Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 279
Filtrar
1.
Cancer Discov ; 14(10): 1768-1770, 2024 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-39363743

RESUMEN

In this issue, Waarts and colleagues developed an advanced ex vivo CRISPR screening platform to identify vulnerabilities in clonal hematopoiesis (CH). This unique system allowed the authors to identify a link between IDH2 and TET2 CH mutations, histone demethylases, and altered cytokine signaling, which enabled targeting by ruxolitinib leading to the elimination of CH clones, offering a possible path for preventing the development of malignancy. See related article by Waarts et al., p. 1860.


Asunto(s)
Hematopoyesis Clonal , Proteínas de Unión al ADN , Dioxigenasas , Isocitrato Deshidrogenasa , Nitrilos , Pirazoles , Pirimidinas , Humanos , Isocitrato Deshidrogenasa/genética , Pirimidinas/farmacología , Pirimidinas/uso terapéutico , Hematopoyesis Clonal/genética , Pirazoles/farmacología , Pirazoles/uso terapéutico , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Mutación
2.
J Clin Invest ; 134(19)2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39352380

RESUMEN

Therapy-related clonal hematopoiesis (t-CH) is defined as clonal hematopoiesis detected in individuals previously treated with chemotherapy and/or radiation therapy. With the increased use of genetic analysis in oncological care, the detection of t-CH among cancer patients is becoming increasingly common. t-CH arises through the selective bottleneck imposed by chemotherapies and potentially through direct mutagenesis from chemotherapies, resulting in a distinct mutational landscape enriched with mutations in DNA damage-response pathway genes such as TP53, PPM1D, and CHEK2. Emerging evidence sheds light on the mechanisms of t-CH development and potential strategies to mitigate its emergence. Due to its unique characteristics that predominantly affect cancer patients, t-CH has clinical implications distinct from those of CH in the general population. This Review discusses the potential mechanisms of t-CH development, its mutational landscape, mutant-drug relationships, and its clinical significance. We highlight the distinct nature of t-CH and call for intensified research in this field.


Asunto(s)
Hematopoyesis Clonal , Mutación , Neoplasias , Humanos , Hematopoyesis Clonal/genética , Neoplasias/genética , Neoplasias/terapia , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Quinasa de Punto de Control 2/genética , Quinasa de Punto de Control 2/metabolismo , Proteína Fosfatasa 2C/genética , Proteína Fosfatasa 2C/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
3.
J Clin Invest ; 134(19)2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39352379

RESUMEN

Clonal hematopoiesis of indeterminate potential (CHIP) has emerged as a previously unrecognized, potent, age-related, and common risk factor for atherosclerosis. Somatic mutations in certain known leukemia driver genes give rise to clones of mutant cells in peripheral blood. The increased risk of developing hematologic malignancy does not, on its own, explain excess mortality in individuals with CHIP. Cardiovascular disease accounts for much of this gap. Experimental evidence supports the causality of certain CHIP mutations in accelerated atherosclerosis. CHIP due to mutations in different driver genes varies in their promotion of atherosclerotic events and in the region of augmented atherosclerotic involvement. For example, CHIP due to mutations in DNMT3a appears less atherogenic than CHIP that arises from TET2 or JAK2, forms of CHIP that incite inflammation. The recognition of certain CHIP mutations as promoters of atherosclerotic risk has opened new insights into understanding of the pathophysiology of this disease. The accentuated cardiovascular risk and involvement of distinct pathways of various forms of CHIP also inform novel approaches to allocation of targeted therapies, affording a step toward personalized medicine.


