1
|
Fattizzo B, Marchetti A, Bosi A, Gurnari C, Giannotta JA, Pedone GL, Rossi E, Carrai V, Guido A, Brioschi F, Carpenedo M, Crugnola M, Caramazza D, Leuzzi L, Marchetti M, Merati G, Malato S, Vianello F, Patriarca A, Awada H, Bortolotti M, Canzi M, Bolli N, Capecchi M, Chen F, Artoni A, Maciejewski JP, Barcellini W. Clonal hematopoiesis in patients with autoimmune thrombocytopenia: an international multicenter study. Blood Adv 2025; 9:488-495. [PMID: 39536292 DOI: 10.1182/bloodadvances.2024014984] [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: 10/01/2024] [Revised: 10/26/2024] [Accepted: 10/26/2024] [Indexed: 11/16/2024] Open
Abstract
ABSTRACT Diagnostic boundaries between immune thrombocytopenia (ITP) and other thrombocytopenic states, such as thrombocytopenic myelodysplastic syndromes, may be difficult to establish, and the detection of somatic mutations by next-generation sequencing (NGS) may be of aid. Here, we aimed at characterizing the prevalence and clinical significance of clonal hematopoiesis in ITP. In this multicentric retrospective observational study, we enrolled 167 adult patients with ITP, followed at 13 centers in Italy, United Kingdom, and the United States. Patients underwent NGS evaluation after a median of 3.6 years from ITP onset, and 83% had received at least 1 therapy line, for a median of 2 lines (range, 0-9); 51 of 167 patients (30%) had at least 1 mutation. After exclusion of germ line variants and polymorphisms, 31 of 167 (18.5%) were defined as having clonal hemopoiesis. Most commonly mutated genes were TET2, DNMT3A, SRSF2, and ASXL1 (median variant allele frequency, 29%); 19 of 31 patients (68%) had high-risk variants, and 8 had multiple mutations. Mutated patients were more frequently older males and showed a shorter time from first to second-line therapy, particularly with thrombopoietin receptor agonist (TPO-RA). Additionally, clonal hematopoiesis was associated with increased thrombotic risk (26% vs 8% in NGS-negative cases; P = .01), independently from TPO-RA exposure, though with an age effect. These data demonstrated the prevalence of clonal hematopoiesis in 18% of adult patients with ITP, which is associated with older age, relapsed/refractory disease, and high risk of thrombotic complications.
Collapse
Affiliation(s)
- Bruno Fattizzo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy
| | - Alfredo Marchetti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy
| | - Alessandro Bosi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy
| | - Carmelo Gurnari
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH
| | | | - Giacinto Luca Pedone
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy
| | - Elena Rossi
- Policlinico Universitario Agostino Gemelli Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | | | - Andrea Guido
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy
| | | | - Monica Carpenedo
- Niguarda Cancer Center, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | | | - Livia Leuzzi
- SC Oncologia, SS Oncoematologia, PO Fatebenefratelli, ASST Fatebenefratelli Sacco, Milan, Italy
| | - Monia Marchetti
- Azienda Ospedaliera Nazionale SS Antonio e Biagio e Cesare Arrigo, Alessandria, Italy
| | | | - Simona Malato
- Hematology and Bone Marrow Transplantation Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Fabrizio Vianello
- Department of Medicine, Division of Hematology, University of Padova, Italy
| | - Andrea Patriarca
- Division of Hematology, Department of Translational Medicine, University of Eastern Piedmont and AOU Maggiore della Carità, Novara, Italy
| | - Hussein Awada
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH
| | - Marta Bortolotti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Marta Canzi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy
| | - Niccolò Bolli
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Oncology and Haemato-Oncology, University of Milan, Milan, Italy
| | - Marco Capecchi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Division of Hematology, Gruppo Ospedaliero Moncucco, Lugano, Switzerland
| | - Frederick Chen
- Barts ITP Centre, Royal London Hospital, Bartshealth NHS Trust, London, United Kingdom
- Blizard institute, Queen Mary University London, London, United Kingdom
| | - Andrea Artoni
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, OH
| | - Wilma Barcellini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
2
|
Raddatz MA, Pershad Y, Parker AC, Bick AG. Clonal Hematopoiesis of Indeterminate Potential and Cardiovascular Health. Cardiol Clin 2025; 43:13-23. [PMID: 39551555 DOI: 10.1016/j.ccl.2024.08.004] [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] [Indexed: 11/19/2024]
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is an age-related phenomenon in which somatic mutations lead to clonal expansion of hematopoietic stem cells without the development of hematologic abnormalities. A growing body of literature demonstrates an association between CHIP and cardiovascular disease. This pathophysiology demonstrates a novel connection between global inflammation and cardiovascular morbidity. While there is limited consensus addressing the cardiovascular care of these patients, risk factor optimization and disease surveillance are advisable. Investigation into possible therapies is ongoing and provides promise for the treatment of inflammation contributing to cardiovascular disease in patients with and without CHIP.
Collapse
Affiliation(s)
- Michael A Raddatz
- Division of Cardiology, Department of Medicine, University of California, Los Angeles, 650 Charles E. Young Dr. South, A2-237 CHS, Los Angeles, CA 90095, USA
| | - Yash Pershad
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, 550 Robinson Research Building, Nashville, TN 37232, USA
| | - Alyssa C Parker
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, 550 Robinson Research Building, Nashville, TN 37232, USA
| | - Alexander G Bick
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University School of Medicine, 550 Robinson Research Building, Nashville, TN 37232, USA.
| |
Collapse
|
3
|
Xi Z, Feng H, Chen K, Guo X, Zhu D, Zheng J, Li Y. Clonal hematopoiesis of indeterminate potential is a risk factor of gastric cancer: A Prospective Cohort in UK Biobank study. Transl Oncol 2025; 52:102242. [PMID: 39675251 PMCID: PMC11713744 DOI: 10.1016/j.tranon.2024.102242] [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: 08/29/2024] [Revised: 11/18/2024] [Accepted: 12/07/2024] [Indexed: 12/17/2024] Open
Abstract
IMPORTANCE Gastric cancer is often diagnosed at an advanced stage and at order age, identification of high-risk population is needed for detection of early-stage gastric cancer. OBJECTIVE To examine whether clonal hematopoiesis of indeterminate potential (CHIP) is a risk factor of gastric cancer. DESIGN This cohort study used data from the UK Biobank collected from baseline (2006-2010) to the end of follow-up in March 2024. SETTING Data on age, sex, race, alcohol consumption, smoking status and type 2 diabetes were collected at baseline interview. Previous and diagnosed cancer or diseases were collected from self-reported and in-hospital records. PARTICIPANTS Participants with no previous cancer or hematologic disorders were selected. Participants with gastric cancer cases were aged 60.7 (S.D. 6.62), 71.8 % male; controls were aged 56.1 (S.D. 8.11), 47.4 % male. EXPOSURES Whole-exome sequencing was performed on blood samples collected at baseline. A CHIP status was identified based on the mutations on 43 CHIP-related genes. MAIN OUTCOMES AND MEASURES Odds ratio (OR) of CHIP with gastric cancer risk was estimated using multivariable logistic regression models. Participants were grouped based on age and CHIP status to examine if there are differences in the cumulative incidence of gastric cancer. RESULTS Among 402,253 participants, 1,070 incident gastric cancer cases were identified (mean age, 60.7 ± 6.62 years). The prevalence of CHIP at baseline was associated with an increased risk of gastric cancer (cases: 6.54 % vs. controls 5.14 %; OR without adjustment, 1.29; 95 % CI, 1.004 to 1.63). The stratified OR (95 % CI) of individuals aged ≥ 57 was 1.33 (1.02 to 1.72) for overall CHIP, whereas the OR for younger individuals was 0.79 (0.37 to 1.44). CHIP involving DNMT3A (OR, 1.81; 95 % CI, 1.05 to 2.88; P = 0.0193) and ASXL1 (OR, 2.43; 95 % CI, 0.95 to 4.99; P = 0.032) was associated with an increased risk of gastric cancer. These positive associations remained significantly in sensitivity analyses adjusted by known risk factors. Compared to younger individuals and non-CHIP carriers, older participants with CHIP exhibited a significantly higher cumulative incidence of gastric cancer (P < 0.0001). CONCLUSIONS AND RELEVANCE CHIP is associated with gastric cancer in the elderly and contributes to the positive association between DNM3A and ASXL1 mutations and risk of gastric cancer.
Collapse
Affiliation(s)
- Zhihui Xi
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, PR China; Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, PR China
| | - Huolun Feng
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, PR China; Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, PR China.
| | - Kunling Chen
- Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China
| | - Xin Guo
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, PR China
| | - Dandan Zhu
- Guangdong Center for Clinical Laboratory, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, PR China
| | - Jiabin Zheng
- Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, PR China.
| | - Yong Li
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, PR China; Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
4
|
Yao CY, Ko TY, Yang LT, Takeuchi M, Yeh CF, Lin MS, Chen YH, Kuo CY, Hsu CL, Chou WC, Kao HL. Clonal Hematopoiesis Is Associated With Adverse Clinical Outcomes and Left Ventricular Remodeling in Aortic Stenosis. JACC. ADVANCES 2025; 4:101532. [PMID: 39886300 PMCID: PMC11780101 DOI: 10.1016/j.jacadv.2024.101532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2024] [Revised: 11/05/2024] [Accepted: 11/18/2024] [Indexed: 02/01/2025]
Abstract
Background Clonal hematopoiesis of indeterminate potential (CHIP) has been linked to intensified systemic inflammation and represents a novel risk factor for atherosclerotic cardiovascular diseases, including aortic stenosis (AS). Objectives This study aimed to assess the clinical impact of CHIP in a cohort of severe AS patients undergoing transcatheter aortic valve implantation (TAVI). Methods We enrolled 110 severe AS patients in this retrospective study. Targeted next-generation sequencing was employed to detect somatic mutations with a variant allele frequency >2% in 16 genes most frequently associated with CHIP. Correlative analyses on clinical, laboratory, and echocardiographic parameters were also performed. The primary endpoint was post-TAVI heart failure hospitalization. Multivariate Cox regression model was used to account for confounding effects of relevant clinical factors. Results CHIP was detected in 40 (36.4%) patients in our cohort. The most commonly mutated genes were DNMT3A, TET2, and ASXL1. With a median follow-up of 55.2 months, patients carrying CHIP had a significantly higher heart failure hospitalization rate (adjusted HR: 3.060; 95% CI: 1.090-8.589; P = 0.034) than those without CHIP. Additionally, patients harboring CHIP had higher serum ferritin levels, as well as echocardiographic evidence of left ventricular hypertrophy and diastolic dysfunction. Conclusions Our study supports the adverse clinical impact of CHIP in AS patients undergoing TAVI, which could be attributed to systemic inflammation and maladaptive LV remodeling. Prospective trials are anticipated to validate our findings and provide further evidence that CHIP holds the potential of being an actionable therapeutic target in AS.
Collapse
Affiliation(s)
- Chi-Yuan Yao
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Tsung-Yu Ko
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Li-Tan Yang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Masaaki Takeuchi
- Department of Cardiology, Tobata General Hospital, Kitakyushu, Japan
| | - Chih-Fan Yeh
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Mao-Shin Lin
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ying-Hsien Chen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Ching-Ying Kuo
- Department of Clinical Laboratory Sciences and Medical Biotechnology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chia-Lang Hsu
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, Taiwan
- Graduate Institute of Medical Genomics and Proteomics, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Chien Chou
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Department of Laboratory Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Hsien-Li Kao
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Clinical Medicine, College of Medicine, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
5
|
Kohutek ZA, Caslin HL, Fehrenbach DJ, Heimlich JB, Brown JD, Madhur MS, Ferrell PB, Doran AC. Bone Marrow Niche in Cardiometabolic Disease: Mechanisms and Therapeutic Potential. Circ Res 2025; 136:325-353. [PMID: 39883790 DOI: 10.1161/circresaha.124.323778] [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] [Indexed: 02/01/2025]
Abstract
Cardiovascular and cardiometabolic diseases are leading causes of morbidity and mortality worldwide, driven in part by chronic inflammation. Emerging research suggests that the bone marrow microenvironment, or marrow niche, plays a critical role in both immune system regulation and disease progression. The bone marrow niche is essential for maintaining hematopoietic stem cells (HSCs) and orchestrating hematopoiesis. Under normal conditions, this niche ensures a return to immune homeostasis after acute stress. However, in the setting of inflammatory conditions such as those seen in cardiometabolic diseases, it becomes dysregulated, leading to enhanced myelopoiesis and immune activation. This review explores the reciprocal relationship between the bone marrow niche and cardiometabolic diseases, highlighting how alterations in the niche contribute to disease development and progression. The niche regulates HSCs through complex interactions with stromal cells, endothelial cells, and signaling molecules. However, in the setting of chronic diseases such as hypertension, atherosclerosis, and diabetes, inflammatory signals disrupt the balance between HSC self-renewal and differentiation, promoting the excessive production of proinflammatory myeloid cells that exacerbate the disease. Key mechanisms discussed include the effects of hyperlipidemia, hyperglycemia, and sympathetic nervous system activation on HSC proliferation and differentiation. Furthermore, the review emphasizes the role of epigenetic modifications and metabolic reprogramming in creating trained immunity, a phenomenon whereby HSCs acquire long-term proinflammatory characteristics that sustain disease states. Finally, we explore therapeutic strategies aimed at targeting the bone marrow niche to mitigate chronic inflammation and its sequelae. Novel interventions that modulate hematopoiesis and restore niche homeostasis hold promise for the treatment of cardiometabolic diseases. By interrupting the vicious cycle of inflammation and marrow dysregulation, such therapies may offer new avenues for reducing cardiovascular risk and improving patient outcomes.
Collapse
Affiliation(s)
- Zachary A Kohutek
- Department of Radiation Oncology (Z.A.K.), Vanderbilt University Medical Center, Nashville, TN
| | - Heather L Caslin
- Department of Health and Human Performance, University of Houston, TX (H.L.C.)
| | - Daniel J Fehrenbach
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis (D.J.F., M.S.M.)
| | - J Brett Heimlich
- Division of Cardiovascular Medicine, Department of Medicine (J.B.H., J.D.B., A.C.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Jonathan D Brown
- Division of Cardiovascular Medicine, Department of Medicine (J.B.H., J.D.B., A.C.D.), Vanderbilt University Medical Center, Nashville, TN
| | - Meena S Madhur
- Division of Clinical Pharmacology, Department of Medicine, Indiana University School of Medicine, Indianapolis (D.J.F., M.S.M.)
| | - P Brent Ferrell
- Division of Hematology and Oncology, Department of Medicine (P.B.F.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Nashville, TN (P.B.F., A.C.D.)
| | - Amanda C Doran
- Division of Cardiovascular Medicine, Department of Medicine (J.B.H., J.D.B., A.C.D.), Vanderbilt University Medical Center, Nashville, TN
- Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University, Nashville, TN (P.B.F., A.C.D.)
| |
Collapse
|
6
|
Bentivegna S, Almosailleakh M, Zhao LP, Schuster MB, Benquet S, Balhuizen A, Munch-Petersen HF, Sjö LD, Andersen MH, Dulphy N, Porse B, Grønbæk K. A non-conditioned bone marrow transplantation mouse model to study clonal hematopoiesis and myeloid malignancies. Exp Hematol Oncol 2025; 14:10. [PMID: 39885602 PMCID: PMC11781034 DOI: 10.1186/s40164-025-00598-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 01/15/2025] [Indexed: 02/01/2025] Open
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) is a condition where blood or bone marrow cells carry mutations associated with hematological malignancies. Individuals with CHIP have an increased risk of developing hematological malignancies, atherosclerotic cardiovascular disease, and all-cause mortality. Bone marrow transplantation (BMT) of cells carrying CHIP mutations into irradiated mice are useful procedures to investigate the dynamics of clonal expansion and potential therapeutic strategies, but myeloablative conditioning can induce confounding effects. We established a non-conditioned BMT model using C57BL/6J-KitW-41J/J (W41) recipient mice to overcome the unwanted effects of irradiation. Conditional Tet2 deletion using tamoxifen was used to obtain Tet2-/- cells from donor mice. Total BM Tet2-/- cells were transplanted into W41 recipients, and longitudinal and terminal analyses at 10 months post-BMT were performed. We showed that W41 mice can be used for BMT procedures without myeloablative pre-conditioning. The transplantation of Tet2-/- BM cells led to a progressive expansion of the donor cells in W41 recipients. By modulating the numbers of Tet2-/- cells transplanted, recipient mice developed features of clonal hematopoiesis or myeloid malignancies. In conclusion, our model is an alternative to conventional irradiation-based transplantation models to study mechanisms underlying malignant hematopoiesis without confounding effects derived from pre-conditioning regimen.
Collapse
Affiliation(s)
- Sofia Bentivegna
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Marwa Almosailleakh
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Lin-Pierre Zhao
- Hôpital Saint-Louis, Assistance Publique des Hôpitaux de Paris (APHP), Paris, France
- INSERM UMR 1160, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
| | - Mikkel Bruhn Schuster
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
- The Finsen Laboratory, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sébastien Benquet
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | - Alexander Balhuizen
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
| | | | - Lene Dissing Sjö
- Department of Pathology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Genetics, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy (CCIT-DK), Department of Oncology, Copenhagen University Hospital, Herlev, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Nicolas Dulphy
- INSERM UMR 1160, Institut de Recherche Saint-Louis, Université Paris Cité, Paris, France
- Laboratoire d'Immunologie et d'Histocompatibilité, Assistance Publique des Hôpitaux de Paris (APHP), Hôpital Saint-Louis, Paris, France
- Institut Carnot OPALE, Institut de Recherche Saint-Louis, Hôpital Saint-Louis, Paris, France
| | - Bo Porse
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark
- The Finsen Laboratory, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Kirsten Grønbæk
- Department of Hematology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark.
- Biotech Research and Innovation Center (BRIC), University of Copenhagen, Copenhagen, Denmark.
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.
| |
Collapse
|
7
|
Mohammed Ismail W, Fernandez JA, Binder M, Lasho TL, Kim M, Geyer SM, Mazzone A, Finke CM, Mangaonkar AA, Lee JH, Wang L, Kim KH, Simon VA, Rakhshan Rohakthar F, Munankarmy A, Byeon SK, Schwager SM, Harrington JJ, Snyder MR, Robertson KD, Pandey A, Wieben ED, Chia N, Gaspar-Maia A, Patnaik MM. Single-cell multiomics reveal divergent effects of DNMT3A- and TET2-mutant clonal hematopoiesis in inflammatory response. Blood Adv 2025; 9:402-416. [PMID: 39631069 DOI: 10.1182/bloodadvances.2024014467] [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: 08/07/2024] [Revised: 10/28/2024] [Accepted: 11/14/2024] [Indexed: 12/07/2024] Open
Abstract
ABSTRACT DNMT3A and TET2 are epigenetic regulator genes commonly mutated in age-related clonal hematopoiesis (CH). Despite having opposed epigenetic functions, these mutations are associated with increased all-cause mortality and a low risk for progression to hematologic neoplasms. Although individual impacts on the epigenome have been described using different model systems, the phenotypic complexity in humans remains to be elucidated. Here, we make use of a natural inflammatory response occurring during coronavirus disease 2019 (COVID-19), to understand the association of these mutations with inflammatory morbidity (acute respiratory distress syndrome [ARDS]) and mortality. We demonstrate the age-independent, negative impact of DNMT3A mutant (DNMT3Amt) CH on COVID-19-related ARDS and mortality. Using single-cell proteogenomics we show that DNMT3A mutations involve myeloid and lymphoid lineage cells. Using single-cell multiomics sequencing, we identify cell-specific gene expression changes associated with DNMT3A mutations, along with significant epigenomic deregulation affecting enhancer accessibility, resulting in overexpression of interleukin-32 (IL-32), a proinflammatory cytokine that can result in inflammasome activation in monocytes and macrophages. Finally, we show with single-cell resolution that the loss of function of DNMT3A is directly associated with increased chromatin accessibility in mutant cells. Hence, we demonstrate the negative prognostic impact of DNMT3Amt CH on COVID-19-related ARDS and mortality. DNMT3Amt CH in the context of COVID-19, was associated with inflammatory transcriptional priming, resulting in overexpression of IL32. This overexpression was secondary to increased chromatic accessibility, specific to DNMT3Amt CH cells. DNMT3Amt CH can thus serve as a potential biomarker for adverse outcomes in COVID-19.
Collapse
Affiliation(s)
- Wazim Mohammed Ismail
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Jenna A Fernandez
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Moritz Binder
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Terra L Lasho
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Minsuk Kim
- Department of Laboratory Medicine and Pathology, Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Susan M Geyer
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
| | - Amelia Mazzone
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Christy M Finke
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | | | - Jeong-Heon Lee
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Liguo Wang
- Department of Quantitative Health Sciences, Mayo Clinic, Rochester, MN
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
| | - Kwan Hyun Kim
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Vernadette A Simon
- Medical Genome Facility, Genome Analysis Core, Mayo Clinic, Rochester, MN
| | | | - Amik Munankarmy
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Seul Kee Byeon
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Susan M Schwager
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Jonathan J Harrington
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Melissa R Snyder
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Keith D Robertson
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Akhilesh Pandey
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Eric D Wieben
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Rochester, MN
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN
| | - Nicholas Chia
- Department of Laboratory Medicine and Pathology, Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
| | - Alexandre Gaspar-Maia
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN
| | - Mrinal M Patnaik
- Department of Laboratory Medicine and Pathology, Epigenomics Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| |
Collapse
|
8
|
Todorovski A, Wang TF, Carrier M, Xu Y. CHIP away at the marrow-clot connection: inflammation, clonal hematopoiesis, and thromboembolic disease. Blood Adv 2025; 9:343-353. [PMID: 39561373 DOI: 10.1182/bloodadvances.2024014430] [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: 08/13/2024] [Revised: 10/11/2024] [Accepted: 11/03/2024] [Indexed: 11/21/2024] Open
Abstract
ABSTRACT Both the incidence and prognosis of arterial atherothrombosis and venous thromboembolism are strongly correlated with increasing age. Over the past decade, clonal hematopoiesis of indeterminate potential (CHIP) has been identified as a novel biomarker for cardiovascular disease. Driven by somatic mutations in the hematopoietic system, the epidemiology of CHIP is highly age dependent: among individuals aged ≥70 years in the general population, estimated prevalence of CHIP exceeds 10%. Several additional risk factors for CHIP have emerged in recent years, including smoking, receipt of anticancer therapy, and germ line predispositions. CHIP carriers consistently have higher risk of incident arterial atherothrombosis, even after accounting for traditional cardiovascular risk factors. However, the magnitude of this association varies across studies. In addition, individuals with established cardiovascular disease and CHIP have higher risks of recurrence and all-cause mortality than their non-CHIP counterparts. An association between CHIP carriership and incident venous thromboembolism has recently been made, although additional studies are needed to confirm this finding. No approved therapy exists to modify the cardiovascular risk among CHIP carriers. However, canakinumab showed promise in a post-hoc analyses of patients with TET2-mutated CHIP, and other anti-inflammasome agents are actively under development or evaluation. In this review, we provide an overview of CHIP as a mediator of thromboembolic diseases and discuss emerging therapeutics aimed at intervening on this thrombo-inflammatory nexus.
Collapse
Affiliation(s)
- Angela Todorovski
- Department of Medicine, University of Ottawa and Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Tzu-Fei Wang
- Department of Medicine, University of Ottawa and Ottawa Hospital Research Institute, Ottawa, ON, Canada
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Marc Carrier
- Department of Medicine, University of Ottawa and Ottawa Hospital Research Institute, Ottawa, ON, Canada
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Yan Xu
- Department of Medicine, University of Ottawa and Ottawa Hospital Research Institute, Ottawa, ON, Canada
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| |
Collapse
|
9
|
Tan Z, Zhang X, Feng J, Zhao Y, Hu H, Wu D, Yu Q, Zhang Y, Wu L, Hu T, Yan Z, Ye B, Liu W. Hematopoietic stem cell transplantation and immunosuppressive therapy: implications of clonal haematopoiesis. Ann Hematol 2025:10.1007/s00277-024-06152-6. [PMID: 39873798 DOI: 10.1007/s00277-024-06152-6] [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: 05/21/2024] [Accepted: 12/16/2024] [Indexed: 01/30/2025]
Abstract
Aplastic anemia (AA) is a life-threatening bone marrow failure syndrome. The advent of next-generation sequencing (NGS) has shed light on the link between somatic mutations (SM) and the efficacy of immunosuppressive therapy (IST) in AA patients. However, the relationship between SM and hematopoietic stem cell transplantation (HSCT) has not been extensively explored. In this retrospective analysis, we examined 166 AA patients who received HSCT or IST at our institution between May 2019 and December 2023. NGS was conducted on 66 genes within bone marrow cells to investigate the correlation between SM and the prognosis and therapeutic response in AA patients, as well as to assess the impact of mutation types on HSCT outcomes. Clinical data were gathered from 166 AA patients, comprising 84 males and 82 females, with a median age of 32 years (ranging from 9 to 75 years). In our study, a total of 151 somatic mutations were identified across 84 patients (50.6%), with 42 patients (25.3%) presenting a single mutation and 26 patients (15.7%) harboring two mutations. The top five genes with the highest mutation frequency were BCOR/BCORL1 (12.6%), ASXL1 (8.6%), TET2 (6.6%), CEBPA (5.3%), and GATA2 (4.6%). We stratified patients into SM and No-SM groups based on the presence of mutations and further divided them into HSCT and IST groups to assess the influence of mutation types on treatment response and survival within and between these groups. The findings were as follows: 1.Patients in the HSCT group exhibited a higher treatment response (OR 85.9% vs. 68.4%, p < 0.05), although there was no significant difference in survival. 2.Patients with favorable mutations, such as PIGA and BCOR/BCORL1, experienced significantly improved response and survival compared to those with unfavorable mutations like ASXL1, DNMT3A, and TET2 (OR 93.7% vs. 72%, p < 0.05) (3-year OS 93.7% vs. 80%, p > 0.05). 3.The HSCT-Favorable group demonstrated superior response rates (OR 100% vs. 67.7%, p < 0.05) and longer survival (3-year OS 100% vs. 67.7%, p < 0.05) compared to the IST-Favorable group. This study underscores that AA patients carrying favorable mutations, particularly BCOR/BCORL1, tend to have a more robust response and better prognosis than those without mutations or those with unfavorable mutations, such as ASXL1/DNMT3A. These findings are especially pertinent to HSCT, highlighting the importance of NGS prior to initiating treatment.
Collapse
Affiliation(s)
- Zhengwei Tan
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xinhe Zhang
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jia Feng
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yuechao Zhao
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Huijin Hu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Dijiong Wu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Qinghong Yu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Yu Zhang
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Liqiang Wu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Tonglin Hu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Zhengsong Yan
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Baodong Ye
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China
| | - Wenbin Liu
- Department of Hematology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, China.
| |
Collapse
|
10
|
Senguttuvan NB, Subramanian V, Tr M, Sankaranarayanan K, Venkatesan V, Sadagopan T. Clonal hematopoiesis of indeterminate potential and cardiovascular diseases: A review. Indian Heart J 2025:S0019-4832(25)00007-0. [PMID: 39863253 DOI: 10.1016/j.ihj.2025.01.006] [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: 07/24/2024] [Revised: 10/08/2024] [Accepted: 01/23/2025] [Indexed: 01/27/2025] Open
Abstract
Cardiovascular disease (CVD) is a major driver of mortality and declining health worldwide. Cardiovascular diseases (CVD) is the most common cause of morbidity and mortality globally. Although dyslipidemia, smoking, diabetes, hypertension and obesity are some well-known causes of CVD, the overlapping genetic pathways between other diseases and those affecting cardiovascular health have been overlooked. In the past decade, mutations in TET2, DNMT3A, ASXL1, and JAK2 are found to cause clonal hematopoiesis of intermediate potential (CHIP), a disease associated with age-related haematological malignancies without the presence of cytopenias or dysplasia. Coronary artery disease, heart failure, aortic stenosis, and arrhythmias have been shown to be associated with the presence of CHIP mutations. Addressing the association between CHIP could significantly reduce residual risk patients with CVD. The link between CHIP and CVD can potentially be addressed through inhibitors of inflammasomes, antagonists in the interleukin pathway, or direct antagonists of CHIP mutations.
Collapse
Affiliation(s)
| | - Vinodhini Subramanian
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education & Research (SRIHER), Chennai, India
| | - Muralidharan Tr
- Department of Cardiology, SRM medical college, Chennai, Chennai, India
| | - Kavitha Sankaranarayanan
- Ion Channel Biology Laboratory, AU-KBC Research Centre, MIT Campus of Anna University, Chennai, Tamil Nadu, 600044, India
| | - Vettriselvi Venkatesan
- Department of Human Genetics, Sri Ramachandra Institute of Higher Education & Research (SRIHER), Chennai, India.
| | - Thanikachalam Sadagopan
- Department of Cardiology, Sri Ramachandra Institute of Higher Education & Research (SRIHER), Chennai, India
| |
Collapse
|
11
|
Brown JA, Bashir H, Zeng MY. Lifelong partners: Gut microbiota-immune cell interactions from infancy to old age. Mucosal Immunol 2025:S1933-0219(25)00006-6. [PMID: 39862964 DOI: 10.1016/j.mucimm.2025.01.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025]
Abstract
Our immune system and gut microbiota are intricately coupled from birth, both going through maturation during early life and senescence during aging almost in a synchronized fashion. The symbiotic relationship between the human host and microbiota is critically dependent on a healthy immune system to keep our microbiota in check, while the microbiota provides essential functions to promote the development and fitness of our immune system. The partnership between our immune system and microbiota is particularly important during early life, when microbial ligands and metabolites shape the development of the immune cells and immune tolerance; during aging, having sufficient beneficial gut bacteria is critical for the maintenance of intact mucosal barriers, immune metabolic fitness, and strong immunity against pathogens. The immune system during childhood is programmed, with the support of the microbiota, to develop robust immune tolerance, and limit autoimmunity and metabolic dysregulation, which are prevalent during aging. This review comprehensively explores the mechanistic underpinnings of gut microbiota-immune cell interactions during infancy and old age, with the goal to gain a better understanding of potential strategies to leverage the gut microbiota to combat age-related immune decline.
Collapse
Affiliation(s)
- Julia A Brown
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Hilal Bashir
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States
| | - Melody Y Zeng
- Gale and Ira Drukier Institute for Children's Health, Weill Cornell Medicine, New York, NY 10065, United States; Department of Pediatrics, Weill Cornell Medicine, New York, NY 10065, United States; Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School, New York, NY 10065, United States.
| |
Collapse
|
12
|
Debureaux PE, Poulain S, Harel S, Passet M, Templé M, Friedrich C, Forgeard N, Elessa D, Plas W, Chat L, Lazarian G, Willems L, Royer B, Talbot A, Vaugeois T, Theves F, Terré A, Brignier A, Malphettes M, Krzisch D, Frenzel L, Davi F, Bravetti C, Nguyen-Khac F, Dupuis J, Cuccuini W, Bouscary D, Hermine O, Roos-Weil D, Kosmider O, Clappier E, Espéli M, Balabanian K, Arnulf B. Inflammatory Waldenström macroglobulinemia is associated with clonal hematopoiesis: a multicentric cohort. Blood 2025; 145:450-454. [PMID: 39571148 DOI: 10.1182/blood.2024025738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 10/23/2024] [Indexed: 01/24/2025] Open
Abstract
ABSTRACT Inflammatory form of Waldenström macroglobulinemia (iWM) predicts outcomes after immuno-chemotherapy and Bruton tyrosine kinase inhibitors, but its origin is unknown. Here, we unravel increased clonal hematopoiesis in patients with iWM (61% vs 23% in noninflammatory WM), suggesting a contribution of environmental cells to iWM.
Collapse
Affiliation(s)
- Pierre-Edouard Debureaux
- Institut de Recherche Saint-Louis, University Paris Cité, INSERM U1160, Paris, France
- OPALE Carnot Institute, Saint-Louis Hospital, Paris, France
- Department of Immuno-Hematology, Saint-Louis Hospital, Paris, France
| | - Stéphanie Poulain
- Hematology Laboratory, Lille Unité Mixte de Recherche en Santé Hospital, Lille, France
| | - Stéphanie Harel
- Department of Immuno-Hematology, Saint-Louis Hospital, Paris, France
| | - Marie Passet
- Institut de Recherche Saint-Louis, University Paris Cité, INSERM U944/Unité Mixte de Recherche 7212, Paris, France
- Hematology Laboratory, Saint-Louis Hospital, Paris, France
| | - Marie Templé
- Hematobiology Unit, Cochin Hospital, Paris, France
| | | | - Nathalie Forgeard
- Department of Immuno-Hematology, Saint-Louis Hospital, Paris, France
| | - Dikelele Elessa
- Department of Immuno-Hematology, Saint-Louis Hospital, Paris, France
| | - William Plas
- Hematology Laboratory, Saint-Louis Hospital, Paris, France
| | - Laureen Chat
- Hematology Laboratory, Saint-Louis Hospital, Paris, France
| | | | | | - Bruno Royer
- Department of Immuno-Hematology, Saint-Louis Hospital, Paris, France
| | - Alexis Talbot
- Department of Immuno-Hematology, Saint-Louis Hospital, Paris, France
| | - Tristan Vaugeois
- Department of Immuno-Hematology, Saint-Louis Hospital, Paris, France
| | - Floriane Theves
- Department of Immuno-Hematology, Saint-Louis Hospital, Paris, France
| | | | | | | | - Daphné Krzisch
- Immuno-Oncology Unit, Saint-Louis Hospital, Paris, France
- Université Paris Cité, INSERM U1153, Paris, France
| | | | - Frédéric Davi
- Department of Hematology, Pitié-Salpêtrière Hospital, Paris, France
- Sorbonne University, Paris, France
| | | | - Florence Nguyen-Khac
- Laboratory of Cytogenetic, Pitié-Salpêtrière Hospital, Paris, France
- Sorbonne Université, INSERM Unité Mixte de Recherche en Santé 1138, Drug Resistance in Hematological Malignancies Team, Centre de Recherche des Cordeliers, Paris, France
| | - Jehan Dupuis
- Department of Lymphoid Malignancies, Henri Mondor Hospital, Paris, France
| | - Wendy Cuccuini
- Hematology Laboratory, Saint-Louis Hospital, Paris, France
| | | | | | - Damien Roos-Weil
- Department of Hematology, Pitié-Salpêtrière Hospital, Paris, France
- Sorbonne Université, INSERM Unité Mixte de Recherche en Santé 1138, Drug Resistance in Hematological Malignancies Team, Centre de Recherche des Cordeliers, Paris, France
| | | | - Emmanuelle Clappier
- Institut de Recherche Saint-Louis, University Paris Cité, INSERM U944/Unité Mixte de Recherche 7212, Paris, France
- Hematology Laboratory, Saint-Louis Hospital, Paris, France
| | - Marion Espéli
- Institut de Recherche Saint-Louis, University Paris Cité, INSERM U1160, Paris, France
- OPALE Carnot Institute, Saint-Louis Hospital, Paris, France
| | - Karl Balabanian
- Institut de Recherche Saint-Louis, University Paris Cité, INSERM U1160, Paris, France
- OPALE Carnot Institute, Saint-Louis Hospital, Paris, France
| | - Bertrand Arnulf
- Department of Immuno-Hematology, Saint-Louis Hospital, Paris, France
| |
Collapse
|
13
|
Qiu JY, Huang SS, Liu C, Ding D, Xu YH, Mao YM, Yuan YD. Genetic evidence for the causal effect of clonal hematopoiesis on pulmonary arterial hypertension. BMC Cardiovasc Disord 2025; 25:38. [PMID: 39849426 PMCID: PMC11755826 DOI: 10.1186/s12872-025-04475-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/03/2025] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is a severe and progressive cardiovascular disease. While potential links between clonal hematopoiesis (CH) and cardiovascular diseases have been identified, the causal relationship between CH and PAH remains unclear. This study aims to investigate the causal effect of CH on the risk of PAH using a two-sample Mendelian randomization (MR) approach. METHODS We utilized genetic variants associated with CH as instrumental variables, identified from two large genome-wide association studies (GWAS) involving 359,088 participants in the discovery cohort and 184,121 participants in the validation cohort, all of European descent. We obtained GWAS summary statistics for PAH. The inverse-variance weighted (IVW) method was employed as the primary analysis, complemented by sensitivity analyses to assess the robustness of our findings. A bidirectional MR analysis was conducted to estimate the causation between CH and PAH. RESULTS Our results indicate a causal effect of CH on the risk of PAH in the discovery cohort, with TET2 showing an IVW odds ratio (OR) of 1.200 (95% CI: 1.001-1.438, P = 0.049). Sensitivity analysis did not reveal significant pleiotropy or heterogeneity. In the validation cohort, we found that TET2 remains a risk factor for PAH (OR = 2.3E + 08, 95% CI 17.007-3.1E + 15, P = 0.022). Additionally, no causal relationship was found between other CH genes, such as DNMT3A and PAH (P > 0.05). The reverse MR analysis provided no evidence of causal effects of PAH on CH. CONCLUSION These findings showed that individuals with CH due to TET2 mutations may have a higher risk of developing PAH, suggesting that the CH patients may be tested for TET2 gene mutations.
Collapse
Affiliation(s)
- Jia-Yong Qiu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, Hebei, China
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, College of Clinical Medicine of Henan, University of Science and Technology, 24 Jinhua Road, Luoyang, China
| | - Shen-Shen Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, College of Clinical Medicine of Henan, University of Science and Technology, 24 Jinhua Road, Luoyang, China
| | - Chao Liu
- Department of Cardiology, Guangdong Provincial People's Hospital, Guangdong Cardiovascular Institute, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Dong Ding
- Institute of Clinical Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, National Infrastructures for Translational Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yan-Hong Xu
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, Hebei, China
| | - Yi-Min Mao
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, College of Clinical Medicine of Henan, University of Science and Technology, 24 Jinhua Road, Luoyang, China.
| | - Ya-Dong Yuan
- Department of Respiratory and Critical Care Medicine, The Second Hospital of Hebei Medical University, 215 Heping West Road, Shijiazhuang, Hebei, China.
| |
Collapse
|
14
|
Zhao X, Li J, Yan S, Tan Y, Chen R, Li N, Zhou J, Liu C, Zhou P, Chen Y, Yan H, Zhao H, Song L. Clonal hematopoiesis of indeterminate potential and type 2 diabetes mellitus among patients with STEMI: from a prospective cohort study combing bidirectional Mendelian randomization. Cardiovasc Diabetol 2025; 24:28. [PMID: 39844252 PMCID: PMC11756161 DOI: 10.1186/s12933-025-02588-w] [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: 12/10/2024] [Accepted: 01/08/2025] [Indexed: 01/24/2025] Open
Abstract
AIM Both clonal hematopoiesis of indeterminate potential (CHIP) and type 2 diabetes mellitus (T2DM) are conditions closely associated with advancing age. This study delves into the possible implications and prognostic significance of CHIP and T2DM in patients diagnosed with ST-segment elevation myocardial infarction (STEMI). METHODS Deep-targeted sequencing employing a unique molecular identifier (UMI) for the analysis of 42 CHIP mutations-achieving an impressive mean depth of coverage at 1000 × -was conducted on a cohort of 1430 patients diagnosed with acute myocardial infarction (473 patients with T2DM and 930 non-DM subjects). Variant allele fraction ≥ 2.0% indicated the presence of CHIP mutations. The association between CHIP and T2DM was evaluated by the comparison of (i) the prevalence of CHIP mutations among individuals with diabetes versus those without, (ii) the clinical characteristics delineated by CHIP mutations within the cohort of diabetic patients and (iii) the prognostic significance and correlation of CHIP mutations with mortality rates in T2DM subjects. Furthermore, a two-sample bidirectional Mendelian randomization study was performed using genetic instruments from the genome-wide association study for TET2 mutation CH from the UK Biobank (UKB) (2041 cases,173,918 controls) to investigate the causal relationship with T2DM from the FinnGen consortium (65,085 cases and 335,112 controls), and vice versa. RESULTS (i) Most commonly CHIP mutations exhibiting a variant allele fraction of ≥ 2.0% were identified in 50/473 (10.6%) patients with T2DM, demonstrating a greater prevalence compared to non-DM subjects [69/930 (7.4%); P < 0.05] across various age groups. (ii) After multivariable adjustment, the mortality of any CHIP mutations were 2.03-fold higher in DM [adjusted hazard ratio (HR) 2.03; 95% confidence interval (CI) 1.07-3.84, P < 0.05]. (iii) In gene-specific analyses, TET2 somatic mutation presented the highest association with mortality among T2DM (adjusted HR 5.24; 95% CI 2.02-13.61, P = 0.001). ASXL1 CHIP mutation which displayed a striking correlation with cardiac death (HR: 3.14; 95% CI 1.24-7.93; P < 0.05) with consistent associations observed among T2DM subgroup (HR: 4.51; 95% CI 1.30-15.6; P < 0.05). (iv) The correlation between PCSK9 and the Tet2-CHIP mutation was observed in both the T2DM cohort (correlation = 0.1215, P = 0.011) and the overall enrolled cohort (correlation = 0.0578, P = 0.0382). (v) Bidirectional Mendelian randomization studies indicated that the development of T2DM increases the propensity for CHIP. However, CHIP does not subsequently accelerate the onset of T2DM. CONCLUSION CHIP mutations, particularly TET2, are more prevalent in patients with T2DM compared to individuals without diabetes. The presence of these mutations is associated with adverse clinical outcomes, notably increased mortality rates. Moreover, bidirectional Mendelian randomization analyses provide supporting evidence for a potential causal relationship between TET2-related CHIP and the development of T2DM.
Collapse
Affiliation(s)
- Xiaoxiao Zhao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, No.167, Beijing, 100037, China
| | - Jiannan Li
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, No.167, Beijing, 100037, China
| | - Shaodi Yan
- Fuwai Hospital, Chinese Academy of Medical Sciences, 12 Langshan Rd, Shenzhen, 518000, China
| | - Yu Tan
- Department of Cardiovascular Medicine, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Runzhen Chen
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, No.167, Beijing, 100037, China
| | - Nan Li
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, No.167, Beijing, 100037, China
| | - Jinying Zhou
- Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Cardiovascular Diseases, Shanghai, China
| | - Chen Liu
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, No.167, Beijing, 100037, China
| | - Peng Zhou
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, No.167, Beijing, 100037, China
| | - Yi Chen
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, No.167, Beijing, 100037, China
| | - Hongbing Yan
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, No.167, Beijing, 100037, China.
- Fuwai Hospital, Chinese Academy of Medical Sciences, 12 Langshan Rd, Shenzhen, 518000, China.
| | - Hanjun Zhao
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, No.167, Beijing, 100037, China.
| | - Li Song
- Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Peking Union Medical College & Chinese Academy of Medical Sciences, No.167, Beijing, 100037, China.
| |
Collapse
|
15
|
Choi YJ, Choi J, Kang Y, Shin S, Lee ST, Choi JR. Evaluation of plasma cell sorting methods in multiple myeloma patients: flow cytometry versus magnetic beads. Cancer Cell Int 2025; 25:16. [PMID: 39825397 PMCID: PMC11740577 DOI: 10.1186/s12935-025-03647-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/09/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND The prognosis of a plasma cell neoplasm (PCN) varies depending on the presence of genetic abnormalities. However, detecting sensitive genetic mutations poses challenges due to the heterogeneous nature of the cell population in bone marrow aspiration. The established gold standard for cell sorting is fluorescence-activated cell sorting (FACS), which is associated with lengthy processing times, substantial cell quantities, and expensive equipment. Magnetic-activated cell sorting (MACS) can be performed without the need for FACS equipment and allows for rapid sorting of many cells, making it a practical alternative. Our objective is to conduct a comparative analysis of these two sorting techniques to assess whether MACS can viably replace FACS in clinical applications. METHODS Plasma cell purity, fluorescence in situ hybridization (FISH), and next-generation sequencing analyses were performed on FACS- and MACS-sorted bone marrow samples from 31 PCN patients. RESULTS The MACS-sorted samples yielded a higher percentage of plasma cells than FACS-sorted samples under microscopy (p = 0.0156) and flow cytometry (p = 0.0313). FISH performed by two methods in 10 samples showed the same results, and the proportion of abnormal cells was significantly higher in MACS than in FACS (p = 0.001). Wilcoxon matched-pairs signed rank test analysis showed that the median of differences of variant allele frequency (VAF) of two methods (VAF of MACS minus VAF of FACS) in the DNMT3A, TET2, and ASXL1 (DTA) group was - 0.006555 (p = 0.0020), while that in the non-DTA group was 0.002805 (p = 0.0019). Ten copy number variants (CNVs) were found in both FACS- and MACS-sorted samples, eight were identified only in MACS-sorted samples, and one was detected only in FACS-sorted samples. CONCLUSION Our study demonstrates that MACS is a viable alternative for plasma cell sorting in bone marrow samples of patients with PCN.
Collapse
Affiliation(s)
- Yu Jeong Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
| | - Jaeguk Choi
- Department of Laboratory Medicine, Severance Hospital, Seoul, Korea
| | - Yehyun Kang
- Graduate School of Medical Science, Brain Korea 21 PLUS Project, Yonsei University College of Medicine, Seoul, Korea
| | - Saeam Shin
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
| | - Seung-Tae Lee
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea
- Dxome Co. Ltd, Seongnam-si, Gyeonggi-do, Korea
| | - Jong Rak Choi
- Department of Laboratory Medicine, Yonsei University College of Medicine, 50-1 Yonsei-ro, Seodaemun-gu, Seoul, 03722, Korea.
- Dxome Co. Ltd, Seongnam-si, Gyeonggi-do, Korea.
| |
Collapse
|
16
|
Mayerhofer C, Freedman RA, Parsons HA, Partridge AH, Miller PG. Clonal Hematopoiesis in Women With Breast Cancer. J Clin Oncol 2025:JCO2401848. [PMID: 39823557 DOI: 10.1200/jco-24-01848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/25/2024] [Accepted: 12/17/2024] [Indexed: 01/19/2025] Open
Abstract
PURPOSE Clonal hematopoiesis (CH) has been associated with a variety of adverse outcomes, most notably hematologic malignancy and ischemic cardiovascular disease. A series of recent studies also suggest that CH may play a role in the outcomes of patients with solid tumors, including breast cancer. Here, we review the clinical and biological data that underlie potential connections between CH, inflammation, and breast cancer, with a focus on the prevalence and impact of clonal hematopoiesis of indeterminate potential in patients with breast cancer. METHODS We summarize data from multiple studies, including a series of cohorts of patients with breast cancer, to assess the prevalence of CH, the relationship between CH and exposure to cytotoxic therapy, and the correlation between CH and breast cancer-specific outcomes. RESULTS Our findings indicate that CH is prevalent among patients with breast cancer, particularly those treated with cytotoxic therapies. However, there are no definitive data to support an association between the presence of CH and breast cancer-specific outcomes. CONCLUSION Current data do not support routine CH testing in patients with breast cancer, nor should the presence of CH influence decisions regarding breast cancer therapy in most patients. However, larger, long-term studies are necessary to further define the implications of CH in patients with breast cancer and guide clinical decision making.
Collapse
Affiliation(s)
- Christina Mayerhofer
- Department of Stem Cell and Regenerative Biology, Harvard Stem Cell Institute, Cambridge, MA
- Center for Regenerative Medicine, Massachusetts General Hospital, Boston, MA
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA
| | - Rachel A Freedman
- Harvard Medical School, Boston, MA
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Heather A Parsons
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA
- Harvard Medical School, Boston, MA
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Ann H Partridge
- Harvard Medical School, Boston, MA
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Breast Oncology Program, Dana-Farber Brigham Cancer Center, Boston, MA
| | - Peter G Miller
- Broad Institute of the Massachusetts Institute of Technology and Harvard University, Cambridge, MA
- Harvard Medical School, Boston, MA
- Center for Cancer Research, Massachusetts General Hospital, Boston, MA
| |
Collapse
|
17
|
Casso-Chapa B, Vazquez González NA, Le NT, Palaskas NL, Nead KT, Eutsey LP, Samanthapudi VSK, Osborn AM, Lee J, Mejia G, Hoang O, Lin SH, Deswal A, Herrmann J, Wang G, Kirkland JL, Krishnan S, Wehrens XHT, Chini EN, Yusuf SW, Iliescu CA, Jain A, Burks JK, Seeley E, Lorenzi PL, Chau KM, Ostos Mendoza KC, Grumbach IM, Brookes PS, Hanssen NMJ, de Winther MPJ, Yvan-Charvet L, Kotla S, Schadler K, Abe JI. Reevaluating Anti-Inflammatory Therapy: Targeting Senescence to Balance Anti-Cancer Efficacy and Vascular Disease. Arterioscler Thromb Vasc Biol 2025. [PMID: 39817327 DOI: 10.1161/atvbaha.124.319870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Modulating immune function is a critical strategy in cancer and atherosclerosis treatments. For cancer, boosting or maintaining the immune system is crucial to prevent tumor growth. However, in vascular disease, mitigating immune responses can decrease inflammation and slow atherosclerosis progression. Anti-inflammatory therapy, therefore, presents a unique dilemma for cancer survivors: while it may decrease cardiovascular risk, it might also promote cancer growth and metastasis by suppressing the immune response. Senescence presents a potentially targetable solution to this challenge; senescence increases the risk of both cancer therapy resistance and vascular disease. Exercise, notably, shows promise in delaying this premature senescence, potentially improving cancer outcomes and lowering vascular disease risk post-treatment. This review focuses on the long-term impact of cancer therapies on vascular health. We underscore the importance of modulating senescence to balance cancer treatment's effectiveness and its vascular impact, and we emphasize investigating the role of exercise-mediated suppression of senescence in improving cancer survivorship.
Collapse
Affiliation(s)
- Bernardo Casso-Chapa
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Norma Alicia Vazquez González
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Nhat-Tu Le
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (N.-T.L., G.W., K.M.C.)
| | - Nicolas L Palaskas
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Kevin T Nead
- Department of Epidemiology, The University of Texas MD Anderson Cancer Center, Houston. (K.T.N.)
| | - Lydia P Eutsey
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Venkata S K Samanthapudi
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Abigail M Osborn
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Jonghae Lee
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston. (J.L., K.S.)
| | - Gilbert Mejia
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Oanh Hoang
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Steven H Lin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston. (S.H.L.)
| | - Anita Deswal
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Joerg Herrmann
- Division of Preventive Cardiology, Department of Cardiovascular Medicine, Cardio-Oncology Clinic, Mayo Clinic, Rochester, MN (J.H.)
| | - Guangyu Wang
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (N.-T.L., G.W., K.M.C.)
| | - James L Kirkland
- Division of Endocrinology and Metabolism, Department of Medicine, Center for Advanced Gerotherapeutics, Cedars-Sinai Medical Center, Los Angeles, CA (J.L.K.)
| | - Sunil Krishnan
- Department of Neurosurgery, The University of Texas Health Science Center at Houston (S. Krishnan)
| | - Xander H T Wehrens
- Department of Integrative Physiology, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX. (X.H.T.W.)
- Department of Medicine, Cardiovascular Research Institute, Baylor College of Medicine, Houston, TX. (X.H.T.W.)
| | - Eduardo N Chini
- Department of Anesthesiology and Perioperative Medicine, Mayo Clinic, Jacksonville, FL (E.N.C.)
| | - Syed Wamique Yusuf
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Cezar A Iliescu
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Abhishek Jain
- Department of Biomedical Engineering, Texas A&M University, College Station (A.J.)
| | - Jared K Burks
- Department of Leukemia, Division of Center Medicine, The University of Texas MD Anderson Cancer Center, Houston. (J.K.B.)
| | - Erin Seeley
- Department of Chemistry, University of Texas at Austin (E.S.)
| | - Philip L Lorenzi
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston. (P.L.L.)
| | - Khanh M Chau
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, TX (N.-T.L., G.W., K.M.C.)
| | - Keila Carolina Ostos Mendoza
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | | | - Paul S Brookes
- Department of Anesthesiology and Perioperative Medicine, University of Rochester, NY (P.S.B.)
| | - Nordin M J Hanssen
- Department of (Experimental) Vascular and Internal Medicine, Amsterdam UMC, the Netherlands (N.M.J.H.)
- Diabeter Centrum Amsterdam, the Netherlands (N.M.J.H.)
| | - Menno P J de Winther
- Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Atherosclerosis and Ischemic Syndromes, Amsterdam Institute for Immunology and Infectious Diseases, Inflammatory Diseases Amsterdam UMC, University of Amsterdam, the Netherlands (M.P.J.d.W.)
| | - Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale U1065, Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire, Fédération Hospitalo-Universitaire Oncoage, IHU RespirERA Respiratory Health, Environment and Ageing, Nice, France (L.Y.-C.)
| | - Sivareddy Kotla
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| | - Keri Schadler
- Department of Pediatric Research, The University of Texas MD Anderson Cancer Center, Houston. (J.L., K.S.)
| | - Jun-Ichi Abe
- Department of Cardiology, The University of Texas MD Anderson Cancer Center, Houston. (B.C.-C., N.A.V.G., N.L.P., L.P.E., V.S.K.S., A.M.O., J.L., G.M., O.H., A.D., S.W.Y., C.A.I., K.C.O.M., S. Kotla, J.-i.A.)
| |
Collapse
|
18
|
Regan JA, Kwee LC, Nafissi NA, Bick AG, Kraus WE, Natarajan P, Jaiswal S, Shah SH. Clonal Hematopoiesis Associates with Prevalent and Incident Cardiometabolic Disease in High-Risk Individuals. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2025:2025.01.14.25320566. [PMID: 39867361 PMCID: PMC11759844 DOI: 10.1101/2025.01.14.25320566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Background Clonal hematopoiesis of indeterminate potential (CHIP) is the age-related presence of expanded somatic clones secondary to leukemogenic driver mutations and is associated with cardiovascular (CV) disease and mortality. We sought to evaluate relationships between CHIP with cardiometabolic diseases and incident outcomes in high-risk individuals. Methods CHIP genotyping was performed in 8469 individuals referred for cardiac catheterization at Duke University (CATHGEN study) to identify variants present at a variant allele fraction (VAF) ≥2%. Associations were tested among any CHIP variant, large CHIP clones (VAF ≥10%) and individual CHIP genes with prevalent cardiometabolic traits. Cox proportional hazard models tested CHIP associations with time-to-overall mortality and Fine-Gray analyses tested CHIP associations with incident cardiovascular outcomes. Results We identified 463 CHIP variants in 427 individuals (5.0%) of which 268 (3.2%) harbored large CHIP clones. CHIP and large CHIP were associated with lower odds of obesity (OR 0.79 [95% CI 0.65-0.98], p=0.03; OR 0.76 [95% CI 0.57-0.99], p=0.04, respectively). CHIP was associated with prevalent HF (OR 1.25 [95% CI 1.01 - 1.55], p=0.04; especially for non- DNMT3A CHIP (OR 1.38 [95% CI 1.04-1.82], p=0.02). CHIP was also associated with incident events: Non- DNMT3A CHIP was associated with increased risk of time-to-HF hospitalization (HR 1.29 [95% CI 1.02-1.63], p=0.03). Conclusions In high-risk individuals referred for cardiac catheterization, large CHIP and non- DNTM3A CHIP were associated with obesity, prevalent HF, incident CV events. These findings strengthen the importance of CHIP as a biomarker for CV disease and highlight the contributing risk of large CHIP clones and non- DNMT3A CHIP variants. Condensed Abstract CHIP, the presence of somatic expanded mutations in myeloid driver genes in hematopoietic cells, is an emerging CVD biomarker. Using whole exome sequencing of peripheral blood derived DNA from participants in the CATHGEN cohort, we identified significant associations with obesity, prevalent HF, incident mortality, HF hospitalization and AF after adjusting for established clinical risk factors. These findings add strength to the growing literature of CHIP as a CVD biomarker, emphasizing large CHIP and non- DNMT3A CHIP variants for driving risk. Future studies should aim to further elucidate gene-specific risk and the inflammatory and metabolic mechanisms possibly mediating these relationships. Clinical Perspective What Is New?: In a cohort with high prevalence of CAD, CHIP is inversely associated with obesity and associated with higher odds of prevalent HF and subsequent mortality, even after adjustment for relevant clinical comorbidities. Risk of incident events of mortality, HF hospitalization and AF were driven by large CHIP variants (VAF≥10%) and CHIP variants in genes other than DNMT3A . What are the Clinical Implications?: Though more research is needed, as the evidence around the risk associated with specific CHIP variants continues to grow, clinicians should be prepared to provide gene- specific counseling for cardiometabolic disease risk.
Collapse
|
19
|
Tremblay D, Kremyanskaya M, Mascarenhas J, Hoffman R. Diagnosis and Treatment of Polycythemia Vera: A Review. JAMA 2025; 333:153-160. [PMID: 39556352 DOI: 10.1001/jama.2024.20377] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Importance Polycythemia vera (PV), a myeloproliferative neoplasm characterized by an increased red blood cell mass and increased risk of thrombosis, affects approximately 65 000 people in the US, with an annual incidence of 0.5 to 4.0 cases per 100 000 persons. Observations Erythrocytosis (hemoglobin >16.5 mg/dL in men or >16.0 mg/dL in women) is a required diagnostic criterion, although thrombocytosis (53%) and leukocytosis (49%) are common. Patients may have pruritus (33%), erythromelalgia (5.3%), transient visual changes (14%), and splenomegaly (36%) with abdominal discomfort. More than 95% of patients have a JAK2 gene variant, which helps distinguish PV from secondary causes of erythrocytosis, such as tobacco smoking or sleep apnea. Among 7 cohorts (1545 individuals), the median survival from diagnosis was 14.1 to 27.6 years. Prior to or at the time of PV diagnosis, arterial thrombosis occurred in 16% of patients and 7% had venous thrombotic events, which could involve unusual sites, such as splanchnic veins. PV is also associated with an increased bleeding risk, especially in patients with acquired von Willebrand disease, which can occur with extreme thrombocytosis (platelet count, ≥1000 × 109/L). All patients with PV should receive therapeutic phlebotomy (goal hematocrit, <45%) and low-dose aspirin (if no contraindications). Patients who are at higher risk of thrombosis include those aged 60 years or older or with a prior thrombosis. These patients and those with persistent PV symptoms may benefit from cytoreductive therapy with hydroxyurea or interferon to lower thrombosis risk and decrease symptoms. Ruxolitinib is a Janus kinase inhibitor that can alleviate pruritus and decrease splenomegaly in patients who are intolerant of or resistant to hydroxyurea. About 12.7% of patients with PV develop myelofibrosis and 6.8% develop acute myeloid leukemia. Conclusions and Relevance PV is a myeloproliferative neoplasm characterized by erythrocytosis and is almost universally associated with a JAK2 gene variant. PV is associated with an increased risk of arterial and venous thrombosis, hemorrhage, myelofibrosis, and acute myeloid leukemia. To decrease the risk of thrombosis, all patients with PV should be treated with aspirin and therapeutic phlebotomy to maintain a hematocrit of less than 45%. Cytoreductive therapies, such as hydroxyurea or interferon, are recommended for patients at high risk of thrombosis.
Collapse
Affiliation(s)
- Douglas Tremblay
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Marina Kremyanskaya
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - John Mascarenhas
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Ronald Hoffman
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York
| |
Collapse
|
20
|
Santisteban MM, Iadecola C. The pathobiology of neurovascular aging. Neuron 2025; 113:49-70. [PMID: 39788087 DOI: 10.1016/j.neuron.2024.12.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 01/12/2025]
Abstract
As global life expectancy increases, age-related brain diseases such as stroke and dementia have become leading causes of death and disability. The aging of the neurovasculature is a critical determinant of brain aging and disease risk. Neurovascular cells are particularly vulnerable to aging, which induces significant structural and functional changes in arterial, venous, and lymphatic vessels. Consequently, neurovascular aging impairs oxygen and glucose delivery to active brain regions, disrupts endothelial transport mechanisms essential for blood-brain exchange, compromises proteostasis by reducing the clearance of potentially toxic proteins, weakens immune surveillance and privilege, and deprives the brain of key growth factors required for repair and renewal. In this review, we examine the effects of neurovascular aging on brain function and its role in stroke, vascular cognitive impairment, and Alzheimer's disease. Finally, we discuss key unanswered questions that must be addressed to develop neurovascular strategies aimed at promoting healthy brain aging.
Collapse
Affiliation(s)
- Monica M Santisteban
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Costantino Iadecola
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
21
|
Esai Selvan M, Nathan DI, Guisado D, Collatuzzo G, Iruvanti S, Boffetta P, Mascarenhas J, Hoffman R, Cohen LJ, Marcellino BK, Gümüş ZH. Clonal Hematopoiesis of Indeterminate Potential in Crohn's Disease and Ulcerative Colitis. Inflamm Bowel Dis 2025:izae312. [PMID: 39761417 DOI: 10.1093/ibd/izae312] [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: 07/30/2024] [Indexed: 01/15/2025]
Abstract
BACKGROUND Clonal hematopoiesis of indeterminate potential (CHIP) is the presence of somatic mutations in myeloid and lymphoid malignancy genes in the blood cells of individuals without a hematologic malignancy. Inflammation is hypothesized to be a key mediator in the progression of CHIP to hematologic malignancy and patients with CHIP have a high prevalence of inflammatory diseases. This study aimed to identify the prevalence and characteristics of CHIP in patients with inflammatory bowel disease (IBD). METHODS We analyzed whole-exome sequencing data from 587 Crohn's disease (CD), 441 ulcerative colitis (UC), and 293 non-IBD controls to assess CHIP prevalence and used logistic regression to study associations with clinical outcomes. RESULTS Older UC patients (age > 45) harbored increased myeloid-CHIP mutations compared to younger patients (age ≤ 45) (P = .01). Lymphoid-CHIP was more prevalent in older IBD patients (P = .007). Young CD patients were found to have myeloid-CHIP with high-risk features. Inflammatory bowel disease patients with CHIP exhibited unique mutational profiles compared to controls. Steroid use was associated with increased CHIP (P = .05), while anti-TNF therapy was associated with decreased myeloid-CHIP (P = .03). Pathway enrichment analyses indicated an overlap between CHIP genes, IBD phenotypes, and inflammatory pathways. CONCLUSIONS Our findings underscore a connection between IBD and CHIP pathophysiology. Patients with IBD and CHIP had unique risk profiles, especially among older UC patients and younger CD patients. These findings suggest distinct evolutionary pathways for CHIP in IBD and necessitate awareness among IBD providers and hematologists to identify patients potentially at risk for CHIP-related complications including malignancy, cardiovascular disease, and acceleration of their inflammatory disease.
Collapse
Affiliation(s)
- Myvizhi Esai Selvan
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, Box 1498, New York, NY 10029, USA
| | - Daniel I Nathan
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, Box 1079, New York, NY 10029, USA
| | - Daniela Guisado
- Division of Pediatric Gastroenterology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, Box 1069, New York, NY 10029, USA
| | - Giulia Collatuzzo
- Department of Medical and Surgical Sciences, University of Bologna, Via Zamboni, 33 - 40126, Bologna, Italy
| | - Sushruta Iruvanti
- Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, Box 1498, New York, NY 10029, USA
| | - Paolo Boffetta
- Department of Medical and Surgical Sciences, University of Bologna, Via Zamboni, 33 - 40126, Bologna, Italy
- Stony Brook Department of Family, Population and Preventive Medicine, Renaissance School of Medicine, 101 Nicolls Road Health Sciences Center, Level 4, Stony Brook, NY 11794, USA
| | - John Mascarenhas
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, Box 1079, New York, NY 10029, USA
| | - Ronald Hoffman
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, Box 1079, New York, NY 10029, USA
| | - Louis J Cohen
- Division of Gastroenterology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, Box 1069, New York, NY 10029, USA
| | - Bridget K Marcellino
- Tisch Cancer Institute, Division of Hematology and Medical Oncology, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, Box 1079, New York, NY 10029, USA
| | - Zeynep H Gümüş
- Department of Genetics and Genomics, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, Box 1498, New York, NY 10029, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, Box 1630, New York, NY 10029, USA
| |
Collapse
|
22
|
Yang S, Penna V, Lavine KJ. Functional diversity of cardiac macrophages in health and disease. Nat Rev Cardiol 2025:10.1038/s41569-024-01109-8. [PMID: 39743564 DOI: 10.1038/s41569-024-01109-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/21/2024] [Indexed: 01/04/2025]
Abstract
Macrophages make up a substantial portion of the stromal compartment of the heart in health and disease. In the past decade, the origins of these cardiac macrophages have been established as two broad populations derived from either embryonic or definitive haematopoiesis and that can be distinguished by the expression of CC-motif chemokine receptor 2 (CCR2). These cardiac macrophage populations are transcriptionally distinct and have differing cell surface markers and divergent roles in cardiac homeostasis and disease. Embryonic-derived CCR2- macrophages are a tissue-resident population that participates in tissue development, repair and maintenance, whereas CCR2+ macrophages are derived from definitive haematopoiesis and contribute to inflammation and tissue damage. Studies from the past 5 years have leveraged single-cell RNA sequencing technologies to expand our understanding of cardiac macrophage diversity, particularly of the monocyte-derived macrophage populations that reside in the injured and diseased heart. Emerging technologies in spatial transcriptomics have enabled the identification of distinct disease-associated cellular neighbourhoods consisting of macrophages, other immune cells and fibroblasts, highlighting the involvement of macrophages in cell-cell communication. Together, these discoveries lend new insights into the role of specific macrophage populations in the pathogenesis of cardiac disease, which can pave the way for the identification of new therapeutic targets and the development of diagnostic tools. In this Review, we discuss the developmental origin of cardiac macrophages and describe newly identified cell states and associated cellular neighbourhoods in the steady state and injury settings. We also discuss various contributions and effector functions of cardiac macrophages in homeostasis and disease.
Collapse
Affiliation(s)
- Steven Yang
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Vinay Penna
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA
| | - Kory J Lavine
- Center for Cardiovascular Research, Division of Cardiology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Pathology and Immunology, Washington University School of Medicine, Saint Louis, MO, USA.
- Department of Developmental Biology, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
23
|
Yvan-Charvet L, Barouillet T, Borowczyk C. Haematometabolism rewiring in atherosclerotic cardiovascular disease. Nat Rev Cardiol 2025:10.1038/s41569-024-01108-9. [PMID: 39743562 DOI: 10.1038/s41569-024-01108-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 01/04/2025]
Abstract
Atherosclerotic cardiovascular diseases are the most frequent cause of death worldwide. The clinical complications of atherosclerosis are closely linked to the haematopoietic and immune systems, which maintain homeostatic functions and vital processes in the body. The nodes linking metabolism and inflammation are receiving increasing attention because they are inextricably linked to inflammatory manifestations of non-communicable diseases, including atherosclerosis. Although metabolism and inflammation are essential to survival and involve all tissues, we still know little about how these processes influence each other. In an effort to understand these mechanisms, in this Review we explore whether and how potent cardiovascular risk factors and metabolic modifiers of atherosclerosis influence the molecular and cellular machinery of 'haematometabolism' (metabolic-dependent haematopoietic stem cell skewing) and 'efferotabolism' (metabolic-dependent efferocyte reprogramming). These changes might ultimately propagate a quantitative and qualitative drift of the macrophage supply chain and affect the clinical manifestations of atherosclerosis. Refining our understanding of the different metabolic requirements of these processes could open the possibility of developing therapeutics targeting haematometabolism that, in conjunction with improved dietary habits, help rebalance and promote efficient haematopoiesis and efferocytosis and decrease the risk of atherosclerosis complications.
Collapse
Affiliation(s)
- Laurent Yvan-Charvet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| | - Thibault Barouillet
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France
| | - Coraline Borowczyk
- Institut National de la Santé et de la Recherche Médicale (Inserm) U1065, Nice, France.
- Université Côte d'Azur, Centre Méditerranéen de Médecine Moléculaire (C3M), Nice, France.
- Fédération Hospitalo-Universitaire (FHU) Oncoage, IHU ResprERA Respiratory Health, Environment and Ageing (RespirERA), Nice, France.
| |
Collapse
|
24
|
Supriami K, Urbut SM, Tello-Ayala JR, Unlu O, Friedman SF, Abou-Karam R, Koyama S, Uddin MM, Pomerantsev E, Lu MT, Honigberg MC, Aragam KG, Doshi-Velez F, Patel AP, Natarajan P, Ellinor PT, Fahed AC. Genomic Drivers of Coronary Artery Disease and Risk of Future Outcomes After Coronary Angiography. JAMA Netw Open 2025; 8:e2455368. [PMID: 39836422 PMCID: PMC11751748 DOI: 10.1001/jamanetworkopen.2024.55368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 11/11/2024] [Indexed: 01/22/2025] Open
Abstract
Importance Disease characteristics of genetically mediated coronary artery disease (CAD) on coronary angiography and the association of genomic risk with outcomes after coronary angiography are not well understood. Objective To assess the angiographic characteristics and risk of post-coronary angiography outcomes of patients with genomic drivers of CAD: familial hypercholesterolemia (FH), high polygenic risk score (PRS), and clonal hematopoiesis of indeterminate potential (CHIP). Design, Setting, and Participants A retrospective cohort study of 3518 Mass General Brigham Biobank participants with genomic information who underwent coronary angiography was conducted between July 18, 2000, and August 1, 2023. Exposures The presence of a genomic risk factor of CAD, defined as FH variant, high CAD PRS, or CHIP driver variation. Main Outcomes and Measures Coronary artery disease presentation (stable or acute), angiographic CAD characteristics (severity and burden), angiographic outcomes (repeat angiogram, revascularization, and in-stent restenosis), and clinical outcomes (heart failure and all-cause mortality). Results Among 3518 participants (2467 [70.1%] male; median age, 64.0 [IQR, 55.0-72.0] years), 1509 (42.9%) had at least 1 genomic driver of CAD (26 FH, 1191 high CAD PRS, and 466 CHIP) that was associated with the presentation of acute coronary syndromes (adjusted odds ratio, 2.67; 95% CI, 2.19-3.26) and with the presence, burden, and severity of angiographic CAD. This association was driven by FH and CAD PRS. One SD of CAD PRS was associated with a 12.51-point higher Gensini score. During 9 years of follow-up, there was an increased risk among FH carriers for a repeat angiogram (adjusted hazard ratio [AHR], 1.70; 95% CI, 1.02-2.83), and revascularization (AHR, 1.97; 95% CI, 1.02-3.80), and among people with high CAD PRS (repeat angiogram: AHR, 1.79; 95% CI, 1.45-2.22; revascularization: AHR, 1.85; 95% CI, 1.37-2.50; and in-stent restenosis: AHR, 3.89; 95% CI, 2.16-7.01). CHIP carriers had no significant increase in angiographic outcomes but were at higher risk of heart failure (AHR, 1.58; 95% CI, 1.04-2.40) and all-cause mortality (AHR, 1.78; 95% CI, 1.47-2.16). Conclusions and Relevance The findings of this study suggest that germline monogenic and polygenic risk are associated with acute coronary syndromes presentation, severity and burden of atherosclerosis, and risk of repeat angiogram, revascularization, and in-stent restenosis. CHIP variant status is associated with incident heart failure and mortality after coronary angiography.
Collapse
Affiliation(s)
- Kelvin Supriami
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
| | - Sarah M. Urbut
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
| | - José R. Tello-Ayala
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Ozan Unlu
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Brigham and Women’s Hospital, Boston, Massachusetts
- Department of Biomedical Informatics, Harvard Medical School, Boston, Massachusetts
| | - Samuel F. Friedman
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
| | - Roukoz Abou-Karam
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
| | - Satoshi Koyama
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
| | - Md Mesbah Uddin
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
| | - Eugene Pomerantsev
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
| | - Michael T. Lu
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Imaging Research Center, Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston
| | - Michael C. Honigberg
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
| | - Krishna G. Aragam
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
| | - Finale Doshi-Velez
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, Massachusetts
| | - Aniruddh P. Patel
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
| | - Pradeep Natarajan
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
- Personalized Medicine, Mass General Brigham, Boston, Massachusetts
| | - Patrick T. Ellinor
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
| | - Akl C. Fahed
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Cardiovascular Disease Initiative, Broad Institute of MIT and Harvard, Cambridge, Massachusetts
- Division of Cardiology, Massachusetts General Hospital, Boston
- Center for Genomic Medicine, Massachusetts General Hospital, Boston
| |
Collapse
|
25
|
Agarwal S, Qamar U, Khan MS, Al-Juhaishi T, Naqash AR, Guha A, Yang EH, Barac A, Ul Abideen Asad Z. Trends and disparities in cardiovascular disease-related mortality among adults with myeloproliferative neoplasms in USA. EUROPEAN HEART JOURNAL OPEN 2025; 5:oeae101. [PMID: 39781540 PMCID: PMC11707680 DOI: 10.1093/ehjopen/oeae101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/02/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025]
Abstract
Aims We aimed to perform a retrospective cohort study using the Centers for Disease Control and Prevention's (CDC's) Wide-Ranging Online Data for Epidemiologic Research (WONDER) database to analyse the trends in cardiovascular disease (CVD)-related mortality in patients with myeloproliferative neoplasms (MPNs) from 1999 to 2020. Methods and results We analysed the death certificate data from the CDC WONDER database from 1999 to 2020 for CVD with co-morbid myeloproliferative disorders in the US population. Age-adjusted mortality rates (AAMRs) and 95% confidence intervals (CIs) were computed per 1 million population by standardizing crude mortality rates to the 2000 US census population. To assess annual national mortality trends, we employed the Joinpoint regression model, calculating the annual per cent change in AAMR and corresponding 95% CIs. A total of 15 269 deaths related to CVD occurred in patients with co-morbid MPNs from 1999 to 2020. Overall, there was a decreasing trend in CVD-related AAMRs throughout these years. Males contributed to 51% of total deaths, and their AAMR was persistently higher than women throughout the study. Non-Hispanic (NH) Whites had the highest overall AAMR, followed by NH Blacks, NH American Indians or Alaska Natives, Hispanics or Latinos, and NH Asian or Pacific Islanders. Conclusion Our findings indicate a significant decline with notable gender, racial/ethnic, and regional differences in CVD-related mortality among patients with MPN over the past two decades. We emphasize the importance of a collaborative approach between oncologists and cardiologists in managing these patients, highlighting the potential benefits of integrating cardio-oncology services to enhance patient outcomes.
Collapse
Affiliation(s)
- Siddharth Agarwal
- Department of Cardiovascular Disease, Mayo Clinic, Rochester, MN 55905, USA
| | - Usama Qamar
- Department of Medicine, King Edward Medical University, Lahore 54000, Pakistan
| | - Muhammad Shahzeb Khan
- Department of Cardiovascular Disease, Duke University School of Medicine, Durham, NC 27710, USA
| | - Taha Al-Juhaishi
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 63117, USA
| | - Abdul Rafeh Naqash
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 63117, USA
| | - Avirup Guha
- Cardio-Oncology Program, Medical College of Georgia at Augusta University, Augusta, GA 30912, USA
| | - Eric H Yang
- UCLA Cardio-Oncology Program, Division of Cardiology, Department of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | - Ana Barac
- Inova Schar Cancer Institute and Inova Heart and Vascular Institute, Fairfax, VA 22042, USA
| | - Zain Ul Abideen Asad
- Department of Medicine, University of Oklahoma Health Sciences Center, Oklahoma City, OK 63117, USA
| |
Collapse
|
26
|
Kristiansen MH, Larsen MK, Massarenti L, Skov V, Kjær L, Enevold C, Ostrowski SR, Nielsen CH, Hasselbalch HC, Wienecke T. Thromboinflammation in ischemic cerebrovascular patients with the JAK2V617F mutation. Thromb Res 2025; 245:109236. [PMID: 39652998 DOI: 10.1016/j.thromres.2024.109236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 10/23/2024] [Accepted: 12/03/2024] [Indexed: 12/30/2024]
Abstract
BACKGROUND The JAK2V617F mutation is a driver of Philadelphia chromosome-negative myeloproliferative neoplasms (MPN) and is also implicated in cardiovascular diseases. Thrombosis in MPN involves JAK2V617F-associated platelet activation and endothelial dysfunction, all potentially influenced by chronic inflammation. Whether the mutation affects thromboinflammatory markers similarly in non-MPN patients remains unclear. METHOD We conducted a study involving 63 ischemic cerebrovascular patients with the JAK2V617F mutation, matched with 63 patients without the mutation. Serum samples were analyzed for 12 thromboinflammatory markers during the acute phase and at three months follow-up. RESULTS Overall, there was no significant difference in thromboinflammatory markers between cases and controls. However, subgroup analysis of patients with a JAK2V617F allele burden ≥1 % (n = 15) showed higher levels of Vascular Cell Adhesion Molecule-1 (VCAM-1) at baseline (p = 0.018), and elevated Interleukin-10 (IL-10) (p = 0.004) and Tumor Necrosis Factor α (TNF-α) (p = 0.018) at follow-up compared to controls. Regression analysis revealed an association between higher JAK2V617F allele burden and increased VCAM-1 at baseline (p < 0.001), and higher VCAM-1 (p = 0.012), IL-10 (p = 0.003), and TNF-α (p = 0.034) at follow-up. CONCLUSION In ischemic cerebrovascular patients, the JAK2V617F mutation is associated with elevated markers of endothelial dysfunction and chronic inflammation. This underscores the role of inflammation in thrombosis driven by the JAK2V617F mutation.
Collapse
Affiliation(s)
- Marie Hvelplund Kristiansen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Neurology, Zealand University Hospital, Roskilde, Denmark.
| | - Morten Kranker Larsen
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Laura Massarenti
- Section for Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Vibe Skov
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Lasse Kjær
- Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Christian Enevold
- Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Sisse Rye Ostrowski
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Immunology, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Claus Henrik Nielsen
- Section for Oral Biology and Immunopathology, Department of Odontology, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Institute for Inflammation Research, Center for Rheumatology and Spine Diseases, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Hans Carl Hasselbalch
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Hematology, Zealand University Hospital, Roskilde, Denmark
| | - Troels Wienecke
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Department of Neurology, Zealand University Hospital, Roskilde, Denmark
| |
Collapse
|
27
|
Shaban D, Najm N, Droin L, Nijnik A. Hematopoietic Stem Cell Fates and the Cellular Hierarchy of Mammalian Hematopoiesis: from Transplantation Models to New Insights from in Situ Analyses. Stem Cell Rev Rep 2025; 21:28-44. [PMID: 39222178 DOI: 10.1007/s12015-024-10782-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/17/2024] [Indexed: 09/04/2024]
Abstract
Hematopoiesis is the process that generates the cells of the blood and immune system from hematopoietic stem and progenitor cells (HSPCs) and represents the system with the most rapid cell turnover in a mammalian organism. HSPC differentiation trajectories, their underlying molecular mechanisms, and their dysfunctions in hematologic disorders are the focal research questions of experimental hematology. While HSPC transplantations in murine models are the traditional tool in this research field, recent advances in genome editing and next generation sequencing resulted in the development of many fundamentally new approaches for the analyses of mammalian hematopoiesis in situ and at single cell resolution. The current review will cover many recent developments in this field in murine models, from the bulk lineage tracing studies of HSPC differentiation to the barcoding of individual HSPCs with Cre-recombinase, Sleeping Beauty transposase, or CRISPR/Cas9 tools, to map hematopoietic cell fates, together with their transcriptional and epigenetic states. We also address studies of the clonal dynamics of human hematopoiesis, from the tracing of HSPC clonal behaviours based on viral integration sites in gene therapy patients to the recent analyses of unperturbed human hematopoiesis based on naturally accrued mutations in either nuclear or mitochondrial genomes. Such studies are revolutionizing our understanding of HSPC biology and hematopoiesis both under homeostatic conditions and in the response to various forms of physiological stress, reveal the mechanisms responsible for the decline of hematopoietic function with age, and in the future may advance the understanding and management of the diverse disorders of hematopoiesis.
Collapse
Affiliation(s)
- Dania Shaban
- Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC, H3G 0B1, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Nay Najm
- Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC, H3G 0B1, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Lucie Droin
- Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC, H3G 0B1, Canada
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada
| | - Anastasia Nijnik
- Department of Physiology, McGill University, 368 Bellini Life Sciences Complex, 3649 Promenade Sir William Osler, Montreal, QC, H3G 0B1, Canada.
- McGill University Research Centre on Complex Traits, McGill University, Montreal, QC, Canada.
| |
Collapse
|
28
|
Krishnan T, Solar Vasconcelos JP, Titmuss E, Vanner RJ, Schaeffer DF, Karsan A, Lim H, Ho C, Gill S, Yip S, Chia SK, Kennecke HF, Jonker DJ, Chen EX, Renouf DJ, O’Callaghan CJ, Loree JM. Clonal Hematopoiesis of Indeterminate Potential and its Association with Treatment Outcomes and Adverse Events in Patients with Solid Tumors. CANCER RESEARCH COMMUNICATIONS 2025; 5:66-73. [PMID: 39636207 PMCID: PMC11713863 DOI: 10.1158/2767-9764.crc-24-0522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 11/14/2024] [Accepted: 12/03/2024] [Indexed: 12/07/2024]
Abstract
SIGNIFICANCE Liquid biopsy is increasingly being used in oncology for tumor molecular characterization. CHIP is a common incidental finding in cfDNA, and its prevalence increases with age. This study builds on growing evidence of common CHIP variants in patients with solid tumors. The results suggest a possible clinical impact of CHIP on treatment outcomes from immunotherapy or chemotherapy. This may have implications for treatment selection for patients with solid tumors.
Collapse
Affiliation(s)
- Tharani Krishnan
- Medical Oncology Department, BC Cancer–Vancouver, Vancouver, Canada
| | | | - Emma Titmuss
- Medical Oncology Department, BC Cancer–Vancouver, Vancouver, Canada
| | - Robert J. Vanner
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | | | - Aly Karsan
- Department of Hematology, BC Cancer–Vancouver, Vancouver, Canada
| | - Howard Lim
- Medical Oncology Department, BC Cancer–Vancouver, Vancouver, Canada
| | - Cheryl Ho
- Medical Oncology Department, BC Cancer–Vancouver, Vancouver, Canada
| | - Sharlene Gill
- Medical Oncology Department, BC Cancer–Vancouver, Vancouver, Canada
| | - Stephen Yip
- Pathology Department, Vancouver General Hospital, Vancouver, Canada
| | - Stephen K. Chia
- Medical Oncology Department, BC Cancer–Vancouver, Vancouver, Canada
| | - Hagen F. Kennecke
- Medical Oncology Department, Providence Cancer Institute, Portland, Oregon
| | - Derek J. Jonker
- Medical Oncology Department, Ottawa Hospital Cancer Centre, Ottawa, Canada
| | - Eric X. Chen
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada
| | - Daniel J. Renouf
- Medical Oncology Department, BC Cancer–Vancouver, Vancouver, Canada
| | | | | |
Collapse
|
29
|
Hoermann G, Khoury JD. Can molecular patterns help to classify overlapping entities in myeloid neoplasms? Histopathology 2025; 86:146-157. [PMID: 39428913 PMCID: PMC11648353 DOI: 10.1111/his.15339] [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: 10/22/2024]
Abstract
Myeloid neoplasms include myeloproliferative and myelodysplastic neoplasms and acute myeloid leukaemia. Historically, these diseases have been diagnosed based on clinicopathological features with sometimes arbitrary thresholds that have persisted even as molecular features were gradually incorporated into their classification. As such, although current diagnostic approaches can classify the majority of myeloid neoplasms accurately using a combination of molecular and clinicopathological features, some areas of overlap persist and occasionally pose diagnostic challenges. These include overlap across BCR::ABL1-negative myeloproliferative neoplasms; between clonal cytopenia of undetermined significance and myelodysplastic neoplasms; myelodysplastic/myeloproliferative neoplasms; and, detection of KIT mutations in myeloid neoplasms other than mastocytosis, raising the prospect of systemic mastocytosis. Molecular testing has become state of the art in the diagnostic work-up of myeloid neoplasms, and molecular patterns can inherently help to classify overlapping entities if considered within a framework of haematological presentations. For future development, molecular testing will likely include whole genome and transcriptome sequencing, and primarily molecular classifications of myeloid neoplasms have already been suggested. As such, genetically defined groups should still constitute the basis for our understanding of disease development from early onset to progression, while clinicopathological features could then be used to describe the stage of the disease rather than the specific type of myeloid neoplasm.
Collapse
Affiliation(s)
| | - Joseph D Khoury
- Department of Pathology, Microbiology, and ImmunologyUniversity of Nebraska Medical CenterOmahaUSA
| |
Collapse
|
30
|
Beeraka NM, Basappa B, Nikolenko VN, Mahesh PA. Role of Neurotransmitters in Steady State Hematopoiesis, Aging, and Leukemia. Stem Cell Rev Rep 2025; 21:2-27. [PMID: 38976142 DOI: 10.1007/s12015-024-10761-z] [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] [Accepted: 07/02/2024] [Indexed: 07/09/2024]
Abstract
Haematopoiesis within the bone marrow (BM) represents a complex and dynamic process intricately regulated by neural signaling pathways. This delicate orchestration is susceptible to disruption by factors such as aging, diabetes, and obesity, which can impair the BM niche and consequently affect haematopoiesis. Genetic mutations in Tet2, Dnmt3a, Asxl1, and Jak2 are known to give rise to clonal haematopoiesis of intermediate potential (CHIP), a condition linked to age-related haematological malignancies. Despite these insights, the exact roles of circadian rhythms, sphingosine-1-phosphate (S1P), stromal cell-derived factor-1 (SDF-1), sterile inflammation, and the complement cascade on various BM niche cells remain inadequately understood. Further research is needed to elucidate how BM niche cells contribute to these malignancies through neural regulation and their potential in the development of gene-corrected stem cells. This literature review describes the updated functional aspects of BM niche cells in haematopoiesis within the context of haematological malignancies, with a particular focus on neural signaling and the potential of radiomitigators in acute radiation syndrome. Additionally, it underscores the pressing need for technological advancements in stem cell-based therapies to alleviate the impacts of immunological stressors. Recent studies have illuminated the microheterogeneity and temporal stochasticity of niche cells within the BM during haematopoiesis, emphasizing the updated roles of neural signaling and immunosurveillance. The development of gene-corrected stem cells capable of producing blood, immune cells, and tissue-resident progeny is essential for combating age-related haematological malignancies and overcoming immunological challenges. This review aims to provide a comprehensive overview of these evolving insights and their implications for future therapeutic strategies.
Collapse
Affiliation(s)
- Narasimha M Beeraka
- Herman B. Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, 1044 W. Walnut Street, R4-168, Indianapolis, IN, 46202, USA.
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russia.
- Raghavendra Institute of Pharmaceutical Education and Research (RIPER), Anantapuramu, Chiyyedu, Andhra Pradesh, 515721, India.
| | - Basappa Basappa
- Department of Studies in Organic Chemistry, Laboratory of Chemical Biology, University of Mysore, Mysore, Karnataka, 570006, India
| | - Vladimir N Nikolenko
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University of the Ministry of Health of the Russian Federation (Sechenov University), 8/2 Trubetskaya Str., Moscow, 119991, Russia
| | - P A Mahesh
- Department of Pulmonary Medicine, JSS Medical College, JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India
| |
Collapse
|
31
|
Englisch C, Vostatek R, Schramm T, Binder CJ, Pabinger I, Jäger R, Ay C. Association of clonal haematopoiesis with recurrent venous thromboembolism: A case-control study. Br J Haematol 2025; 206:263-271. [PMID: 39497597 PMCID: PMC11739764 DOI: 10.1111/bjh.19871] [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: 08/12/2024] [Accepted: 10/19/2024] [Indexed: 01/18/2025]
Abstract
Venous thromboembolism (VTE) is the third most common cardiovascular disease. Clonal haematopoiesis (CH) is linked to cardiovascular disease risk, but its potential association with VTE remains poorly understood. We assessed the prevalence of CH in patients with recurrent VTE (n = 107; median age [IQR] 57 [48-63] years, 44.9% female) and matched healthy controls (n = 127; median age [IQR] 53 [45-60] years, 51.2% female) to investigate a putative association of CH with VTE risk. We detected 12 CH-associated mutations in 11 (10.3%) VTE cases and six mutations in 5 (3.9%) controls. Thus, patients with recurrent VTE tended to have higher odds of presenting with CH compared to controls (OR: 2.74, 95% CI: 0.95-9.16). Moreover, the odds of detecting CH were significantly higher in VTE cases in the subgroup of individuals without thrombophilia (OR: 4.58, 95% CI: 1.48-15.99). VTE cases with CH showed elevated platelet counts compared to cases and controls without CH (median [IQR]: 292 [254-298], 223 [198-260] and 220 [185-259] × 109/L; both p < 0.01). Fibrinogen, sP-selectin, D-dimer and hsCRP levels did not differ according to CH status. Overall, we identified a trend for an association between CH and recurrent VTE, particularly in individuals without underlying thrombophilia, warranting further research in this patient group.
Collapse
Affiliation(s)
- Cornelia Englisch
- Division of Hematology and Hemostaseology, Department of Medicine IMedical University of ViennaViennaAustria
| | - Rafaela Vostatek
- Division of Hematology and Hemostaseology, Department of Medicine IMedical University of ViennaViennaAustria
| | - Theresa Schramm
- Division of Hematology and Hemostaseology, Department of Medicine IMedical University of ViennaViennaAustria
| | | | - Ingrid Pabinger
- Division of Hematology and Hemostaseology, Department of Medicine IMedical University of ViennaViennaAustria
| | - Roland Jäger
- Department of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Cihan Ay
- Division of Hematology and Hemostaseology, Department of Medicine IMedical University of ViennaViennaAustria
| |
Collapse
|
32
|
Singh A, Jain AG, Hamilton BK, Adjei A. Care Models for Cancer Survivors. Annu Rev Med 2025; 76:225-241. [PMID: 39661552 DOI: 10.1146/annurev-med-042423-044004] [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: 12/13/2024]
Abstract
The population of cancer survivors is on the rise due to an increase in cancer incidence and a decline in cancer mortality. This growing survivor population creates a number of challenges. Although there have been improvements in care planning for cancer survivors, our healthcare system still lacks the delivery of coordinated care between primary care physicians and specialists. Understanding the needs of cancer survivors can help improve the current care models. In this review we describe existing survivorship care models. We also describe emerging models using some programs at the Cleveland Clinic to highlight various potential approaches.
Collapse
Affiliation(s)
- Abhay Singh
- Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio, USA;
| | - Akriti G Jain
- Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio, USA;
| | | | - Alex Adjei
- Taussig Cancer Center, Cleveland Clinic, Cleveland, Ohio, USA;
| |
Collapse
|
33
|
Cau R, Saba L. Interlinking pathways: a narrative review on the role of IL-6 in cancer and atherosclerosis. Cardiovasc Diagn Ther 2024; 14:1186-1201. [PMID: 39790197 PMCID: PMC11707487 DOI: 10.21037/cdt-24-344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Accepted: 10/18/2024] [Indexed: 01/12/2025]
Abstract
Background and Objective Interleukin-6 (IL-6) plays multifaceted roles in cancer and atherosclerosis. Initially recognized for its role in immune response and inflammation, IL-6 promotes tumor progression via the JAK-STAT and MAP kinase pathways and is associated with poor cancer prognoses. In atherosclerosis, IL-6 contributes to endothelial dysfunction and plaque formation. This review highlights the shared inflammatory mechanisms of IL-6 in both diseases and explores the regulatory dynamics of IL-6 signaling, including gene polymorphisms and epigenetic modifications. Methods Google Scholar, Scopus, and PubMed were searched for English-language articles on IL-6 and those reporting shared pathogenic mechanisms of IL-6 in cancer and atherosclerosis from their inception through June 2024. Key Content and Findings The investigation into IL-6's mechanisms in cancer and atherosclerosis reveals the intricate and interconnected nature of inflammatory processes in chronic diseases. The role of IL-6 in both conditions underscores its centrality in disease pathology, particularly through its involvement in inflammation, immune modulation, and cellular proliferation. This commonality highlights IL-6 as a key player linking these seemingly distinct diseases. Conclusions Given the shared pathogenic mechanism of IL-6 in cancer and atherosclerosis, this narrative review concludes by emphasizing the therapeutic potential of modulating IL-6 in treating both cancer and atherosclerosis. It advocates for personalized treatment strategies that combine targeted therapies with lifestyle modifications. This holistic approach is considered crucial for effective disease management, given the diverse and complex roles IL-6 plays in these widespread conditions.
Collapse
Affiliation(s)
- Riccardo Cau
- Department of Radiology, University of Cagliari, Cagliari, Italy
| | - Luca Saba
- Department of Radiology, University of Cagliari, Cagliari, Italy
| |
Collapse
|
34
|
Fotopoulou F, Rodríguez-Correa E, Dussiau C, Milsom MD. Reconsidering the usual suspects in age-related hematologic disorders: is stem cell dysfunction a root cause of aging? Exp Hematol 2024; 143:104698. [PMID: 39725143 DOI: 10.1016/j.exphem.2024.104698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 12/28/2024]
Abstract
Aging exerts a profound impact on the hematopoietic system, leading to increased susceptibility to infections, autoimmune diseases, chronic inflammation, anemia, thrombotic events, and hematologic malignancies. Within the field of experimental hematology, the functional decline of hematopoietic stem cells (HSCs) is often regarded as a primary driver of age-related hematologic conditions. However, aging is clearly a complex multifaceted process involving not only HSCs but also mature blood cells and their interactions with other tissues. This review reappraises an HSC-centric view of hematopoietic aging by exploring how the entire hematopoietic hierarchy, from stem cells to mature cells, contributes to age-related disorders. It highlights the decline of both innate and adaptive immunity, leading to increased susceptibility to infections and cancer, and the rise of autoimmunity as peripheral immune cells undergo aging-induced changes. It explores the concept of "inflammaging," where persistent, low-grade inflammation driven by old immune cells creates a cycle of tissue damage and disease. Additionally, this review delves into the roles of inflammation and homeostatic regulation in age-related conditions such as thrombotic events and anemia, arguing that these issues arise from broader dysfunctions rather than stemming from HSC functional attrition alone. In summary, this review highlights the importance of taking a holistic approach to studying hematopoietic aging and its related pathologies. By looking beyond just stem cells and considering the full spectrum of age-associated changes, one can better capture the complexity of aging and attempt to develop preventative or rejuvenation strategies that target multiple facets of this process.
Collapse
Affiliation(s)
- Foteini Fotopoulou
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Experimental Hematology Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
| | - Esther Rodríguez-Correa
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Experimental Hematology Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany
| | - Charles Dussiau
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Experimental Hematology Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany; Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Michael D Milsom
- Division of Experimental Hematology, German Cancer Research Center (DKFZ), Heidelberg, Germany; Experimental Hematology Group, Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM), Heidelberg, Germany.
| |
Collapse
|
35
|
Sheets K, Baker JV. HIV and Inflamm-Aging: How Do We Reach the Summit of Healthy Aging? TOPICS IN ANTIVIRAL MEDICINE 2024; 32:589-596. [PMID: 39765238 PMCID: PMC11737810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
People with HIV (PWH) are living longer and experiencing a greater burden of morbidity from non-AIDS-defining conditions. Chronically treated HIV disease is associated with ongoing systemic inflammation that contributes to the development of chronic conditions (eg, cardiovascular disease) and geriatric syndromes (eg, frailty). Apart from HIV disease, a progressive increase in systemic inflammation is a characteristic feature of biologic aging, a process described as "inflammaging." Inflamm-aging is driven by persistent antigen stimulation and stress, leading to an immune profile characterized by elevated levels of blood inflammatory markers and cellular activation and senescence. Chronic HIV disease is hypothesized to accentuate the immune profile of inflamm-aging, in part through viral persistence in lymphatic tissues, permanent injury impairing immune recovery, the presence of copathogens, gut dysbiosis and microbial translocation, and chromosomal and genetic alterations associated with immune activation. Few strategies exist for safe and effective modulation of systemic inflammation among older PWH. The strongest current evidence supports aggressive management of modifiable risk factors such as lipids, blood pressure, and levels of physical activity. Future inflamm-aging research should be directed toward advancing the implementation of proven approaches, such as physical activity, as well as studying novel mechanisms of, and treatments for, inflamm-aging among PWH.
Collapse
Affiliation(s)
- Kerry Sheets
- Hennepin Healthcare, Minneapolis, Minnesota, and University of Minnesota, Minneapolis
| | - Jason V. Baker
- Hennepin Healthcare, Minneapolis, Minnesota, and University of Minnesota, Minneapolis
| |
Collapse
|
36
|
Regan JA, Shah SH. CHIP-ing Away at Post-Transplant Outcomes: the Role of Somatic Mutations in Heart Transplant Outcomes. J Card Fail 2024:S1071-9164(24)00977-1. [PMID: 39710296 DOI: 10.1016/j.cardfail.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/24/2024]
Affiliation(s)
- Jessica A Regan
- Division of Cardiology, Duke University School of Medicine, Durham, North Carolina; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina
| | - Svati H Shah
- Division of Cardiology, Duke University School of Medicine, Durham, North Carolina; Duke Molecular Physiology Institute, Duke University School of Medicine, Durham, North Carolina; Duke Center for Precision Health, Duke University School of Medicine, Durham, North Carolina.
| |
Collapse
|
37
|
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
|
38
|
Stiehl T. Stem cell graft dose and composition could impact on the expansion of donor-derived clones after allogeneic hematopoietic stem cell transplantation - a virtual clinical trial. Front Immunol 2024; 15:1321336. [PMID: 39737169 PMCID: PMC11682905 DOI: 10.3389/fimmu.2024.1321336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 09/10/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Hematopoietic stem cell transplantation is a potentially curative intervention for a broad range of diseases. However, there is evidence that malignant or pre-malignant clones contained in the transplant can expand in the recipient and trigger donor-derived malignancies. This observation has gained much attention in the context of clonal hematopoiesis, a medical condition where significant amounts of healthy blood cells are derived from a small number of hematopoietic stem cell clones. In many cases the dominating clones carry mutations conferring a growth advantage and thus could undergo malignant transformation in the recipient. Since clonal hematopoiesis exists in a significant proportion of potential stem cell donors, a more detailed understanding of its role for stem cell transplantation is required. Methods We propose mechanistic computational models and perform virtual clinical trials to investigate clonal dynamics during and after allogenic hematopoietic stem cell transplantation. Different mechanisms of clonal expansion are considered, including mutation-related changes of stem cell proliferation and self-renewal, aberrant response of mutated cells to systemic signals, and self-sustaining chronic inflammation triggered by the mutated cells. Results Model simulations suggest that an aberrant response of mutated cells to systemic signals is sufficient to explain the frequently observed quick expansion of the mutated clone shortly after transplantation which is followed by a stabilization of the mutated cell number at a constant value. In contrary, a mutation-related increase of self-renewal or self-sustaining chronic inflammation lead to ongoing clonal expansion. Our virtual clinical trials suggest that a low number of transplanted stem cells per kg of body weight increases the transplantation-related expansion of donor-derived clones, whereas the transplanted progenitor dose or growth factor support after transplantation have no impact on clonal dynamics. Furthermore, in our simulations the change of the donors' variant allele frequencies in the year before stem cell donation is associated with the expansion of donor-derived clones in the recipient. Discussion This in silico study provides insights in the mechanisms leading to clonal expansion and identifies questions that could be addressed in future clinical trials.
Collapse
Affiliation(s)
- Thomas Stiehl
- Aachen Medical School, Institute for Computational Biomedicine & Disease Modeling,
RWTH Aachen University, Aachen, Germany
- Department for Science and Environment, Roskilde University,
Roskilde, Denmark
| |
Collapse
|
39
|
Fawaz S, Marti S, Dufossee M, Pucheu Y, Gaufroy A, Broitman J, Bidet A, Soumare A, Munsch G, Tzourio C, Debette S, Trégouët DA, James C, Mansier O, Couffinhal T. Evaluation of clonal hematopoiesis and mosaic loss of Y chromosome in cardiovascular risk: An analysis in prospective studies. eLife 2024; 13:RP96150. [PMID: 39665621 PMCID: PMC11637461 DOI: 10.7554/elife.96150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024] Open
Abstract
Background Clonal hematopoiesis of indeterminate potential (CHIP) was initially linked to a twofold increase in atherothrombotic events. However, recent investigations have revealed a more nuanced picture, suggesting that CHIP may confer only a modest rise in myocardial infarction (MI) risk. This observed lower risk might be influenced by yet unidentified factors that modulate the pathological effects of CHIP. Mosaic loss of the Y chromosome (mLOY), a common marker of clonal hematopoiesis in men, has emerged as a potential candidate for modulating cardiovascular risk associated with CHIP. In this study, we aimed to ascertain the risk linked to each somatic mutation or mLOY and explore whether mLOY could exert an influence on the cardiovascular risk associated with CHIP. Methods We conducted an examination for the presence of CHIP and mLOY using targeted high-throughput sequencing and digital PCR in a cohort of 446 individuals. Among them, 149 patients from the CHAth study had experienced a first MI at the time of inclusion (MI(+) subjects), while 297 individuals from the Three-City cohort had no history of cardiovascular events (CVE) at the time of inclusion (MI(-) subjects). All subjects underwent thorough cardiovascular phenotyping, including a direct assessment of atherosclerotic burden. Our investigation aimed to determine whether mLOY could modulate inflammation, atherosclerosis burden, and atherothrombotic risk associated with CHIP. Results CHIP and mLOY were detected with a substantial prevalence (45.1% and 37.7%, respectively), and their occurrence was similar between MI(+) and MI(-) subjects. Notably, nearly 40% of CHIP(+) male subjects also exhibited mLOY. Interestingly, neither CHIP nor mLOY independently resulted in significant increases in plasma hs-CRP levels, atherosclerotic burden, or MI incidence. Moreover, mLOY did not amplify or diminish inflammation, atherosclerosis, or MI incidence among CHIP(+) male subjects. Conversely, in MI(-) male subjects, CHIP heightened the risk of MI over a 5 y period, particularly in those lacking mLOY. Conclusions Our study highlights the high prevalence of CHIP and mLOY in elderly individuals. Importantly, our results demonstrate that neither CHIP nor mLOY in isolation substantially contributes to inflammation, atherosclerosis, or MI incidence. Furthermore, we find that mLOY does not exert a significant influence on the modulation of inflammation, atherosclerosis burden, or atherothrombotic risk associated with CHIP. However, CHIP may accelerate the occurrence of MI, especially when unaccompanied by mLOY. These findings underscore the complexity of the interplay between CHIP, mLOY, and cardiovascular risk, suggesting that large-scale studies with thousands more patients may be necessary to elucidate subtle correlations. Funding This study was supported by the Fondation Cœur & Recherche (the Société Française de Cardiologie), the Fédération Française de Cardiologie, ERA-CVD (« CHEMICAL » consortium, JTC 2019) and the Fondation Université de Bordeaux. The laboratory of Hematology of the University Hospital of Bordeaux benefitted of a convention with the Nouvelle Aquitaine Region (2018-1R30113-8473520) for the acquisition of the Nextseq 550Dx sequencer used in this study. Clinical trial number NCT04581057.
Collapse
Affiliation(s)
- Sami Fawaz
- CHU de Bordeaux, Service des Maladies Coronaires et VasculairesPessacFrance
| | | | - Melody Dufossee
- Univ. Bordeaux, INSERM, Biologie des maladies cardiovasculairesPessacFrance
| | - Yann Pucheu
- CHU de Bordeaux, Service des Maladies Coronaires et VasculairesPessacFrance
| | - Astrid Gaufroy
- CHU de Bordeaux, Service des Maladies Coronaires et VasculairesPessacFrance
| | - Jean Broitman
- CHU de Bordeaux, Service des Maladies Coronaires et VasculairesPessacFrance
| | - Audrey Bidet
- CHU de Bordeaux, Laboratoire d'hematologiePessacFrance
| | - Aicha Soumare
- Univ. Bordeaux, Bordeaux Population Health Research Center, INSERMBordeauxFrance
| | - Gaëlle Munsch
- Univ. Bordeaux, Bordeaux Population Health Research Center, INSERMBordeauxFrance
| | - Christophe Tzourio
- Univ. Bordeaux, Bordeaux Population Health Research Center, INSERMBordeauxFrance
| | - Stephanie Debette
- Univ. Bordeaux, Bordeaux Population Health Research Center, INSERMBordeauxFrance
| | | | - Chloe James
- CHU de Bordeaux, Laboratoire d'hematologiePessacFrance
- Univ. Bordeaux, INSERM, Biologie des maladies cardiovasculairesPessacFrance
| | - Olivier Mansier
- CHU de Bordeaux, Laboratoire d'hematologiePessacFrance
- Univ. Bordeaux, INSERM, Biologie des maladies cardiovasculairesPessacFrance
| | - Thierry Couffinhal
- CHU de Bordeaux, Service des Maladies Coronaires et VasculairesPessacFrance
- Univ. Bordeaux, INSERM, Biologie des maladies cardiovasculairesPessacFrance
| |
Collapse
|
40
|
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
|
41
|
Guedon AF, Ouafdi A, Belfeki N, Dellal A, Ghriss N, Scheen M, Haidar F, Espitia O, Scoazec JY, Fain O, Marzac C, Hermine O, Solary E, Rossignol J, Mekinian A. Higher risk profile among patients with TET2-mutated giant cell arteritis: a cluster analysis. RMD Open 2024; 10:e004694. [PMID: 39658053 PMCID: PMC11647381 DOI: 10.1136/rmdopen-2024-004694] [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: 06/26/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVE We aimed to assess the prevalence of clonal haematopoiesis (CH) in patients with giant cell arteritis (GCA) compared with controls and individuals with other autoimmune diseases (AIDs) and to identify high-risk clinical/genetic profiles that could influence disease outcomes. METHODS In a prospective observational study at three hospitals, we included 49 patients diagnosed with GCA, 48 patients with other AIDs and 27 control participants. We used next-generation sequencing to detect clonal haematopoiesis (CH) among them. RESULTS CH was detected in 55.1% of patients with GCA, 59.3% of controls and 18.8% of patients with other AIDs. The most commonly mutated genes in GCA and control groups were DNMT3A and TET2. No significant differences in CH prevalence were found between patients with GCA and controls or other AID when adjusted for age and sex. Cluster analysis revealed two distinct groups within the patients with GCA, one of which displayed a higher prevalence of TET2 and JAK2 variants, and was associated with worse prognosis. CONCLUSIONS CH is prevalent among patients with GCA but does not differ significantly from controls or other autoimmune conditions. However, specific genetic profiles, particularly mutations in TET2 and JAK2, are associated with a higher risk cluster within the GCA cohort. This observation highlights the interest of detecting CH in patients with GCA in both routine practice and clinical trials for better risk stratification. Further prospective studies are needed to determine if management tailored to the genetic profile would improve outcomes.
Collapse
Affiliation(s)
- Alexis F Guedon
- Institut Pierre Louis d'Epidémiologie et de Santé Publique, Sorbonne Université, Institut National de la Santé et de la Recherche Médicale, Paris, France
- Sorbonne Université Service de médecine interne, AP-HP, Hôpital Saint Antoine, Paris, France
| | - Asmaa Ouafdi
- Sorbonne Université Service de médecine interne, AP-HP, Hôpital Saint Antoine, Paris, France
| | - Nabil Belfeki
- Hôpital de Melun, Service de Médecine Interne, Melun, France
| | - Azeddine Dellal
- Hôpital Montfermeil, Service de Médecine Interne et Rhumatologie, Montfermeil, France
| | - Nouha Ghriss
- Hôpital de Melun, Service de Médecine Interne, Melun, France
| | | | | | - Olivier Espitia
- Nantes Université, CHU Nantes, Department of Internal and Vascular Medicine, l'institut du thorax, INSERM UMR1087/CNRS UMR 6291, Team III Vascular & Pulmonary diseases, Nantes, France
| | | | - Olivier Fain
- Sorbonne Université Service de médecine interne, AP-HP, Hôpital Saint Antoine, Paris, France
| | | | - Olivier Hermine
- CEREMAST, Paris University, Imagine Institute, INSERM U1163, Necker-Enfants Malades University Hospital, Paris, France
| | | | | | - Arsène Mekinian
- Sorbonne Université Service de médecine interne, AP-HP, Hôpital Saint Antoine, Paris, France
| |
Collapse
|
42
|
Vijenthira A, Volpe VO, Sekar A, Santos Azevedo R, Mikhaleva M, Gibson CJ, Martindale SP, Fardoun R, Tyekucheva S, Ren Y, Fernandes SM, Knisbacher BA, Hahn CK, Getz G, Wu CJ, Davids MS, Brown JR. Myeloid clonal hematopoiesis of indeterminate potential in patients with chronic lymphocytic leukemia. Blood Adv 2024; 8:5949-5956. [PMID: 39321421 PMCID: PMC11629178 DOI: 10.1182/bloodadvances.2024013414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/26/2024] [Accepted: 09/15/2024] [Indexed: 09/27/2024] Open
Abstract
ABSTRACT Clonal hematopoiesis of indeterminate potential (CHIP) in patients with chronic lymphocytic leukemia (CLL) has not been extensively characterized. The objective of this study was to describe the prevalence of myeloid CHIP (M-CHIP) in patients with CLL, and to determine its association with time to first treatment (TTFT) and overall survival (OS). We retrospectively analyzed data from patients participating in a prospective CLL database at the Dana-Farber Cancer Institute who had standard-of-care targeted 95-gene next-generation sequencing (NGS) performed. A schema was devised to classify mutations as M-CHIP related. M-CHIP was analyzed as a binary (present/absent) and categorical (≥2 vs 1 vs 0 mutations) predictor. We included 966 patients (median age at time of NGS, 65 years; 38% female). Overall, 747 (77%) patients had NGS performed before CLL treatment, whereas 219 (23%) had it performed after receiving treatment. Median follow-up time from NGS was 1.9 years. The prevalence of M-CHIP in untreated (12%) and treated (24%) patients with CLL was similar to that described in previous literature. M-CHIP prevalence appeared to increase with age in untreated patients, but appeared consistent across age in treated patients, suggesting that treatment (85% had prior chemotherapy) may have an impact on M-CHIP emergence even in younger patients. The presence of ≥2 M-CHIP mutations was associated with OS, even accounting for prior treatment and age, but was driven by a small subset of patients (n = 28). M-CHIP was not associated with TTFT. These findings support continued work into characterizing the effects of M-CHIP in patients with CLL.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/mortality
- Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis
- Female
- Aged
- Male
- Clonal Hematopoiesis
- Middle Aged
- Mutation
- High-Throughput Nucleotide Sequencing
- Retrospective Studies
- Aged, 80 and over
- Adult
- Prognosis
Collapse
Affiliation(s)
- Abi Vijenthira
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada
- Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Virginia O. Volpe
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Aswin Sekar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | | | - Mariia Mikhaleva
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Christopher J. Gibson
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | | | - Rayan Fardoun
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Svitlana Tyekucheva
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA
| | - Yue Ren
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | | | - Binyamin A. Knisbacher
- The Mina and Everard Goodman Faculty of Life Sciences, Ramat Gan, Israel
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Cynthia K. Hahn
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Gad Getz
- Department of Medicine, Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Pathology, Massachusetts General Hospital, Boston, MA
- Krantz Family Center for Cancer Research and Dept. of Pathology, Massachusetts General Hospital, Boston, MA
| | - Catherine J. Wu
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Matthew S. Davids
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| | - Jennifer R. Brown
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA
- Department of Medicine, Harvard Medical School, Boston, MA
| |
Collapse
|
43
|
Chapple ILC, Hirschfeld J, Cockwell P, Dietrich T, Sharma P. Interplay between periodontitis and chronic kidney disease. Nat Rev Nephrol 2024:10.1038/s41581-024-00910-5. [PMID: 39658571 DOI: 10.1038/s41581-024-00910-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2024] [Indexed: 12/12/2024]
Abstract
Periodontitis is a ubiquitous chronic inflammatory disease affecting the supporting tissues of the teeth and is a major cause of multiple tooth loss. Despite being preventable, periodontitis and dental caries are responsible for more years lost to disability than any other human condition. The most severe form of periodontitis affects 1 billion individuals, and its prevalence is increasing globally. Periodontitis arises from a dysregulated and hyperactive inflammatory response to dysbiosis in the periodontal microbiome. This response has systemic effects associated with premature mortality and elevated risk of several systemic non-communicable diseases (NCDs), including atheromatous cardiovascular disease, type 2 diabetes and chronic kidney disease (CKD). This risk association between periodontitis and NCDs is independent of their shared common risk factors, suggesting that periodontitis is a non-traditional risk factor for NCDs such as CKD. As periodontitis progresses, the immune cells and mediators underpinning its pathophysiology leak into the systemic circulation through the ulcerated oral mucosal lining, inducing in a systemic inflammatory profile that closely mirrors that observed in patients with CKD. The relationship between periodontitis and CKD seems to be bi-directional, but large-scale intervention studies are required to clarify causality and could lead to new care pathways for managing each condition as an exposure for the other.
Collapse
Affiliation(s)
- Iain L C Chapple
- Periodontal Research Group, Institute of Clinical Sciences, University of Birmingham and Birmingham Community Healthcare NHS Foundation Trust, Birmingham, UK.
- NIHR Birmingham Biomedical Research Centre in Inflammation, Birmingham, UK.
| | - Josefine Hirschfeld
- Periodontal Research Group, Institute of Clinical Sciences, University of Birmingham and Birmingham Community Healthcare NHS Foundation Trust, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre in Inflammation, Birmingham, UK
| | - Paul Cockwell
- Department of Nephrology, University Hospital Birmingham, Birmingham, UK
| | - Thomas Dietrich
- Periodontal Research Group, Institute of Clinical Sciences, University of Birmingham and Birmingham Community Healthcare NHS Foundation Trust, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre in Inflammation, Birmingham, UK
| | - Praveen Sharma
- Periodontal Research Group, Institute of Clinical Sciences, University of Birmingham and Birmingham Community Healthcare NHS Foundation Trust, Birmingham, UK
- NIHR Birmingham Biomedical Research Centre in Inflammation, Birmingham, UK
| |
Collapse
|
44
|
Zhang Q, Yim R, Lee P, Chin L, Li V, Gill H. Implications of Clonal Hematopoiesis in Hematological and Non-Hematological Disorders. Cancers (Basel) 2024; 16:4118. [PMID: 39682303 DOI: 10.3390/cancers16234118] [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: 10/21/2024] [Revised: 12/02/2024] [Accepted: 12/06/2024] [Indexed: 12/18/2024] Open
Abstract
Clonal hematopoiesis (CH) is associated with an increased risk of developing myeloid neoplasms (MNs) such as myelodysplastic neoplasm (MDS) and acute myeloid leukemia (AML). In general, CH comprises clonal hematopoiesis of indeterminate potential (CHIP) and clonal cytopenia of undetermined significance (CCUS). It is an age-related phenomenon characterized by the presence of somatic mutations in hematopoietic stem cells (HSCs) and hematopoietic stem and progenitor cells (HSPCs) that acquire a fitness advantage under selection pressure. Individuals with CHIP have an absolute risk of 0.5-1.0% per year for progressing to MDS or AML. Inflammation, smoking, cytotoxic therapy, and radiation can promote the process of clonal expansion and leukemic transformation. Of note, exposure to chemotherapy or radiation for patients with solid tumors or lymphomas can increase the risk of therapy-related MN. Beyond hematological malignancies, CH also serves as an independent risk factor for heart disease, stroke, chronic obstructive pulmonary disease, and chronic kidney disease. Prognostic models such as the CH risk score and MN-prediction models can provide a framework for risk stratification and clinical management of CHIP/CCUS and identify high-risk individuals who may benefit from close surveillance. For CH or related disorders, therapeutic strategies targeting specific CH-associated mutations and specific selection pressure may have a potential role in the future.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Rita Yim
- Department of Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Paul Lee
- Department of Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Lynn Chin
- Department of Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Vivian Li
- Department of Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Harinder Gill
- Department of Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
45
|
Sim H, Park HJ, Park GH, Kim YJ, Park WY, Lee SH, Choi M. Increased inflammatory signature in myeloid cells of non-small cell lung cancer patients with high clonal hematopoiesis burden. eLife 2024; 13:RP96951. [PMID: 39641768 PMCID: PMC11623926 DOI: 10.7554/elife.96951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Clonal hematopoiesis of indeterminate potential (CHIP) allows estimation of clonal dynamics and documentation of somatic mutations in the hematopoietic system. Recent studies utilizing large cohorts of the general population and patients have revealed significant associations of CHIP burden with age and disease status, including in cancer and chronic diseases. An increasing number of cancer patients are treated with immune checkpoint inhibitors (ICIs), but the association of ICI response in non-small cell lung cancer (NSCLC) patients with CHIP burden remains to be determined. We collected blood samples from 100 metastatic NSCLC patients before and after ICI for high-depth sequencing of the CHIP panel and 63 samples for blood single-cell RNA sequencing. Whole exome sequencing was performed in an independent replication cohort of 180 patients. The impact of CHIP status on the immunotherapy response was not significant. However, metastatic lung cancer patients showed higher CHIP prevalence (44/100 for patients vs. 5/42 for controls; p = 0.01). In addition, lung squamous cell carcinoma (LUSC) patients showed increased burden of larger clones compared to lung adenocarcinoma (LUAD) patients (8/43 for LUSC vs. 2/50 for LUAD; p = 0.04). Furthermore, single-cell RNA-seq analysis of the matched patients showed significant enrichment of inflammatory pathways mediated by NF-κB in myeloid clusters of the severe CHIP group. Our findings suggest minimal involvement of CHIP mutation and clonal dynamics during immunotherapy but a possible role of CHIP as an indicator of immunologic response in NSCLC patients.
Collapse
Affiliation(s)
- Hyungtai Sim
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
| | - Hyun Jung Park
- Samsung Genome Institute, Samsung Medical CenterSeoulRepublic of Korea
- Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National UniversitySeoulRepublic of Korea
| | - Geun-Ho Park
- Samsung Genome Institute, Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoulRepublic of Korea
| | - Yeon Jeong Kim
- Samsung Genome Institute, Samsung Medical CenterSeoulRepublic of Korea
| | - Woong-Yang Park
- Samsung Genome Institute, Samsung Medical CenterSeoulRepublic of Korea
| | - Se-Hoon Lee
- Samsung Genome Institute, Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of MedicineSeoulRepublic of Korea
- Department of Health Sciences and Technology, Samsung Advanced Institute for Health Science & Technology (SAIHST), Sungkyunkwan UniversitySeoulRepublic of Korea
| | - Murim Choi
- Department of Biomedical Sciences, Seoul National University College of MedicineSeoulRepublic of Korea
| |
Collapse
|
46
|
Blaser MC, Bäck M, Lüscher TF, Aikawa E. Calcific aortic stenosis: omics-based target discovery and therapy development. Eur Heart J 2024:ehae829. [PMID: 39656785 DOI: 10.1093/eurheartj/ehae829] [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] [Received: 07/19/2024] [Revised: 10/01/2024] [Accepted: 11/14/2024] [Indexed: 12/17/2024] Open
Abstract
Calcific aortic valve disease (CAVD) resulting in aortic stenosis (AS) is the most common form of valvular heart disease, affecting 2% of those over age 65. Those who develop symptomatic severe AS have an average further lifespan of <2 years without valve replacement, and three-quarters of these patients will develop heart failure, undergo valve replacement, or die within 5 years. There are no approved pharmaceutical therapies for AS, due primarily to a limited understanding of the molecular mechanisms that direct CAVD progression in the complex haemodynamic environment. Here, advances in efforts to understand the pathogenesis of CAVD and to identify putative drug targets derived from recent multi-omics studies [including (epi)genomics, transcriptomics, proteomics, and metabolomics] of blood and valvular tissues are reviewed. The recent explosion of single-cell omics-based studies in CAVD and the pathobiological and potential drug discovery insights gained from the application of omics to this disease area are a primary focus. Lastly, the translation of knowledge gained in valvular pathobiology into clinical therapies is addressed, with a particular emphasis on treatment regimens that consider sex-specific, renal, and lipid-mediated contributors to CAVD, and ongoing Phase I/II/III trials aimed at the prevention/treatment of AS are described.
Collapse
Affiliation(s)
- Mark C Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, 17th Floor, Boston, MA 02115, USA
| | - Magnus Bäck
- Department of Medicine, Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Division of Valvular and Coronary Disease, Heart and Vascular Theme, Karolinska University Hospital, Stockholm, Sweden
| | - Thomas F Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, Switzerland
- Heart Division, Royal Brompton and Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women's Hospital, Harvard Medical School, 3 Blackfan Street, 17th Floor, Boston, MA 02115, USA
- Center for Excellence in Vascular Biology, Brigham and Women's Hospital, Harvard Medical School, 77 Avenue Louis Pasteur, NRB 741, Boston, MA 02115, USA
| |
Collapse
|
47
|
Vilahur G, Fuster V. Interplay between platelets and coagulation: from protective haemostasis to pathological arterial thrombosis. Eur Heart J 2024:ehae776. [PMID: 39673717 DOI: 10.1093/eurheartj/ehae776] [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: 06/25/2024] [Revised: 08/28/2024] [Accepted: 10/27/2024] [Indexed: 12/16/2024] Open
Abstract
Haemostasis refers to the physiological process aimed at repairing vessel injury and preventing bleeding. It involves four interlinked stages culminating in the formation of a platelet-fibrin haemostatic plug that is eventually dissolved once the vessel heals. In contrast, arterial thrombosis is a pathological condition resulting from atheroma exposure, triggering the formation of a platelet-rich thrombus that may obstruct blood flow, leading to the clinical manifestations of ischaemic cardiovascular disease. The following review will provide a comprehensive overview of the finely regulated endogenous antithrombotic mechanisms responsible for maintaining the haemostatic balance and preventing intravascular thrombosis. Thereafter, it will further detail the different stages and mechanisms governing the intricate interplay between the vessel, platelets, and the coagulation cascade in haemostasis, highlighting the most recent advances in platelet biology and function, to further elucidate the differential traits and players contributing to pathological arterial thrombus growth. The review will also delve into the impact of emerging cardiovascular risk factors on tilting the haemostatic balance towards a pro-thrombotic state, thereby increasing the patient's vulnerability to thrombotic events. Finally, it will underscore the importance of early screening for subclinical atherosclerosis through advanced imaging technologies capable of quantifying plaque burden and metabolic activity since they may set the stage for an increased thrombotic risk. Implementing proactive interventions to halt atherosclerosis progression or inducing its regression at early stages is crucial for preserving haemostasis and reducing the likelihood of ischaemic atherothrombotic disease.
Collapse
Affiliation(s)
- Gemma Vilahur
- Research Institute, Hospital de la Santa Creu i Sant Pau, IIB-Sant Pau, Carrer Sant Quintí 77-79, Barcelona 08041, Spain
- CiberCV, Institute Carlos III, Madrid 28029, Spain
| | - Valentin Fuster
- Mount Sinai Fuster Heart Hospital, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
48
|
Klauzen P, Basovich L, Shishkova D, Markova V, Malashicheva A. Macrophages in Calcific Aortic Valve Disease: Paracrine and Juxtacrine Disease Drivers. Biomolecules 2024; 14:1547. [PMID: 39766254 PMCID: PMC11673549 DOI: 10.3390/biom14121547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/27/2024] [Accepted: 11/29/2024] [Indexed: 01/11/2025] Open
Abstract
A significant role in the pathogenesis of CAVD is played by innate immunity cells, such as macrophages. In stenotic valves, macrophages have enhanced inflammatory activity, and the population's balance is shifted toward pro-inflammatory ones. Pro-inflammatory macrophages release cytokines, chemokines, and microRNA, which can directly affect the resident valvular cells and cause valve calcification. In CAVD patients, macrophages may have more pronounced pro-inflammatory properties, enhanced not only by paracrine signals but also by juxtacrine Notch signaling and epigenetic factors, which influence the maturation of macrophages' progenitors. In this review, we observe the accumulated data on the involvement of macrophages in CAVD development via paracrine and juxtacrine interactions.
Collapse
Affiliation(s)
- Polina Klauzen
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| | - Liubov Basovich
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| | - Daria Shishkova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo 650002, Russia; (D.S.); (V.M.)
| | - Victoria Markova
- Department of Experimental Medicine, Research Institute for Complex Issues of Cardiovascular Diseases, Kemerovo 650002, Russia; (D.S.); (V.M.)
| | - Anna Malashicheva
- Laboratory of Regenerative Biomedicine, Institute of Cytology, Russian Academy of Sciences, Saint-Petersburg 194064, Russia.; (L.B.)
| |
Collapse
|
49
|
Ullrich F, Bröckelmann PJ, Turki AT, Khan AM, Chiru ED, Vetter M, von Tresckow B, Wirth R, Cordoba R, Ortiz-Maldonado V, Fülöp T, Neuendorff NR. Impact of immunological aging on T cell-mediated therapies in older adults with multiple myeloma and lymphoma. J Immunother Cancer 2024; 12:e009462. [PMID: 39622581 PMCID: PMC11624774 DOI: 10.1136/jitc-2024-009462] [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: 04/26/2024] [Accepted: 10/24/2024] [Indexed: 12/09/2024] Open
Abstract
The treatment landscape for lymphoma and multiple myeloma, which disproportionally affect older adults, has been transformed by the advent of T cell-mediated immunotherapies, including immune checkpoint inhibition, T cell-engaging bispecific antibodies, and chimeric antigen receptor (CAR) T cell therapy, during the last decade. These treatment modalities re-enable the patient's own immune system to combat malignant cells and offer the potential for sustained remissions and cure for various diseases.Age profoundly affects the physiological function of the immune system. The process of biological aging is largely driven by inflammatory signaling, which is reciprocally fueled by aging-related alterations of physiology and metabolism. In the T cell compartment, aging contributes to T cell senescence and exhaustion, increased abundance of terminally differentiated cells, a corresponding attrition in naïve T cell numbers, and a decrease in the breadth of the receptor repertoire. Furthermore, inflammatory signaling drives aging-related pathologies and contributes to frailty in older individuals. Thus, there is growing evidence of biological aging modulating the efficacy and toxicity of T cell-mediated immunotherapies.Here, we review the available evidence from biological and clinical studies focusing on the relationship between T cell-mediated treatment of hematologic malignancies and age. We discuss biological features potentially impacting clinical outcomes in various scenarios, and potential strategies to improve the safety and efficacy of immune checkpoint inhibitors, T cell-engaging bispecific antibodies, and CAR-T cell therapy in older patients.
Collapse
Affiliation(s)
- Fabian Ullrich
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK partner site Essen), University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Paul J Bröckelmann
- Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD) and German Hodgkin Study Group (GHSG), University of Cologne, Cologne, Germany
- Max Planck Institute for Biology of Ageing, Cologne, Nordrhein-Westfalen, Germany
| | - Amin T Turki
- Department of Hematology and Oncology, University Hospital Marien Hospital Herne, Herne, Nordrhein-Westfalen, Germany
- Institute for Artificial Intelligence in Medicine, University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Abdullah M Khan
- Division of Hematology, The Ohio State University Comprehensive Cancer Center Arthur G James Cancer Hospital and Richard J Solove Research Institute, Columbus, Ohio, USA
| | - Elena-Diana Chiru
- Cancer Center Baselland, University of Basel Faculty of Medicine, Basel, Liestal, Switzerland
| | - Marcus Vetter
- Cancer Center Baselland, University of Basel Faculty of Medicine, Basel, Liestal, Switzerland
| | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK partner site Essen), University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Rainer Wirth
- Department of Geriatrics, Ruhr University Bochum, University Hospital Marien Hospital Herne, Herne, Germany
| | - Raul Cordoba
- Department of Hematology, Lymphoma Unit, Hospital Universitario Fundacion Jimenez Diaz, Madrid, Spain
| | - Valentín Ortiz-Maldonado
- Department of Hematology, Oncoimmunotherapy Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Tamas Fülöp
- Department of Medicine, Division of Geriatrics, Research Center on Aging, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Nina Rosa Neuendorff
- Department of Geriatrics, Ruhr University Bochum, University Hospital Marien Hospital Herne, Herne, Germany
| |
Collapse
|
50
|
Sato N, Goyama S, Chang YH, Miyawaki M, Fujino T, Koide S, Denda T, Liu X, Ueda K, Yamamoto K, Asada S, Takeda R, Yonezawa T, Tanaka Y, Honda H, Ota Y, Shibata T, Sekiya M, Isobe T, Lamagna C, Masuda E, Iwama A, Shimano H, Inoue JI, Miyake K, Kitamura T. Clonal hematopoiesis-related mutant ASXL1 promotes atherosclerosis in mice via dysregulated innate immunity. NATURE CARDIOVASCULAR RESEARCH 2024; 3:1568-1583. [PMID: 39653824 DOI: 10.1038/s44161-024-00579-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 10/30/2024] [Indexed: 12/13/2024]
Abstract
Certain somatic mutations provide a fitness advantage to hematopoietic stem cells and lead to clonal expansion of mutant blood cells, known as clonal hematopoiesis (CH). Among the most common CH mutations, ASXL1 mutations pose the highest risk for cardiovascular diseases (CVDs), yet the mechanisms by which they contribute to CVDs are unclear. Here we show that hematopoietic cells harboring C-terminally truncated ASXL1 mutant (ASXL1-MT) accelerate the development of atherosclerosis in Ldlr-/- mice. Transcriptome analyses of plaque cells showed that monocytes and macrophages expressing ASXL1-MT exhibit inflammatory signatures. Mechanistically, we demonstrate that wild-type ASXL1 has an unexpected non-epigenetic role by suppressing innate immune signaling through the inhibition of IRAK1-TAK1 interaction in the cytoplasm. This regulatory function is lost in ASXL1-MT, resulting in NF-κB activation. Inhibition of IRAK1/4 alleviated atherosclerosis driven by ASXL1-MT and decreased inflammatory monocytes. The present work provides a mechanistic and cellular explanation linking ASXL1 mutations, CH and CVDs.
Collapse
Affiliation(s)
- Naru Sato
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-Ku, Japan
| | - Susumu Goyama
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Yu-Hsuan Chang
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
- Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-Ku, Japan
| | - Masashi Miyawaki
- Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-Ku, Japan
| | - Takeshi Fujino
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Shuhei Koide
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Tamami Denda
- Department of Pathology, The Institute of Medical Science Research Hospital, University of Tokyo, Tokyo, Japan
| | - Xiaoxiao Liu
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Koji Ueda
- Project for Personalized Cancer Medicine, Cancer Precision Medicine Centre, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Keita Yamamoto
- Division of Molecular Oncology, Department of Computational Biology and Medical Sciences, Graduate School of Frontier Sciences, University of Tokyo, Tokyo, Japan
| | - Shuhei Asada
- The Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - Reina Takeda
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Taishi Yonezawa
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Yosuke Tanaka
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hiroaki Honda
- The Institute of Laboratory Animals, Tokyo Women's Medical University, Tokyo, Japan
| | - Yasunori Ota
- Department of Pathology, The Institute of Medical Science Research Hospital, University of Tokyo, Tokyo, Japan
| | - Takuma Shibata
- Department of Microbiology and Immunology, Division of Infectious Genetics, University of Tokyo, Tokyo, Japan
| | - Motohiro Sekiya
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Tomoya Isobe
- Department of Hematology, Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | | | - Esteban Masuda
- Rigel Pharmaceuticals, Inc., South San Francisco, CA, USA
| | - Atsushi Iwama
- Division of Stem Cell and Molecular Medicine, Centre for Stem Cell Biology and Regenerative Medicine, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Hitoshi Shimano
- Department of Internal Medicine (Endocrinology and Metabolism), Faculty of Medicine, University of Tsukuba, Ibaraki, Japan
| | - Jun-Ichiro Inoue
- Research Platform Office, The Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | - Kensuke Miyake
- Department of Microbiology and Immunology, Division of Infectious Genetics, University of Tokyo, Tokyo, Japan
| | - Toshio Kitamura
- Division of Cellular Therapy, The Institute of Medical Science, University of Tokyo, Tokyo, Japan.
- Molecular Pharmacology of Malignant Diseases, Graduate School of Pharmaceutical Sciences, University of Tokyo, Bunkyo-Ku, Japan.
- Institute of Biomedical Research and Innovation, Foundation for Biomedical Research and Innovation at Kobe, Kobe, Japan.
| |
Collapse
|