1
|
Tóth DM, Szeri F, Ashaber M, Muazu M, Székvölgyi L, Arányi T. Tissue-specific roles of de novo DNA methyltransferases. Epigenetics Chromatin 2025; 18:5. [PMID: 39819598 PMCID: PMC11740433 DOI: 10.1186/s13072-024-00566-2] [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: 09/10/2024] [Accepted: 12/23/2024] [Indexed: 01/19/2025] Open
Abstract
DNA methylation, catalyzed by DNA methyltransferases (DNMT), plays pivotal role in regulating embryonic development, gene expression, adaption to environmental stress, and maintaining genome integrity. DNMT family consists of DNMT1, DNMT3A, DNMT3B, and the enzymatically inactive DNMT3L. DNMT3A and DNMT3B establish novel methylation patterns maintained by DNMT1 during replication. Genetic variants of DNMT3A and DNMT3B cause rare diseases such as Tatton-Brown-Rahman and ICF syndromes. Additionally, somatic mutations cause common conditions such as osteoarthritis, osteoporosis, clonal hematopoiesis of indeterminate potential (CHIP), hematologic malignancies, and cancer. While DNMTs have been extensively studied in vitro, in early development and in disease, their detailed physiologic roles remain less understood as in vivo investigations are hindered by the embryonic or perinatal lethality of the knockout mice. To circumvent this problem, tissue-specific Dnmt3a and Dnmt3b knockouts were engineered. This review explores their diverse molecular roles across various organs and cell types and characterizes the phenotype of the knockout mice. We provide a comprehensive collection of over forty tissue-specific knockout models generated by cre recombinase. We highlight the distinct functions of DNMT3A and DNMT3B in germ cells, early development, uterus, hematopoietic differentiation, musculoskeletal development, visceral organs, and nervous system. Our findings indicate that DNMT3A primarily regulates hematopoietic differentiation, while DNMT3B is crucial for cartilage homeostasis and ossification. We emphasize the context-dependent roles of DNMT3A and DNMT3B and demonstrate that they also complement DNMT1 maintenance methyltransferase activity. Overall, the expression patterns of DNMTs across tissues provide insights into potential therapeutic applications for treating neurologic diseases, cancer, and osteoporosis.
Collapse
Affiliation(s)
- Dániel Márton Tóth
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary.
| | - Flóra Szeri
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
| | - Mária Ashaber
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Muhyiddeen Muazu
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary
| | - Lóránt Székvölgyi
- Department of Molecular and Nanopharmaceutics, Genome Architecture and Recombination Research Group, Faculty of Pharmacy, MTA-DE Momentum, University of Debrecen, Debrecen, Hungary.
| | - Tamás Arányi
- Department of Molecular Biology, Semmelweis University, Budapest, Hungary.
- Institute of Molecular Life Sciences, HUN-REN Research Centre for Natural Sciences, Budapest, Hungary.
| |
Collapse
|
2
|
Franco S, Godley LA. Genetic and environmental risks for clonal hematopoiesis and cancer. J Exp Med 2025; 222:e20230931. [PMID: 39626264 PMCID: PMC11614460 DOI: 10.1084/jem.20230931] [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: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 12/11/2024] Open
Abstract
Somatic variants accumulate in all organs with age, with a positive selection of clonal populations that provide a fitness advantage during times of heightened cellular stress leading to clonal expansion. Easily measured within the hematopoietic compartment, clonal hematopoiesis (CH) is now recognized as a common process in which hematopoietic clones with somatic variants associated with hematopoietic neoplasms exist within the blood or bone marrow of individuals without evidence of malignancy. Most cases of CH involve a limited number of genes, most commonly DNMT3A, TET2, and ASXL1. CH confers risk for solid and hematopoietic malignancies as well as cardiovascular and numerous inflammatory diseases and offers opportunities for cancer prevention. Here, we explore the genetic and environmental factors that predispose individuals to CH with unique variant signatures and discuss how CH drives cancer progression with the goals of improving individual cancer risk stratification, identifying key intervention opportunities, and understanding how CH impacts therapeutic strategies and outcomes.
Collapse
Affiliation(s)
| | - Lucy A. Godley
- Department of Medicine, Northwestern Medicine, Chicago, IL, USA
- Division of Hematology/Oncology, Department of Medicine, Robert H. Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| |
Collapse
|
3
|
Kirino Y. Clinical Challenges of Emerging Acquired Autoinflammatory Diseases, Including VEXAS Syndrome. Intern Med 2025; 64:25-30. [PMID: 38296470 DOI: 10.2169/internalmedicine.3219-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2025] Open
Abstract
Vacuoles, E1-ubiquitin-activating enzyme, X-linked, autoinflammatory, somatic (VEXAS) syndrome, caused by an acquired mutation in the ubiquitin-activating enzyme ubiquitin-like modifier activating enzyme 1 (UBA1), was discovered in 2020. Since then, many cases have been reported worldwide. Recently, we performed UBA1 genetic testing in suspected cases of VEXAS throughout Japan and investigated the clinical features of these cases. Most cases were elderly patients in their 70s with clinical features consistent with VEXAS syndrome, such as myelodysplastic syndrome, high-grade fever, skin rash, chondritis, and pulmonary infiltration. However, approximately half of the analyzed patients were negative for the UBA1 variant. As the concept of "acquired autoinflammatory diseases," including VEXAS syndrome, has gained popularity, the number of suspected cases is expected to increase. Currently, there are no established diagnostic or treatment guidelines for these conditions, and they need to be urgently developed. This review summarizes the clinical problems faced by patients with acquired autoinflammatory diseases, including VEXAS.
Collapse
Affiliation(s)
- Yohei Kirino
- Department of Stem Cell and Immune Regulation, Yokohama City University, Graduate School of Medicine, Japan
| |
Collapse
|
4
|
Sobacchi C, Menale C, Crisafulli L, Ficara F. Role of RANKL Signaling in Bone Homeostasis. Physiology (Bethesda) 2025; 40:0. [PMID: 39255276 DOI: 10.1152/physiol.00031.2024] [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: 06/28/2024] [Revised: 08/19/2024] [Accepted: 09/02/2024] [Indexed: 09/12/2024] Open
Abstract
RANKL and its cognate receptor RANK are crucial regulators of bone metabolism in physiological as well as in pathological conditions. Here we go through the works that unveiled the paramount role of this signaling pathway. We focus on the RANKL cytokine, whose alterations are responsible for rare and common bone diseases. We describe recent insights on the regulation of RANKL expression, which provide new hints for the pharmacological regulation of this molecule. Based on the multiple functions exerted by RANKL (within and outside the bone tissue), we advise caution regarding the potential unintended consequences of its inhibition.
Collapse
Affiliation(s)
- Cristina Sobacchi
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| | - Ciro Menale
- Department of Clinical Medicine and Surgery, University of Naples "Federico II," Naples, Italy
| | - Laura Crisafulli
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| | - Francesca Ficara
- Milan Unit, Institute of Genetic and Biomedical Research, National Research Council, Milan, Italy
- Istituto di Ricovero e Cura a Carattere Scientifico Humanitas Research Hospital, Milan, Italy
| |
Collapse
|
5
|
Evans MA, Chavkin NW, Sano S, Sun H, Sardana T, Ravi R, Doviak H, Wang Y, Yura Y, Polizio AH, Horitani K, Ogawa H, Hirschi KK, Walsh K. Tet2-mediated clonal hematopoiesis modestly improves neurological deficits and is associated with inflammation resolution in the subacute phase of experimental stroke. Front Cell Neurosci 2024; 18:1487867. [PMID: 39742155 PMCID: PMC11685025 DOI: 10.3389/fncel.2024.1487867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 11/12/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Recent work has revealed that clonal hematopoiesis (CH) is associated with a higher risk of numerous age-related diseases, including ischemic stroke, however little is known about whether it influences stroke outcome independent of its widespread effects on cardiovascular disease. Studies suggest that leukocytes carrying CH driver mutations have an enhanced inflammatory profile, which could conceivably exacerbate brain injury after a stroke. Methods Using a competitive bone marrow transplant model of Tet2-mediated CH, we tested the hypothesis that CH would lead to a poorer outcome after ischemic stroke by augmenting brain inflammation. Stroke was induced in mice by middle cerebral artery occlusion and neurological outcome was assessed at acute (24 h) and subacute (14 d) timepoints. Brains were collected at both time points for histological, immunofluorescence and gene expression assays. Results Unexpectedly, Tet2-mediated CH had no effect on acute stroke outcome but led to a reduction in neurological deficits during the subacute phase. This improved neurological outcome was associated with lower levels of brain inflammation as evidenced by lower transcript levels of various inflammatory molecules alongside reduced astrogliosis. Discussion These findings suggest that Tet2-mediated CH may have beneficial effects on outcome after stroke, contrasting with the conventional understanding of CH whereby leukocytes with driver mutations promote disease by exacerbating inflammation.
Collapse
Affiliation(s)
- Megan A. Evans
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Nicholas W. Chavkin
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Soichi Sano
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Hanna Sun
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Taneesha Sardana
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Ramya Ravi
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Heather Doviak
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Ying Wang
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Yoshimitsu Yura
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Ariel H. Polizio
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Keita Horitani
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Hayato Ogawa
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Karen K. Hirschi
- Department of Cell Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Kenneth Walsh
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
6
|
Luo Q, Zhou L, Luo D, Yu L. Clonal hematopoiesis of indeterminate potential (CHIP): A potential contributor to lymphoma. Crit Rev Oncol Hematol 2024; 206:104589. [PMID: 39667716 DOI: 10.1016/j.critrevonc.2024.104589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/14/2024] Open
Abstract
Clonal hematopoiesis (CH) typically refers to the clonal expansion of hematopoietic stem cells (HSCs) due to genetic mutations, serving as the pathogenic basis for various diseases. Clonal hematopoiesis of indeterminate potential (CHIP) is a subtype of CH, emerging as a significant risk factor for myeloid malignancies and cardiovascular diseases, which has attracted increasing attention. However, recent research has unveiled previously overlooked links between CHIP and lymphoma. This paper reviews the relationship between CHIP and lymphoma, focusing on the role and mechanism of TET2 and DNMT3A-mediated CHIP in lymphoma from the perspective of laboratory research and clinical observation. Additionally, we explore the therapeutic implications of targeting CHIP genes and inflammatory pathways in lymphoma. Our findings underscore the multifaceted influence of CHIP on lymphoma development and provide a promising avenue for therapeutic interventions in CHIP mediated lymphoma.
Collapse
Affiliation(s)
- QingQing Luo
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Provincial Key Laboratory of Hematological Diseases (2024SSY06052), Nanchang, Jiangxi, China
| | - LiLi Zhou
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Provincial Key Laboratory of Hematological Diseases (2024SSY06052), Nanchang, Jiangxi, China
| | - DaYa Luo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, China.
| | - Li Yu
- Department of Hematology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Jiangxi Provincial Key Laboratory of Hematological Diseases (2024SSY06052), Nanchang, Jiangxi, China.
| |
Collapse
|
7
|
Sano S, Walsh K. Can dental problems be indicative of clonal hematopoiesis? Cell Res 2024; 34:824-825. [PMID: 39054344 PMCID: PMC11614864 DOI: 10.1038/s41422-024-01006-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/27/2024] Open
Affiliation(s)
- Soichi Sano
- Laboratory of Cardiovascular Mosaicism, National Cerebral and Cardiovascular Center, Osaka, Japan
| | - Kenneth Walsh
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
8
|
Prummel KD, Woods K, Kholmatov M, Schmitt EC, Vlachou EP, Poschmann G, Stühler K, Wehner R, Schmitz M, Winter S, Oelschlaegel U, Schwartz LS, Moura PL, Hellström-Lindberg E, Theobald M, Trowbridge JJ, Platzbecker U, Zaugg JB, Guezguez B. Inflammatory Mesenchymal Stromal Cells and IFN-responsive T cells are key mediators of human bone marrow niche remodeling in CHIP and MDS. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.27.625734. [PMID: 39651275 PMCID: PMC11623587 DOI: 10.1101/2024.11.27.625734] [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
Somatic mutations in hematopoietic stem/progenitor cells (HSPCs) can lead to clonal hematopoiesis of indeterminate potential (CHIP), potentially progressing to myelodysplastic syndromes (MDS). Here, we investigated how CHIP and MDS remodel the human bone marrow (BM) niche relative to healthy elderly donors, using single cell and anatomical analyses in a large BM cohort. We found distinct inflammatory remodeling of the BM in CHIP and MDS. Furthermore, the stromal compartment progressively lost its HSPC-supportive adipogenic CXCL12-abundant reticular cells while an inflammatory mesenchymal stroma cell (iMSCs) population emerged in CHIP, which expanded in MDS. iMSCs exhibited distinct functional signatures in CHIP and MDS, retaining residual HSPC-support and angiogenic activity in MDS, corresponding with an increase in microvasculature in the MDS niche. Additionally, an IFN-responsive T cell population was linked to fueling inflammation in the stroma. Overall, these findings open new avenues for early intervention in hematological malignancies.
Collapse
|
9
|
Yu Z, Vromman A, Nguyen NQH, Schuermans A, Rentz T, Vellarikkal SK, Uddin MM, Niroula A, Griffin G, Honigberg MC, Lin AE, Gibson CJ, Katz DH, Tahir U, Fang S, Haidermota S, Ganesh S, Antoine T, Weinstock J, Austin TR, Ramachandran VS, Peloso GM, Hornsby W, Ganz P, Manson JE, Haring B, Kooperberg CL, Reiner AP, Bis JC, Psaty BM, Min YI, Correa A, Lange LA, Post WS, Rotter JI, Rich SS, Wilson JG, Ebert BL, Yu B, Ballantyne CM, Coresh J, Sankaran VG, Bick AG, Jaiswal S, Gerszten RE, Libby P, Gupta RM, Natarajan P. Human Plasma Proteomic Profile of Clonal Hematopoiesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.07.25.550557. [PMID: 39554199 PMCID: PMC11565774 DOI: 10.1101/2023.07.25.550557] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Plasma proteomic profiles associated with subclinical somatic mutations in blood cells may offer novel insights into downstream clinical consequences. Here, we explore such patterns in clonal hematopoiesis of indeterminate potential (CHIP), which is linked to several cancer and non-cancer outcomes, including coronary artery disease (CAD). Among 61,833 ancestrally diverse participants (3,881 with CHIP) from NHLBI TOPMed and UK Biobank with blood-based DNA sequencing and proteomic measurements (1,148 proteins by SomaScan in TOPMed and 2,917 proteins by Olink in UK Biobank), we identified 32 and 345 unique proteins from TOPMed and UK Biobank, respectively, associated with the most prevalent driver genes ( DNMT3A , TET2 , and ASXL1 ). These associations showed substantial heterogeneity by driver genes, sex, and race, and were enriched for immune response and inflammation pathways. Mendelian randomization in humans, coupled with ELISA in hematopoietic Tet2 -/- vs wild-type mice validation, disentangled causal proteomic perturbations from TET2 CHIP. Lastly, we identified plasma proteins shared between CHIP and CAD.
Collapse
|
10
|
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
|
11
|
Verdonschot JAJ, Fuster JJ, Walsh K, Heymans SRB. The emerging role of clonal haematopoiesis in the pathogenesis of dilated cardiomyopathy. Eur Heart J 2024; 45:ehae682. [PMID: 39417710 PMCID: PMC11638724 DOI: 10.1093/eurheartj/ehae682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/30/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
The increased sensitivity of novel DNA sequencing techniques has made it possible to identify somatic mutations in small circulating clones of haematopoietic stem cells. When the mutation affects a 'driver' gene, the mutant clone gains a competitive advantage and has the potential to expand over time, a phenomenon referred to as clonal haematopoiesis (CH), which is emerging as a new risk factor for various non-haematological conditions, most notably cardiovascular disease (e.g. heart failure). Dilated cardiomyopathy (DCM) is a form of non-ischaemic heart failure that is characterized by a heterogeneous aetiology. The first evidence is arising that CH plays an important role in the disease course in patients with DCM, and a strong association of CH with multiple aetiologies of DCM has been described (e.g. inflammation, chemotherapy, and atrial fibrillation). The myocardial inflammation induced by CH may be an important trigger for DCM development for an already susceptible heart, e.g. in the presence of genetic variants, environmental triggers, and comorbidities. Studies investigating the role of CH in the pathogenesis of DCM are expected to increase rapidly. To move the field forward, it will be important to report the methodology and results in a standardized manner, so results can be combined and compared. The accurate measurement of CH in patients with DCM can provide guidance of specific (anti-inflammatory) therapies, as mutations in the CH driver genes prime the inflammasome pathway.
Collapse
Affiliation(s)
- Job A J Verdonschot
- Department of Clinical Genetics, Maastricht University Medical Center+, Maastricht, the Netherlands
- Department of Cardiology, Maastricht University, Cardiovascular Research Institute Maastricht (CARIM), P.O. Box 616, 6200 MD Maastricht, the Netherlands
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart)
| | - Jose J Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), C. de Melchor Fernández Almagro, 3, Fuencarral-El Pardo, 28029 Madrid, Spain
- CIBER en Enfermedades Cardiovasculares (CIBER-CV), Av. Monforte de Lemos, 3-5. Pabellón 11, Planta 0, 28029 Madrid, Spain
| | - Kenneth Walsh
- Division of Cardiovascular Medicine and Robert M. Berne Cardiovascular Research Center, Hematovascular Biology Center, University of Virginia School of Medicine, 415 Lane Rd, Suite 1010, PO Box 801394, Charlottesville, VA, USA
| | - Stephane R B Heymans
- Department of Cardiology, Maastricht University, Cardiovascular Research Institute Maastricht (CARIM), P.O. Box 616, 6200 MD Maastricht, the Netherlands
- European Reference Network for Rare, Low Prevalence and Complex Diseases of the Heart (ERN GUARD-Heart)
- Department of Cardiovascular Science, Katholieke Universiteit Leuven, Herestraat 49, 3000 Leuven, Belgium
| |
Collapse
|
12
|
Tiedje V, Vela PS, Yang JL, Untch BR, Boucai L, Stonestrom AJ, Costa AB, Expósito SF, Srivastava A, Kerpelev M, Greenberg J, Wereski M, Kulick A, Chen K, Qin T, Im SY, Krishnan A, Martinez Benitez AR, Pluvinet R, Sahin M, Menghrajani K, Krishnamoorthy GP, de Stanchina E, Zehir A, Satija R, Knauf J, Bowman RL, Esteller M, Devlin S, Berger MF, Koche RP, Fagin JA, Levine RL. Targetable treatment resistance in thyroid cancer with clonal hematopoiesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.10.617685. [PMID: 39415999 PMCID: PMC11483059 DOI: 10.1101/2024.10.10.617685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2024]
Abstract
Anaplastic thyroid cancer (ATC) is a clinically aggressive malignancy with a dismal prognosis. Combined BRAF/MEK inhibition offers significant therapeutic benefit in patients with BRAF V600E -mutant ATCs. However, relapses are common and overall survival remains poor. Compared with differentiated thyroid cancer, a hallmark of ATCs is significant infiltration with myeloid cells, particularly macrophages. ATCs are most common in the aging population, which also has an increased incidence of TET2 -mutant clonal hematopoiesis (CH). CH-mutant macrophages have been shown to accelerate CH-associated pathophysiology including atherosclerosis. However, the clinical and mechanistic contribution of CH-mutant clones to solid tumour biology, prognosis and therapeutic response has not been elucidated. Here we show that TET2 -mutant CH is enriched in the tumour microenvironment of patients with solid tumours and associated with adverse prognosis in ATC patients. We find that Tet2 -mutant macrophages selectively infiltrate mouse Braf V600E -mutant ATC and that their overexpression of Tgfβ-family ligands mediates resistance to BRAF/MEK inhibition. Importantly, inhibition of Tgfβ signaling restores sensitivity to MAPK pathway inhibition, opening a path for synergistic strategies to improve outcomes of patients with ATCs and concurrent CH.
Collapse
|
13
|
Takahashi K, Nakada D, Goodell M. Distinct landscape and clinical implications of therapy-related clonal hematopoiesis. J Clin Invest 2024; 134:e180069. [PMID: 39352380 PMCID: PMC11444158 DOI: 10.1172/jci180069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024] Open
Abstract
Therapy-related clonal hematopoiesis (t-CH) is defined as clonal hematopoiesis detected in individuals previously treated with chemotherapy and/or radiation therapy. With the increased use of genetic analysis in oncological care, the detection of t-CH among cancer patients is becoming increasingly common. t-CH arises through the selective bottleneck imposed by chemotherapies and potentially through direct mutagenesis from chemotherapies, resulting in a distinct mutational landscape enriched with mutations in DNA damage-response pathway genes such as TP53, PPM1D, and CHEK2. Emerging evidence sheds light on the mechanisms of t-CH development and potential strategies to mitigate its emergence. Due to its unique characteristics that predominantly affect cancer patients, t-CH has clinical implications distinct from those of CH in the general population. This Review discusses the potential mechanisms of t-CH development, its mutational landscape, mutant-drug relationships, and its clinical significance. We highlight the distinct nature of t-CH and call for intensified research in this field.
Collapse
Affiliation(s)
- Koichi Takahashi
- Departments of Leukemia and Genomic Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Margaret Goodell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
14
|
Díez-Díez M, Ramos-Neble BL, de la Barrera J, Silla-Castro JC, Quintas A, Vázquez E, Rey-Martín MA, Izzi B, Sánchez-García L, García-Lunar I, Mendieta G, Mass V, Gómez-López N, Espadas C, González G, Quesada AJ, García-Álvarez A, Fernández-Ortiz A, Lara-Pezzi E, Dopazo A, Sánchez-Cabo F, Ibáñez B, Andrés V, Fuster V, Fuster JJ. Unidirectional association of clonal hematopoiesis with atherosclerosis development. Nat Med 2024; 30:2857-2866. [PMID: 39215150 PMCID: PMC11485253 DOI: 10.1038/s41591-024-03213-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Accepted: 07/25/2024] [Indexed: 09/04/2024]
Abstract
Clonal hematopoiesis, a condition in which acquired somatic mutations in hematopoietic stem cells lead to the outgrowth of a mutant hematopoietic clone, is associated with a higher risk of hematological cancer and a growing list of nonhematological disorders, most notably atherosclerosis and associated cardiovascular disease. However, whether accelerated atherosclerosis is a cause or a consequence of clonal hematopoiesis remains a matter of debate. Some studies support a direct contribution of certain clonal hematopoiesis-related mutations to atherosclerosis via exacerbation of inflammatory responses, whereas others suggest that clonal hematopoiesis is a symptom rather than a cause of atherosclerosis, as atherosclerosis or related traits may accelerate the expansion of mutant hematopoietic clones. Here we combine high-sensitivity DNA sequencing in blood and noninvasive vascular imaging to investigate the interplay between clonal hematopoiesis and atherosclerosis in a longitudinal cohort of healthy middle-aged individuals. We found that the presence of a clonal hematopoiesis-related mutation confers an increased risk of developing de novo femoral atherosclerosis over a 6-year period, whereas neither the presence nor the extent of atherosclerosis affects mutant cell expansion during this timeframe. These findings indicate that clonal hematopoiesis unidirectionally promotes atherosclerosis, which should help translate the growing understanding of this condition into strategies for the prevention of atherosclerotic cardiovascular disease in individuals exhibiting clonal hematopoiesis.
Collapse
Affiliation(s)
- Miriam Díez-Díez
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | | | - J C Silla-Castro
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Ana Quintas
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Enrique Vázquez
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | - Benedetta Izzi
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | - Inés García-Lunar
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain
- Cardiology Department, University Hospital La Moraleja, Madrid, Spain
| | - Guiomar Mendieta
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- Servicio de Cardiología, Institut Clínic Cardiovascular, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain
| | - Virginia Mass
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | - Cristina Espadas
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | - Gema González
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
| | | | - Ana García-Álvarez
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain
- Servicio de Cardiología, Institut Clínic Cardiovascular, Hospital Clínic de Barcelona, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
| | - Antonio Fernández-Ortiz
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain
- Hospital Clínico San Carlos, Universidad Complutense, IdISSC, Madrid, Spain
| | - Enrique Lara-Pezzi
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Ana Dopazo
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Fátima Sánchez-Cabo
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Borja Ibáñez
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain
- Cardiology Department, IIS-Fundación Jiménez Díaz University Hospital, Madrid, Spain
| | - Vicente Andrés
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain
| | - Valentín Fuster
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.
- Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| | - José J Fuster
- Centro Nacional de Investigaciones Cardiovasculares, Madrid, Spain.
- CIBER de Enfermedades Cardiovasculares, Madrid, Spain.
| |
Collapse
|
15
|
Yalcinkaya M, Tall AR. Genetic and epigenetic regulation of inflammasomes: Role in atherosclerosis. Atherosclerosis 2024; 396:118541. [PMID: 39111028 PMCID: PMC11374466 DOI: 10.1016/j.atherosclerosis.2024.118541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/05/2024] [Accepted: 07/10/2024] [Indexed: 09/06/2024]
Abstract
The cardiovascular complications of atherosclerosis are thought to arise from an inflammatory response to the accumulation of cholesterol-rich lipoproteins in the arterial wall. The positive outcome of CANTOS (Canakinumab Anti-inflammatory Thrombosis Outcome Study) provided key evidence to support this concept and suggested that inflammasomes and IL-1β are important inflammatory mediators in human atherosclerotic cardiovascular diseases (ACVD). In specific settings NLRP3 or AIM2 inflammasomes can induce inflammatory responses in the arterial wall and promote the formation of unstable atherosclerotic plaques. Clonal hematopoiesis (CH) has recently emerged as a major independent risk factor for ACVD. CH mutations arise during ageing and commonly involves variants in genes mediating epigenetic modifications (TET2, DNMT3A, ASXL1) or cytokine signaling (JAK2). Accumulating evidence points to the role of inflammasomes in the progression of CH-induced ACVD events and has shed light on the regulatory pathways and possible therapeutic approaches that specifically target inflammasomes in atherosclerosis. Epigenetic dynamics play a vital role in regulating the generation and activation of inflammasome components by causing changes in DNA methylation patterns and chromatin assembly. This review examines the genetic and epigenetic regulation of inflammasomes, the intersection of macrophage cholesterol accumulation with inflammasome activation and their roles in atherosclerosis. Understanding the involvement of inflammasomes in atherosclerosis pathogenesis may lead to customized treatments that reduce the burden of ACVD.
Collapse
Affiliation(s)
- Mustafa Yalcinkaya
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| | - Alan R Tall
- Division of Molecular Medicine, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA.
| |
Collapse
|
16
|
Liu J, Wang B, Chen H, Yu X, Cao X, Zhang H. Osteoclast-derived exosomes influence osteoblast differentiation in osteoporosis progression via the lncRNA AW011738/ miR-24-2-5p/ TREM1 axis. Biomed Pharmacother 2024; 178:117231. [PMID: 39094542 DOI: 10.1016/j.biopha.2024.117231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/29/2024] [Accepted: 07/30/2024] [Indexed: 08/04/2024] Open
Abstract
AIMS To investigate the molecular mechanism of osteoclast-derived exosomes in osteoporosis. MAIN METHODS RANKL induced osteoclast model was screened for significantly differentially expressed lncRNAs and mRNAs by whole RNA sequencing. Exosomes were characterized using electron microscopy, western blotting and nanosight. Overexpression or knockdown of AW011738 was performed to explore its function. The degree of osteoporosis in an osteoporosis model was assessed by mirco-CT. The osteoclast model, osteoblast differentiation ability and the molecular mechanism of lncRNA AW011738/miR-24-2-5p/TREM1 axis in osteoporosis were assessed by dual luciferase reporter gene assay, Western blotting (WB), immunofluorescence and ALP staining. Bioinformatics was used to predict interactions of key osteoporosis-related genes with miRNAs, transcription factors, and potential drugs after upregulation of AW011738. To predict the protein-protein interaction (PPI) network associated with key genes, GO and KEGG analyses were performed on the key genes. The ssGSVA was used to predict changes in the immune microenvironment. KEY FINDINGS Osteoclast-derived exosomes containing lncRNA AW011738 decreased the osteogenesis-related markers and accelerated bone loss in OVX mice. Osteoclast (si-AW011738)-derived exosomes showed a significant increase in biomarkers of osteoblast differentiation in vitro compared to the si-NC group. As analyzed by mirco-CT, tail vein injected si-AW011738 OVX mice were less osteoporotic than the control group. AW011738 inhibited osteoblast differentiation by regulating TREM1 expression through microRNA. Meanwhile, overexpression of miR-24-2-5p inhibited TREM1 expression to promote osteoblast differentiation. SIGNIFICANCE Osteoclast-derived exosomes containing lncRNA AW011738 inhibit osteogenesis in MC3T3-E1 cells through the lncRNA AW011738/miR-24-2-5p/TREM1 axis and exacerbate osteoporosis in OVX mice.
Collapse
Affiliation(s)
- Jingcheng Liu
- Department of Orthopedics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Binyu Wang
- Department of Orthopedics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Hongtao Chen
- Department Of Orthopedics, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu 210008, China.
| | - Xiao Yu
- Department of Orthopedics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Xiaojian Cao
- Department of Orthopedics, The First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| | - Hongxiu Zhang
- Department of Obstetrics and Gynecology, the First Affiliated Hospital with Nanjing Medical University, Nanjing, Jiangsu 210029, China.
| |
Collapse
|
17
|
Weidner H, Baschant U, Ledesma-Colunga MG, Basiak K, Tsourdi E, Sockel K, Götze KS, Rivière J, Platzbecker U, Hofbauer LC, Rauner M. Bone marrow transplantation reduces FGF-23 levels and restores bone formation in myelodysplastic neoplasms. Leukemia 2024; 38:1853-1857. [PMID: 38906963 PMCID: PMC11286507 DOI: 10.1038/s41375-024-02315-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/11/2024] [Accepted: 06/12/2024] [Indexed: 06/23/2024]
Affiliation(s)
- Heike Weidner
- Department of Medicine III & Center for Healthy Aging, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III & Center for Healthy Aging, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Maria G Ledesma-Colunga
- Department of Medicine III & Center for Healthy Aging, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Karolina Basiak
- Department of Medicine III & Center for Healthy Aging, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Elena Tsourdi
- Department of Medicine III & Center for Healthy Aging, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Katja Sockel
- Department of Medicine I, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Katharina S Götze
- Department of Medicine III, Technical University of Munich, School of Medicine and Health, Munich, Germany
- German Cancer Consortium (DKTK), partner sites Dresden and Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Bavarian Center for Cancer Research (BZKF), Munich, Germany
| | - Jennifer Rivière
- Department of Medicine III, Technical University of Munich, School of Medicine and Health, Munich, Germany
| | - Uwe Platzbecker
- German Cancer Consortium (DKTK), partner sites Dresden and Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Medical Clinic and Policlinic I, Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Lorenz C Hofbauer
- Department of Medicine III & Center for Healthy Aging, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
- German Cancer Consortium (DKTK), partner sites Dresden and Munich and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Martina Rauner
- Department of Medicine III & Center for Healthy Aging, Medical Faculty and University Hospital Carl Gustav Carus, Dresden University of Technology, Dresden, Germany.
| |
Collapse
|
18
|
Petrone G, Turker I, Natarajan P, Bolton KL. Clinical and Therapeutic Implications of Clonal Hematopoiesis. Annu Rev Genomics Hum Genet 2024; 25:329-351. [PMID: 39190914 DOI: 10.1146/annurev-genom-120722-100409] [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: 08/29/2024]
Abstract
Clonal hematopoiesis (CH) is an age-related process whereby hematopoietic stem and progenitor cells (HSPCs) acquire mutations that lead to a proliferative advantage and clonal expansion. The most commonly mutated genes are epigenetic regulators, DNA damage response genes, and splicing factors, which are essential to maintain functional HSPCs and are frequently involved in the development of hematologic malignancies. Established risk factors for CH, including age, prior cytotoxic therapy, and smoking, increase the risk of acquiring CH and/or may increase CH fitness. CH has emerged as a novel risk factor in many age-related diseases, such as hematologic malignancies, cardiovascular disease, diabetes, and autoimmune disorders, among others. Future characterization of the mechanisms driving CH evolution will be critical to develop preventative and therapeutic approaches.
Collapse
Affiliation(s)
- Giulia Petrone
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Isik Turker
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Pradeep Natarajan
- Cardiovascular Research Center and Center for Genomic Medicine, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Kelly L Bolton
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
19
|
Singh J, Li N, Ashrafi E, Thao LTP, Curtis DJ, Wood EM, McQuilten ZK. Clonal hematopoiesis of indeterminate potential as a prognostic factor: a systematic review and meta-analysis. Blood Adv 2024; 8:3771-3784. [PMID: 38838228 PMCID: PMC11298876 DOI: 10.1182/bloodadvances.2024013228] [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: 03/21/2024] [Revised: 05/06/2024] [Accepted: 05/20/2024] [Indexed: 06/07/2024] Open
Abstract
ABSTRACT With advances in sequencing, individuals with clonal hematopoiesis of indeterminate potential (CHIP) are increasingly being identified, making it essential to understand its prognostic implications. We conducted a systematic review of studies comparing the risk of clinical outcomes in individuals with and without CHIP. We searched MEDLINE and EMBASE and included original research reporting an outcome risk measure in individuals with CHIP, adjusted for the effect of age. From the 3305 studies screened, we included 88 studies with 45 to 470 960 participants. Most studies had a low-to-moderate risk of bias in all domains of the Quality in Prognostic Factor Studies tool. Random-effects meta-analyses were performed for outcomes reported in at least 3 studies. CHIP conferred an increased risk of all-cause mortality (hazard ratio [HR], 1.34; 95% confidence interval, 1.19-1.50), cancer mortality (HR, 1.46; 1.13-1.88), composite cardiovascular events (HR, 1.40; 1.19-1.65), coronary heart disease (HR, 1.76; 1.27-2.44), stroke (HR, 1.16; 1.05-1.28), heart failure (HR, 1.27; 1.15-1.41), hematologic malignancy (HR, 4.28; 2.29-7.98), lung cancer (HR, 1.40; 1.27-1.54), renal impairment (HR, 1.25; 1.18-1.33) and severe COVID-19 (odds ratio [OR], 1.46; 1.18-1.80). CHIP was not associated with cardiovascular mortality (HR, 1.09; 0.97-1.22), except in the subgroup analysis restricted to larger clones (HR, 1.31; 1.12-1.54). Isolated DNMT3A mutations did not increase the risk of myeloid malignancy, all-cause mortality, or renal impairment. The reasons for heterogeneity between studies included differences in definitions and measurements of CHIP and the outcomes, and populations studied. In summary, CHIP is associated with diverse clinical outcomes, with clone size, specific gene, and inherent patient characteristics important mediators of risk.
Collapse
Affiliation(s)
- Jasmine Singh
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
- Department of Haematology, Fiona Stanley Hospital, Perth, Australia
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
| | - Nancy Li
- Department of Haematology, Eastern Health, Melbourne, Australia
| | - Elham Ashrafi
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - Le Thi Phuong Thao
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
| | - David J. Curtis
- Australian Centre for Blood Diseases, Monash University, Melbourne, Australia
- Department of Clinical Haematology, Alfred Health, Melbourne, Australia
| | - Erica M. Wood
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
- Department of Haematology, Monash Health, Melbourne, Australia
| | - Zoe K. McQuilten
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Australia
- Department of Haematology, Monash Health, Melbourne, Australia
| |
Collapse
|
20
|
Heimlich JB, Bhat P, Parker AC, Jenkins MT, Vlasschaert C, Ulloa J, Van Amburg JC, Potts CR, Olson S, Silver AJ, Ahmad A, Sharber B, Brown D, Hu N, van Galen P, Savona MR, Bick AG, Ferrell PB. Multiomic profiling of human clonal hematopoiesis reveals genotype and cell-specific inflammatory pathway activation. Blood Adv 2024; 8:3665-3678. [PMID: 38507736 PMCID: PMC11284682 DOI: 10.1182/bloodadvances.2023011445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 02/08/2024] [Accepted: 02/08/2024] [Indexed: 03/22/2024] Open
Abstract
ABSTRACT Clonal hematopoiesis (CH) is an age-associated phenomenon that increases the risk of hematologic malignancy and cardiovascular disease. CH is thought to enhance disease risk through inflammation in the peripheral blood.1 Here, we profile peripheral blood gene expression in 66 968 single cells from a cohort of 17 patients with CH and 7 controls. Using a novel mitochondrial DNA barcoding approach, we were able to identify and separately compare mutant Tet methylcytosine dioxygenase 2 (TET2) and DNA methyltransferase 3A (DNMT3A) cells with nonmutant counterparts. We discovered the vast majority of mutated cells were in the myeloid compartment. Additionally, patients harboring DNMT3A and TET2 CH mutations possessed a proinflammatory profile in CD14+ monocytes through previously unrecognized pathways such as galectin and macrophage inhibitory factor. We also found that T cells from patients with CH, although mostly unmutated, had decreased expression of GTPase of the immunity associated protein genes, which are critical to T-cell development, suggesting that CH impairs T-cell function.
Collapse
Affiliation(s)
- J. Brett Heimlich
- Division of Cardiovascular Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Pawan Bhat
- Vanderbilt University School of Medicine, Nashville, TN
| | | | | | | | - Jessica Ulloa
- Division of Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Joseph C. Van Amburg
- Division of Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Chad R. Potts
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Sydney Olson
- Division of Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | | | - Ayesha Ahmad
- Division of Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Brian Sharber
- Division of Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Donovan Brown
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Ningning Hu
- Division of Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Peter van Galen
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Ludwig Center at Harvard, Harvard Medical School, Boston, MA
| | - Michael R. Savona
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt-Ingram Cancer Center, Program in Cancer Biology, and Center for Immunobiology, Nashville, TN
- Center for Immunobiology, Vanderbilt University School of Medicine, Nashville, TN
| | - Alexander G. Bick
- Division of Genomic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - P. Brent Ferrell
- Division of Hematology and Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
21
|
Wang H, Divaris K, Pan B, Li X, Lim JH, Saha G, Barovic M, Giannakou D, Korostoff JM, Bing Y, Sen S, Moss K, Wu D, Beck JD, Ballantyne CM, Natarajan P, North KE, Netea MG, Chavakis T, Hajishengallis G. Clonal hematopoiesis driven by mutated DNMT3A promotes inflammatory bone loss. Cell 2024; 187:3690-3711.e19. [PMID: 38838669 PMCID: PMC11246233 DOI: 10.1016/j.cell.2024.05.003] [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: 08/23/2023] [Revised: 02/19/2024] [Accepted: 05/01/2024] [Indexed: 06/07/2024]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) arises from aging-associated acquired mutations in hematopoietic progenitors, which display clonal expansion and produce phenotypically altered leukocytes. We associated CHIP-DNMT3A mutations with a higher prevalence of periodontitis and gingival inflammation among 4,946 community-dwelling adults. To model DNMT3A-driven CHIP, we used mice with the heterozygous loss-of-function mutation R878H, equivalent to the human hotspot mutation R882H. Partial transplantation with Dnmt3aR878H/+ bone marrow (BM) cells resulted in clonal expansion of mutant cells into both myeloid and lymphoid lineages and an elevated abundance of osteoclast precursors in the BM and osteoclastogenic macrophages in the periphery. DNMT3A-driven clonal hematopoiesis in recipient mice promoted naturally occurring periodontitis and aggravated experimentally induced periodontitis and arthritis, associated with enhanced osteoclastogenesis, IL-17-dependent inflammation and neutrophil responses, and impaired regulatory T cell immunosuppressive activity. DNMT3A-driven clonal hematopoiesis and, subsequently, periodontitis were suppressed by rapamycin treatment. DNMT3A-driven CHIP represents a treatable state of maladaptive hematopoiesis promoting inflammatory bone loss.
Collapse
Affiliation(s)
- Hui Wang
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kimon Divaris
- Division of Pediatric and Public Health, Adams School of Dentistry, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA; Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Bohu Pan
- Division of Bioinformatics and Biostatistics, National Center for Toxicological Research, US Food and Drug Administration, Jefferson, AR 72079, USA
| | - Xiaofei Li
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Shanghai Jiao Tong University, School of Life Sciences and Biotechnology, Sheng Yushou Center of Cell Biology and Immunology, Shanghai 200240, China
| | - Jong-Hyung Lim
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gundappa Saha
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Marko Barovic
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität Dresden, 01307 Dresden, Germany
| | - Danai Giannakou
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität Dresden, 01307 Dresden, Germany
| | - Jonathan M Korostoff
- Department of Periodontics, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yu Bing
- Human Genetics Center, Department of Epidemiology, School of Public Health, The University of Texas Health Science Center at Houston, Houston, TX 77030, USA
| | - Souvik Sen
- Department of Neurology, University of South Carolina, Columbia, SC 29209, USA; Center for the Study of Aphasia Recovery, University of South Carolina, Columbia, SC 29209, USA
| | - Kevin Moss
- Department of Biostatistics and Health Data Sciences, School of Medicine, Indiana University, Indianapolis, IN 46202, USA; Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Di Wu
- Division of Oral and Craniofacial Health Sciences, Adams School of Dentistry, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA; Department of Biostatistics, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - James D Beck
- Division of Comprehensive Oral Health-Periodontology, Adams School of Dentistry, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | | | - Pradeep Natarajan
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Program in Medical and Population Genetics, Broad Institute of Harvard and MIT, Cambridge, MA 02141, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Kari E North
- Department of Epidemiology, Gillings School of Global Public Health, University of North Carolina, Chapel Hill, Chapel Hill, NC 27599, USA
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, 6525 XZ Nijmegen, the Netherlands; Department of Immunology and Metabolism, LIMES, University of Bonn, 53115 Bonn, Germany
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital, Technische Universität Dresden, 01307 Dresden, Germany
| | - George Hajishengallis
- Department of Basic and Translational Sciences, Laboratory of Innate Immunity and Inflammation, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
22
|
Lyu T, Qiu X, Wang Y, Zhang L, Dai Y, Wang X, Zhao S, Xiang M, Cui L, Cheng S, Liu Y, Gu H, Jiang Y, Meng X, Wang Y, Zhao X, Wang X, Li Q, Wang M, Jiang Y, Xu Z, Huang X, Li H, Wang Y, Li Z. DNMT3A dysfunction promotes neuroinflammation and exacerbates acute ischemic stroke. MedComm (Beijing) 2024; 5:e652. [PMID: 39006763 PMCID: PMC11246610 DOI: 10.1002/mco2.652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 05/18/2024] [Accepted: 05/30/2024] [Indexed: 07/16/2024] Open
Abstract
Somatic mutations related to clonal hematopoiesis of indeterminate potential (CHIP) are risk factors for stroke. The impact of DNMT3A, the most mutated gene in CHIP, on clinical functional outcomes of acute ischemic stroke (AIS) remains unclear. In a well-characterized cohort of 8524 ischemic stroke patients, we demonstrated that DNMT3A-driven CHIP was significantly associated with neurological disability in these patients. With a stroke mouse model of transient middle cerebral artery occlusion (tMCAO), we demonstrated that DNMT3A protein levels in the brain penumbra increased. The DNMT3A inhibitor RG108 administration amplified neutrophil proliferation in the blood, promoted neutrophil infiltration into the brain penumbra, and exaggerated proinflammatory activation in tMCAO male mice. DNMT3A inhibition also significantly increased infarct volume and worsened neurobehavioral function in tMCAO male mice. In conclusion, DNMT3A somatic mutations are associated with worsened neurological disability in some patients with AIS, potentially through increased neutrophil proliferation and infiltration in the ischemic brain region. These findings suggest a possible mechanism for proinflammatory activation and tissue damage in the affected brain tissue, highlighting the need for further research in this area.
Collapse
|
23
|
Maeda H, Kakiuchi N. Clonal expansion in normal tissues. Cancer Sci 2024; 115:2117-2124. [PMID: 38623936 PMCID: PMC11247609 DOI: 10.1111/cas.16183] [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: 02/07/2024] [Revised: 03/24/2024] [Accepted: 04/01/2024] [Indexed: 04/17/2024] Open
Abstract
Cancer originates from a single ancestral cell that acquires a driver mutation, which confers a growth or survival advantage, followed by the acquisition of additional driver mutations by descendant cells. Recently, it has become evident that somatic cell mutations accumulate in normal tissues with aging and exposure to environmental factors, such as alcohol, smoking, and UV rays, increases the mutation rate. Clones harboring driver mutations expand with age, leading to tissue remodeling. Lineage analysis of myeloproliferative neoplasms and der(1;16)-positive breast cancer revealed that driver mutations were acquired early in our lives and that the development of cancer takes decades, unveiling the previously unknown early process of cancer development. Evidence that clonal hematopoiesis affects various diseases, including nonneoplastic diseases, highlights the potential role of the identification and functional analysis of mutated clones in unraveling unknown pathologies. In this review, we summarize the recent updates on clonal expansion in normal tissues and the natural history of cancer revealed through lineage analysis of noncancerous and cancerous tissues.
Collapse
Affiliation(s)
- Hirona Maeda
- Department of Pathology and Tumor Biology, Graduate School of MedicineKyoto UniversityKyotoJapan
- Department of Diagnostic PathologyKyoto University HospitalKyotoJapan
| | - Nobuyuki Kakiuchi
- Department of Pathology and Tumor Biology, Graduate School of MedicineKyoto UniversityKyotoJapan
- The Hakubi Center for Advanced ResearchKyoto UniversityKyotoJapan
- Department of Gastroenterology and Hepatology, Graduate School of MedicineKyoto UniversityKyotoJapan
| |
Collapse
|
24
|
Mack T, Vlasschaert C, von Beck K, Silver AJ, Heimlich JB, Poisner H, Condon HR, Ulloa J, Sochacki AL, Spaulding TP, Kishtagari A, Bejan CA, Xu Y, Savona MR, Jones A, Bick AG. Cost-Effective and Scalable Clonal Hematopoiesis Assay Provides Insight into Clonal Dynamics. J Mol Diagn 2024; 26:563-573. [PMID: 38588769 PMCID: PMC11536471 DOI: 10.1016/j.jmoldx.2024.03.007] [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: 11/09/2023] [Revised: 03/06/2024] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is a common age-related phenomenon in which hematopoietic stem cells acquire mutations in a select set of genes commonly mutated in myeloid neoplasia which then expand clonally. Current sequencing assays to detect CHIP mutations are not optimized for the detection of these variants and can be cost-prohibitive when applied to large cohorts or to serial sequencing. In this study, an affordable (approximately US $8 per sample), accurate, and scalable sequencing assay for CHIP is introduced and validated. The efficacy of the assay was demonstrated by identifying CHIP mutations in a cohort of 456 individuals with DNA collected at multiple time points in Vanderbilt University's biobank and quantifying clonal expansion rates over time. A total of 101 individuals with CHIP/clonal cytopenia of undetermined significance were identified, and individual-level clonal expansion rate was calculated using the variant allele fraction at both time points. Differences in clonal expansion rate by driver gene were observed, but there was also significant individual-level heterogeneity, emphasizing the multifactorial nature of clonal expansion. Additionally, mutation co-occurrence and clonal competition between multiple driver mutations were explored.
Collapse
Affiliation(s)
- Taralynn Mack
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Kelly von Beck
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Alexander J Silver
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - J Brett Heimlich
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hannah Poisner
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Henry R Condon
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Jessica Ulloa
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Andrew L Sochacki
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Travis P Spaulding
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Ashwin Kishtagari
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Cosmin A Bejan
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Yaomin Xu
- Department of Biomedical Informatics, Vanderbilt University Medical Center, Nashville, Tennessee; Department of Biostatistics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Michael R Savona
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, Tennessee; Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, Tennessee; Center for Immunobiology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Angela Jones
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Alexander G Bick
- Vanderbilt Genetics Institute, Vanderbilt University School of Medicine, Nashville, Tennessee; Division of Genetic Medicine, Vanderbilt University Medical Center, Nashville, Tennessee.
| |
Collapse
|
25
|
Vlasschaert C, Pan Y, Chen J, Akwo E, Rao V, Hixson JE, Chong M, Uddin MM, Yu Z, Jiang M, Peng F, Cao S, Wang Y, Kim DK, Hung AM, He J, Tamura MK, Cohen DL, He J, Li C, Bhat Z, Rao P, Xie D, Bick AG, Kestenbaum B, Paré G, Rauh MJ, Levin A, Natarajan P, Lash JP, Zhang MZ, Harris RC, Robinson-Cohen C, Lanktree MB, Kelly TN. Clonal hematopoiesis of indeterminate potential contributes to accelerated chronic kidney disease progression. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.19.24309181. [PMID: 38946975 PMCID: PMC11213124 DOI: 10.1101/2024.06.19.24309181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Clonal hematopoiesis of indeterminate potential (CHIP) is a common inflammatory condition of aging that causes myriad end-organ damage. We have recently shown associations for CHIP with acute kidney injury and with kidney function decline in the general population, with stronger associations for CHIP driven by mutations in genes other than DNMT3A (non- DNMT3A CHIP). Longitudinal kidney function endpoints in individuals with pre-existing chronic kidney disease (CKD) and CHIP have been examined in two previous studies, which reported conflicting findings and were limited by small sample sizes. Methods In this study, we examined the prospective associations between CHIP and CKD progression events in four cohorts of CKD patients (total N = 5,772). The primary outcome was a composite of 50% kidney function decline or kidney failure. The slope of eGFR decline was examined as a secondary outcome. Mendelian randomization techniques were then used to investigate potential causal effects of CHIP on eGFR decline. Finally, kidney function was assessed in adenine-fed CKD model mice having received a bone marrow transplant recapitulating Tet2 -CHIP compared to controls transplanted wild-type bone marrow. Results Across all cohorts, the average age was 66.4 years, the average baseline eGFR was 42.6 ml/min/1.73m 2 , and 24% had CHIP. Upon meta-analysis, non- DNMT3A CHIP was associated with a 59% higher relative risk of incident CKD progression (HR 1.59, 95% CI: 1.02-2.47). This association was more pronounced among individuals with diabetes (HR 1.29, 95% CI: 1.03-1.62) and with baseline eGFR ≥ 30 ml/min/1.73m (HR 1.80, 95% CI: 1.11-2.90). Additionally, the annualized slope of eGFR decline was steeper among non- DNMT3A CHIP carriers, relative to non-carriers (β -0.61 ± 0.31 ml/min/1.73m 2 , p = 0.04). Mendelian randomization analyses suggested a causal role for CHIP in eGFR decline among individuals with diabetes. In a dietary adenine mouse model of CKD, Tet2 -CHIP was associated with lower GFR as well as greater kidney inflammation, tubular injury, and tubulointerstitial fibrosis. Conclusion Non- DNMT3A CHIP is a potentially targetable novel risk factor for CKD progression.
Collapse
|
26
|
Arends CM, Kopp K, Hablesreiter R, Estrada N, Christen F, Moll UM, Zeillinger R, Schmitt WD, Sehouli J, Kulbe H, Fleischmann M, Ray-Coquard I, Zeimet A, Raspagliesi F, Zamagni C, Vergote I, Lorusso D, Concin N, Bullinger L, Braicu EI, Damm F. Dynamics of clonal hematopoiesis under DNA-damaging treatment in patients with ovarian cancer. Leukemia 2024; 38:1378-1389. [PMID: 38637689 PMCID: PMC11147769 DOI: 10.1038/s41375-024-02253-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 04/20/2024]
Abstract
Clonal hematopoiesis (CH) driven by mutations in the DNA damage response (DDR) pathway is frequent in patients with cancer and is associated with a higher risk of therapy-related myeloid neoplasms (t-MNs). Here, we analyzed 423 serial whole blood and plasma samples from 103 patients with relapsed high-grade ovarian cancer receiving carboplatin, poly(ADP-ribose) polymerase inhibitor (PARPi) and heat shock protein 90 inhibitor (HSP90i) treatment within the phase II EUDARIO trial using error-corrected sequencing of 72 genes. DDR-driven CH was detected in 35% of patients and was associated with longer duration of prior PARPi treatment. TP53- and PPM1D-mutated clones exhibited substantially higher clonal expansion rates than DNMT3A- or TET2-mutated clones during treatment. Expansion of DDR clones correlated with HSP90i exposure across the three study arms and was partially abrogated by the presence of germline mutations related to homologous recombination deficiency. Single-cell DNA sequencing of selected samples revealed clonal exclusivity of DDR mutations, and identified DDR-mutated clones as the origin of t-MN in two investigated cases. Together, these results provide unique insights into the architecture and the preferential selection of DDR-mutated hematopoietic clones under intense DNA-damaging treatment. Specifically, PARPi and HSP90i therapies pose an independent risk for the expansion of DDR-CH in a dose-dependent manner.
Collapse
Affiliation(s)
- Christopher Maximilian Arends
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Klara Kopp
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Raphael Hablesreiter
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Natalia Estrada
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Friederike Christen
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Ute Martha Moll
- Department of Pathology, Stony Brook University Cancer Center, Stony Brook, NY, 11794, USA
| | - Robert Zeillinger
- Department of Obstetrics and Gynaecology, Molecular Oncology Group, Comprehensive Cancer Center-Gynaecologic Cancer Unit, Medical University of Vienna, Vienna, Austria
| | - Wolfgang Daniel Schmitt
- Department of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin, Berlin, Germany
| | - Jalid Sehouli
- Department of Gynaecology, European Competence Center for Ovarian Cancer, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow Klinikum, Berlin, Germany
- North Eastern German Society for Gynecological Cancer. Tumor Bank Ovarian Cancer Network, Berlin, Germany
| | - Hagen Kulbe
- Department of Gynaecology, European Competence Center for Ovarian Cancer, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow Klinikum, Berlin, Germany
- North Eastern German Society for Gynecological Cancer. Tumor Bank Ovarian Cancer Network, Berlin, Germany
| | - Maximilian Fleischmann
- Klinik für Innere Medizin II, Abteilung Hämatologie und Onkologie, Universitätsklinikum Jena, Jena, Germany
| | - Isabelle Ray-Coquard
- Centre Anticancereux Léon Bérard, University Claude Bernard Lyon, GINECO Group, Lyon, France
| | - Alain Zeimet
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Austrian AGO, Innsbruck, Austria
| | | | - Claudio Zamagni
- Division of Oncology, IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Ignace Vergote
- Division of Gynecological Oncology, Department of Gynecology and Obstetrics, Leuven Cancer Institute, Katholieke Universiteit Leuven, Leuven, Belgium
- Belgium and Luxembourg Gynaecological Oncology Group (BGOG), Leuven, Belgium
| | | | - Nicole Concin
- Department of Obstetrics and Gynecology, Medical University of Innsbruck, Austrian AGO, Innsbruck, Austria
| | - Lars Bullinger
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Elena Ioana Braicu
- Department of Gynaecology, European Competence Center for Ovarian Cancer, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Campus Virchow Klinikum, Berlin, Germany
- North Eastern German Society for Gynecological Cancer. Tumor Bank Ovarian Cancer Network, Berlin, Germany
| | - Frederik Damm
- Department of Hematology, Oncology, and Cancer Immunology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
27
|
Winter S, Götze KS, Hecker JS, Metzeler KH, Guezguez B, Woods K, Medyouf H, Schäffer A, Schmitz M, Wehner R, Glauche I, Roeder I, Rauner M, Hofbauer LC, Platzbecker U. Clonal hematopoiesis and its impact on the aging osteo-hematopoietic niche. Leukemia 2024; 38:936-946. [PMID: 38514772 PMCID: PMC11073997 DOI: 10.1038/s41375-024-02226-6] [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: 11/16/2023] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/23/2024]
Abstract
Clonal hematopoiesis (CH) defines a premalignant state predominantly found in older persons that increases the risk of developing hematologic malignancies and age-related inflammatory diseases. However, the risk for malignant transformation or non-malignant disorders is variable and difficult to predict, and defining the clinical relevance of specific candidate driver mutations in individual carriers has proved to be challenging. In addition to the cell-intrinsic mechanisms, mutant cells rely on and alter cell-extrinsic factors from the bone marrow (BM) niche, which complicates the prediction of a mutant cell's fate in a shifting pre-malignant microenvironment. Therefore, identifying the insidious and potentially broad impact of driver mutations on supportive niches and immune function in CH aims to understand the subtle differences that enable driver mutations to yield different clinical outcomes. Here, we review the changes in the aging BM niche and the emerging evidence supporting the concept that CH can progressively alter components of the local BM microenvironment. These alterations may have profound implications for the functionality of the osteo-hematopoietic niche and overall bone health, consequently fostering a conducive environment for the continued development and progression of CH. We also provide an overview of the latest technology developments to study the spatiotemporal dependencies in the CH BM niche, ideally in the context of longitudinal studies following CH over time. Finally, we discuss aspects of CH carrier management in clinical practice, based on work from our group and others.
Collapse
Affiliation(s)
- Susann Winter
- Department of Internal Medicine I, University Hospital Carl Gustav Carus, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Katharina S Götze
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine III, Technical University of Munich (TUM), School of Medicine and Health, Munich, Germany
- German MDS Study Group (D-MDS), Leipzig, Germany
| | - Judith S Hecker
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Medicine III, Technical University of Munich (TUM), School of Medicine and Health, Munich, Germany
- TranslaTUM, Center for Translational Cancer Research, Technical University of Munich (TUM), Munich, Germany
| | - Klaus H Metzeler
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University of Leipzig Medical Center, Leipzig, Germany
| | - Borhane Guezguez
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology and Oncology, University Medical Center Mainz, Mainz, Germany
| | - Kevin Woods
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Department of Hematology and Oncology, University Medical Center Mainz, Mainz, Germany
| | - Hind Medyouf
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
- Frankfurt Cancer Institute, Frankfurt am Main, Germany
| | - Alexander Schäffer
- Institute for Tumor Biology and Experimental Therapy, Georg-Speyer-Haus, Frankfurt am Main, Germany
| | - Marc Schmitz
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Rebekka Wehner
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
| | - Ingmar Glauche
- Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Ingo Roeder
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany
- National Center for Tumor Diseases (NCT), Dresden, Germany: German Cancer Research Center (DKFZ), Heidelberg, Germany; Faculty of Medicine and University Hospital Carl Gustav Carus, TU Dresden, Dresden, Germany; Helmholtz-Zentrum Dresden-Rossendorf (HZDR), Dresden, Germany
- Institute for Medical Informatics and Biometry, Faculty of Medicine Carl Gustav Carus, TU Dresden, Dresden, Germany
| | - Martina Rauner
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine III, and Center for Healthy Aging, University Medical Center, TU Dresden, Dresden, Germany
| | - Lorenz C Hofbauer
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- Division of Endocrinology, Diabetes and Bone Diseases, Department of Medicine III, and Center for Healthy Aging, University Medical Center, TU Dresden, Dresden, Germany.
| | - Uwe Platzbecker
- German Cancer Consortium (DKTK), CHOICE Consortium, Partner Sites Dresden/Munich/Frankfurt/Mainz, and German Cancer Research Center (DKFZ), Heidelberg, Germany.
- German MDS Study Group (D-MDS), Leipzig, Germany.
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University of Leipzig Medical Center, Leipzig, Germany.
| |
Collapse
|
28
|
Kapadia CD, Goodell MA. Tissue mosaicism following stem cell aging: blood as an exemplar. NATURE AGING 2024; 4:295-308. [PMID: 38438628 DOI: 10.1038/s43587-024-00589-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Accepted: 02/07/2024] [Indexed: 03/06/2024]
Abstract
Loss of stem cell regenerative potential underlies aging of all tissues. Somatic mosaicism, the emergence of cellular patchworks within tissues, increases with age and has been observed in every organ yet examined. In the hematopoietic system, as in most tissues, stem cell aging through a variety of mechanisms occurs in lockstep with the emergence of somatic mosaicism. Here, we draw on insights from aging hematopoiesis to illustrate fundamental principles of stem cell aging and somatic mosaicism. We describe the generalizable changes intrinsic to aged stem cells and their milieu that provide the backdrop for somatic mosaicism to emerge. We discuss genetic and nongenetic mechanisms that can result in tissue somatic mosaicism and existing methodologies to detect such clonal outgrowths. Finally, we propose potential avenues to modify mosaicism during aging, with the ultimate aim of increasing tissue resiliency.
Collapse
Affiliation(s)
- Chiraag D Kapadia
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Margaret A Goodell
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA.
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
29
|
Bucala R, Tsao BP. The Emerging Spectrum of Somatic Mutation in Rheumatic Disease: Clonal Hematopoiesis Connects Aging With Giant Cell Arteritis. Arthritis Rheumatol 2024; 76:351-353. [PMID: 37961826 PMCID: PMC11253228 DOI: 10.1002/art.42745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 11/01/2023] [Indexed: 11/15/2023]
Affiliation(s)
| | - Betty P Tsao
- Medical University of South Carolina, Charleston
| |
Collapse
|
30
|
Jiang Z, Qi G, He X, Yu Y, Cao Y, Zhang C, Zou W, Yuan H. Ferroptosis in Osteocytes as a Target for Protection Against Postmenopausal Osteoporosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307388. [PMID: 38233202 PMCID: PMC10966575 DOI: 10.1002/advs.202307388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 12/18/2023] [Indexed: 01/19/2024]
Abstract
Ferroptosis is a necrotic form of iron-dependent regulatory cell death. Estrogen withdrawal can interfere with iron metabolism, which is responsible for the pathogenesis of postmenopausal osteoporosis (PMOP). Here, it is demonstrated that estrogen withdrawal induces iron accumulation in the skeleton and the ferroptosis of osteocytes, leading to reduced bone mineral density. Furthermore, the facilitatory effect of ferroptosis of osteocytes is verified in the occurrence and development of postmenopausal osteoporosis is associated with over activated osteoclastogenesis using a direct osteocyte/osteoclast coculture system and glutathione peroxidase 4 (GPX4) knockout ovariectomized mice. In addition, the nuclear factor erythroid derived 2-related factor-2 (Nrf2) signaling pathway is confirmed to be a crucial factor in the ferroptosis of osteocytic cells. Nrf2 regulates the expression of nuclear factor kappa-B ligand (RANKL) by regulating the DNA methylation level of the RANKL promoter mediated by DNA methyltransferase 3a (Dnmt3a), which is as an important mechanism in osteocytic ferroptosis-mediated osteoclastogenesis. Taken together, this data suggests that osteocytic ferroptosis is involved in PMOP and can be targeted to tune bone homeostasis.
Collapse
Affiliation(s)
- Zengxin Jiang
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Guobin Qi
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Xuecheng He
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yifan Yu
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Yuting Cao
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Changqing Zhang
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| | - Weiguo Zou
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
- State Key Laboratory of Cell BiologyCAS Center for Excellence in Molecular Cell SciencesShanghai Institute of Biochemistry and Cell BiologyChinese Academy of SciencesUniversity of Chinese Academy of SciencesShanghai200031China
| | - Hengfeng Yuan
- Department of OrthopaedicsShanghai Jiaotong University Affiliated Sixth People's HospitalNo. 600 Yishan RoadShanghai200233China
- Institute of Microsurgery on ExtremitiesShanghai Jiao Tong University Affiliated Sixth People's HospitalShanghai200233China
| |
Collapse
|
31
|
Vlasschaert C, Lanktree MB, Rauh MJ, Kelly TN, Natarajan P. Clonal haematopoiesis, ageing and kidney disease. Nat Rev Nephrol 2024; 20:161-174. [PMID: 37884787 PMCID: PMC10922936 DOI: 10.1038/s41581-023-00778-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/29/2023] [Indexed: 10/28/2023]
Abstract
Clonal haematopoiesis of indeterminate potential (CHIP) is a preclinical condition wherein a sizeable proportion of an individual's circulating blood cells are derived from a single mutated haematopoietic stem cell. CHIP occurs frequently with ageing - more than 10% of individuals over 65 years of age are affected - and is associated with an increased risk of disease across several organ systems and premature death. Emerging evidence suggests that CHIP has a role in kidney health, including associations with predisposition to acute kidney injury, impaired recovery from acute kidney injury and kidney function decline, both in the general population and among those with chronic kidney disease. Beyond its direct effect on the kidney, CHIP elevates the susceptibility of individuals to various conditions that can detrimentally affect the kidneys, including cardiovascular disease, obesity and insulin resistance, liver disease, gout, osteoporosis and certain autoimmune diseases. Aberrant pro-inflammatory signalling, telomere attrition and epigenetic ageing are potential causal pathophysiological pathways and mediators that underlie CHIP-related disease risk. Experimental animal models have shown that inhibition of inflammatory cytokine signalling can ameliorate many of the pathological effects of CHIP, and assessment of the efficacy and safety of this class of medications for human CHIP-associated pathology is ongoing.
Collapse
Affiliation(s)
| | - Matthew B Lanktree
- Department of Medicine and Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
- St. Joseph's Healthcare Hamilton, Hamilton, Ontario, Canada
- Population Health Research Institute, Hamilton, Ontario, Canada
| | - Michael J Rauh
- Department of Pathology and Molecular Medicine, Kingston, Ontario, Canada
| | - Tanika N Kelly
- Division of Nephrology, Department of Medicine, College of Medicine, University of Illinois at Chicago, Chicago, IL, USA
| | - Pradeep Natarajan
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Program in Medical and Population Genetics and Cardiovascular Disease Initiative, Broad Institute of Harvard and MIT, Cambridge, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
32
|
Liu C, Zhou YP, Lian TY, Li RN, Ma JS, Yang YJ, Zhang SJ, Li XM, Qiu LH, Qiu BC, Ren LY, Wang J, Han ZY, Li JH, Wang L, Xu XQ, Sun K, Chen LF, Cheng CY, Zhang ZJ, Jing ZC. Clonal Hematopoiesis of Indeterminate Potential in Chronic Thromboembolic Pulmonary Hypertension: A Multicenter Study. Hypertension 2024; 81:372-382. [PMID: 38116660 DOI: 10.1161/hypertensionaha.123.22274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Accepted: 12/05/2023] [Indexed: 12/21/2023]
Abstract
BACKGROUND The pathogenesis of chronic thromboembolic pulmonary hypertension (CTEPH) is multifactorial and growing evidence has indicated that hematological disorders are involved. Clonal hematopoiesis of indeterminate potential (CHIP) has recently been associated with an increased risk of both hematological malignancies and cardiovascular diseases. However, the prevalence and clinical relevance of CHIP in patients with CTEPH remains unclear. METHODS Using stepwise calling on next-generation sequencing data from 499 patients with CTEPH referred to 3 centers between October 2006 and December 2021, CHIP mutations were identified. We associated CHIP with all-cause mortality in patients with CTEPH. To provide insights into potential mechanisms, the associations between CHIP and inflammatory markers were also determined. RESULTS In total, 47 (9.4%) patients with CTEPH carried at least 1 CHIP mutation at a variant allele frequency of ≥2%. The most common mutations were in DNMT3A, TET2, RUNX1, and ASXL1. During follow-up (mean, 55 months), deaths occurred in 22 (46.8%) and 104 (23.0%) patients in the CHIP and non-CHIP groups, respectively (P<0.001, log-rank test). The association of CHIP with mortality remained robust in the fully adjusted model (hazard ratio, 2.190 [95% CI, 1.257-3.816]; P=0.006). Moreover, patients with CHIP mutations showed higher circulating interleukin-1β and interleukin-6 and lower interleukin-4 and IgG galactosylation levels. CONCLUSIONS This is the first study to show that CHIP mutations occurred in 9.4% of patients with CTEPH are associated with a severe inflammatory state and confer a poorer prognosis in long-term follow-up.
Collapse
Affiliation(s)
- Chao Liu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu-Ping Zhou
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tian-Yu Lian
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China (T.-Y.L., S.-J.Z., C.-Y.C., Z.-C.J)
| | - Ruo-Nan Li
- School of Pharmacy, Henan University, Zhengzhou, China (R.-N.L., J.-S.M.)
| | - Jing-Si Ma
- School of Pharmacy, Henan University, Zhengzhou, China (R.-N.L., J.-S.M.)
| | - Yin-Jian Yang
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (Y.-J.Y., K.S., Z.-J.Z.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Si-Jin Zhang
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China (T.-Y.L., S.-J.Z., C.-Y.C., Z.-C.J)
| | - Xian-Mei Li
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lu-Hong Qiu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bao-Chen Qiu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Li-Yan Ren
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Wang
- Department of Medical Laboratory, Weifang Medical University, China (J.W.)
| | - Zhi-Yan Han
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital (Z.-Y.H., J.-H.L.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jing-Hui Li
- State Key Laboratory of Cardiovascular Disease, FuWai Hospital (Z.-Y.H., J.-H.L.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lan Wang
- Department of Cardio-Pulmonary Circulation, Shanghai Pulmonary Hospital, Tongji University, China (L.W.)
| | - Xi-Qi Xu
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Kai Sun
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (Y.-J.Y., K.S., Z.-J.Z.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lian-Feng Chen
- Department of Cardiology, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (C.L., Y.-P.Z., X.-M.L., L.-H.Q., B.-C.Q., L.-Y.R., X.-Q.X., L.-F.C.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chun-Yan Cheng
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China (T.-Y.L., S.-J.Z., C.-Y.C., Z.-C.J)
| | - Ze-Jian Zhang
- State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (Y.-J.Y., K.S., Z.-J.Z.), Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhi-Cheng Jing
- Department of Cardiology, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China (T.-Y.L., S.-J.Z., C.-Y.C., Z.-C.J)
| |
Collapse
|
33
|
Wang H, Yan H, Chen W, Tang H, Pei Y, Shan Q, Cang J, Miao C, Tan L, Tan L. Association of clonal haematopoiesis with severe postoperative complications in patients undergoing radical oesophagectomy. Br J Anaesth 2024; 132:277-284. [PMID: 38044238 DOI: 10.1016/j.bja.2023.10.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 10/11/2023] [Accepted: 10/15/2023] [Indexed: 12/05/2023] Open
Abstract
BACKGROUND Clonal haematopoiesis (CH) is an age-associated clonal expansion of blood cells driven by leukaemia-associated somatic mutations. Although CH has been reported to be a risk factor for leukaemia and a number of non-haematopoietic diseases, its role in perioperative medicine remains unexplored. METHODS This was a single-centre, prospective, observational study. Patients undergoing radical oesophagectomy were enrolled, and peripheral blood samples were collected for DNA sequencing. Patients with haematopoietic somatic mutations (variant allele frequencies ≥1%) in the DNMT3A gene, TET2 gene, or both were defined as CH carriers. The primary outcome was the incidence of severe postoperative complications (Clavien-Dindo classification ≥3). The secondary outcomes included the major types of postoperative complications, mortality, and other common perioperative variables. RESULTS Clonal haematopoiesis was found in 21.2% (33/156) of the patients (mean age: 66 yr [range: 26-79 yr]; 83% males). Some 14/33 (42.4%) patients with CH had severe postoperative complications, compared with patients without CH carriers (28/123 [22.8%]; P=0.024). Multivariable logistic regression analysis showed that CH was associated with an increased risk of developing severe postoperative complications (odds ratio, 3.63; 95% confidence interval, 1.37-9.66; P=0.010). Among the major postoperative complications, the incidence of pulmonary complications was significantly higher in the patients with CH than in those without CH (15 in 33 [45.5%] vs 30 in 123 [24.4%], P=0.018). CONCLUSIONS Clonal haematopoiesis was associated with a higher incidence of severe postoperative complications in patients undergoing radical oesophagectomy, suggesting that clonal haematopoiesis can play an important role in perioperative medicine. CLINICAL TRIAL REGISTRATION ChiCTR2100044175 (Chinese Clinical Trial Registry, http://www.chictr.org.cn/showproj.aspx?proj=123193).
Collapse
Affiliation(s)
- Hao Wang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Huan Yan
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wannan Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Han Tang
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yanzi Pei
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Qi Shan
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jing Cang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Changhong Miao
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lijie Tan
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Li Tan
- Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Centre, and Shanghai Key Laboratory of Medical Epigenetics, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
34
|
Su Y, Yu G, Li D, Lu Y, Ren C, Xu Y, Yang Y, Zhang K, Ma T, Li Z. Identification of mitophagy-related biomarkers in human osteoporosis based on a machine learning model. Front Physiol 2024; 14:1289976. [PMID: 38260098 PMCID: PMC10800828 DOI: 10.3389/fphys.2023.1289976] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 12/21/2023] [Indexed: 01/24/2024] Open
Abstract
Background: Osteoporosis (OP) is a chronic bone metabolic disease and a serious global public health problem. Several studies have shown that mitophagy plays an important role in bone metabolism disorders; however, its role in osteoporosis remains unclear. Methods: The Gene Expression Omnibus (GEO) database was used to download GSE56815, a dataset containing low and high BMD, and differentially expressed genes (DEGs) were analyzed. Mitochondrial autophagy-related genes (MRG) were downloaded from the existing literature, and highly correlated MRG were screened by bioinformatics methods. The results from both were taken as differentially expressed (DE)-MRG, and Gene Ontology (GO) analysis and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were performed. Protein-protein interaction network (PPI) analysis, support vector machine recursive feature elimination (SVM-RFE), and Boruta method were used to identify DE-MRG. A receiver operating characteristic curve (ROC) was drawn, a nomogram model was constructed to determine its diagnostic value, and a variety of bioinformatics methods were used to verify the relationship between these related genes and OP, including GO and KEGG analysis, IP pathway analysis, and single-sample Gene Set Enrichment Analysis (ssGSEA). In addition, a hub gene-related network was constructed and potential drugs for the treatment of OP were predicted. Finally, the specific genes were verified by real-time quantitative polymerase chain reaction (RT-qPCR). Results: In total, 548 DEGs were identified in the GSE56815 dataset. The weighted gene co-expression network analysis(WGCNA) identified 2291 key module genes, and 91 DE-MRG were obtained by combining the two. The PPI network revealed that the target gene for AKT1 interacted with most proteins. Three MRG (NELFB, SFSWAP, and MAP3K3) were identified as hub genes, with areas under the curve (AUC) 0.75, 0.71, and 0.70, respectively. The nomogram model has high diagnostic value. GO and KEGG analysis showed that ribosome pathway and cellular ribosome pathway may be the pathways regulating the progression of OP. IPA showed that MAP3K3 was associated with six pathways, including GNRH Signaling. The ssGSEA indicated that NELFB was highly correlated with iDCs (cor = -0.390, p < 0.001). The regulatory network showed a complex relationship between miRNA, transcription factor(TF) and hub genes. In addition, 4 drugs such as vinclozolin were predicted to be potential therapeutic drugs for OP. In RT-qPCR verification, the hub gene NELFB was consistent with the results of bioinformatics analysis. Conclusion: Mitophagy plays an important role in the development of osteoporosis. The identification of three mitophagy-related genes may contribute to the early diagnosis, mechanism research and treatment of OP.
Collapse
Affiliation(s)
- Yu Su
- Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Gangying Yu
- Department of International Ward (Orthopedic), Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dongchen Li
- Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yao Lu
- Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Cheng Ren
- Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yibo Xu
- Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yanling Yang
- Basic Medical College of Yan’an University, Yan’an, China
| | - Kun Zhang
- Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Teng Ma
- Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Zhong Li
- Honghui Hospital, Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
35
|
Park E, Evans MA, Walsh K. Regulators of clonal hematopoiesis and physiological consequences of this condition. THE JOURNAL OF CARDIOVASCULAR AGING 2024; 4:3. [PMID: 39119355 PMCID: PMC11309374 DOI: 10.20517/jca.2023.39] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Clonal hematopoiesis (CH) is a prevalent condition that results from somatic mutations in hematopoietic stem cells. When these mutations occur in "driver" genes, they can potentially confer fitness advantages to the affected cells, leading to a clonal expansion. While most clonal expansions of mutant cells are generally considered to be asymptomatic since they do not impact overall blood cell numbers, CH carriers face long-term risks of all-cause mortality and age-associated diseases, including cardiovascular disease and hematological malignancies. While considerable research has focused on understanding the association between CH and these diseases, less attention has been given to exploring the regulatory factors that contribute to the expansion of the driver gene clone. This review focuses on the association between environmental stressors and inherited genetic risk factors in the context of CH development. A better understanding of how these stressors impact CH development will facilitate mechanistic studies and potentially lead to new therapeutic avenues to treat individuals with this condition.
Collapse
Affiliation(s)
- Eunbee Park
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Megan A. Evans
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| | - Kenneth Walsh
- Department of Biochemistry and Molecular Genetics, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
- Hematovascular Biology Center, Robert M. Berne Cardiovascular Research Center, University of Virginia School of Medicine, Charlottesville, VA 22908, USA
| |
Collapse
|
36
|
Weeks LD, Ebert BL. Causes and consequences of clonal hematopoiesis. Blood 2023; 142:2235-2246. [PMID: 37931207 PMCID: PMC10862247 DOI: 10.1182/blood.2023022222] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/08/2023] Open
Abstract
ABSTRACT Clonal hematopoiesis (CH) is described as the outsized contribution of expanded clones of hematopoietic stem and progenitor cells (HSPCs) to blood cell production. The prevalence of CH increases dramatically with age. CH can be caused by somatic mutations in individual genes or by gains and/or losses of larger chromosomal segments. CH is a premalignant state; the somatic mutations detected in CH are the initiating mutations for hematologic malignancies, and CH is a strong predictor of the development of blood cancers. Moreover, CH is associated with nonmalignant disorders and increased overall mortality. The somatic mutations that drive clonal expansion of HSPCs can alter the function of terminally differentiated blood cells, including the release of elevated levels of inflammatory cytokines. These cytokines may then contribute to a broad range of inflammatory disorders that increase in prevalence with age. Specific somatic mutations in the peripheral blood in coordination with blood count parameters can powerfully predict the development of hematologic malignancies and overall mortality in CH. In this review, we summarize the current understanding of CH nosology and origins. We provide an overview of available tools for risk stratification and discuss management strategies for patients with CH presenting to hematology clinics.
Collapse
Affiliation(s)
- Lachelle D. Weeks
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Center for Early Detection and Interception of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Center for Early Detection and Interception of Blood Cancers, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Howard Hughes Medical Institute, Boston, MA
| |
Collapse
|
37
|
Cacic AM, Schulz FI, Germing U, Dietrich S, Gattermann N. Molecular and clinical aspects relevant for counseling individuals with clonal hematopoiesis of indeterminate potential. Front Oncol 2023; 13:1303785. [PMID: 38162500 PMCID: PMC10754976 DOI: 10.3389/fonc.2023.1303785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/28/2023] [Indexed: 01/03/2024] Open
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) has fascinated the medical community for some time. Discovered about a decade ago, this phenomenon links age-related alterations in hematopoiesis not only to the later development of hematological malignancies but also to an increased risk of early-onset cardiovascular disease and some other disorders. CHIP is detected in the blood and is characterized by clonally expanded somatic mutations in cancer-associated genes, predisposing to the development of hematologic neoplasms such as MDS and AML. CHIP-associated mutations often involve DNA damage repair genes and are frequently observed following prior cytotoxic cancer therapy. Genetic predisposition seems to be a contributing factor. It came as a surprise that CHIP significantly elevates the risk of myocardial infarction and stroke, and also contributes to heart failure and pulmonary hypertension. Meanwhile, evidence of mutant clonal macrophages in vessel walls and organ parenchyma helps to explain the pathophysiology. Besides aging, there are some risk factors promoting the appearance of CHIP, such as smoking, chronic inflammation, chronic sleep deprivation, and high birth weight. This article describes fundamental aspects of CHIP and explains its association with hematologic malignancies, cardiovascular disorders, and other medical conditions, while also exploring potential progress in the clinical management of affected individuals. While it is important to diagnose conditions that can lead to adverse, but potentially preventable, effects, it is equally important not to stress patients by confronting them with disconcerting findings that cannot be remedied. Individuals with diagnosed or suspected CHIP should receive counseling in a specialized outpatient clinic, where professionals from relevant medical specialties may help them to avoid the development of CHIP-related health problems. Unfortunately, useful treatments and clinical guidelines for managing CHIP are still largely lacking. However, there are some promising approaches regarding the management of cardiovascular disease risk. In the future, strategies aimed at restoration of gene function or inhibition of inflammatory mediators may become an option.
Collapse
Affiliation(s)
- Anna Maria Cacic
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Felicitas Isabel Schulz
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Sascha Dietrich
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| | - Norbert Gattermann
- Department of Hematology, Oncology and Clinical Immunology, Medical Faculty and University Hospital Düsseldorf, Heinrich Heine Universität Düsseldorf, Düsseldorf, Germany
- Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD), Düsseldorf, Germany
| |
Collapse
|
38
|
Beeler JS, Bolton KL. How low can you go?: Methodologic considerations in clonal hematopoiesis variant calling. Leuk Res 2023; 135:107419. [PMID: 37956474 DOI: 10.1016/j.leukres.2023.107419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023]
Abstract
Clonal hematopoiesis (CH) is defined by the presence of an expanded clonal hematopoietic cell population due to an acquired mutation conferring a selective growth advantage and is known to predispose to hematologic malignancy. In this review, we discuss sequencing methods for CH detection in bulk sequencing data and corresponding bioinformatic approaches for variant calling, filtering, and curation. We detail practical recommendations for CH calling. Finally, we discuss how improvements in CH sequencing and bioinformatic approaches will enable the characterization of CH trajectories, its impact on human health, and therapeutic approaches to mitigate its adverse effects.
Collapse
Affiliation(s)
- J Scott Beeler
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Kelly L Bolton
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
39
|
Mack T, Vlasschaert C, von Beck K, Silver AJ, Heimlich JB, Poisner H, Condon HR, Ulloa J, Sochacki AL, Spaulding TP, Kishtagari A, Bejan CA, Xu Y, Savona MR, Jones A, Bick A. Cost-effective and scalable clonal hematopoiesis assay provides insight into clonal dynamics. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.11.08.23298270. [PMID: 37986782 PMCID: PMC10659520 DOI: 10.1101/2023.11.08.23298270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is a common age-related phenomenon that occurs when hematopoietic stem cells acquire mutations in a select set of genes commonly mutated in myeloid neoplasia which then expand clonally. Current sequencing assays to detect CHIP are not optimized for the detection of these variants and can be cost-prohibitive when applied to large cohorts or serial sequencing. Here, we present and validate a CHIP targeted sequencing assay that is affordable (∼$8/sample), accurate and highly scalable. To demonstrate the utility of this assay, we detected CHIP in a cohort of 456 individuals with DNA collected at multiple timepoints in the Vanderbilt BioVU biobank and quantified clonal expansion rates over time. A total of 101 individuals with CHIP were identified, and individual-level clonal expansion rate was calculated using the variant allele fraction (VAF) at both timepoints. Differences in clonal expansion rate by driver gene were observed, but there was also significant individual-level heterogeneity, emphasizing the multifactorial nature of clonal expansion. We further describe the mutation co-occurrence and clonal competition between multiple driver mutations.
Collapse
|
40
|
Feng Y, Yuan Q, Newsome RC, Robinson T, Bowman RL, Zuniga AN, Hall KN, Bernsten CM, Shabashvili DE, Krajcik KI, Gunaratne C, Zaroogian ZJ, Venugopal K, Casellas Roman HL, Levine RL, Chatila WK, Yaeger R, Riva A, Jobin C, Kopinke D, Avram D, Guryanova OA. Hematopoietic-specific heterozygous loss of Dnmt3a exacerbates colitis-associated colon cancer. J Exp Med 2023; 220:e20230011. [PMID: 37615936 PMCID: PMC10450614 DOI: 10.1084/jem.20230011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 06/12/2023] [Accepted: 08/02/2023] [Indexed: 08/25/2023] Open
Abstract
Clonal hematopoiesis (CH) is defined as clonal expansion of mutant hematopoietic stem cells absent diagnosis of a hematologic malignancy. Presence of CH in solid tumor patients, including colon cancer, correlates with shorter survival. We hypothesized that bone marrow-derived cells with heterozygous loss-of-function mutations of DNMT3A, the most common genetic alteration in CH, contribute to the pathogenesis of colon cancer. In a mouse model that combines colitis-associated colon cancer (CAC) with experimental CH driven by Dnmt3a+/Δ, we found higher tumor penetrance and increased tumor burden compared with controls. Histopathological analysis revealed accentuated colonic epithelium injury, dysplasia, and adenocarcinoma formation. Transcriptome profiling of colon tumors identified enrichment of gene signatures associated with carcinogenesis, including angiogenesis. Treatment with the angiogenesis inhibitor axitinib eliminated the colon tumor-promoting effect of experimental CH driven by Dnmt3a haploinsufficiency and rebalanced hematopoiesis. This study provides conceptually novel insights into non-tumor-cell-autonomous effects of hematopoietic alterations on colon carcinogenesis and identifies potential therapeutic strategies.
Collapse
Affiliation(s)
- Yang Feng
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Qingchen Yuan
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Rachel C. Newsome
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Troy Robinson
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Robert L. Bowman
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ashley N. Zuniga
- Department of Anatomy and Cell Biology, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Kendra N. Hall
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Cassandra M. Bernsten
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Daniil E. Shabashvili
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Kathryn I. Krajcik
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Chamara Gunaratne
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Zachary J. Zaroogian
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Kartika Venugopal
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Heidi L. Casellas Roman
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Ross L. Levine
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Walid K. Chatila
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Rona Yaeger
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Alberto Riva
- Interdisciplinary Center for Biotechnology Research, University of Florida, Gainesville, FL, USA
- University of FloridaHealth Cancer Center, Gainesville, FL, USA
| | - Christian Jobin
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of FloridaCollege of Medicine, Gainesville, FL, USA
- University of FloridaHealth Cancer Center, Gainesville, FL, USA
| | - Daniel Kopinke
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
| | - Dorina Avram
- Department of Anatomy and Cell Biology, University of FloridaCollege of Medicine, Gainesville, FL, USA
- University of FloridaHealth Cancer Center, Gainesville, FL, USA
- Immunology Department, Moffitt Cancer Center, Tampa, FL, USA
| | - Olga A. Guryanova
- Department of Pharmacology and Therapeutics, University of FloridaCollege of Medicine, Gainesville, FL, USA
- University of FloridaHealth Cancer Center, Gainesville, FL, USA
| |
Collapse
|
41
|
Vlasschaert C, Akwo E, Robinson-Cohen C, Cook EK, Lanktree MB, Rauh MJ, Bick AG. Infection risk associated with clonal hematopoiesis of indeterminate potential is partly mediated by hematologic cancer transformation in the UK Biobank. Leukemia 2023; 37:2306-2308. [PMID: 37689824 PMCID: PMC11151170 DOI: 10.1038/s41375-023-02023-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2023] [Revised: 07/25/2023] [Accepted: 09/04/2023] [Indexed: 09/11/2023]
Affiliation(s)
| | - Elvis Akwo
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Cassianne Robinson-Cohen
- Department of Medicine, Division of Nephrology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Elina K Cook
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Matthew B Lanktree
- Department of Medicine, McMaster University, Hamilton, ON, Canada
- Population Health Research Institute (PHRI), Hamilton, ON, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, ON, Canada
| | - Michael J Rauh
- Department of Pathology and Molecular Medicine, Queen's University, Kingston, ON, Canada
| | - Alexander G Bick
- Department of Medicine, Division of Genomic Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
42
|
Willimann R, Chougar C, Wolfe LC, Blanc L, Lipton JM. Defects in Bone and Bone Marrow in Inherited Anemias: the Chicken or the Egg. Curr Osteoporos Rep 2023; 21:527-539. [PMID: 37436584 DOI: 10.1007/s11914-023-00809-3] [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] [Accepted: 06/19/2023] [Indexed: 07/13/2023]
Abstract
PURPOSE OF REVIEW Recently, there has been an increasing number of studies on the crosstalk between the bone and the bone marrow and how it pertains to anemia. Here, we discuss four heritable clinical syndromes contrasting those in which anemia affects bone growth and development, with those in which abnormal bone development results in anemia, highlighting the multifaceted interactions between skeletal development and hematopoiesis. RECENT FINDINGS Anemia results from both inherited and acquired disorders caused by either impaired production or premature destruction of red blood cells or blood loss. The downstream effects on bone development and growth in patients with anemia often constitute an important part of their clinical condition. We will discuss the interdependence of abnormal bone development and growth and hematopoietic abnormalities, with a focus on the erythroid lineage. To illustrate those points, we selected four heritable anemias that arise from either defective hematopoiesis impacting the skeletal system (the hemoglobinopathies β-thalassemia and sickle cell disease) versus defective osteogenesis resulting in impaired hematopoiesis (osteopetrosis). Finally, we will discuss recent findings in Diamond Blackfan anemia, an intrinsic disorder of both the erythron and the bone. By focusing on four representative hereditary hematopoietic disorders, this complex relationship between bone and blood should lead to new areas of research in the field.
Collapse
Affiliation(s)
- Rachel Willimann
- Division of Hematology Oncology and Cellular Therapy, Steven and Alexandra Cohen Children's Medical Center of New York, 269-01 76th Avenue, New Hyde Park, NY, 11040, USA
| | - Christina Chougar
- Division of Hematology Oncology and Cellular Therapy, Steven and Alexandra Cohen Children's Medical Center of New York, 269-01 76th Avenue, New Hyde Park, NY, 11040, USA
- Division of Pediatric Radiology, Steven and Alexandra Cohen Children's Medical Center of New York, 269-01 76th Avenue, New Hyde Park, NY, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Lawrence C Wolfe
- Division of Hematology Oncology and Cellular Therapy, Steven and Alexandra Cohen Children's Medical Center of New York, 269-01 76th Avenue, New Hyde Park, NY, 11040, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
| | - Lionel Blanc
- Division of Hematology Oncology and Cellular Therapy, Steven and Alexandra Cohen Children's Medical Center of New York, 269-01 76th Avenue, New Hyde Park, NY, 11040, USA
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA
- The Feinstein Institutes for Medical Research, Manhasset, NY, USA
| | - Jeffrey M Lipton
- Division of Hematology Oncology and Cellular Therapy, Steven and Alexandra Cohen Children's Medical Center of New York, 269-01 76th Avenue, New Hyde Park, NY, 11040, USA.
- Donald and Barbara Zucker School of Medicine at Hofstra/Northwell, Hempstead, NY, USA.
- The Feinstein Institutes for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
43
|
Rauch PJ, Gopakumar J, Silver AJ, Nachun D, Ahmad H, McConkey M, Nakao T, Bosse M, Rentz T, Vivanco Gonzalez N, Greenwald NF, McCaffrey EF, Khair Z, Gopakumar M, Rodrigues KB, Lin AE, Sinha E, Fefer M, Cohen DN, Vromman A, Shvartz E, Sukhova G, Bendall S, Angelo M, Libby P, Ebert BL, Jaiswal S. Loss-of-function mutations in Dnmt3a and Tet2 lead to accelerated atherosclerosis and concordant macrophage phenotypes. NATURE CARDIOVASCULAR RESEARCH 2023; 2:805-818. [PMID: 39196062 DOI: 10.1038/s44161-023-00326-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 07/27/2023] [Indexed: 08/29/2024]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is defined by the presence of a cancer-associated somatic mutation in white blood cells in the absence of overt hematological malignancy. It arises most commonly from loss-of-function mutations in the epigenetic regulators DNMT3A and TET2. CHIP predisposes to both hematological malignancies and atherosclerotic cardiovascular disease in humans. Here we demonstrate that loss of Dnmt3a in myeloid cells increased murine atherosclerosis to a similar degree as previously seen with loss of Tet2. Loss of Dnmt3a enhanced inflammation in macrophages in vitro and generated a distinct adventitial macrophage population in vivo which merges a resident macrophage profile with an inflammatory cytokine signature. These changes surprisingly phenocopy the effect of loss of Tet2. Our results identify a common pathway promoting heightened innate immune cell activation with loss of either gene, providing a biological basis for the excess atherosclerotic disease burden in carriers of these two most prevalent CHIP mutations.
Collapse
Affiliation(s)
- Philipp J Rauch
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| | | | - Alexander J Silver
- Division of Hematology, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Daniel Nachun
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Herra Ahmad
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Marie McConkey
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
| | - Tetsushi Nakao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Research Center and Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Marc Bosse
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Thiago Rentz
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | - Noah F Greenwald
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Erin F McCaffrey
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Zumana Khair
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Manu Gopakumar
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Kameron B Rodrigues
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Amy E Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Eti Sinha
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Maia Fefer
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Drew N Cohen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amélie Vromman
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Eugenia Shvartz
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Galina Sukhova
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Sean Bendall
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Michael Angelo
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Peter Libby
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Howard Hughes Medical Institute, Boston, MA, USA
| | - Siddhartha Jaiswal
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
44
|
Belizaire R, Wong WJ, Robinette ML, Ebert BL. Clonal haematopoiesis and dysregulation of the immune system. Nat Rev Immunol 2023; 23:595-610. [PMID: 36941354 PMCID: PMC11140722 DOI: 10.1038/s41577-023-00843-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2023] [Indexed: 03/23/2023]
Abstract
Age-related diseases are frequently linked to pathological immune dysfunction, including excessive inflammation, autoreactivity and immunodeficiency. Recent analyses of human genetic data have revealed that somatic mutations and mosaic chromosomal alterations in blood cells - a condition known as clonal haematopoiesis (CH) - are associated with ageing and pathological immune dysfunction. Indeed, large-scale epidemiological studies and experimental mouse models have demonstrated that CH can promote cardiovascular disease, chronic obstructive pulmonary disease, chronic liver disease, osteoporosis and gout. The genes most frequently mutated in CH, the epigenetic regulators TET2 and DNMT3A, implicate increased chemokine expression and inflammasome hyperactivation in myeloid cells as a possible mechanistic connection between CH and age-related diseases. In addition, TET2 and DNMT3A mutations in lymphoid cells have been shown to drive methylation-dependent alterations in differentiation and function. Here we review the observational and mechanistic studies describing the connection between CH and pathological immune dysfunction, the effects of CH-associated genetic alterations on the function of myeloid and lymphoid cells, and the clinical and therapeutic implications of CH as a target for immunomodulation.
Collapse
Affiliation(s)
- Roger Belizaire
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Waihay J Wong
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Michelle L Robinette
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Medicine, Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Benjamin L Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Howard Hughes Medical Institute, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
45
|
Marongiu F, Cheri S, Laconi E. Clones of aging: When better fitness can be dangerous. Eur J Cell Biol 2023; 102:151340. [PMID: 37423036 DOI: 10.1016/j.ejcb.2023.151340] [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: 02/28/2023] [Revised: 05/29/2023] [Accepted: 07/05/2023] [Indexed: 07/11/2023] Open
Abstract
The biological and clinical significance of aberrant clonal expansions in aged tissues is being intensely discussed. Evidence is accruing that these clones often result from the normal dynamics of cell turnover in our tissues. The aged tissue microenvironment is prone to favour the emergence of specific clones with higher fitness partly because of an overall decline in cell intrinsic regenerative potential of surrounding counterparts. Thus, expanding clones in aged tissues need not to be mechanistically associated with the development of cancer, albeit this is a possibility. We suggest that growth pattern is a critical phenotypic attribute that impacts on the fate of such clonal proliferations. The acquisition of a better proliferative fitness, coupled with a defect in tissue pattern formation, could represent a dangerous mix setting the stage for their evolution towards neoplasia.
Collapse
Affiliation(s)
- Fabio Marongiu
- Department of Biomedical Sciences, University of Cagliari, Italy
| | - Samuele Cheri
- Department of Biomedical Sciences, University of Cagliari, Italy
| | - Ezio Laconi
- Department of Biomedical Sciences, University of Cagliari, Italy.
| |
Collapse
|
46
|
Colom Díaz PA, Mistry JJ, Trowbridge JJ. Hematopoietic stem cell aging and leukemia transformation. Blood 2023; 142:533-542. [PMID: 36800569 PMCID: PMC10447482 DOI: 10.1182/blood.2022017933] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 02/19/2023] Open
Abstract
With aging, hematopoietic stem cells (HSCs) have an impaired ability to regenerate, differentiate, and produce an entire repertoire of mature blood and immune cells. Owing to dysfunctional hematopoiesis, the incidence of hematologic malignancies increases among elderly individuals. Here, we provide an update on HSC-intrinsic and -extrinsic factors and processes that were recently discovered to contribute to the functional decline of HSCs during aging. In addition, we discuss the targets and timing of intervention approaches to maintain HSC function during aging and the extent to which these same targets may prevent or delay transformation to hematologic malignancies.
Collapse
|
47
|
Feng J, Hsu PF, Esteva E, Labella R, Wang Y, Khodadadi-Jamayran A, Pucella J, Liu CZ, Arbini AA, Tsirigos A, Kousteni S, Reizis B. Haplodeficiency of the 9p21 tumor suppressor locus causes myeloid disorders driven by the bone marrow microenvironment. Blood 2023; 142:460-476. [PMID: 37267505 DOI: 10.1182/blood.2022018512] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/04/2023] Open
Abstract
The chromosome 9p21 locus comprises several tumor suppressor genes including MTAP, CDKN2A, and CDKN2B, and its homo- or heterozygous deletion is associated with reduced survival in multiple cancer types. We report that mice with germ line monoallelic deletion or induced biallelic deletion of the 9p21-syntenic locus (9p21s) developed a fatal myelodysplastic syndrome/myeloproliferative neoplasm (MDS/MPN)-like disease associated with aberrant trabecular bone formation and/or fibrosis in the bone marrow (BM). Reciprocal BM transfers and conditional targeting of 9p21s suggested that the disease originates in the BM stroma. Single-cell analysis of 9p21s-deficient BM stroma revealed the expansion of chondrocyte and osteogenic precursors, reflected in increased osteogenic differentiation in vitro. It also showed reduced expression of factors maintaining hematopoietic stem/progenitor cells, including Cxcl12. Accordingly, 9p21s-deficient mice showed reduced levels of circulating Cxcl12 and concomitant upregulation of the profibrotic chemokine Cxcl13 and the osteogenesis- and fibrosis-related multifunctional glycoprotein osteopontin/Spp1. Our study highlights the potential of mutations in the BM microenvironment to drive MDS/MPN-like disease.
Collapse
Affiliation(s)
- Jue Feng
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Pei-Feng Hsu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Eduardo Esteva
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Rossella Labella
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY
- Edward P. Evans Center for Myelodysplastic Syndromes, Columbia University Medical Center, New York, NY
| | - Yueyang Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Alireza Khodadadi-Jamayran
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY
| | - Joseph Pucella
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Cynthia Z Liu
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Arnaldo A Arbini
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| | - Aristotelis Tsirigos
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
- Applied Bioinformatics Laboratories, New York University Grossman School of Medicine, New York, NY
| | - Stavroula Kousteni
- Department of Physiology and Cellular Biophysics, College of Physicians and Surgeons, Columbia University Medical Center, New York, NY
- Edward P. Evans Center for Myelodysplastic Syndromes, Columbia University Medical Center, New York, NY
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY
| | - Boris Reizis
- Department of Pathology, New York University Grossman School of Medicine, New York, NY
| |
Collapse
|
48
|
Avagyan S, Zon LI. Clonal hematopoiesis and inflammation - the perpetual cycle. Trends Cell Biol 2023; 33:695-707. [PMID: 36593155 PMCID: PMC10310890 DOI: 10.1016/j.tcb.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 12/04/2022] [Accepted: 12/08/2022] [Indexed: 01/01/2023]
Abstract
Acquired genetic or cytogenetic alterations in a blood stem cell that confer clonal fitness promote its relative expansion leading to clonal hematopoiesis (CH). Despite a largely intact hematopoietic output, CH is associated with a heightened risk of progression to hematologic malignancies and with non-hematologic health manifestations, including cardiovascular disease and overall mortality. We focus on the evidence for the role of inflammation in establishing, maintaining and reciprocally being affected by CH. We describe the known pro-inflammatory signals associated with CH and preclinical studies that elucidated the cellular mechanisms involved. We review the evolving literature on early-onset CH in germline predisposition conditions and the possible role of immune dysregulation in this context.
Collapse
Affiliation(s)
- Serine Avagyan
- Dana-Farber/Boston Children's Hospital Cancer and Blood Disorders Center, Boston, MA, USA.
| | - Leonard I Zon
- Boston Children's Hospital, Boston, MA 02215, USA; Howard Hughes Medical Institute, USA
| |
Collapse
|
49
|
Gibson CJ, Fell G, Sella T, Sperling AS, Snow C, Rosenberg SM, Kirkner G, Patel A, Dillon D, Bick AG, Neuberg D, Partridge AH, Miller PG. Clonal Hematopoiesis in Young Women Treated for Breast Cancer. Clin Cancer Res 2023; 29:2551-2558. [PMID: 37115512 PMCID: PMC10330424 DOI: 10.1158/1078-0432.ccr-23-0050] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/01/2023] [Accepted: 04/25/2023] [Indexed: 04/29/2023]
Abstract
PURPOSE Young women treated for breast cancer with cytotoxic therapies are at risk for clonal hematopoiesis of indeterminate potential (CHIP), a condition in which blood cells carrying a somatic mutation associated with hematologic malignancy comprise at least 4% of the total blood system. CHIP has primarily been studied in older patient cohorts with limited clinical phenotyping. EXPERIMENTAL DESIGN We performed targeted sequencing on longitudinal blood samples to characterize the clonal hematopoietic landscape of 878 women treated for breast cancer enrolled in the prospective Young Women's Breast Cancer Study. RESULTS We identified somatic driver mutations in 252 study subjects (28.7%), but only 24 (2.7%) had clones large enough to meet criteria for CHIP. The most commonly mutated genes were DNMT3A and TET2, similar to mutations observed in noncancer cohorts. At 9-year median follow-up, we found no association between the presence of a somatic blood mutation (regardless of clone size) and adverse breast cancer (distant relapse-free survival) or non-breast cancer-related outcomes in this cohort. A subset of paired blood samples obtained over 4 years showed no evidence of mutant clonal expansion, regardless of genotype. Finally, we identified a subset of patients with likely germline mutations in genes known to contribute to inherited cancer risk, such as TP53 and ATM. CONCLUSIONS Our data show that for young women with early-stage breast cancer, CHIP is uncommon after cytotoxic exposure, is unlikely to contribute to adverse outcomes over the decade-long follow-up and may not require additional monitoring if discovered incidentally.
Collapse
Affiliation(s)
- Christopher J. Gibson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
- Harvard Medical School, Boston, MA
| | - Geoffrey Fell
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA
| | - Tal Sella
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Oncology, Sheba Medical Center, Israel
| | - Adam S. Sperling
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Broad Institute of Harvard and MIT, Cambridge, MA
- Harvard Medical School, Boston, MA
- Division of Hematology, Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - Craig Snow
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | | | - Greg Kirkner
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Ashka Patel
- Department of Pathology, Brigham and Women’s Hospital, Boston MA
| | - Deborah Dillon
- Department of Pathology, Brigham and Women’s Hospital, Boston MA
| | - Alexander G. Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Donna Neuberg
- Department of Data Sciences, Dana-Farber Cancer Institute, Boston, MA
| | - Ann H. Partridge
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Peter G. Miller
- Broad Institute of Harvard and MIT, Cambridge, MA
- Harvard Medical School, Boston, MA
- Center for Cancer Research and Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
50
|
Balandrán JC, Lasry A, Aifantis I. The Role of Inflammation in the Initiation and Progression of Myeloid Neoplasms. Blood Cancer Discov 2023; 4:254-266. [PMID: 37052531 PMCID: PMC10320626 DOI: 10.1158/2643-3230.bcd-22-0176] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 03/10/2023] [Accepted: 03/10/2023] [Indexed: 04/14/2023] Open
Abstract
Myeloid malignancies are devastating hematologic cancers with limited therapeutic options. Inflammation is emerging as a novel driver of myeloid malignancy, with important implications for tumor composition, immune response, therapeutic options, and patient survival. Here, we discuss the role of inflammation in normal and malignant hematopoiesis, from clonal hematopoiesis to full-blown myeloid leukemia. We discuss how inflammation shapes clonal output from hematopoietic stem cells, how inflammation alters the immune microenvironment in the bone marrow, and novel therapies aimed at targeting inflammation in myeloid disease. SIGNIFICANCE Inflammation is emerging as an important factor in myeloid malignancies. Understanding the role of inflammation in myeloid transformation, and the interplay between inflammation and other drivers of leukemogenesis, may yield novel avenues for therapy.
Collapse
Affiliation(s)
- Juan Carlos Balandrán
- Department of Pathology and Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York
| | - Audrey Lasry
- Department of Pathology and Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York
| | - Iannis Aifantis
- Department of Pathology and Laura and Isaac Perlmutter Cancer Center, NYU Grossman School of Medicine, New York, New York
| |
Collapse
|