Asunto(s)
Aterosclerosis , Hematopoyesis Clonal , ADN Metiltransferasa 3A , Dioxigenasas , Mutación , Humanos , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/patología , Hematopoyesis Clonal/genética , ADN Metiltransferasa 3A/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Animales , Janus Quinasa 2/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Factores de Riesgo
5.
J Clin Invest ; 134(19)2024 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-39352393

RESUMEN

Clonal hematopoiesis (CH), the expansion of hematopoietic stem cells and their progeny driven by somatic mutations in leukemia-associated genes, is a common phenomenon that rises in prevalence with advancing age to affect most people older than 70 years. CH remains subclinical in most carriers, but, in a minority, it progresses to a myeloid neoplasm, such as acute myeloid leukemia, myelodysplastic syndrome, or myeloproliferative neoplasm. Over the last decade, advances in our understanding of CH, its molecular landscape, and the risks associated with different driver gene mutations have culminated in recent developments that allow for a more precise estimation of myeloid neoplasia risk in CH carriers. In turn, this is leading to the development of translational and clinical programs to intercept and prevent CH from developing into myeloid neoplasia. Here, we give an overview of the spectrum of CH driver mutations, what is known about their pathophysiology, and how this informs the risk of incident myeloid malignancy.


Asunto(s)
Hematopoyesis Clonal , Neoplasias Hematológicas , Mutación , Humanos , Hematopoyesis Clonal/genética , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Trastornos Mieloproliferativos/metabolismo , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología
6.
Science ; 386(6718): eadl4492, 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39388542

RESUMEN

Epigenetic reinforcement of T cell exhaustion is known to be a major barrier limiting T cell responses during immunotherapy. However, the core epigenetic regulators restricting antitumor immunity during prolonged antigen exposure are not clear. We investigated three commonly mutated epigenetic regulators that promote clonal hematopoiesis to determine whether they affect T cell stemness and response to checkpoint blockade immunotherapy. CD8 T cells lacking Dnmt3a, Tet2, or Asxl1 preserved a progenitor-exhausted (Tpex) population for more than 1 year during chronic antigen exposure without undergoing malignant transformation. Asxl1 controlled the self-renewal capacity of T cells and reduced CD8 T cell differentiation through H2AK119 ubiquitination and epigenetic modification of the polycomb group-repressive deubiquitinase pathway. Asxl1-deficient T cells synergized with anti-PD-L1 immunotherapy to improve tumor control in experimental models and conferred a survival advantage to mutated T cells from treated patients.


Asunto(s)
Linfocitos T CD8-positivos , Hematopoyesis Clonal , ADN Metiltransferasa 3A , Proteínas de Unión al ADN , Dioxigenasas , Epigénesis Genética , Inhibidores de Puntos de Control Inmunológico , Terapia de Inmunosupresión , Proteínas Proto-Oncogénicas , Proteínas Represoras , Agotamiento de Células T , Animales , Humanos , Ratones , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Hematopoyesis Clonal/genética , Dioxigenasas/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , ADN Metiltransferasa 3A/genética , Proteínas de Unión al ADN/genética , Histonas/metabolismo , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Agotamiento de Células T/genética
7.
Int J Mol Sci ; 25(19)2024 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-39408588

RESUMEN

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potential cure for myelodysplastic neoplasms (MDSs) and other hematologic malignancies. This study investigates post-transplantation genetic evolution and telomere dynamics in hematopoietic cells, with a focus on clonal hematopoiesis (CH). We conducted a longitudinal analysis of 21 MDS patients who underwent allo-HSCT between September 2009 and February 2015. Genetic profiles of hematopoietic cells from both recipients and donors were compared at equivalent pre- and post-transplantation time points. Targeted sequencing identified CH-associated mutations, and real-time quantitative PCR measured telomere length. Furthermore, we compared CH incidence between recipients and age-matched controls from the GENIE cohort from routine health checkups. Post-allo-HSCT, 38% of recipients developed somatic mutations not detected before transplantation, indicating de novo CH originating from donor cells. Compared to age-matched healthy controls, recipients showed a significantly higher incidence of CH, suggesting increased susceptibility to genetic changes post-transplant. Telomere length analysis also revealed accelerated shortening in transplanted cells, highlighting the heightened stress and proliferation demands in the new microenvironment. Our findings reveal a notable incidence of donor-derived CH in allo-HSCT recipients, alongside significant telomere attrition. This suggests the potential influence of the marrow microenvironment on genetic and molecular changes in hematopoietic cells.


Asunto(s)
Hematopoyesis Clonal , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas , Telómero , Humanos , Trasplante de Células Madre Hematopoyéticas/métodos , Masculino , Persona de Mediana Edad , Femenino , Hematopoyesis Clonal/genética , Adulto , Células Madre Hematopoyéticas/metabolismo , Telómero/genética , Anciano , Trasplante Homólogo , Mutación , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/terapia , Síndromes Mielodisplásicos/etiología , Médula Ósea/metabolismo
8.
Circ Res ; 135(9): 933-950, 2024 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-39234670

RESUMEN

BACKGROUND: Hypertension incidence increases with age and represents one of the most prevalent risk factors for cardiovascular disease. Clonal events in the hematopoietic system resulting from somatic mutations in driver genes are prevalent in elderly individuals who lack overt hematologic disorders. This condition is referred to as age-related clonal hematopoiesis (CH), and it is a newly recognized risk factor for cardiovascular disease. It is not known whether CH and hypertension in the elderly are causally related and, if so, what are the mechanistic features. METHODS: A murine model of adoptive bone marrow transplantation was employed to examine the interplay between Tet2 (ten-eleven translocation methylcytosine dioxygenase 2) clonal hematopoiesis and hypertension. RESULTS: In this model, a subpressor dose of Ang II (angiotensin II) resulted in elevated systolic and diastolic blood pressure as early as 1 day after challenge. These conditions led to the expansion of Tet2-deficient proinflammatory monocytes and bone marrow progenitor populations. Tet2 deficiency promoted renal CCL5 (C-C motif ligand 5) chemokine expression and macrophage infiltration into the kidney. Consistent with macrophage involvement, Tet2 deficiency in myeloid cells promoted hypertension when mice were treated with a subpressor dose of Ang II. The hematopoietic Tet2-/- condition led to sodium retention, renal inflammasome activation, and elevated levels of IL (interleukin)-1ß and IL-18. Analysis of the sodium transporters indicated NCC (sodium-chloride symporter) and NKCC2 (Na+-K+-Cl- cotransporter 2) activation at residues Thr53 and Ser105, respectively. Administration of the NLRP3 (NLR family pyrin domain containing 3) inflammasome inhibitor MCC950 reversed the hypertensive state, sodium retention, and renal transporter activation. CONCLUSIONS: Tet2-mediated CH sensitizes mice to a hypertensive stimulus. Mechanistically, the expansion of hematopoietic Tet2-deficient cells promotes hypertension due to elevated renal immune cell infiltration and activation of the NLRP3 inflammasome, with consequences on sodium retention. These data indicate that carriers of TET2 CH could be at elevated risk for the development of hypertension and that immune modulators could be useful in treating hypertension in this patient population.


Asunto(s)
Angiotensina II , Hematopoyesis Clonal , Proteínas de Unión al ADN , Dioxigenasas , Hipertensión , Inflamasomas , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas , Animales , Dioxigenasas/metabolismo , Dioxigenasas/genética , Inflamasomas/metabolismo , Inflamasomas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/genética , Hipertensión/metabolismo , Hipertensión/genética , Hipertensión/fisiopatología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Ratones , Hematopoyesis Clonal/genética , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Masculino , Trasplante de Médula Ósea
10.
Nat Commun ; 15(1): 7858, 2024 Sep 09.
Artículo en Inglés | MEDLINE | ID: mdl-39251642

RESUMEN

Clonal hematopoiesis of indeterminate potential (CHIP) is linked to diverse aging-related diseases, including hematologic malignancy and atherosclerotic cardiovascular disease (ASCVD). While CHIP is common among older adults, the underlying factors driving its development are largely unknown. To address this, we performed whole-exome sequencing on 8,374 blood DNA samples collected from 4,187 Atherosclerosis Risk in Communities Study (ARIC) participants over a median follow-up of 21 years. During this period, 735 participants developed incident CHIP. Splicing factor genes (SF3B1, SRSF2, U2AF1, and ZRSR2) and TET2 CHIP grow significantly faster than DNMT3A non-R882 clones. We find that age at baseline and sex significantly influence the incidence of CHIP, while ASCVD and other traditional ASCVD risk factors do not exhibit such associations. Additionally, baseline synonymous passenger mutations are strongly associated with CHIP status and are predictive of new CHIP clone acquisition and clonal growth over extended follow-up, providing valuable insights into clonal dynamics of aging hematopoietic stem and progenitor cells. This study also reveals associations between germline genetic variants and incident CHIP. Our comprehensive longitudinal assessment yields insights into cell-intrinsic and -extrinsic factors contributing to the development and progression of CHIP clones in older adults.


Asunto(s)
Hematopoyesis Clonal , Dioxigenasas , Humanos , Hematopoyesis Clonal/genética , Masculino , Femenino , Anciano , Estudios Longitudinales , Persona de Mediana Edad , Dioxigenasas/genética , ADN Metiltransferasa 3A , Factores de Empalme de ARN/genética , Factores de Empalme de ARN/metabolismo , Aterosclerosis/genética , Factores de Riesgo , Secuenciación del Exoma , Proteínas de Unión al ADN/genética , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/metabolismo , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/citología , Envejecimiento/genética , Incidencia , Mutación
11.
Cancer Discov ; 14(9): 1581-1583, 2024 Sep 04.
Artículo en Inglés | MEDLINE | ID: mdl-39228297

RESUMEN

There is no general consensus on the set of mutations capable of driving the age-related clonal expansions in hematopoietic stem cells known as clonal hematopoiesis, and current variant classifications typically rely on rules derived from expert knowledge. In this issue of Cancer Discovery, Damajo and colleagues trained and validated machine learning models without prior knowledge of clonal hematopoiesis driver mutations to classify somatic mutations in blood for 12 genes in a purely data-driven way. See related article by Demajo et al., p. 1717 (9).


Asunto(s)
Hematopoyesis Clonal , Aprendizaje Automático , Mutación , Humanos , Hematopoyesis Clonal/genética
14.
J Clin Invest ; 134(15)2024 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-39087468

RESUMEN

Clonal hematopoiesis of indeterminate potential (CHIP) is characterized by the selective expansion of hematopoietic stem and progenitor cells (HSPCs) carrying somatic mutations. While CHIP is typically asymptomatic, it has garnered substantial attention due to its association with the pathogenesis of multiple disease conditions, including cardiovascular disease (CVD) and hematological malignancies. In this Review, we will discuss seminal and recent studies that have advanced our understanding of mechanisms that drive selection for mutant HSPCs in the BM niche. Next, we will address recent studies evaluating potential relationships between the clonal dynamics of CHIP and hematopoietic development across the lifespan. Next, we will examine the roles of systemic factors that can influence hematopoietic stem cell (HSC) fitness, including inflammation, and exposures to cytotoxic agents in driving selection for CHIP clones. Furthermore, we will consider how - through their impact on the BM niche - lifestyle factors, including diet, exercise, and psychosocial stressors, might contribute to the process of somatic evolution in the BM that culminates in CHIP. Finally, we will review the role of old age as a major driver of selection in CHIP.


Asunto(s)
Hematopoyesis Clonal , Células Madre Hematopoyéticas , Nicho de Células Madre , Humanos , Células Madre Hematopoyéticas/metabolismo , Hematopoyesis Clonal/genética , Animales , Médula Ósea/metabolismo , Mutación
15.
Atherosclerosis ; 396: 118541, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-39111028

RESUMEN

The cardiovascular complications of atherosclerosis are thought to arise from an inflammatory response to the accumulation of cholesterol-rich lipoproteins in the arterial wall. The positive outcome of CANTOS (Canakinumab Anti-inflammatory Thrombosis Outcome Study) provided key evidence to support this concept and suggested that inflammasomes and IL-1ß are important inflammatory mediators in human atherosclerotic cardiovascular diseases (ACVD). In specific settings NLRP3 or AIM2 inflammasomes can induce inflammatory responses in the arterial wall and promote the formation of unstable atherosclerotic plaques. Clonal hematopoiesis (CH) has recently emerged as a major independent risk factor for ACVD. CH mutations arise during ageing and commonly involves variants in genes mediating epigenetic modifications (TET2, DNMT3A, ASXL1) or cytokine signaling (JAK2). Accumulating evidence points to the role of inflammasomes in the progression of CH-induced ACVD events and has shed light on the regulatory pathways and possible therapeutic approaches that specifically target inflammasomes in atherosclerosis. Epigenetic dynamics play a vital role in regulating the generation and activation of inflammasome components by causing changes in DNA methylation patterns and chromatin assembly. This review examines the genetic and epigenetic regulation of inflammasomes, the intersection of macrophage cholesterol accumulation with inflammasome activation and their roles in atherosclerosis. Understanding the involvement of inflammasomes in atherosclerosis pathogenesis may lead to customized treatments that reduce the burden of ACVD.


Asunto(s)
Aterosclerosis , Epigénesis Genética , Inflamasomas , Humanos , Inflamasomas/metabolismo , Aterosclerosis/genética , Aterosclerosis/metabolismo , Aterosclerosis/inmunología , Animales , Hematopoyesis Clonal/genética , Metilación de ADN , Macrófagos/metabolismo , Macrófagos/inmunología , Colesterol/metabolismo , Transducción de Señal , Predisposición Genética a la Enfermedad
16.
Annu Rev Genomics Hum Genet ; 25(1): 329-351, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-39190914

RESUMEN

Clonal hematopoiesis (CH) is an age-related process whereby hematopoietic stem and progenitor cells (HSPCs) acquire mutations that lead to a proliferative advantage and clonal expansion. The most commonly mutated genes are epigenetic regulators, DNA damage response genes, and splicing factors, which are essential to maintain functional HSPCs and are frequently involved in the development of hematologic malignancies. Established risk factors for CH, including age, prior cytotoxic therapy, and smoking, increase the risk of acquiring CH and/or may increase CH fitness. CH has emerged as a novel risk factor in many age-related diseases, such as hematologic malignancies, cardiovascular disease, diabetes, and autoimmune disorders, among others. Future characterization of the mechanisms driving CH evolution will be critical to develop preventative and therapeutic approaches.


Asunto(s)
Hematopoyesis Clonal , Neoplasias Hematológicas , Humanos , Hematopoyesis Clonal/genética , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Neoplasias Hematológicas/terapia , Neoplasias Hematológicas/metabolismo , Mutación , Células Madre Hematopoyéticas/metabolismo , Epigénesis Genética , Factores de Riesgo , Animales
17.
Nat Genet ; 56(9): 1832-1840, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39192095

RESUMEN

Telomeres protect chromosome ends from damage and their length is linked with human disease and aging. We developed a joint telomere length metric, combining quantitative PCR and whole-genome sequencing measurements from 462,666 UK Biobank participants. This metric increased SNP heritability, suggesting that it better captures genetic regulation of telomere length. Exome-wide rare-variant and gene-level collapsing association studies identified 64 variants and 30 genes significantly associated with telomere length, including allelic series in ACD and RTEL1. Notably, 16% of these genes are known drivers of clonal hematopoiesis-an age-related somatic mosaicism associated with myeloid cancers and several nonmalignant diseases. Somatic variant analyses revealed gene-specific associations with telomere length, including lengthened telomeres in individuals with large SRSF2-mutant clones, compared with shortened telomeres in individuals with clonal expansions driven by other genes. Collectively, our findings demonstrate the impact of rare variants on telomere length, with larger effects observed among genes also associated with clonal hematopoiesis.


Asunto(s)
Bancos de Muestras Biológicas , Polimorfismo de Nucleótido Simple , Telómero , Secuenciación Completa del Genoma , Humanos , Telómero/genética , Reino Unido , Secuenciación Completa del Genoma/métodos , Homeostasis del Telómero/genética , Masculino , Femenino , Hematopoyesis Clonal/genética , Estudio de Asociación del Genoma Completo/métodos , Anciano , ADN Helicasas/genética , Persona de Mediana Edad , Biobanco del Reino Unido
18.
Nat Med ; 30(9): 2641-2647, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39107561

RESUMEN

Clonal hematopoiesis of indeterminate potential (CHIP) has been associated with an increased risk of cardiovascular (CV) disease in the general population. Currently, it is unclear whether this association is observed in large clinical trial cohorts with a high burden of existing CV disease or whether CV therapies can mitigate CHIP-associated CV risk. To address these questions, we studied 63,700 patients from five randomized trials that tested established therapies for CV disease, including treatments targeting the proteins PCSK9, SGLT2, P2Y12 and FXa. During a median follow-up of 2.5 years, 7,453 patients had at least one CV event (CV death, myocardial infarction (MI), ischemic stroke or coronary revascularization). The adjusted hazard ratio (aHR) for CV events for CHIP+ patients was 1.07 (95% CI: 0.99-1.16, P = 0.08), with consistent risk estimates across each component of CV risk. Significant heterogeneity in the risk of MI was observed, such that CHIP+ patients had a 30% increased risk of first MI (aHR = 1.31 (1.05-1.64), P = 0.02) but no increased risk of recurrent MI (aHR = 0.94 (0.79-1.13), Pint = 0.008), as compared to CHIP- patients. Moreover, no significant heterogeneity in treatment effect between individuals with and without CHIP was observed for any of the therapies studied in the five trials. These results indicate that in clinical trial populations, CHIP is associated with incident but not recurrent coronary events and that the presence of CHIP does not appear to identify patients who will derive greater benefit from commonly used CV therapies.


Asunto(s)
Enfermedades Cardiovasculares , Hematopoyesis Clonal , Humanos , Hematopoyesis Clonal/genética , Enfermedades Cardiovasculares/epidemiología , Enfermedades Cardiovasculares/genética , Masculino , Femenino , Persona de Mediana Edad , Anciano , Infarto del Miocardio/epidemiología , Ensayos Clínicos Controlados Aleatorios como Asunto , Resultado del Tratamiento , Factores de Riesgo
20.
J Am Heart Assoc ; 13(17): e034255, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39206728

RESUMEN

BACKGROUND: Cardiac surgery triggers sterile innate immune responses leading to postoperative complications. Clonal hematopoiesis (CH) is associated with short-term inflammation-mediated outcomes after cardiac surgery. The impact of CH on long-term postoperative outcomes remains unknown. METHODS AND RESULTS: In this cohort study, patients undergoing elective cardiac surgery were included from January 2017 to September 2019. Patients were screened for CH using a predefined gene panel of 19 genes. Recorded clinical events were all-cause death, major adverse cardiac and cerebral events including cardiovascular death, myocardial infarction or nonscheduled coronary revascularization, stroke, and hospitalization for acute heart failure. The primary study outcome was time to a composite criterion including all-cause mortality and major adverse cardiac and cerebral events. Among 314 genotyped patients (median age: 67 years; interquartile range 59-74 years), 139 (44%) presented with CH, based on a variant allelic frequency ≥1%. Carriers of CH had a higher proportion of patients with a history of atrial fibrillation (26% for CH versus 17% for non-CH carriers, P=0.022). The most frequently mutated genes were DNMT3A, TET2, and ASXL1. After a median follow-up of 1203 [813-1435] days, the primary outcome occurred in 50 patients. After multivariable adjustment, CH was independently associated with a higher risk for the primary outcome (hazard ratio, 1.88 [95% CI, 1.05-3.41], P=0.035). Most adverse events occurred in patients carrying TET2 variants. CONCLUSIONS: In patients undergoing cardiac surgery, CH is frequent and associated with a 2-fold increased long-term risk for major adverse clinical outcomes. CH is a novel risk factor for long-term postcardiac surgery complications and might be useful to personalize management decisions. REGISTRATION: URL: https://www.clinicaltrials.gov; Unique identifier: NCT03376165.


Asunto(s)
Procedimientos Quirúrgicos Cardíacos , Hematopoyesis Clonal , Complicaciones Posoperatorias , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Procedimientos Quirúrgicos Cardíacos/efectos adversos , Hematopoyesis Clonal/genética , Dioxigenasas/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/genética , Mutación , Complicaciones Posoperatorias/genética , Complicaciones Posoperatorias/epidemiología , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Medición de Riesgo/métodos , Factores de Riesgo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA