Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Nat Cardiovasc Res ; 2: 144-158, 2023 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-36949957

RESUMEN

Somatic mutations in blood indicative of clonal hematopoiesis of indeterminate potential (CHIP) are associated with an increased risk of hematologic malignancy, coronary artery disease, and all-cause mortality. Here we analyze the relation between CHIP status and incident peripheral artery disease (PAD) and atherosclerosis, using whole-exome sequencing and clinical data from the UK Biobank and Mass General Brigham Biobank. CHIP associated with incident PAD and atherosclerotic disease across multiple beds, with increased risk among individuals with CHIP driven by mutation in DNA Damage Repair (DDR) genes such as TP53 and PPM1D. To model the effects of DDR-induced CHIP on atherosclerosis, we used a competitive bone marrow transplantation strategy, and generated atherosclerosis-prone Ldlr-/- chimeric mice carrying 20% p53-deficient hematopoietic cells. The chimeric mice were analyzed 13-weeks post-grafting and showed increased aortic plaque size and accumulation of macrophages within the plaque, driven by increased proliferation of p53-deficient plaque macrophages. In summary, our findings highlight the role of CHIP as a broad driver of atherosclerosis across the entire arterial system beyond the coronary arteries, and provide genetic and experimental support for a direct causal contribution of TP53-mutant CHIP to atherosclerosis.

2.
Circ J ; 87(3): 394-400, 2023 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-34433749

RESUMEN

Accumulating evidence suggests that conventional cardiovascular risk factors are incompletely predictive of cardiovascular disease, as a substantial risk remains even when these factors are apparently managed well. In this context, clonal hematopoiesis has emerged as a new and potent risk factor for atherosclerotic cardiovascular disease and other cardiometabolic conditions. Clonal hematopoiesis typically arises from somatic mutations that confer a competitive advantage to a mutant hematopoietic stem cell, leading to its clonal expansion in the stem cell population and its progeny of blood leukocytes. Human sequencing studies and experiments in mice suggest that clonal hematopoiesis, at least when driven by certain mutations, contributes to accelerated atherosclerosis development. However, the epidemiology, biology and clinical implications of this phenomenon remain incompletely understood. Here, we review the current understanding of the connection between clonal hematopoiesis and atherosclerosis, and highlight knowledge gaps in this area of research.


Asunto(s)
Aterosclerosis , Enfermedades Cardiovasculares , Humanos , Animales , Ratones , Factores de Riesgo , Hematopoyesis Clonal , Enfermedades Cardiovasculares/etiología , Hematopoyesis/genética , Aterosclerosis/genética , Factores de Riesgo de Enfermedad Cardiaca , Mutación
3.
Geroscience ; 45(2): 1231-1236, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-35752705

RESUMEN

Clonal hematopoiesis of indeterminate potential (CHIP), defined as the presence of somatic mutations in cancer-related genes in blood cells in the absence of hematological cancer, has recently emerged as an important risk factor for several age-related conditions, especially cardiovascular disease. CHIP is strongly associated with normal aging, but its role in premature aging syndromes is unknown. Hutchinson-Gilford progeria syndrome (HGPS) is an ultra-rare genetic condition driven by the accumulation of a truncated form of the lamin A protein called progerin. HGPS patients exhibit several features of accelerated aging and typically die from cardiovascular complications in their early teens. Previous studies have shown normal hematological parameters in HGPS patients, except for elevated platelets, and low levels of lamin A expression in hematopoietic cells relative to other cell types in solid tissues, but the prevalence of CHIP in HGPS remains unexplored. To investigate the potential role of CHIP in HGPS, we performed high-sensitivity targeted sequencing of CHIP-related genes in blood DNA samples from a cohort of 47 HGPS patients. As a control, the same sequencing strategy was applied to blood DNA samples from middle-aged and elderly individuals, expected to exhibit a biological age and cardiovascular risk profile similar to HGPS patients. We found that CHIP is not prevalent in HGPS patients, in marked contrast to our observations in individuals who age normally. Thus, our study unveils a major difference between HGPS and normal aging and provides conclusive evidence that CHIP is not frequent in HGPS and, therefore, is unlikely to contribute to the pathophysiology of this accelerated aging syndrome.


