize the updated understanding of age-related changes in hematopoietic stem and progenitor cells (HSPCs) and the mechanisms underlying the aging process in mammalian models, especially in human study. Additionally, we provide insights into potential therapeutic strategies to counteract aging process and enhance HSC regenerative capacity, which will support therapeutic interventions and promote healthy aging.
Collapse
Affiliation(s)
- Xueling Li
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China
| | - Jianwei Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China
| | - Linping Hu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| | - Tao Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China; Tianjin Institutes of Health Science, Tianjin, China.
| |
Collapse
|
25
|
Li H, Côté P, Kuoch M, Ezike J, Frenis K, Afanassiev A, Greenstreet L, Tanaka-Yano M, Tarantino G, Zhang S, Whangbo J, Butty VL, Moiso E, Falchetti M, Lu K, Connelly GG, Morris V, Wang D, Chen AF, Bianchi G, Daley GQ, Garg S, Liu D, Chou ST, Regev A, Lummertz da Rocha E, Schiebinger G, Rowe RG. The dynamics of hematopoiesis over the human lifespan. Nat Methods 2025; 22:422-434. [PMID: 39639169 PMCID: PMC11908799 DOI: 10.1038/s41592-024-02495-0] [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: 11/29/2023] [Accepted: 09/19/2024] [Indexed: 12/07/2024]
Abstract
Over a lifetime, hematopoietic stem cells (HSCs) adjust their lineage output to support age-aligned physiology. In model organisms, stereotypic waves of hematopoiesis have been observed corresponding to defined age-biased HSC hallmarks. However, how the properties of hematopoietic stem and progenitor cells change over the human lifespan remains unclear. To address this gap, we profiled individual transcriptome states of human hematopoietic stem and progenitor cells spanning gestation, maturation and aging. Here we define the gene expression networks dictating age-specific differentiation of HSCs and the dynamics of fate decisions and lineage priming throughout life. We additionally identifiy and functionally validate a fetal-specific HSC state with robust engraftment and multilineage capacity. Furthermore, we observe that classification of acute myeloid leukemia against defined transcriptional age states demonstrates that utilization of early life transcriptional programs associates with poor prognosis. Overall, we provide a disease-relevant framework for heterochronic orientation of stem cell ontogeny along the real time axis of the human lifespan.
Collapse
Affiliation(s)
- Hojun Li
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Department of Pediatrics, University of California, San Diego, CA, USA.
- Division of Hematology/Oncology, Rady Children's Hospital, San Diego, CA, USA.
| | - Parker Côté
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Pediatrics, University of California, San Diego, CA, USA
| | - Michael Kuoch
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jideofor Ezike
- Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, MA, USA
- Klarman Cell Observatory, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Katie Frenis
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Anton Afanassiev
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Laura Greenstreet
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Mayuri Tanaka-Yano
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Giuseppe Tarantino
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stephen Zhang
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Jennifer Whangbo
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
- Vor Biopharma, Cambridge, MA, USA
| | - Vincent L Butty
- Barbara K. Ostrom Bioinformatics Facility, Integrated Genomics and Bioinformatics Core of the Koch Institute, Cambridge, MA, USA
| | - Enrico Moiso
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Marcelo Falchetti
- Departments of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Kate Lu
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Guinevere G Connelly
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Vivian Morris
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Dahai Wang
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Antonia F Chen
- Harvard Medical School, Boston, MA, USA
- Department of Orthopedic Surgery, Brigham and Women's Hospital, Boston, MA, USA
| | - Giada Bianchi
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - George Q Daley
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Salil Garg
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - David Liu
- Harvard Medical School, Boston, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Stella T Chou
- Division of Hematology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
- Department of Pediatrics, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Aviv Regev
- Division of Hematology/Oncology, Rady Children's Hospital, San Diego, CA, USA
- Genentech, South San Francisco, CA, USA
| | - Edroaldo Lummertz da Rocha
- Departments of Microbiology, Immunology and Parasitology, Federal University of Santa Catarina, Florianopolis, Brazil
| | - Geoffrey Schiebinger
- Department of Mathematics, University of British Columbia, Vancouver, British Columbia, Canada
| | - R Grant Rowe
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Division of Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA.
- Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
26
|
Bril V, Gilhus NE. Aging and infectious diseases in myasthenia gravis. J Neurol Sci 2025; 468:123314. [PMID: 39671879 DOI: 10.1016/j.jns.2024.123314] [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: 07/09/2024] [Revised: 11/07/2024] [Accepted: 11/16/2024] [Indexed: 12/15/2024]
Abstract
Over the past 120 years, mortality associated with myasthenia gravis (MG) has steadily decreased while the incidence of MG has increased. While mortality due to MG has been ≤5 % for at least the past 25 years, the prevalence of MG has increased. This increase in prevalence of MG may be due, in part, to improvements in diagnostics but also to an aging global population and immunosenescence as the largest increases in MG prevalence have been in patients ≥65 years old. In fact, a "very late-onset" subtype of MG has been proposed for patients diagnosed at or after age 65 years. These patients are predominantly anti-AChR antibody positive and thymoma negative. Preferred therapeutic options differ based on age at MG onset. Immunosenescence may play a role not only in MG etiology but also in the increased susceptibility of MG patients to infection. Immunosuppressive effects of MG therapies can also increase vulnerability to infection. Despite the improvements in MG treatment, mortality in MG patients remains higher than in the non-MG population. This is partly due to increased vulnerability to infection but also due to infection acting as a precipitating factor for MG exacerbation or crisis. The increased infection risk inherent with MG and the increased risk resulting from some therapies calls for increased diligence in monitoring and treating infections in MG patients.
Collapse
Affiliation(s)
- Vera Bril
- Division of Neurology, Department of Medicine, University Health Network, University of Toronto, Toronto, Ontario, Canada.
| | - Nils Erik Gilhus
- Department of Neurology, Haukeland University Hospital, Bergen, Norway; Department of Clinical Medicine, University of Bergen, Bergen, Norway
| |
Collapse
|
27
|
Katson M, Gorenshtein A, Pepys J, Mina Y, Shelly S. Mortality and prognosis in herpes simplex Virus-1 encephalitis long-term follow up study. J Neurol Sci 2025; 468:123330. [PMID: 39616793 DOI: 10.1016/j.jns.2024.123330] [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: 07/30/2024] [Revised: 10/17/2024] [Accepted: 11/24/2024] [Indexed: 01/13/2025]
Abstract
BACKGROUND AND OBJECTIVES Herpes simplex virus-1 (HSV-1) encephalitis is the most prevalent form of viral encephalitis worldwide. Consensus statements on the rate of mortality are lacking, with most studies emphasizing short-term mortality risks . We aimed to describe variables effecting mortality for HSV-1 encephalitis in a long term well defined HSV cohorts. METHODS This is a retrospective study, encephalitis patients who were HSV-positive (HSV- 1,HSV-2 and VZV) in the cerebrospinal fluid (CSF) in 23 years' time frame were compared. Clinical, electrophysiological, imaging, and laboratory data were analyzed. RESULTS We identified 47 HSV-1, 8 HSV-2 and 216 with VZV patients with a molecular CSF PCR diagnosis. The median age at diagnosis was 63.3 (interquartile range(IQR) 50.42-72.52) for HSV-1, 46.79 (IQR 36.55-55.05) for HSV-2 and 60.33. (IQR 33.78-74.11) for VZV (p = 0.14). The mean follow up time was 6.25 ± 5.92 years for the group as whole. Among HSV-1 patients, during the follow-up period, 26 patients (55.31 %) died. Ten deaths occurred within the first year, with a median age of death of 70.6 [63.53-75.39]. Patients who died were older (70.6 [63.53-75.39 vs. 48.59 [37.88-61.71], p < 0.001), had a longer time to treatment initiation (4.01 ± 5.69 vs. 1.96 ± 3.58 days, p = 0.026), with cancer comorbidities more prevalent (42.3 % vs. 0 %, p < 0.001). Univariate analysis showed older age (HR 1.07, 95 % CI 1.03-1.10, p < 0.01), and cancer comorbidity (HR 5.55, 95 % CI 2.31-13.33, p < 0.001) were associated with significantly higher risk for mortality. Multivariate analysis confirmed that older age (HR 1.096, 95 % CI 1.04-1.15, p < 0.001), cancer comorbidity (HR 11.02, 95 % CI 2.76-43.9, p < 0.001) and lower lymphocyte count (HR 0.97, 95 % CI 0.95-0.99, p = 0.032) influenced mortality risk. The optimal cut-off age to predict mortality based on AUC-ROC curve was 63.29 (AUC = 0.83, sensitivity = 0.76, specificity = 0.80, PPV = 0.83, NNV = 0.73, p < 0.001). Patients above this age cutoff had a significantly greater cumulative incidence of mortality than did those aged 50-63 years (p < 0.01). DISCUSSION Mortality due to HSV-1 was high and highest in patients >63 years or immunocompromised patients. Favorable outcomes were associated with increased lymphocyte levels in CSF, and early antiviral treatment. These finding may help explain the wide discrepancies in reported mortality rates for HSV encephalitis patients.
Collapse
Affiliation(s)
- Mark Katson
- Department of Neurology, Rambam Medical Center, Haifa, Israel
| | - Alon Gorenshtein
- Department of Neurology, Rambam Medical Center, Haifa, Israel; Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel; AI in Neurology Laboratory, Ruth and Bruce Rapaport Faculty of Medicine, Technion Institute of Technology, Haifa 3525408, Israel
| | - Jack Pepys
- Preventive Neurology Unit, Wolfson Institute of Population Health, Queen Mary University of London, London, UK
| | - Yair Mina
- Sackler Faculty of Medicine, Tel-Aviv University, Israel; Department of Neurology, Tel-Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Shahar Shelly
- Department of Neurology, Rambam Medical Center, Haifa, Israel; AI in Neurology Laboratory, Ruth and Bruce Rapaport Faculty of Medicine, Technion Institute of Technology, Haifa 3525408, Israel; Department of Neurology, Mayo Clinic, Rochester, MN, United States of America.
| |
Collapse
|
28
|
Watt SM, Roubelakis MG. Deciphering the Complexities of Adult Human Steady State and Stress-Induced Hematopoiesis: Progress and Challenges. Int J Mol Sci 2025; 26:671. [PMID: 39859383 PMCID: PMC11766050 DOI: 10.3390/ijms26020671] [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: 09/25/2024] [Revised: 01/05/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Human hematopoietic stem cells (HSCs) have traditionally been viewed as self-renewing, multipotent cells with enormous potential in sustaining essential steady state blood and immune cell production throughout life. Indeed, around 86% (1011-1012) of new cells generated daily in a healthy young human adult are of hematopoietic origin. Therapeutically, human HSCs have contributed to over 1.5 million hematopoietic cell transplants (HCTs) globally, making this the most successful regenerative therapy to date. We will commence this review by briefly highlighting selected key achievements (from 1868 to the end of the 20th century) that have contributed to this accomplishment. Much of our knowledge of hematopoiesis is based on small animal models that, despite their enormous importance, do not always recapitulate human hematopoiesis. Given this, we will critically review the progress and challenges faced in identifying adult human HSCs and tracing their lineage differentiation trajectories, referring to murine studies as needed. Moving forward and given that human hematopoiesis is dynamic and can readily adjust to a variety of stressors, we will then discuss recent research advances contributing to understanding (i) which HSPCs maintain daily steady state human hematopoiesis, (ii) where these are located, and (iii) which mechanisms come into play when homeostatic hematopoiesis switches to stress-induced or emergency hematopoiesis.
Collapse
Affiliation(s)
- Suzanne M. Watt
- Stem Cell Research, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford OX3 9BQ, UK
- Myeloma Research Laboratory, Adelaide Medical School, Faculty of Health and Medical Sciences, University of Adelaide, North Terrace, Adelaide 5005, Australia
- Cancer Program, Precision Medicine Theme, South Australian Health and Medical Research Institute, Adelaide 5001, Australia
| | - Maria G. Roubelakis
- Laboratory of Biology, School of Medicine, National and Kapodistrian University of Athens (NKUA), 11527 Athens, Greece;
- Cell and Gene Therapy Laboratory, Centre of Basic Research, Biomedical Research Foundation of the Academy of Athens (BRFAA), 11527 Athens, Greece
| |
Collapse
|
29
|
Ng M, Cerezo-Wallis D, Ng LG, Hidalgo A. Adaptations of neutrophils in cancer. Immunity 2025; 58:40-58. [PMID: 39813993 DOI: 10.1016/j.immuni.2024.12.009] [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: 09/02/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
There is a renewed interest in neutrophil biology, largely instigated by their prominence in cancer. From an immunologist's perspective, a conceptual breakthrough is the realization that prototypical inflammatory, cytotoxic leukocytes can be tamed to promote the survival and growth of other cells. This has sparked interest in defining the biological principles and molecular mechanisms driving the adaptation of neutrophils to cancer. Yet, many questions remain: is this adaptation mediated by reprogramming mature neutrophils inside the tumoral mass, or rather by rewiring granulopoiesis in the bone marrow? Why, in some instances, are neutrophils beneficial and in others detrimental to cancer? How many different functional programs can be induced in neutrophils by tumors, and is this dependent on the type of tumor? This review summarizes what we know about these questions and discusses therapeutic strategies based on our incipient knowledge of how neutrophils adapt to cancer.
Collapse
Affiliation(s)
- Melissa Ng
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research (A(∗)STAR), Singapore, Singapore.
| | - Daniela Cerezo-Wallis
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| | - Lai Guan Ng
- Shanghai Immune Therapy Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Andres Hidalgo
- Vascular Biology and Therapeutics Program and Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.
| |
Collapse
|
30
|
Basurco L, Abellanas MA, Purnapatre M, Antonello P, Schwartz M. Chronological versus immunological aging: Immune rejuvenation to arrest cognitive decline. Neuron 2025; 113:140-153. [PMID: 39788084 DOI: 10.1016/j.neuron.2024.12.004] [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: 09/05/2024] [Revised: 10/11/2024] [Accepted: 12/03/2024] [Indexed: 01/12/2025]
Abstract
The contemporary understanding that the immune response significantly supports higher brain functions has emphasized the notion that the brain's condition is linked in a complex manner to the state of the immune system. It is therefore not surprising that immunity is a key factor in shaping brain aging. In this perspective article, we propose amending the Latin phrase "mens sana in corpore sano" ("a healthy mind in a healthy body") to "a healthy mind in a healthy immune system." Briefly, we discuss the emerging understanding of the pivotal role of the immune system in supporting lifelong brain maintenance, how the aging of the immune system impacts the brain, and how the potential rejuvenation of the immune system could, in turn, help revitalize brain function, with the ultimate ambitious goal of developing an anti-aging immune therapy.
Collapse
Affiliation(s)
- Leyre Basurco
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | | | | | - Paola Antonello
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel
| | - Michal Schwartz
- Department of Brain Sciences, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
31
|
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
|
32
|
Johansson A, Ho NPY, Takizawa H. Microbiome and Hemato-immune Aging. Exp Hematol 2025; 141:104685. [PMID: 39581302 DOI: 10.1016/j.exphem.2024.104685] [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: 09/08/2024] [Revised: 10/17/2024] [Accepted: 11/16/2024] [Indexed: 11/26/2024]
Abstract
The microbiome is a highly complex and diverse symbiotic component that undergoes dynamic changes with the organismal aging. Microbial perturbations, termed dysbiosis, exert strong influence on dysregulating the bone marrow niche and subsequently promoting the aging of hematopoietic and immune system. Accumulating studies have revealed the substantial impact of intestinal microbiome on the initiation and progression of age-related hematologic alteration and diseases, such as clonal hematopoiesis and blood cancers. Current therapeutic approaches to restore the altered microbiome diversity target specific pathobionts and are demonstrated to improve clinical outcomes of antihematologic malignancy treatments. In this review, we discuss the interplay between the microbiome and the hemato-immune system during aging process. We also shed light on the emerging therapeutic strategies to tackle the dysbiosis for amelioration of aging and disease progression.
Collapse
Affiliation(s)
- Alban Johansson
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences, Kumamoto University, Japan
| | - Nicole Pui-Yu Ho
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences, Kumamoto University, Japan
| | - Hitoshi Takizawa
- Laboratory of Stem Cell Stress, International Research Center for Medical Sciences, Kumamoto University, Japan; Center for Metabolic Regulation of Healthy Aging, Kumamoto University, Japan.
| |
Collapse
|
33
|
Sant'Ana AN, Dias CK, Nunes VBS, Farias MG, Alegretti AP, Portela P, Calvache ET, Meirelles MF, Daudt LE, Michalowski MB, Paz AA, Figueiró F. Prognostic value of myeloid-derived suppressor-like cells in acute myeloid leukemia: insights from immunophenotyping and clinical correlations. Immunol Res 2024; 73:11. [PMID: 39673675 DOI: 10.1007/s12026-024-09558-6] [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: 07/08/2024] [Accepted: 11/18/2024] [Indexed: 12/16/2024]
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population that acts on both innate and adaptive immunity, fostering immune escape in tumors and contributing to cancer progression. Despite the lack of definitive markers for immunophenotyping MDSCs, particularly the polymorphonuclear (PMN-MDSC) subset, these cells seem to play a crucial role in acute myeloid leukemia (AML) patients' prognosis. Additionally, the maturation stage of MDSCs remains a subject of debate and is largely unknown within the AML context. In this study, we conducted a retrospective analysis of flow cytometry immunophenotyping data obtained at the diagnosis of AML patients. We explored how the enrichment of neutrophil maturation stages, the frequency of PMN-MDSC-like cells and monocytic MDSC-like population (M-MDSC-like), and the ratios of MDSC-like cells to T lymphocytes correlate with relevant prognostic indicators. Our findings revealed that CD45+CD33lowHLA-DR-CD36+ PMN-MDSC-like cells and mature CD13+CD11b+CD10+ neutrophils correlate poor survival in AML patients. Furthermore, PMN-MDSC-like cells, and their ratio to T lymphocytes, are elevated in patients with adverse-risk stratification. Similarly, the M-MDSC-like population is increased in FLT3-ITD mutation carrier patients. Notably, we observed confirmational evidence of CD36 relevance in the AML context, which has emerged recently as a potential marker for PMN-MDSCs. Our study highlights significant findings associating increased MDSC-like subsets and poor prognostic factors in AML.
Collapse
MESH Headings
- Humans
- Myeloid-Derived Suppressor Cells/immunology
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/diagnosis
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/pathology
- Immunophenotyping
- Prognosis
- Female
- Male
- Middle Aged
- Adult
- Aged
- Neutrophils/immunology
- Retrospective Studies
- Flow Cytometry
- fms-Like Tyrosine Kinase 3/genetics
- fms-Like Tyrosine Kinase 3/metabolism
- Antigens, CD/metabolism
- Aged, 80 and over
- T-Lymphocytes/immunology
- Young Adult
- Mutation
Collapse
Affiliation(s)
- Alexia N Sant'Ana
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, 90035-003, Brazil
| | - Camila K Dias
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, 90035-003, Brazil
| | - Vitória B S Nunes
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, 90035-003, Brazil
| | - Mariela G Farias
- Unidade de Hematologia e Citometria de Fluxo, Serviço de Diagnóstico Laboratorial, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-903, Brazil
| | - Ana P Alegretti
- Setor de Inovação, Serviço de Diagnóstico Laboratorial, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-903, Brazil
| | - Pâmela Portela
- Unidade de Hematologia e Citometria de Fluxo, Serviço de Diagnóstico Laboratorial, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-903, Brazil
| | - Ebellins T Calvache
- Serviço de Hematologia Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-903, Brazil
| | - Maria F Meirelles
- Serviço de Hematologia Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-903, Brazil
| | - Liane E Daudt
- Serviço de Hematologia Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-903, Brazil
- Programa de Pós-Gradução em Saúde da Criança e do Adolescente, UFRGS, Porto Alegre, RS, 90035-003, Brazil
| | - Mariana B Michalowski
- Programa de Pós-Gradução em Saúde da Criança e do Adolescente, UFRGS, Porto Alegre, RS, 90035-003, Brazil
- Serviço de Oncologia Pediátrica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-903, Brazil
| | - Alessandra A Paz
- Serviço de Hematologia Clínica, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, 90035-903, Brazil
| | - Fabrício Figueiró
- Laboratório de Imunobioquímica do Câncer, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, 90035-003, Brazil.
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, 90035-003, Brazil.
| |
Collapse
|
34
|
Salminen A. GDF15/MIC-1: a stress-induced immunosuppressive factor which promotes the aging process. Biogerontology 2024; 26:19. [PMID: 39643709 PMCID: PMC11624233 DOI: 10.1007/s10522-024-10164-0] [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: 09/06/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
The GDF15 protein, a member of the TGF-β superfamily, is a stress-induced multifunctional protein with many of its functions associated with the regulation of the immune system. GDF15 signaling provides a defence against the excessive inflammation induced by diverse stresses and tissue injuries. Given that the aging process is associated with a low-grade inflammatory state, called inflammaging, it is not surprising that the expression of GDF15 gradually increases with aging. In fact, the GDF15 protein is a core factor secreted by senescent cells, a state called senescence-associated secretory phenotype (SASP). Many age-related stresses, e.g., mitochondrial and endoplasmic reticulum stresses as well as inflammatory, metabolic, and oxidative stresses, induce the expression of GDF15. Although GDF15 signaling is an effective anti-inflammatory modulator, there is robust evidence that it is a pro-aging factor promoting the aging process. GDF15 signaling is not only an anti-inflammatory modulator but it is also a potent immunosuppressive enhancer in chronic inflammatory states. The GDF15 protein can stimulate immune responses either non-specifically via receptors of the TGF-β superfamily or specifically through the GFRAL/HPA/glucocorticoid pathway. GDF15 signaling stimulates the immunosuppressive network activating the functions of MDSCs, Tregs, and M2 macrophages and triggering inhibitory immune checkpoint signaling in senescent cells. Immunosuppressive responses not only suppress chronic inflammatory processes but they evoke many detrimental effects in aged tissues, such as cellular senescence, fibrosis, and tissue atrophy/sarcopenia. It seems that the survival functions of GDF15 go awry in persistent inflammation thus promoting the aging process and age-related diseases.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
35
|
Mosialou I, Kousteni S. From brain to blood and back again: Linking chronic stress, myelopoiesis, and depression. Cell Stem Cell 2024; 31:1721-1723. [PMID: 39642860 DOI: 10.1016/j.stem.2024.11.012] [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: 11/07/2024] [Revised: 11/11/2024] [Accepted: 11/11/2024] [Indexed: 12/09/2024]
Abstract
In this issue of Cell Stem Cell, Mou et al. identified a brain-bone marrow axis reinforcing myelopoiesis and neuroinflammation during psychological stress, culminating in depression. The identification of this pathway provides insights into hematopoietic stem cell homeostasis and regulatory neuronal function with potentially significant implications for the treatment of stress-related disorders.
Collapse
Affiliation(s)
- Ioanna Mosialou
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY 10032, USA; Edward P. Evans for Myelodysplastic Syndromes at Columbia University Medical Center, New York, NY 10032, USA
| | - Stavroula Kousteni
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY 10032, USA; Edward P. Evans for Myelodysplastic Syndromes at Columbia University Medical Center, New York, NY 10032, USA; Herbert Irving Comprehensive Cancer Center (HICCC), Columbia University, New York, NY 10032, USA; Columbia Stem Cell Initiative, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
36
|
Jacobsen SEW. Rejuvenating immunity through a balancing stem cell act. Cell Res 2024; 34:826-827. [PMID: 39043851 PMCID: PMC11614912 DOI: 10.1038/s41422-024-01005-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/25/2024] Open
Affiliation(s)
- Sten Eirik W Jacobsen
- Department of Cell and Molecular Biology and Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet and Karolinska University Hospital Huddinge, Stockholm, Sweden.
- Haematopoietic Stem Cell Biology Laboratory and MRC Molecular Haematology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
37
|
Reusswig F, An O, Deppermann C. Platelet life cycle during aging: function, production and clearance. Platelets 2024; 35:2433750. [PMID: 39618096 DOI: 10.1080/09537104.2024.2433750] [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: 09/04/2024] [Revised: 10/10/2024] [Accepted: 11/15/2024] [Indexed: 12/13/2024]
Abstract
Platelets are important players in hemostasis. Alterations in platelet number and/or function lead to life-threatening conditions like thrombosis, myocardial infarction and stroke. During aging, changes at the cellular, organ and systemic level occur that affect platelet counts, platelet functionality, the expression of platelet surface receptors, clearance markers as well as their interactions with immune cells. Understanding how these changes influence platelets can help to prevent the alterations of hemostasis and thrombosis we observe in the elderly. In this review, we highlight the respective changes at important sites of the platelet life cycle: bone marrow, liver and spleen, but also show how alterations in immunity contribute. We point out the necessity for further research on age-related systemic alterations in these systems and their interplay with platelets to better understand the complex processes that cause alterations in the platelet life cycle during aging.
Collapse
Affiliation(s)
- Friedrich Reusswig
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Olga An
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Carsten Deppermann
- Center for Thrombosis and Hemostasis, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
- Research Center for Immune Therapy, Forschungszentrum für Immuntherapie (FZI), University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| |
Collapse
|
38
|
van Bergen MGJM, Kamphuis P, de Graaf AO, Salzbrunn JB, Koorenhof-Scheele TN, van Zeventer IA, Dinmohamed AG, Schuringa JJ, van der Reijden BA, Huls G, Jansen JH. Clonal hematopoiesis and myeloid skewing in older population-based individuals. Am J Hematol 2024; 99:2402-2405. [PMID: 39429216 DOI: 10.1002/ajh.27495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/09/2024] [Accepted: 09/12/2024] [Indexed: 10/22/2024]
Affiliation(s)
- Maaike G J M van Bergen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Priscilla Kamphuis
- Department of Hematology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Aniek O de Graaf
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jonas B Salzbrunn
- Department of Hematology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Theresia N Koorenhof-Scheele
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Isabelle A van Zeventer
- Department of Hematology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Avinash G Dinmohamed
- Department of Research and Development, Netherlands Comprehensive Cancer Organization (IKNL), Utrecht, The Netherlands
- Erasmus MC, Department of Public Health, University Medical Center, Rotterdam, The Netherlands
| | - Jan Jacob Schuringa
- Department of Hematology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Bert A van der Reijden
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Gerwin Huls
- Department of Hematology, University of Groningen, University Medical Center Groningen, The Netherlands
| | - Joop H Jansen
- Department of Laboratory Medicine, Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
39
|
Li Y, Wu A, Jin X, Shen H, Zhao C, Yi X, Nie H, Wang M, Yin S, Zuo H, Ju Z, Jiang Z, Wang H. DDO1002, an NRF2-KEAP1 inhibitor, improves hematopoietic stem cell aging and stress response. LIFE MEDICINE 2024; 3:lnae043. [PMID: 39872153 PMCID: PMC11748272 DOI: 10.1093/lifemedi/lnae043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 12/10/2024] [Indexed: 01/29/2025]
Abstract
Oxidative stress diminishes the functionality of hematopoietic stem cells (HSCs) as age advances, with heightened reactive oxygen species (ROS) levels exacerbating DNA damage, cellular senescence, and hematopoietic impairment. DDO1002, a potent inhibitor of the NRF2-KEAP1 pathway, modulates the expression of antioxidant genes. Yet, the extent to which it mitigates hematopoietic decline post-total body irradiation (TBI) or in the context of aging remains to be elucidated. Our study has elucidated the role of DDO1002 in modulating NRF2 activity, which, in turn, activates the NRF2-driven antioxidant response element (ARE) signaling cascade. This activation can diminish intracellular levels of ROS, thereby attenuating cellular senescence. In addition, DDO1002 has been demonstrated to ameliorate DNA damage and avert HSC apoptosis, underscoring its potential to mitigate hematopoietic injury precipitated by TBI. Competitive transplantation assay revealed that the administration of DDO1002 can improve the reconstitution and self-renewal capacity of HSCs in aged mice. Single-cell sequencing analysis elucidated that DDO1002 treatment attenuated intracellular inflammatory signaling pathways and mitigated ROS pathway in aged HSCs, suggesting its potential to restore the viability of these cells. Consequently, DDO1002 effectively activated the NRF2-ARE pathway, delaying cellular senescence and ameliorating impaired hematopoiesis, thereby demonstrating its potential as a therapeutic agent for age-related hematopoietic disorders.
Collapse
Affiliation(s)
- Yuwen Li
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People’s Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Aiwei Wu
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People’s Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Xinrong Jin
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People’s Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Haiping Shen
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People’s Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Chenyan Zhao
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People’s Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Xiao Yi
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People’s Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Hui Nie
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People’s Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Mingwei Wang
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People’s Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Shouchun Yin
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People’s Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Hongna Zuo
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People’s Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| | - Zhenyu Ju
- MOE Key Laboratory of Regenerative Medicine, Institute of Aging and Regenerative Medicine, Jinan University, Guangzhou 510632, China
| | - Zhenyu Jiang
- Jiang Su Key Laboratory of Drug Design and Optimization and State Key Laboratory of Natural Medicines, Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Hu Wang
- Zhejiang Key Laboratory of Medical Epigenetics, School of Basic Medical Sciences, The Third People’s Hospital of Deqing, Department of Cardiology, Affiliated Hospital of Hangzhou Normal University, Hangzhou Normal University, Hangzhou 311121, China
| |
Collapse
|
40
|
Tharmapalan V, Wagner W. Biomarkers for aging of blood - how transferable are they between mice and humans? Exp Hematol 2024; 140:104600. [PMID: 39128692 DOI: 10.1016/j.exphem.2024.104600] [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: 06/24/2024] [Revised: 07/18/2024] [Accepted: 07/30/2024] [Indexed: 08/13/2024]
Abstract
Aging significantly impacts the hematopoietic system, reducing its regenerative capacity and ability to restore homeostasis after stress. Mouse models have been invaluable in studying this process due to their shorter lifespan and the ability to explore genetic, treatment, and environmental influences on aging. However, not all aspects of aging are mirrored between species. This review compares three key aging biomarkers in the hematopoietic systems of mice and humans: myeloid bias, telomere attrition, and epigenetic clocks. Myeloid bias, marked by an increased fraction of myeloid cells and decreased lymphoid cells, is a significant aging marker in mice but is scarcely observed in humans after childhood. Conversely, telomere length is a robust aging biomarker in humans, whereas mice exhibit significantly different telomere dynamics, making telomere length less reliable in the murine system. Epigenetic clocks, based on DNA methylation changes at specific genomic regions, provide precise estimates of chronologic age in both mice and humans. Notably, age-associated regions in mice and humans occur at homologous genomic locations. Epigenetic clocks, depending on the epigenetic signatures used, also capture aspects of biological aging, offering powerful tools to assess genetic and environmental impacts on aging. Taken together, not all blood aging biomarkers are transferable between mice and humans. When using murine models to extrapolate human aging, it may be advantageous to focus on aging phenomena observed in both species. In conclusion, although mouse models offer significant insights, selecting appropriate biomarkers is crucial for translating findings to human aging.
Collapse
Affiliation(s)
- Vithurithra Tharmapalan
- Institute for Stem Cell Biology, RWTH Aachen University Medical School, Aachen, Germany; Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University Medical Faculty, Aachen, Germany
| | - Wolfgang Wagner
- Institute for Stem Cell Biology, RWTH Aachen University Medical School, Aachen, Germany; Helmholtz-Institute for Biomedical Engineering, RWTH Aachen University Medical Faculty, Aachen, Germany; Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Aachen, Germany.
| |
Collapse
|
41
|
Martinez-Romero J, Fernandez ME, Bernier M, Price NL, Mueller W, Candia J, Camandola S, Meirelles O, Hu YH, Li Z, Asefa N, Deighan A, Vieira Ligo Teixeira C, Palliyaguru DL, Serrano C, Escobar-Velasquez N, Dickinson S, Shiroma EJ, Ferrucci L, Churchill GA, Allison DB, Launer LJ, de Cabo R. A hematology-based clock derived from the Study of Longitudinal Aging in Mice to estimate biological age. NATURE AGING 2024; 4:1882-1896. [PMID: 39424993 DOI: 10.1038/s43587-024-00728-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Accepted: 09/24/2024] [Indexed: 10/21/2024]
Abstract
Biological clocks and other molecular biomarkers of aging are difficult to implement widely in a clinical setting. In this study, we used routinely collected hematological markers to develop an aging clock to predict blood age and determine whether the difference between predicted age and chronologic age (aging gap) is associated with advanced aging in mice. Data from 2,562 mice of both sexes and three strains were drawn from two longitudinal studies of aging. Eight hematological variables and two metabolic indices were collected longitudinally (12,010 observations). Blood age was predicted using a deep neural network. Blood age was significantly correlated with chronological age, and aging gap was positively associated with mortality risk and frailty. Platelets were identified as the strongest age predictor by the deep neural network. An aging clock based on routinely collected blood measures has the potential to provide a practical clinical tool to better understand individual variability in the aging process.
Collapse
Affiliation(s)
- Jorge Martinez-Romero
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD, USA
| | | | - Michel Bernier
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Nathan L Price
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - William Mueller
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Julián Candia
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Simonetta Camandola
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | - Osorio Meirelles
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD, USA
| | - Yi-Han Hu
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD, USA
| | - Zhiguang Li
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD, USA
| | - Nigus Asefa
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD, USA
| | | | | | | | - Carlos Serrano
- Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
| | | | - Stephanie Dickinson
- Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
| | - Eric J Shiroma
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD, USA
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA
| | | | - David B Allison
- Indiana University School of Public Health-Bloomington, Bloomington, IN, USA
| | - Lenore J Launer
- Laboratory of Epidemiology and Population Sciences, National Institute on Aging, Baltimore, MD, USA
| | - Rafael de Cabo
- Translational Gerontology Branch, National Institute on Aging, Baltimore, MD, USA.
| |
Collapse
|
42
|
Safi R, Mohsen-Kanson T, Kouzi F, El-Saghir J, Dermesrobian V, Zugasti I, Zibara K, Menéndez P, El Hajj H, El-Sabban M. Direct Interaction Between CD34 + Hematopoietic Stem Cells and Mesenchymal Stem Cells Reciprocally Preserves Stemness. Cancers (Basel) 2024; 16:3972. [PMID: 39682159 DOI: 10.3390/cancers16233972] [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: 09/28/2024] [Revised: 11/14/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES A specialized microenvironment in the bone marrow, composed of stromal cells including mesenchymal stem cells (MSCs), supports hematopoietic stem cell (HSC) self-renewal, and differentiation bands play an important role in leukemia development and progression. The reciprocal direct interaction between MSCs and CD34+ HSCs under physiological and pathological conditions is yet to be fully characterized. METHODS Here, we established a direct co-culture model between MSCs and CD34+ HSCs or MSCs and acute myeloid leukemia cells (THP-1, Molm-13, and primary cells from patients) to study heterocellular communication. RESULTS Following MSCs-CD34+ HSCs co-culture, the expression of adhesion markers N-Cadherin and connexin 43 increased in both cell types, forming gap junction channels. Moreover, the clonogenic potential of CD34+ HSCs was increased. However, direct contact of acute myeloid leukemia cells with MSCs reduced the expression levels of connexin 43 and N-Cadherin in MSCs. The impairment in gap junction formation may potentially be due to a defect in the acute myeloid leukemia-derived MSCs. Interestingly, CD34+ HSCs and acute myeloid leukemia cell lines attenuated MSC osteoblastic differentiation upon prolonged direct cell-cell contact. CONCLUSIONS In conclusion, under physiological conditions, connexin 43 and N-Cadherin interaction preserves stemness of both CD34+ HSCs and MSCs, a process that is compromised in acute myeloid leukemia, pointing to the possible role of gap junctions in modulating stemness.
Collapse
Affiliation(s)
- Rémi Safi
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Josep Carreras Leukemia Research Institute, 08916 Barcelona, Spain
| | - Tala Mohsen-Kanson
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Farah Kouzi
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Jamal El-Saghir
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Division of Nephrology, Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Vera Dermesrobian
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
- Laboratory of Adaptive Immunity, Department of Microbiology, Immunology and Transplantation, KU Leuven, 3000 Leuven, Belgium
| | - Inés Zugasti
- Department of Hematology, Hospital Clínic Barcelona, 08036 Barcelona, Spain
| | - Kazem Zibara
- Faculty of Science, Lebanese University, Zahle 1801, Lebanon
- Faculty of Science, Lebanese University, Hadath 40016, Lebanon
| | - Pablo Menéndez
- Josep Carreras Leukemia Research Institute, 08916 Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), 08010 Barcelona, Spain
- Consorcio Investigación Biomédica en Red de Cancer, CIBER-ONC, ISCIII, 28029 Barcelona, Spain
- Spanish Network for Advanced Cell Therapies (TERAV), 08028 Barcelona, Spain
| | - Hiba El Hajj
- Department of Experimental Pathology, Immunology and Microbiology, Faculty of Medicine, American University of Beirut, Beirut 1107, Lebanon
| | - Marwan El-Sabban
- Department of Anatomy, Cell Biology and Physiological Sciences, American University of Beirut, Beirut 1107, Lebanon
| |
Collapse
|
43
|
Zhang S, Ayemoba CE, Di Staulo AM, Joves K, Patel CM, Leung EHW, Ong SG, Nerlov C, Maryanovich M, Chronis C, Pinho S. Platelet Factor 4 (PF4) Regulates Hematopoietic Stem Cell Aging. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625252. [PMID: 39651177 PMCID: PMC11623642 DOI: 10.1101/2024.11.25.625252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Hematopoietic stem cells (HSCs) responsible for blood cell production and their bone marrow regulatory niches undergo age-related changes, impacting immune responses and predisposing individuals to hematologic malignancies. Here, we show that the age-related alterations of the megakaryocytic niche and associated downregulation of Platelet Factor 4 (PF4) are pivotal mechanisms driving HSC aging. PF4-deficient mice display several phenotypes reminiscent of accelerated HSC aging, including lymphopenia, increased myeloid output, and DNA damage, mimicking physiologically aged HSCs. Remarkably, recombinant PF4 administration restored old HSCs to youthful functional phenotypes characterized by improved cell polarity, reduced DNA damage, enhanced in vivo reconstitution capacity, and balanced lineage output. Mechanistically, we identified LDLR and CXCR3 as the HSC receptors transmitting the PF4 signal, with double knockout mice showing exacerbated HSC aging phenotypes similar to PF4-deficient mice. Furthermore, human HSCs across various age groups also respond to the youthful PF4 signaling, highlighting its potential for rejuvenating aged hematopoietic systems. These findings pave the way for targeted therapies aimed at reversing age-related HSC decline with potential implications in the prevention or improvement of the course of age-related hematopoietic diseases. Key Points Age-related attrition of the megakaryocytic niche and associated PF4 downregulation is a central mechanism in HSC aging.PF4 supplementation, acting on LDLR and CXCR3 receptors, rejuvenates the function of aged HSCs.
Collapse
|
44
|
Venkataraman A, Kordic I, Li J, Zhang N, Bharadwaj NS, Fang Z, Das S, Coskun AF. Decoding senescence of aging single cells at the nexus of biomaterials, microfluidics, and spatial omics. NPJ AGING 2024; 10:57. [PMID: 39592596 PMCID: PMC11599402 DOI: 10.1038/s41514-024-00178-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 11/05/2024] [Indexed: 11/28/2024]
Abstract
Aging has profound effects on the body, most notably an increase in the prevalence of several diseases. An important aging hallmark is the presence of senescent cells that no longer multiply nor die off properly. Another characteristic is an altered immune system that fails to properly self-surveil. In this multi-player aging process, cellular senescence induces a change in the secretory phenotype, known as senescence-associated secretory phenotype (SASP), of many cells with the intention of recruiting immune cells to accelerate the clearance of these damaged senescent cells. However, the SASP phenotype results in inducing secondary senescence of nearby cells, resulting in those cells becoming senescent, and improper immune activation resulting in a state of chronic inflammation, called inflammaging, in many diseases. Senescence in immune cells, termed immunosenescence, results in further dysregulation of the immune system. An interdisciplinary approach is needed to physiologically assess aging changes of the immune system at the cellular and tissue level. Thus, the intersection of biomaterials, microfluidics, and spatial omics has great potential to collectively model aging and immunosenescence. Each of these approaches mimics unique aspects of the body undergoes as a part of aging. This perspective highlights the key aspects of how biomaterials provide non-cellular cues to cell aging, microfluidics recapitulate flow-induced and multi-cellular dynamics, and spatial omics analyses dissect the coordination of several biomarkers of senescence as a function of cell interactions in distinct tissue environments. An overview of how senescence and immune dysregulation play a role in organ aging, cancer, wound healing, Alzheimer's, and osteoporosis is included. To illuminate the societal impact of aging, an increasing trend in anti-senescence and anti-aging interventions, including pharmacological interventions, medical procedures, and lifestyle changes is discussed, including further context of senescence.
Collapse
Affiliation(s)
- Abhijeet Venkataraman
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Ivan Kordic
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - JiaXun Li
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Nicholas Zhang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nivik Sanjay Bharadwaj
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Zhou Fang
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Machine Learning Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA
| | - Sandip Das
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Ahmet F Coskun
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Dr NW, Atlanta, GA, 30332, USA.
- Interdisciplinary Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
45
|
Matteini F, Montserrat‐Vazquez S, Florian MC. Rejuvenating aged stem cells: therapeutic strategies to extend health and lifespan. FEBS Lett 2024; 598:2776-2787. [PMID: 38604982 PMCID: PMC11586596 DOI: 10.1002/1873-3468.14865] [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: 10/23/2023] [Revised: 02/03/2024] [Accepted: 03/07/2024] [Indexed: 04/13/2024]
Abstract
Aging is associated with a global decline in stem cell function. To date, several strategies have been proposed to rejuvenate aged stem cells: most of these result in functional improvement of the tissue where the stem cells reside, but the impact on the lifespan of the whole organism has been less clearly established. Here, we review some of the most recent work dealing with interventions that improve the regenerative capacity of aged somatic stem cells in mammals and that might have important translational possibilities. Overall, we underscore that somatic stem cell rejuvenation represents a strategy to improve tissue homeostasis upon aging and present some recent approaches with the potential to affect health span and lifespan of the whole organism.
Collapse
Affiliation(s)
- Francesca Matteini
- Stem Cell Aging Group, Regenerative Medicine ProgramThe Bellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P‐CMR[C])BarcelonaSpain
| | - Sara Montserrat‐Vazquez
- Stem Cell Aging Group, Regenerative Medicine ProgramThe Bellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P‐CMR[C])BarcelonaSpain
| | - M. Carolina Florian
- Stem Cell Aging Group, Regenerative Medicine ProgramThe Bellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
- Program for Advancing the Clinical Translation of Regenerative Medicine of Catalonia (P‐CMR[C])BarcelonaSpain
- Center for Networked Biomedical Research on Bioengineering, Biomaterials and Nanomedicine (CIBER‐BBN)MadridSpain
- The Catalan Institution for Research and Advanced Studies (ICREA)BarcelonaSpain
| |
Collapse
|
46
|
Ajoolabady A, Pratico D, Tang D, Zhou S, Franceschi C, Ren J. Immunosenescence and inflammaging: Mechanisms and role in diseases. Ageing Res Rev 2024; 101:102540. [PMID: 39395575 DOI: 10.1016/j.arr.2024.102540] [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: 04/28/2024] [Revised: 10/02/2024] [Accepted: 10/03/2024] [Indexed: 10/14/2024]
Abstract
Age-related changes initiate a cascade of cellular and molecular alterations that lead to immune system dysfunction or abnormal activation, predisposing individuals to age-related diseases. This phenomenon, commonly referred to as immunosenescence, highlighting aging-associated progressive decline of the immune system. Moreover, mounting evidence suggests that immunosenescence contributes to a related pathological phenomenon known as inflammaging. Inflammaging refers to chronic, low-grade, and systemic inflammation associated with aging, occurring despite the absence of overt stimuli. In the body, inflammation is typically activated in response to overt stimuli such as bacterial/microbial invasion or a pathological state, however, inflammaging occurrence and its underpinning mechanisms seem to be independent and in the absence of such stimuli. Despite recent advancements in molecular characterization and the scrutiny of disease relevance, these two interconnected concepts have remained largely unexplored and unrecognized. In this comprehensive review, we aim to shed light on the mechanistic and cellular aspects of immunosenescence and inflammaging, as well as their pivotal roles in the pathogenesis of aging-related diseases, including cancer, infections, dementia, and neurodegenerative disorders.
Collapse
Affiliation(s)
- Amir Ajoolabady
- Department of Biomedical Engineering, University of Alabama at Birmingham, AL 35294, USA
| | - Domenico Pratico
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA 19140, USA
| | - Daolin Tang
- Department of Surgery, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Shuqin Zhou
- Department of Emergency, Shanghai Tenth People's Hospital, School of Medicine Tongji University, Shanghai 200072, China
| | - Claudio Franceschi
- IRCCS Institute of Neurological Sciences of Bologna, Bologna, Italy; Department of Applied Mathematics and Laboratory of Systems Biology of Aging, Lobachevsky University, Nizhny Novgorod, Russia.
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China; National Clinical Research Center for Interventional Medicine, Shanghai 200032, China.
| |
Collapse
|
47
|
van der Werf I, Sneifer J, Jamieson C. RNA Modifications Shape Hematopoietic Stem Cell Aging: Beyond the Code. FEBS Lett 2024; 598:2774-2775. [PMID: 39252150 PMCID: PMC11586592 DOI: 10.1002/1873-3468.15014] [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: 01/18/2024] [Revised: 02/21/2024] [Accepted: 03/07/2024] [Indexed: 09/11/2024]
Abstract
Hematopoietic system aging is characterized by both hematopoietic stem cell (HSC) and niche degeneration resulting in myeloid lineage-biased differentiation, reduced B cell and T cell lymphopoiesis, increased HSC mobilization, and fat deposition in the bone marrow. Both alterations in RNA splicing and editing during HSC aging contribute to increased myeloid lineage skewing and inflammation-responsive transcription factors, underscoring the importance of epitranscriptomic mechanisms in the acquisition of an age-related phenotype.
Collapse
Affiliation(s)
- Inge van der Werf
- Sanford Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, California, 92037, USA
| | - Jenna Sneifer
- Sanford Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, California, 92037, USA
| | - Catriona Jamieson
- Sanford Stem Cell Institute, Division of Regenerative Medicine, Department of Medicine, Moores Cancer Center, University of California, San Diego, La Jolla, California, 92037, USA
| |
Collapse
|
48
|
Park MD, Berichel JL, Hamon P, Wilk CM, Belabed M, Yatim N, Saffon A, Boumelha J, Falcomatà C, Tepper A, Hegde S, Mattiuz R, Soong BY, LaMarche NM, Rentzeperis F, Troncoso L, Halasz L, Hennequin C, Chin T, Chen EP, Reid AM, Su M, Cahn AR, Koekkoek LL, Venturini N, Wood-isenberg S, D’souza D, Chen R, Dawson T, Nie K, Chen Z, Kim-Schulze S, Casanova-Acebes M, Swirski FK, Downward J, Vabret N, Brown BD, Marron TU, Merad M. Hematopoietic aging promotes cancer by fueling IL-1⍺-driven emergency myelopoiesis. Science 2024; 386:eadn0327. [PMID: 39236155 PMCID: PMC7616710 DOI: 10.1126/science.adn0327] [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: 11/19/2023] [Revised: 06/18/2024] [Accepted: 08/22/2024] [Indexed: 09/07/2024]
Abstract
Age is a major risk factor for cancer, but how aging impacts tumor control remains unclear. In this study, we establish that aging of the immune system, regardless of the age of the stroma and tumor, drives lung cancer progression. Hematopoietic aging enhances emergency myelopoiesis, resulting in the local accumulation of myeloid progenitor-like cells in lung tumors. These cells are a major source of interleukin (IL)-1⍺, which drives the enhanced myeloid response. The age-associated decline of DNA methyltransferase 3A enhances IL-1⍺ production, and disrupting IL-1 receptor 1 signaling early during tumor development normalized myelopoiesis and slowed the growth of lung, colonic, and pancreatic tumors. In human tumors, we identified an enrichment for IL-1⍺-expressing monocyte-derived macrophages linked to age, poorer survival, and recurrence, unraveling how aging promotes cancer and offering actionable therapeutic strategies.
Collapse
Affiliation(s)
- Matthew D. Park
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Jessica Le Berichel
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Pauline Hamon
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - C. Matthias Wilk
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Meriem Belabed
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Nader Yatim
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Alexis Saffon
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- INSERM U932, Immunity and Cancer, Institut Curie, Paris-Cité University; Paris, France
| | - Jesse Boumelha
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Chiara Falcomatà
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Alexander Tepper
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Samarth Hegde
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Raphaël Mattiuz
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Brian Y. Soong
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Nelson M. LaMarche
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Frederika Rentzeperis
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Leanna Troncoso
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Laszlo Halasz
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Clotilde Hennequin
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Theodore Chin
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Earnest P. Chen
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Amanda M. Reid
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Matthew Su
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Ashley Reid Cahn
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Laura L. Koekkoek
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Brain and Body Research Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Nicholas Venturini
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Shira Wood-isenberg
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Darwin D’souza
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Rachel Chen
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Travis Dawson
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Kai Nie
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Zhihong Chen
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Seunghee Kim-Schulze
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Maria Casanova-Acebes
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Filip K. Swirski
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Brain and Body Research Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Cardiovascular Research Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Julian Downward
- Oncogene Biology Laboratory, Francis Crick Institute; London, UK
- Lung Cancer Group, Division of Molecular Pathology, Institute of Cancer Research; London, UK
| | - Nicolas Vabret
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Brian D. Brown
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Thomas U. Marron
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Division of Hematology/Oncology, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Center for Thoracic Oncology, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| | - Miriam Merad
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
- Human Immune Monitoring Center, Icahn School of Medicine at Mount Sinai; New York, NY10029, USA
| |
Collapse
|
49
|
Wrona MV, Ghosh R, Coll K, Chun C, Yousefzadeh MJ. The 3 I's of immunity and aging: immunosenescence, inflammaging, and immune resilience. FRONTIERS IN AGING 2024; 5:1490302. [PMID: 39478807 PMCID: PMC11521913 DOI: 10.3389/fragi.2024.1490302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 09/23/2024] [Indexed: 11/02/2024]
Abstract
As we age, our immune system's ability to effectively respond to pathogens declines, a phenomenon known as immunosenescence. This age-related deterioration affects both innate and adaptive immunity, compromising immune function and leading to chronic inflammation that accelerates aging. Immunosenescence is characterized by alterations in immune cell populations and impaired functionality, resulting in increased susceptibility to infections, diminished vaccine efficacy, and higher prevalence of age-related diseases. Chronic low-grade inflammation further exacerbates these issues, contributing to a decline in overall health and resilience. This review delves into the characteristics of immunosenescence and examines the various intrinsic and extrinsic factors contributing to immune aging and how the hallmarks of aging and cell fates can play a crucial role in this process. Additionally, it discusses the impact of sex, age, social determinants, and gut microbiota health on immune aging, illustrating the complex interplay of these factors in altering immune function. Furthermore, the concept of immune resilience is explored, focusing on the metrics for assessing immune health and identifying strategies to enhance immune function. These strategies include lifestyle interventions such as diet, regular physical activity, stress management, and the use of gerotherapeutics and other approaches. Understanding and mitigating the effects of immunosenescence are crucial for developing interventions that support robust immune responses in aged individuals.
Collapse
Affiliation(s)
- Marianna V. Wrona
- Columbia University in the City of New York, New York, NY, United States
| | - Rituparna Ghosh
- Columbia Center for Human Longevity, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| | - Kaitlyn Coll
- Florida International University, Miami, FL, United States
| | - Connor Chun
- Bronx High School of Science, New York, NY, United States
| | - Matthew J. Yousefzadeh
- Columbia University in the City of New York, New York, NY, United States
- Columbia Center for Human Longevity, Columbia University Medical Center, New York, NY, United States
- Columbia Center for Translational Immunology, Columbia University Medical Center, New York, NY, United States
- Department of Medicine, Columbia University Medical Center, New York, NY, United States
| |
Collapse
|
50
|
Gorelov R, Hochedlinger K. A cellular identity crisis? Plasticity changes during aging and rejuvenation. Genes Dev 2024; 38:823-842. [PMID: 39293862 PMCID: PMC11535162 DOI: 10.1101/gad.351728.124] [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: 09/20/2024]
Abstract
Cellular plasticity in adult multicellular organisms is a protective mechanism that allows certain tissues to regenerate in response to injury. Considering that aging involves exposure to repeated injuries over a lifetime, it is conceivable that cell identity itself is more malleable-and potentially erroneous-with age. In this review, we summarize and critically discuss the available evidence that cells undergo age-related shifts in identity, with an emphasis on those that contribute to age-associated pathologies, including neurodegeneration and cancer. Specifically, we focus on reported instances of programs associated with dedifferentiation, biased differentiation, acquisition of features from alternative lineages, and entry into a preneoplastic state. As some of the most promising approaches to rejuvenate cells reportedly also elicit transient changes to cell identity, we further discuss whether cell state change and rejuvenation can be uncoupled to yield more tractable therapeutic strategies.
Collapse
Affiliation(s)
- Rebecca Gorelov
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Konrad Hochedlinger
- Department of Molecular Biology, Massachusetts General Hospital, Boston, Massachusetts 02114, USA;
- Cancer Center, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, Massachusetts 02114, USA
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts 02138, USA
- Department of Genetics, Harvard Medical School, Boston, Massachusetts 02115, USA
| |
Collapse
|