Asunto(s)
Enfermedades Cardiovasculares , Progeria , Humanos , Persona de Mediana Edad , Anciano , Adolescente , Progeria/genética , Hematopoyesis Clonal , Lamina Tipo A/genética , Envejecimiento/genética , Envejecimiento/metabolismo
4.
JACC CardioOncol ; 5(6): 715-731, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38205010

RESUMEN

Despite improvements in cancer survival, cancer therapy-related cardiovascular toxicity has risen to become a prominent clinical challenge. This has led to the growth of the burgeoning field of cardio-oncology, which aims to advance the cardiovascular health of cancer patients and survivors, through actionable and translatable science. In these Global Cardio-Oncology Symposium 2023 scientific symposium proceedings, we present a focused review on the mechanisms that contribute to common cardiovascular toxicities discussed at this meeting, the ongoing international collaborative efforts to improve patient outcomes, and the bidirectional challenges of translating basic research to clinical care. We acknowledge that there are many additional therapies that are of significance but were not topics of discussion at this symposium. We hope that through this symposium-based review we can highlight the knowledge gaps and clinical priorities to inform the design of future studies that aim to prevent and mitigate cardiovascular disease in cancer patients and survivors.

5.
Nat Cardiovasc Res ; 1(2): 116-124, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36337911

RESUMEN

Clonal hematopoiesis arises from somatic mutations that provide a fitness advantage to hematopoietic stem cells and the outgrowth of clones of blood cells. Clonal hematopoiesis commonly involves mutations in genes that are involved in epigenetic modifications, signaling and DNA damage repair. Clonal hematopoiesis has emerged as a major independent risk factor in atherosclerotic cardiovascular disease, thrombosis and heart failure. Studies in mouse models of clonal hematopoiesis have shown an increase in atherosclerosis, thrombosis and heart failure, involving increased myeloid cell inflammatory responses and inflammasome activation. Although increased inflammatory responses have emerged as a common underlying principle, some recent studies indicate mutation-specific effects. The discovery of the association of clonal hematopoiesis with cardiovascular disease and the recent demonstration of benefit of anti-inflammatory treatments in human cardiovascular disease converge to suggest that anti-inflammatory treatments should be directed to individuals with clonal hematopoiesis. Such treatments could target specific inflammasomes, common downstream mediators such as IL-1ß and IL-6, or mutations linked to clonal hematopoiesis.

6.
Thromb Res ; 213 Suppl 1: S107-S112, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-36210552

RESUMEN

Cancer genomes have long been known to carry a high number of somatic mutations distributed across many genes. However, recent sequencing studies have unveiled that non-cancerous cells also carry a considerable number of somatic mutations, which are acquired continuously through the lifespan. Accordingly, the pathophysiological relevance of somatic mutagenesis beyond cancer has become a topic of intensive research. Human genetic studies and experiments in mice have shown that some somatic mutations in the hematopoietic system provide a competitive advantage to the mutant cell and allow its clonal expansion. This phenomenon, termed clonal hematopoiesis, is typically driven by mutations in known oncogenes and tumor suppressor genes, and it is associated with a higher risk of hematological malignancies. Unexpectedly, accumulating genetic and experimental evidence strongly suggest that clonal hematopoiesis, at least when driven by certain mutations, also contributes causally to the development of cardiovascular disease and, therefore, represents a new cardiovascular risk factor. While clonal hematopoiesis is relatively common in healthy individuals, especially among the elderly, it is particularly frequent in cancer patients and survivors. Hence, it has emerged as a candidate contributor to the increased risk of cardiovascular complications in cancer patients. This review summarizes our current understanding of the connection between clonal hematopoiesis and cardiovascular disease, with a special focus on the available evidence linking clonal hematopoiesis to cardiovascular disorders that are frequent in cancer patients and survivors.


Asunto(s)
Enfermedades Cardiovasculares , Neoplasias , Anciano , Animales , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Hematopoyesis Clonal/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/patología , Humanos , Ratones , Mutación , Neoplasias/complicaciones , Neoplasias/genética , Neoplasias/patología , Sobrevivientes
7.
Eur Heart J ; 43(19): 1809-1828, 2022 05 14.
Artículo en Inglés | MEDLINE | ID: mdl-35567559

RESUMEN

AIMS: Experimental studies suggest that increased bone marrow (BM) activity is involved in the association between cardiovascular risk factors and inflammation in atherosclerosis. However, human data to support this association are sparse. The purpose was to study the association between cardiovascular risk factors, BM activation, and subclinical atherosclerosis. METHODS AND RESULTS: Whole body vascular 18F-fluorodeoxyglucose positron emission tomography/magnetic resonance imaging (18F-FDG PET/MRI) was performed in 745 apparently healthy individuals [median age 50.5 (46.8-53.6) years, 83.8% men] from the Progression of Early Subclinical Atherosclerosis (PESA) study. Bone marrow activation (defined as BM 18F-FDG uptake above the median maximal standardized uptake value) was assessed in the lumbar vertebrae (L3-L4). Systemic inflammation was indexed from circulating biomarkers. Early atherosclerosis was evaluated by arterial metabolic activity by 18F-FDG uptake in five vascular territories. Late atherosclerosis was evaluated by fully formed plaques on MRI. Subjects with BM activation were more frequently men (87.6 vs. 80.0%, P = 0.005) and more frequently had metabolic syndrome (MetS) (22.2 vs. 6.7%, P < 0.001). Bone marrow activation was significantly associated with all MetS components. Bone marrow activation was also associated with increased haematopoiesis-characterized by significantly elevated leucocyte (mainly neutrophil and monocytes) and erythrocyte counts-and with markers of systemic inflammation including high-sensitivity C-reactive protein, ferritin, fibrinogen, P-selectin, and vascular cell adhesion molecule-1. The associations between BM activation and MetS (and its components) and increased erythropoiesis were maintained in the subgroup of participants with no systemic inflammation. Bone marrow activation was significantly associated with high arterial metabolic activity (18F-FDG uptake). The co-occurrence of BM activation and arterial 18F-FDG uptake was associated with more advanced atherosclerosis (i.e. plaque presence and burden). CONCLUSION: In apparently healthy individuals, BM 18F-FDG uptake is associated with MetS and its components, even in the absence of systemic inflammation, and with elevated counts of circulating leucocytes. Bone marrow activation is associated with early atherosclerosis, characterized by high arterial metabolic activity. Bone marrow activation appears to be an early phenomenon in atherosclerosis development.[Progression of Early Subclinical Atherosclerosis (PESA); NCT01410318].


Asunto(s)
Aterosclerosis , Síndrome Metabólico , Placa Aterosclerótica , Aterosclerosis/metabolismo , Biomarcadores/metabolismo , Médula Ósea , Femenino , Fluorodesoxiglucosa F18 , Humanos , Inflamación/metabolismo , Masculino , Síndrome Metabólico/metabolismo , Persona de Mediana Edad , Placa Aterosclerótica/patología , Tomografía Computarizada por Tomografía de Emisión de Positrones/métodos , Tomografía de Emisión de Positrones , Radiofármacos
8.
J Am Coll Cardiol ; 77(14): 1747-1759, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33832602

RESUMEN

BACKGROUND: Clonal hematopoiesis driven by somatic mutations in hematopoietic cells, frequently called clonal hematopoiesis of indeterminate potential (CHIP), has been associated with adverse cardiovascular outcomes in population-based studies and in patients with ischemic heart failure (HF) and reduced left ventricular ejection fraction (LVEF). Yet, the impact of CHIP on HF progression, including nonischemic etiology, is unknown. OBJECTIVES: The purpose of this study was to assess the clinical impact of clonal hematopoiesis on HF progression irrespective of its etiology. METHODS: The study cohort comprised 62 patients with HF and LVEF <45% (age 74 ± 7 years, 74% men, 52% nonischemic, and LVEF 30 ± 8%). Deep sequencing was used to detect CHIP mutations with a variant allelic fraction >2% in 54 genes. Patients were followed for at least 3.5 years for various adverse events including death, HF-related death, and HF hospitalization. RESULTS: CHIP mutations were detected in 24 (38.7%) patients, without significant differences in all-cause mortality (p = 0.151). After adjusting for risk factors, patients with mutations in either DNA methyltransferase 3 alpha (DNMT3A) or Tet methylcytosine dioxygenase 2 (TET2) exhibited accelerated HF progression in terms of death (hazard ratio [HR]: 2.79; 95% confidence interval [CI]: 1.31 to 5.92; p = 0.008), death or HF hospitalization (HR: 3.84; 95% CI: 1.84 to 8.04; p < 0.001) and HF-related death or HF hospitalization (HR: 4.41; 95% CI: 2.15 to 9.03; p < 0.001). In single gene-specific analyses, somatic mutations in DNMT3A or TET2 retained prognostic significance with regard to HF-related death or HF hospitalization (HR: 4.50; 95% CI: 2.07 to 9.74; p < 0.001, for DNMT3A mutations; HR: 3.18; 95% CI: 1.52 to 6.66; p = 0.002, for TET2 mutations). This association remained significant irrespective of ischemic/nonischemic etiology. CONCLUSIONS: Somatic mutations that drive clonal hematopoiesis are common among HF patients with reduced LVEF and are associated with accelerated HF progression regardless of etiology.


Asunto(s)
Hematopoyesis Clonal/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Proteínas de Unión al ADN/genética , Insuficiencia Cardíaca , Proteínas Proto-Oncogénicas/genética , Disfunción Ventricular Izquierda , Anciano , Causas de Muerte , ADN Metiltransferasa 3A , Dioxigenasas , Progresión de la Enfermedad , Femenino , Insuficiencia Cardíaca/diagnóstico , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/mortalidad , Insuficiencia Cardíaca/fisiopatología , Hospitalización/estadística & datos numéricos , Humanos , Masculino , Mortalidad , Mutación , Pronóstico , Estudios Prospectivos , España/epidemiología , Disfunción Ventricular Izquierda/etiología , Disfunción Ventricular Izquierda/fisiopatología
9.
Cell Rep ; 33(4): 108326, 2020 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-33113366

RESUMEN

Human aging is frequently accompanied by the acquisition of somatic mutations in the hematopoietic system that induce clonal hematopoiesis, leading to the development of a mutant clone of hematopoietic progenitors and leukocytes. This somatic-mutation-driven clonal hematopoiesis has been associated with an increased incidence of cardiovascular disease and type 2 diabetes, but whether this epidemiological association reflects a direct, causal contribution of mutant hematopoietic and immune cells to age-related metabolic abnormalities remains unexplored. Here, we show that inactivating mutations in the epigenetic regulator TET2, which lead to clonal hematopoiesis, aggravate age- and obesity-related insulin resistance in mice. This metabolic dysfunction is paralleled by increased expression of the pro-inflammatory cytokine IL-1ß in white adipose tissue, and it is suppressed by pharmacological inhibition of NLRP3 inflammasome-mediated IL-1ß production. These findings support a causal contribution of somatic TET2 mutations to insulin resistance and type 2 diabetes.


Asunto(s)
Hematopoyesis Clonal/genética , Proteínas de Unión al ADN/metabolismo , Dioxigenasas/metabolismo , Resistencia a la Insulina/genética , Obesidad/genética , Envejecimiento , Animales , Humanos , Ratones
10.
JCI Insight ; 5(6)2020 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-32154790

RESUMEN

Clonal hematopoiesis of indeterminate potential is prevalent in elderly individuals and associated with increased risks of all-cause mortality and cardiovascular disease. However, mouse models to study the dynamics of clonal hematopoiesis and its consequences on the cardiovascular system under homeostatic conditions are lacking. We developed a model of clonal hematopoiesis using adoptive transfer of unfractionated ten-eleven translocation 2-mutant (Tet2-mutant) bone marrow cells into nonirradiated mice. Consistent with age-related clonal hematopoiesis observed in humans, these mice displayed a progressive expansion of Tet2-deficient cells in multiple hematopoietic stem and progenitor cell fractions and blood cell lineages. The expansion of the Tet2-mutant fraction was also observed in bone marrow-derived CCR2+ myeloid cell populations within the heart, but there was a negligible impact on the yolk sac-derived CCR2- cardiac-resident macrophage population. Transcriptome profiling revealed an enhanced inflammatory signature in the donor-derived macrophages isolated from the heart. Mice receiving Tet2-deficient bone marrow cells spontaneously developed age-related cardiac dysfunction characterized by greater hypertrophy and fibrosis. Altogether, we show that Tet2-mediated hematopoiesis contributes to cardiac dysfunction in a nonconditioned setting that faithfully models human clonal hematopoiesis in unperturbed bone marrow. Our data support clinical findings that clonal hematopoiesis per se may contribute to diminished health span.


Asunto(s)
Hematopoyesis Clonal/fisiología , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Cardiopatías , Proteínas Proto-Oncogénicas/metabolismo , Traslado Adoptivo , Envejecimiento/patología , Animales , Dioxigenasas , Células Madre Hematopoyéticas , Macrófagos , Ratones
11.
Atherosclerosis ; 297: 120-126, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32109665

RESUMEN

The accumulation of acquired mutations is an inevitable consequence of the aging process, but its pathophysiological relevance has remained largely unexplored beyond cancer. Most of these mutations have little or no functional consequences, but in a few rare instances, a mutation may arise that confers a competitive advantage to a stem cell, leading to its clonal expansion. When such a mutation occurs in hematopoietic stem cells, it leads to a situation of clonal hematopoiesis, which has the potential to affect multiple tissues beyond the bone marrow, as the clonal expansion of the mutant stem cell is extended to circulating blood cells and tissue-infiltrating immune cells. Recent genomics and experimental studies have provided support to the notion that this somatic mutation-driven clonal hematopoiesis contributes to vascular inflammation and the development of atherosclerosis and related cardiovascular and cerebrovascular ischemic events. Here, we review our current understanding of this emerging cardiovascular risk modifier and the mechanisms underlying its connection to atherosclerosis development.


Asunto(s)
Aterosclerosis/genética , Hematopoyesis Clonal/genética , Células Madre Hematopoyéticas/patología , Mutación , Animales , Aterosclerosis/sangre , Aterosclerosis/patología , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/genética , Dioxigenasas , Predisposición Genética a la Enfermedad , Factores de Riesgo de Enfermedad Cardiaca , Células Madre Hematopoyéticas/metabolismo , Humanos , Fenotipo , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Medición de Riesgo
12.
J Am Coll Cardiol ; 74(4): 578-586, 2019 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-31345433

RESUMEN

The incidence of cardiovascular diseases increases with age and is also correlated with increased inflammatory burden. Recently, human genetics provided a new paradigm linking aging, inflammation, and atherosclerotic cardiovascular disease (ASCVD). Next-generation genetic sequencing of whole blood-derived DNA in humans showed that clonal expansion of hematopoietic cells with somatic mutations in leukemogenic genes was associated with age and correlated with increased mortality. This phenomenon, termed clonal hematopoiesis of indeterminate potential (CHIP), was associated with hematologic malignancy as well as ASCVD independently of age and other traditional risk factors. Because the implication of CHIP with ASCVD, genetic loss-of-function studies of Tet2 and Dnmt3a in murine models have supported a mechanistic role for CHIP in promoting vascular disease. Despite the potential contribution of CHIP to myriad cardiovascular and aging-related diseases, the epidemiology and biology surrounding this phenomenon remains incompletely appreciated and understood, especially as applied to clinical practice and prognostication. Here, the authors review this emerging key risk factor, defining its discovery, relationship to cardiovascular diseases, preclinical evidence for causality, and implications for risk prediction and mitigation.


Asunto(s)
Envejecimiento , Enfermedades Cardiovasculares/etiología , Hematopoyesis/genética , Células Madre Hematopoyéticas , Mutación , Aterosclerosis/etiología , Enfermedades Cardiovasculares/genética , Humanos , Neoplasias/complicaciones , Neoplasias/genética , Factores de Riesgo
13.
J Am Coll Cardiol ; 71(8): 875-886, 2018 02 27.
Artículo en Inglés | MEDLINE | ID: mdl-29471939

RESUMEN

BACKGROUND: Recent studies have shown that hematopoietic stem cells can undergo clonal expansion secondary to somatic mutations in leukemia-related genes, thus leading to an age-dependent accumulation of mutant leukocytes in the blood. This somatic mutation-related clonal hematopoiesis is common in healthy older individuals, but it has been associated with an increased incidence of future cardiovascular disease. The epigenetic regulator TET2 is frequently mutated in blood cells of individuals exhibiting clonal hematopoiesis. OBJECTIVES: This study investigated whether Tet2 mutations within hematopoietic cells can contribute to heart failure in 2 models of cardiac injury. METHODS: Heart failure was induced in mice by pressure overload, achieved by transverse aortic constriction or chronic ischemia induced by the permanent ligation of the left anterior descending artery. Competitive bone marrow transplantation strategies with Tet2-deficient cells were used to mimic TET2 mutation-driven clonal hematopoiesis. Alternatively, Tet2 was specifically ablated in myeloid cells using Cre recombinase expressed from the LysM promoter. RESULTS: In both experimental heart failure models, hematopoietic or myeloid Tet2 deficiency worsened cardiac remodeling and function, in parallel with increased interleukin-1beta (IL-1ß) expression. Treatment with a selective NLRP3 inflammasome inhibitor protected against the development of heart failure and eliminated the differences in cardiac parameters between Tet2-deficient and wild-type mice. CONCLUSIONS: Tet2 deficiency in hematopoietic cells is associated with greater cardiac dysfunction in murine models of heart failure as a result of elevated IL-1ß signaling. These data suggest that individuals with TET2-mediated clonal hematopoiesis may be at greater risk of developing heart failure and respond better to IL-1ß-NLRP3 inflammasome inhibition.


Asunto(s)
Proteínas de Unión al ADN/deficiencia , Insuficiencia Cardíaca/metabolismo , Hematopoyesis/fisiología , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Animales , Células Cultivadas , Proteínas de Unión al ADN/genética , Dioxigenasas , Furanos , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/prevención & control , Hematopoyesis/efectos de los fármacos , Compuestos Heterocíclicos de 4 o más Anillos/farmacología , Compuestos Heterocíclicos de 4 o más Anillos/uso terapéutico , Indenos , Inflamasomas/antagonistas & inhibidores , Interleucina-1beta/antagonistas & inhibidores , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/genética , Sulfonamidas , Sulfonas/farmacología , Sulfonas/uso terapéutico
14.
Circ Res ; 122(3): 523-532, 2018 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-29420212

RESUMEN

Increasing evidence shows that conventional cardiovascular risk factors are incompletely predictive of cardiovascular disease, particularly in elderly individuals, suggesting that there may still be unidentified causal risk factors. Although the accumulation of somatic DNA mutations is a hallmark of aging, its relevance in cardiovascular disease or other age-related conditions has been, with the exception of cancer, largely unexplored. Here, we review recent clinical and preclinical studies that have identified acquired mutations in hematopoietic stem cells and subsequent clonal hematopoiesis as a new cardiovascular risk factor and a potential major driver of atherosclerosis. Understanding the mechanisms underlying the connection between somatic mutation-driven clonal hematopoiesis and cardiovascular disease will be highly relevant in the context of personalized medicine, as it may provide key information for the design of diagnostic, preventive, or therapeutic strategies tailored to the effects of specific somatic mutations.


Asunto(s)
Envejecimiento/genética , Enfermedades Cardiovasculares/etiología , Hematopoyesis/genética , Células Madre Hematopoyéticas/patología , Mutación , Anciano , Envejecimiento/patología , Animales , Aterosclerosis/etiología , Aterosclerosis/genética , Aterosclerosis/patología , Aterosclerosis/terapia , Trasplante de Médula Ósea , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/patología , Causalidad , Células Clonales/patología , ADN (Citosina-5-)-Metiltransferasas/genética , ADN (Citosina-5-)-Metiltransferasas/fisiología , ADN Metiltransferasa 3A , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/fisiología , Dioxigenasas , Genes Relacionados con las Neoplasias , Estudios de Asociación Genética , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/patología , Humanos , Janus Quinasa 2/deficiencia , Janus Quinasa 2/genética , Janus Quinasa 2/fisiología , Ratones , Dinámica Poblacional , Medicina de Precisión , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/fisiología , Factores de Riesgo
15.
J Mol Cell Cardiol ; 116: 5-15, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29408196

RESUMEN

Phosphorylation at serine 10 (S10) is the major posttranslational modification of the tumor suppressor p27, and is reduced in both human and mouse atherosclerosis. Moreover, a lack of p27-phospho-S10 in apolipoprotein E-null mice (apoE-/-) leads to increased high-fat diet-induced atherosclerosis associated with endothelial dysfunction and augmented leukocyte recruitment. In this study, we analyzed whether p27-phospho-S10 modulates additional endothelial functions and associated pathologies. Defective p27-phospho-S10 increases COX-2 activity in mouse aortic endothelial cells without affecting other key regulators of vascular reactivity, reduces endothelium-dependent dilation, and increases arterial contractility. Lack of p27-phospho-S10 also elevates aortic COX-2 expression and thromboxane A2 production, increases aortic lumen diameter, and aggravates angiotensin II-induced abdominal aortic aneurysm development in apoE-/- mice. All these abnormal responses linked to defective p27-phospho-S10 are blunted by pharmacological inhibition of COX-2. These results demonstrate that defective p27-phospho-S10 modifies endothelial behavior and promotes aneurysm formation via COX-2 activation.


Asunto(s)
Aneurisma de la Aorta Abdominal/metabolismo , Aneurisma de la Aorta Abdominal/fisiopatología , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Ciclooxigenasa 2/metabolismo , Fosfoserina/metabolismo , Acetilcolina/farmacología , Angiotensina II , Animales , Aorta/patología , Presión Sanguínea/efectos de los fármacos , Células Endoteliales/metabolismo , Activación Enzimática , Ratones Endogámicos C57BL , Fosforilación , Tromboxanos/metabolismo , Vasodilatación , Remodelación Ventricular/efectos de los fármacos
16.
Sci Rep ; 7(1): 17326, 2017 12 11.
Artículo en Inglés | MEDLINE | ID: mdl-29229927

RESUMEN

The accumulation of visceral adiposity is strongly associated with systemic inflammation and increased cardiometabolic risk. WNT5A, a non-canonical WNT ligand, has been shown to promote adipose tissue inflammation and insulin resistance in animal studies. Among other non-canonical pathways, WNT5A activates planar cell polarity (PCP) signaling. The current study investigated the potential contribution of non-canonical WNT5A/PCP signaling to visceral adipose tissue (VAT) inflammation and associated metabolic dysfunction in individuals with obesity. VAT and subcutaneous adipose tissue (SAT) samples obtained from subjects undergoing bariatric surgery were analyzed by qRT-PCR for expression of WNT/PCP genes. In vitro experiments were conducted with preadipocytes isolated from VAT and SAT biopsies. The expression of 23 out of 33 PCP genes was enriched in VAT compared to SAT. Strong positive expression correlations of individual PCP genes were observed in VAT. WNT5A expression in VAT, but not in SAT, correlated with indexes of JNK signaling activity, IL6, waist-to-hip ratio and hsCRP. In vitro, WNT5A promoted the expression of IL6 in human preadipocytes. In conclusion, elevated non-canonical WNT5A signaling in VAT contributes to the exacerbated IL-6 production in this depot and the low-grade systemic inflammation typically associated with visceral adiposity.


Asunto(s)
Regulación de la Expresión Génica , Paniculitis/metabolismo , Grasa Subcutánea/metabolismo , Vía de Señalización Wnt , Adulto , Femenino , Humanos , Inflamación/metabolismo , Inflamación/patología , Masculino , Paniculitis/patología , Grasa Subcutánea/patología
17.
Science ; 355(6327): 842-847, 2017 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-28104796

RESUMEN

Human aging is associated with an increased frequency of somatic mutations in hematopoietic cells. Several of these recurrent mutations, including those in the gene encoding the epigenetic modifier enzyme TET2, promote expansion of the mutant blood cells. This clonal hematopoiesis correlates with an increased risk of atherosclerotic cardiovascular disease. We studied the effects of the expansion of Tet2-mutant cells in atherosclerosis-prone, low-density lipoprotein receptor-deficient (Ldlr-/-) mice. We found that partial bone marrow reconstitution with TET2-deficient cells was sufficient for their clonal expansion and led to a marked increase in atherosclerotic plaque size. TET2-deficient macrophages exhibited an increase in NLRP3 inflammasome-mediated interleukin-1ß secretion. An NLRP3 inhibitor showed greater atheroprotective activity in chimeric mice reconstituted with TET2-deficient cells than in nonchimeric mice. These results support the hypothesis that somatic TET2 mutations in blood cells play a causal role in atherosclerosis.


Asunto(s)
Aterosclerosis/genética , Proteínas de Unión al ADN/genética , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Proteínas Proto-Oncogénicas/genética , Animales , Dioxigenasas , Inflamasomas/metabolismo , Macrófagos , Ratones , Ratones Endogámicos C57BL , Mutación , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Placa Aterosclerótica/genética , Receptores de LDL/genética
18.
Arterioscler Thromb Vasc Biol ; 36(3): 561-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26800561

RESUMEN

OBJECTIVE: Endothelial dysfunction is linked to insulin resistance, inflammatory activation, and increased cardiovascular risk in diabetes mellitus; however, the mechanisms remain incompletely understood. Recent studies have identified proinflammatory signaling of wingless-type family member (Wnt) 5a through c-jun N-terminal kinase (JNK) as a regulator of metabolic dysfunction with potential relevance to vascular function. We sought to gain evidence that increased activation of Wnt5a-JNK signaling contributes to impaired endothelial function in patients with diabetes mellitus. APPROACH AND RESULTS: We measured flow-mediated dilation of the brachial artery and characterized freshly isolated endothelial cells by protein expression, eNOS activation, and nitric oxide production in 85 subjects with type 2 diabetes mellitus (n=42) and age- and sex-matched nondiabetic controls (n=43) and in human aortic endothelial cells treated with Wnt5a. Endothelial cells from patients with diabetes mellitus displayed 1.3-fold higher Wnt5a levels (P=0.01) along with 1.4-fold higher JNK activation (P<0.01) without a difference in total JNK levels. Higher JNK activation was associated with lower flow-mediated dilation, consistent with endothelial dysfunction (r=0.53, P=0.02). Inhibition of Wnt5a and JNK signaling restored insulin and A23187-mediated eNOS activation and improved nitric oxide production in endothelial cells from patients with diabetes mellitus. In endothelial cells from nondiabetic controls, rWnt5a treatment inhibited eNOS activation replicating the diabetic endothelial phenotype. In human aortic endothelial cells, Wnt5a-induced impairment of eNOS activation and nitric oxide production was reversed by Wnt5a and JNK inhibition. CONCLUSIONS: Our findings demonstrate that noncanonical Wnt5a signaling and JNK activity contribute to vascular insulin resistance and endothelial dysfunction and may represent a novel therapeutic opportunity to protect the vasculature in patients with diabetes mellitus.


Asunto(s)
Arteria Braquial/enzimología , Diabetes Mellitus Tipo 2/enzimología , Células Endoteliales/enzimología , Endotelio Vascular/enzimología , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Vasodilatación , Proteínas Wnt/metabolismo , Vía de Señalización Wnt , Adulto , Anciano , Arteria Braquial/efectos de los fármacos , Arteria Braquial/fisiopatología , Estudios de Casos y Controles , Células Cultivadas , Diabetes Mellitus Tipo 2/fisiopatología , Células Endoteliales/efectos de los fármacos , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/fisiopatología , Activación Enzimática , Femenino , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Masculino , Persona de Mediana Edad , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo III/metabolismo , Estrés Oxidativo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/farmacología , Vasodilatación/efectos de los fármacos , Proteínas Wnt/antagonistas & inhibidores , Proteínas Wnt/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Proteína Wnt-5a
19.
J Biol Chem ; 291(6): 2566-75, 2016 Feb 05.
Artículo en Inglés | MEDLINE | ID: mdl-26631720

RESUMEN

Wnt signaling has diverse actions in cardiovascular development and disease processes. Secreted frizzled-related protein 5 (Sfrp5) has been shown to function as an extracellular inhibitor of non-canonical Wnt signaling that is expressed at relatively high levels in white adipose tissue. The aim of this study was to investigate the role of Sfrp5 in the heart under ischemic stress. Sfrp5 KO and WT mice were subjected to ischemia/reperfusion (I/R). Although Sfrp5-KO mice exhibited no detectable phenotype when compared with WT control at baseline, they displayed larger infarct sizes, enhanced cardiac myocyte apoptosis, and diminished cardiac function following I/R. The ischemic lesions of Sfrp5-KO mice had greater infiltration of Wnt5a-positive macrophages and greater inflammatory cytokine and chemokine gene expression when compared with WT mice. In bone marrow-derived macrophages, Wnt5a promoted JNK activation and increased inflammatory gene expression, whereas treatment with Sfrp5 blocked these effects. These results indicate that Sfrp5 functions to antagonize inflammatory responses after I/R in the heart, possibly through a mechanism involving non-canonical Wnt5a/JNK signaling.


Asunto(s)
Proteínas de la Membrana/metabolismo , Isquemia Miocárdica/metabolismo , Miocarditis/metabolismo , Miocardio/metabolismo , Miocitos Cardíacos/metabolismo , Transducción de Señal , Animales , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , MAP Quinasa Quinasa 4/genética , MAP Quinasa Quinasa 4/metabolismo , Macrófagos/metabolismo , Macrófagos/patología , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Isquemia Miocárdica/genética , Isquemia Miocárdica/patología , Miocarditis/genética , Miocarditis/patología , Miocardio/patología , Miocitos Cardíacos/patología , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteína Wnt-5a
20.
Diabetes ; 64(4): 1235-48, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25352637

RESUMEN

Adipose tissue dysfunction plays a pivotal role in the development of insulin resistance in obese individuals. Cell culture studies and gain-of-function mouse models suggest that canonical Wnt proteins modulate adipose tissue expansion. However, no genetic evidence supports a role for endogenous Wnt proteins in adipose tissue dysfunction, and the role of noncanonical Wnt signaling remains largely unexplored. Here we provide evidence from human, mouse, and cell culture studies showing that Wnt5a-mediated, noncanonical Wnt signaling contributes to obesity-associated metabolic dysfunction by increasing adipose tissue inflammation. Wnt5a expression is significantly upregulated in human visceral fat compared with subcutaneous fat in obese individuals. In obese mice, Wnt5a ablation ameliorates insulin resistance, in parallel with reductions in adipose tissue inflammation. Conversely, Wnt5a overexpression in myeloid cells augments adipose tissue inflammation and leads to greater impairments in glucose homeostasis. Wnt5a ablation or overexpression did not affect fat mass or adipocyte size. Mechanistically, Wnt5a promotes the expression of proinflammatory cytokines by macrophages in a Jun NH2-terminal kinase-dependent manner, leading to defective insulin signaling in adipocytes. Exogenous interleukin-6 administration restores insulin resistance in obese Wnt5a-deficient mice, suggesting a central role for this cytokine in Wnt5a-mediated metabolic dysfunction. Taken together, these results demonstrate that noncanonical Wnt signaling contributes to obesity-induced insulin resistance independent of adipose tissue expansion.


Asunto(s)
Grasa Intraabdominal/metabolismo , Obesidad/metabolismo , Grasa Subcutánea/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Glucosa/metabolismo , Humanos , Inflamación/metabolismo , Resistencia a la Insulina , Macrófagos/metabolismo , Ratones , Fosforilación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA