Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 927
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 170(3): 522-533.e15, 2017 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-28753427

RESUMEN

Genome-wide association studies (GWASs) implicate the PHACTR1 locus (6p24) in risk for five vascular diseases, including coronary artery disease, migraine headache, cervical artery dissection, fibromuscular dysplasia, and hypertension. Through genetic fine mapping, we prioritized rs9349379, a common SNP in the third intron of the PHACTR1 gene, as the putative causal variant. Epigenomic data from human tissue revealed an enhancer signature at rs9349379 exclusively in aorta, suggesting a regulatory function for this SNP in the vasculature. CRISPR-edited stem cell-derived endothelial cells demonstrate rs9349379 regulates expression of endothelin 1 (EDN1), a gene located 600 kb upstream of PHACTR1. The known physiologic effects of EDN1 on the vasculature may explain the pattern of risk for the five associated diseases. Overall, these data illustrate the integration of genetic, phenotypic, and epigenetic analysis to identify the biologic mechanism by which a common, non-coding variant can distally regulate a gene and contribute to the pathogenesis of multiple vascular diseases.


Asunto(s)
Enfermedad de la Arteria Coronaria/genética , Endotelina-1/genética , Predisposición Genética a la Enfermedad , Polimorfismo de Nucleótido Simple , Enfermedades Vasculares/genética , Acetilación , Células Cultivadas , Cromatina/metabolismo , Mapeo Cromosómico , Cromosomas Humanos Par 6 , Células Endoteliales/citología , Endotelina-1/sangre , Epigenómica , Edición Génica , Expresión Génica , Estudio de Asociación del Genoma Completo , Histonas/metabolismo , Humanos , Músculo Liso Vascular/citología
2.
Circ Res ; 135(1): 159-173, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38900856

RESUMEN

Over the past several centuries, the integration of contemporary medical techniques and innovative technologies, like genetic sequencing, have played a pivotal role in enhancing our comprehension of congenital vascular and lymphatic disorders. Nonetheless, the uncommon and complex characteristics of these disorders, especially considering their formation during the intrauterine stage, present significant obstacles in diagnosis and treatment. Here, we review the intricacies of these congenital abnormalities, offering an in-depth examination of key diagnostic approaches, genetic factors, and therapeutic methods.


Asunto(s)
Enfermedades Linfáticas , Humanos , Enfermedades Linfáticas/terapia , Enfermedades Linfáticas/genética , Enfermedades Vasculares/congénito , Enfermedades Vasculares/genética , Enfermedades Vasculares/terapia , Enfermedades Vasculares/diagnóstico , Animales , Malformaciones Vasculares/genética , Malformaciones Vasculares/terapia , Vasos Linfáticos/anomalías , Predisposición Genética a la Enfermedad
3.
Proc Natl Acad Sci U S A ; 119(25): e2202327119, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35696583

RESUMEN

Pediatric patients with constitutively active mutations in the cytosolic double-stranded-DNA-sensing adaptor STING develop an autoinflammatory syndrome known as STING-associated vasculopathy with onset in infancy (SAVI). SAVI patients have elevated interferon-stimulated gene expression and suffer from interstitial lung disease (ILD) with lymphocyte predominate bronchus-associated lymphoid tissue (BALT). Mice harboring SAVI mutations (STING V154M [VM]) that recapitulate human disease also develop lymphocyte-rich BALT. Ablation of either T or B lymphocytes prolongs the survival of SAVI mice, but lung immune aggregates persist, indicating that T cells and B cells can independently be recruited as BALT. VM T cells produced IFNγ, and IFNγR deficiency prolonged the survival of SAVI mice; however, T-cell-dependent recruitment of infiltrating myeloid cells to the lung was IFNγ independent. Lethally irradiated VM recipients fully reconstituted with wild type bone-marrow-derived cells still developed ILD, pointing to a critical role for VM-expressing radioresistant parenchymal and/or stromal cells in the recruitment and activation of pathogenic lymphocytes. We identified lung endothelial cells as radioresistant cells that express STING. Transcriptional analysis of VM endothelial cells revealed up-regulation of chemokines, proinflammatory cytokines, and genes associated with antigen presentation. Together, our data show that VM-expressing radioresistant cells play a key role in the initiation of lung disease in VM mice and provide insights for the treatment of SAVI patients, with implications for ILD associated with other connective tissue disorders.


Asunto(s)
Células Endoteliales , Enfermedades Pulmonares Intersticiales , Proteínas de la Membrana , Linfocitos T , Enfermedades Vasculares , Animales , Niño , Células Endoteliales/inmunología , Células Endoteliales/efectos de la radiación , Mutación con Ganancia de Función , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Enfermedades Pulmonares Intersticiales/genética , Enfermedades Pulmonares Intersticiales/inmunología , Depleción Linfocítica , Tejido Linfoide/inmunología , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Tolerancia a Radiación , Linfocitos T/inmunología , Enfermedades Vasculares/genética , Enfermedades Vasculares/inmunología
4.
Curr Opin Lipidol ; 35(2): 78-84, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38054895

RESUMEN

PURPOSE OF REVIEW: Apolipoprotein E (apoE) plays a pivotal role in lipid metabolism in the peripheral circulation and in the brain. This has been recognized for decades; however, the importance of the full spectrum of variation in the APOE gene has been less investigated. This review focusses on current progresses in this field with main focus on apoE in dyslipidemia and vascular disease. RECENT FINDINGS: Whereas ε4 is the risk increasing allele for Alzheimer disease, ε2 is associated with increased risk for age-related macular degeneration. Rare functional ε2-like variants in APOE have previously been reported to have protective associations for Alzheimer disease but recent findings suggest a simultaneous high risk of age-related macular degeneration, in line with observations for the ε2 allele. SUMMARY: ApoE plays an important and well established role in dyslipidemia, vascular disease, and dementia. Recent evidence from large general population studies now also suggests that apoE is involved in age-related macular degeneration. ApoE-targeted therapeutics are being developed for multiple purposes; this heralds a promising change in the approach to disease processes involving apoE. The different risk profile for dementia and age-related macular degeneration should, however, be kept in mind when developing drugs targeting mechanisms resembling these variants.


Asunto(s)
Enfermedad de Alzheimer , Dislipidemias , Degeneración Macular , Enfermedades Vasculares , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/epidemiología , Genotipo , Apolipoproteínas E/genética , Alelos , Enfermedades Vasculares/genética , Degeneración Macular/genética , Dislipidemias/genética
5.
Physiol Rev ; 97(4): 1555-1617, 2017 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-28954852

RESUMEN

The cushioning function of large arteries encompasses distension during systole and recoil during diastole which transforms pulsatile flow into a steady flow in the microcirculation. Arterial stiffness, the inverse of distensibility, has been implicated in various etiologies of chronic common and monogenic cardiovascular diseases and is a major cause of morbidity and mortality globally. The first components that contribute to arterial stiffening are extracellular matrix (ECM) proteins that support the mechanical load, while the second important components are vascular smooth muscle cells (VSMCs), which not only regulate actomyosin interactions for contraction but mediate also mechanotransduction in cell-ECM homeostasis. Eventually, VSMC plasticity and signaling in both conductance and resistance arteries are highly relevant to the physiology of normal and early vascular aging. This review summarizes current concepts of central pressure and tensile pulsatile circumferential stress as key mechanical determinants of arterial wall remodeling, cell-ECM interactions depending mainly on the architecture of cytoskeletal proteins and focal adhesion, the large/small arteries cross-talk that gives rise to target organ damage, and inflammatory pathways leading to calcification or atherosclerosis. We further speculate on the contribution of cellular stiffness along the arterial tree to vascular wall stiffness. In addition, this review provides the latest advances in the identification of gene variants affecting arterial stiffening. Now that important hemodynamic and molecular mechanisms of arterial stiffness have been elucidated, and the complex interplay between ECM, cells, and sensors identified, further research should study their potential to halt or to reverse the development of arterial stiffness.


Asunto(s)
Envejecimiento/metabolismo , Matriz Extracelular/metabolismo , Músculo Liso Vascular/metabolismo , Miocitos del Músculo Liso/metabolismo , Enfermedades Vasculares/metabolismo , Rigidez Vascular , Factores de Edad , Envejecimiento/genética , Envejecimiento/patología , Animales , Presión Arterial , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Humanos , Mecanotransducción Celular , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Flujo Pulsátil , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología , Enfermedades Vasculares/fisiopatología
6.
Neurogenetics ; 25(3): 157-164, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38625441

RESUMEN

Vascular risk factors, including diabetes, hypertension, hyperlipidemia, and obesity, pose significant health threats with implications extending to neuropsychiatric disorders such as stroke and Alzheimer's disease. The Asian population, in particular, appears to be disproportionately affected due to unique genetic predispositions, as well as epigenetic factors such as dietary patterns and lifestyle habits. Existing management strategies often fall short of addressing these specific needs, leading to greater challenges in prevention and treatment. This review highlights a significant gap in our understanding of the impact of genetic screening in the early detection and tailored treatment of vascular risk factors among the Asian population. Apolipoprotein, a key player in cholesterol metabolism, is primarily associated with dyslipidemia, yet emerging evidence suggests its involvement in conditions such as diabetes, hypertension, and obesity. While genetic variants of vascular risk are ethnic-dependent, current evidence indicates that epigenetics also exhibits ethnic specificity. Understanding the interplay between Apolipoprotein and genetics, particularly within diverse ethnic backgrounds, has the potential to refine risk stratification and enhance precision in management. For Caucasian carrying the APOA5 rs662799 C variant, pharmacological interventions are recommended, as dietary interventions may not be sufficient. In contrast, for Asian populations with the same genetic variant, dietary modifications are initially advised. Should dyslipidemia persist, the consideration of pharmaceutical agents such as statins is recommended.


Asunto(s)
Pueblo Asiatico , Predisposición Genética a la Enfermedad , Humanos , Pueblo Asiatico/genética , Factores de Riesgo , Variación Genética , Apolipoproteínas/genética , Apolipoproteína A-V/genética , Enfermedades Vasculares/genética
7.
Curr Atheroscler Rep ; 26(7): 331-340, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38761354

RESUMEN

PURPOSE OF REVIEW: Spontaneous coronary artery dissection (SCAD) has been increasingly recognized as a significant cause of acute myocardial infarction (AMI) in young and middle-aged women and arises through mechanisms independent of atherosclerosis. SCAD has a multifactorial etiology that includes environmental, individual, and genetic factors distinct from those typically associated with coronary artery disease. Here, we summarize the current understanding of the genetic factors contributing to the development of SCAD and highlight those factors which differentiate SCAD from atherosclerotic coronary artery disease. RECENT FINDINGS: Recent studies have revealed several associated variants with varying effect sizes for SCAD, giving rise to a complex genetic architecture. Associated genes highlight an important role for arterial cells and their extracellular matrix in the pathogenesis of SCAD, as well as notable genetic overlap between SCAD and other systemic arteriopathies such as fibromuscular dysplasia and vascular connective tissue diseases. Further investigation of individual variants (including in the associated gene PHACTR1) along with polygenic score analysis have demonstrated an inverse genetic relationship between SCAD and atherosclerosis as distinct causes of AMI. SCAD represents an increasingly recognized cause of AMI with opposing clinical and genetic risk factors from that of AMI due to atherosclerosis, and it is often associated with complex underlying genetic conditions. Genetic study of SCAD on a larger scale and with more diverse cohorts will not only further our evolving understanding of a newly defined genetic spectrum for AMI, but it will also inform the clinical utility of integrating genetic testing in AMI prevention and management moving forward.


Asunto(s)
Anomalías de los Vasos Coronarios , Predisposición Genética a la Enfermedad , Infarto del Miocardio , Enfermedades Vasculares , Humanos , Infarto del Miocardio/genética , Anomalías de los Vasos Coronarios/genética , Anomalías de los Vasos Coronarios/complicaciones , Enfermedades Vasculares/genética , Enfermedades Vasculares/congénito , Factores de Riesgo , Aterosclerosis/genética , Aterosclerosis/complicaciones , Enfermedad de la Arteria Coronaria/genética
8.
Arterioscler Thromb Vasc Biol ; 43(1): 15-29, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36412195

RESUMEN

Cardiovascular disease is the most common cause of death worldwide, especially beyond the age of 65 years, with the vast majority of morbidity and mortality due to myocardial infarction and stroke. Vascular pathology stems from a combination of genetic risk, environmental factors, and the biologic changes associated with aging. The pathogenesis underlying the development of vascular aging, and vascular calcification with aging, in particular, is still not fully understood. Accumulating data suggests that genetic risk, likely compounded by epigenetic modifications, environmental factors, including diabetes and chronic kidney disease, and the plasticity of vascular smooth muscle cells to acquire an osteogenic phenotype are major determinants of age-associated vascular calcification. Understanding the molecular mechanisms underlying genetic and modifiable risk factors in regulating age-associated vascular pathology may inspire strategies to promote healthy vascular aging. This article summarizes current knowledge of concepts and mechanisms of age-associated vascular disease, with an emphasis on vascular calcification.


Asunto(s)
Enfermedades Cardiovasculares , Calcificación Vascular , Enfermedades Vasculares , Humanos , Calcificación Vascular/patología , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología , Músculo Liso Vascular/patología , Enfermedades Cardiovasculares/patología , Miocitos del Músculo Liso/patología
9.
Int J Mol Sci ; 25(10)2024 May 18.
Artículo en Inglés | MEDLINE | ID: mdl-38791563

RESUMEN

Chronic venous disease (CVD) comprises a spectrum of morphofunctional disorders affecting the venous system, affecting approximately 1 in 3 women during gestation. Emerging evidence highlights diverse maternofetal implications stemming from CVD, particularly impacting the placenta. While systemic inflammation has been associated with pregnancy-related CVD, preliminary findings suggest a potential link between this condition and exacerbated inflammation in the placental tissue. Inflammasomes are major orchestrators of immune responses and inflammation in different organs and systems. Notwithstanding the relevance of inflammasomes, specifically the NLRP3 (nucleotide-binding domain, leucine-rich-containing family, pyrin domain-containing-3)- which has been demonstrated in the placentas of women with different obstetric complications, the precise involvement of this component in the placentas of women with CVD remains to be explored. This study employs immunohistochemistry and real-time PCR (RT-qPCR) to examine the gene and protein expression of key components in both canonical and non-canonical pathways of the NLRP3 inflammasome (NLRP3, ASC-apoptosis-associated speck-like protein containing a C-terminal caspase recruitment domain-caspase 1, caspase 5, caspase 8, and interleukin 1ß) within the placental tissue of women affected by CVD. Our findings reveal a substantial upregulation of these components in CVD-affected placentas, indicating a potential pathophysiological role of the NLRP3 inflammasome in the development of this condition. Subsequent investigations should focus on assessing translational interventions addressing this dysregulation in affected patient populations.


Asunto(s)
Inflamasomas , Proteína con Dominio Pirina 3 de la Familia NLR , Placenta , Adulto , Femenino , Humanos , Embarazo , Enfermedad Crónica , Inflamasomas/metabolismo , Interleucina-1beta/metabolismo , Interleucina-1beta/genética , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Placenta/metabolismo , Placenta/patología , Complicaciones Cardiovasculares del Embarazo/genética , Complicaciones Cardiovasculares del Embarazo/patología , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología
10.
Circ Res ; 128(12): 1958-1972, 2021 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-34110898

RESUMEN

Multifocal fibromuscular dysplasia (FMD) and spontaneous coronary artery dissection are both sex-biased diseases disproportionately affecting women over men in a 9:1 ratio. Traditionally known in the context of renovascular hypertension, recent advances in knowledge about FMD have demonstrated that FMD is a systemic arteriopathy presenting as arterial stenosis, aneurysm, and dissection in virtually any arterial bed. FMD is also characterized by major cardiovascular presentations including hypertension, stroke, and myocardial infarction. Similar to FMD, spontaneous coronary artery dissection is associated with a high prevalence of extracoronary vascular abnormalities, including FMD, aneurysm, and extracoronary dissection, and recent studies have also found genetic associations between the two diseases. This review will summarize the relationship between FMD and spontaneous coronary artery dissection with a focus on common clinical associations, histopathologic mechanisms, genetic susceptibilities, and the biology of these diseases. The current status of disease models and critical future research directions will also be addressed.


Asunto(s)
Anomalías de los Vasos Coronarios , Displasia Fibromuscular , Factores Sexuales , Enfermedades Vasculares/congénito , Aneurisma/etiología , Disección Aórtica/etiología , Angiografía , Constricción Patológica/etiología , Anomalías de los Vasos Coronarios/diagnóstico por imagen , Anomalías de los Vasos Coronarios/epidemiología , Anomalías de los Vasos Coronarios/genética , Anomalías de los Vasos Coronarios/patología , Femenino , Displasia Fibromuscular/diagnóstico por imagen , Displasia Fibromuscular/epidemiología , Displasia Fibromuscular/genética , Displasia Fibromuscular/patología , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Hipertensión/etiología , Masculino , Infarto del Miocardio/etiología , Accidente Cerebrovascular/etiología , Enfermedades Vasculares/diagnóstico por imagen , Enfermedades Vasculares/epidemiología , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología
11.
PLoS Genet ; 16(1): e1008538, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31917787

RESUMEN

Genome-wide association studies have identified multiple novel genomic loci associated with vascular diseases. Many of these loci are common non-coding variants that affect the expression of disease-relevant genes within coronary vascular cells. To identify such genes on a genome-wide level, we performed deep transcriptomic analysis of genotyped primary human coronary artery smooth muscle cells (HCASMCs) and coronary endothelial cells (HCAECs) from the same subjects, including splicing Quantitative Trait Loci (sQTL), allele-specific expression (ASE), and colocalization analyses. We identified sQTLs for TARS2, YAP1, CFDP1, and STAT6 in HCASMCs and HCAECs, and 233 ASE genes, a subset of which are also GTEx eGenes in arterial tissues. Colocalization of GWAS association signals for coronary artery disease (CAD), migraine, stroke and abdominal aortic aneurysm with GTEx eGenes in aorta, coronary artery and tibial artery discovered novel candidate risk genes for these diseases. At the CAD and stroke locus tagged by rs2107595 we demonstrate colocalization with expression of the proximal gene TWIST1. We show that disrupting the rs2107595 locus alters TWIST1 expression and that the risk allele has increased binding of the NOTCH signaling protein RBPJ. Finally, we provide data that TWIST1 expression influences vascular SMC phenotypes, including proliferation and calcification, as a potential mechanism supporting a role for TWIST1 in CAD.


Asunto(s)
Vasos Coronarios/metabolismo , Células Endoteliales/metabolismo , Miocitos del Músculo Liso/metabolismo , Proteínas Nucleares/genética , Proteína 1 Relacionada con Twist/genética , Enfermedades Vasculares/genética , Células Cultivadas , Vasos Coronarios/citología , Endotelio Vascular/citología , Endotelio Vascular/metabolismo , Humanos , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/metabolismo , Proteínas Nucleares/metabolismo , Polimorfismo de Nucleótido Simple , Unión Proteica , Transcriptoma , Proteína 1 Relacionada con Twist/metabolismo
12.
Curr Cardiol Rep ; 25(12): 1735-1743, 2023 12.
Artículo en Inglés | MEDLINE | ID: mdl-37979122

RESUMEN

PURPOSE OF REVIEW: Spontaneous coronary artery dissection (SCAD) is a significant cause of acute myocardial infarction that is increasingly recognized in young and middle-aged women. The etiology of SCAD is likely multifactorial and may include the interaction of environmental and individual factors. Here, we summarize the current understanding of the genetic factors contributing to the development of SCAD. RECENT FINDINGS: The molecular findings underlying SCAD have been demonstrated to include a combination of rare DNA sequence variants with large effects, common variants contributing to a complex genetic architecture, and variants with intermediate impact. The genes associated with SCAD highlight the role of arterial cells and their extracellular matrix in the pathogenesis of the disease and shed light on the relationship between SCAD and other disorders, including fibromuscular dysplasia and connective tissue diseases. While up to 10% of affected individuals may harbor a rare variant with large effect, SCAD most often presents as a complex genetic condition. Analyses of larger and more diverse cohorts will continue to improve our understanding of risk susceptibility loci and will also enable consideration of the clinical utility of genetic testing strategies in the management of SCAD.


Asunto(s)
Anomalías de los Vasos Coronarios , Infarto del Miocardio , Enfermedades Vasculares , Persona de Mediana Edad , Humanos , Femenino , Vasos Coronarios/patología , Enfermedades Vasculares/genética , Infarto del Miocardio/complicaciones , Anomalías de los Vasos Coronarios/genética , Angiografía Coronaria , Factores de Riesgo
13.
Am J Hum Genet ; 104(3): 503-519, 2019 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-30827500

RESUMEN

Although the use of model systems for studying the mechanism of mutations that have a large effect is common, we highlight here the ways that zebrafish-model-system studies of a gene, GRIK5, that contributes to the polygenic liability to develop eye diseases have helped to illuminate a mechanism that implicates vascular biology in eye disease. A gene-expression prediction derived from a reference transcriptome panel applied to BioVU, a large electronic health record (EHR)-linked biobank at Vanderbilt University Medical Center, implicated reduced GRIK5 expression in diverse eye diseases. We tested the function of GRIK5 by depletion of its ortholog in zebrafish, and we observed reduced blood vessel numbers and integrity in the eye and increased vascular permeability. Analyses of EHRs in >2.6 million Vanderbilt subjects revealed significant comorbidity of eye and vascular diseases (relative risks 2-15); this comorbidity was confirmed in 150 million individuals from a large insurance claims dataset. Subsequent studies in >60,000 genotyped BioVU participants confirmed the association of reduced genetically predicted expression of GRIK5 with comorbid vascular and eye diseases. Our studies pioneer an approach that allows a rapid iteration of the discovery of gene-phenotype relationships to the primary genetic mechanism contributing to the pathophysiology of human disease. Our findings also add dimension to the understanding of the biology driven by glutamate receptors such as GRIK5 (also referred to as GLUK5 in protein form) and to mechanisms contributing to human eye diseases.


Asunto(s)
Bancos de Muestras Biológicas , Registros Electrónicos de Salud , Embrión no Mamífero/patología , Oftalmopatías/patología , Regulación de la Expresión Génica , Receptores de Ácido Kaínico/genética , Enfermedades Vasculares/patología , Animales , Embrión no Mamífero/metabolismo , Oftalmopatías/genética , Oftalmopatías/metabolismo , Genotipo , Humanos , Fenómica , Fenotipo , Receptores de Ácido Kaínico/metabolismo , Enfermedades Vasculares/genética , Enfermedades Vasculares/metabolismo , Pez Cebra
14.
Am J Med Genet A ; 188(5): 1448-1456, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35092149

RESUMEN

Spontaneous coronary artery dissection (SCAD) is a potential precipitant of myocardial infarction and sudden death for which the etiology is poorly understood. Mendelian vascular and connective tissue disorders underlying thoracic aortic disease (TAD), have been reported in ~5% of individuals with SCAD. We therefore hypothesized that patients with TAD are at elevated risk for SCAD. We queried registries enrolling patients with TAD to define the incidence of SCAD. Of 7568 individuals enrolled, 11 (0.15%) were found to have SCAD. Of the sequenced cases (9/11), pathogenic variants were identified (N = 9), including COL3A1 (N = 3), FBN1 (N = 2), TGFBR2 (N = 2), TGFBR1 (N = 1), and PRKG1 (N = 1). Individuals with SCAD had an increased frequency of iliac artery dissection (25.0% vs. 5.1%, p = 0.047). The prevalence of SCAD among individuals with TAD is low. The identification of pathogenic variants in genes previously described in individuals with SCAD, particularly those underlying vascular Ehlers-Danlos, Marfan syndrome, and Loeys-Dietz syndrome, is consistent with prior reports from clinical SCAD series. Further research is needed to identify specific genetic influences on SCAD risk.


Asunto(s)
Anomalías de los Vasos Coronarios , Síndrome de Ehlers-Danlos , Síndrome de Loeys-Dietz , Enfermedades Vasculares , Anomalías de los Vasos Coronarios/epidemiología , Anomalías de los Vasos Coronarios/genética , Síndrome de Ehlers-Danlos/genética , Predisposición Genética a la Enfermedad , Humanos , Síndrome de Loeys-Dietz/complicaciones , Síndrome de Loeys-Dietz/epidemiología , Síndrome de Loeys-Dietz/genética , Factores de Riesgo , Enfermedades Vasculares/congénito , Enfermedades Vasculares/epidemiología , Enfermedades Vasculares/genética
15.
Circ Res ; 126(9): 1127-1145, 2020 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-32324505

RESUMEN

The advent of deep sequencing technologies led to the identification of a considerable amount of noncoding RNA transcripts, which are increasingly recognized for their functions in controlling cardiovascular diseases. MicroRNAs have already been studied for a decade, leading to the identification of several vasculoprotective and detrimental species, which might be considered for therapeutic targeting. Other noncoding RNAs such as circular RNAs, YRNAs, or long noncoding RNAs are currently gaining increasing attention, and first studies provide insights into their functions as mediators or antagonists of vascular diseases in vivo. The present review article will provide an overview of the different types of noncoding RNAs controlling the vasculature and focus on the developing field of long noncoding RNAs.


Asunto(s)
MicroARNs/genética , ARN Circular/genética , ARN Largo no Codificante/genética , Enfermedades Vasculares/genética , Animales , Regulación de la Expresión Génica , Humanos , MicroARNs/metabolismo , ARN Circular/metabolismo , ARN Largo no Codificante/metabolismo , Transducción de Señal , Enfermedades Vasculares/metabolismo , Enfermedades Vasculares/patología , Remodelación Vascular
16.
Proc Natl Acad Sci U S A ; 116(16): 7941-7950, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30944222

RESUMEN

Studies over the past decade have revealed a central role for innate immune sensors in autoimmune and autoinflammatory diseases. cGAS, a cytosolic DNA sensor, detects both foreign and host DNA and generates a second-messenger cGAMP, which in turn binds and activates stimulator of IFN genes (STING), leading to induction of type I interferons and inflammatory cytokines. Recently, gain-of-function mutations in STING have been identified in patients with STING-associated vasculopathy with onset in infancy (SAVI). SAVI patients present with early-onset systemic inflammation and interstitial lung disease, resulting in pulmonary fibrosis and respiratory failure. Here, we describe two independent SAVI mouse models, harboring the two most common mutations found in patients. A direct comparison of these strains reveals a hierarchy of immune abnormalities, lung inflammation and fibrosis, which do not depend on either IFN-α/ß receptor signaling or mixed lineage kinase domain-like pseudokinase (MLKL)-dependent necroptotic cell death pathways. Furthermore, radiation chimera experiments reveal how bone marrow from the V154M mutant mice transfer disease to the WT host, whereas the N153S does not, indicating mutation-specific disease outcomes. Moreover, using radiation chimeras we find that T cell lymphopenia depends on T cell-intrinsic expression of the SAVI mutation. Collectively, these mutant mice recapitulate many of the disease features seen in SAVI patients and highlight mutation-specific functions of STING that shed light on the heterogeneity observed in SAVI patients.


Asunto(s)
Modelos Animales de Enfermedad , Interferón Tipo I/metabolismo , Enfermedades Vasculares , Animales , Muerte Celular/inmunología , Citocinas/metabolismo , Mutación con Ganancia de Función , Inflamación/inmunología , Inflamación/fisiopatología , Ratones , Enfermedades Vasculares/genética , Enfermedades Vasculares/inmunología , Enfermedades Vasculares/fisiopatología
17.
Int J Neurosci ; 132(11): 1132-1136, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-33345688

RESUMEN

OBJECTIVE: This study investigated the relationship between the MTRA2576G polymorphism of the key enzyme in homocysteine metabolism and deep vein thrombosis (DVT), chronic venous insufficiency (CVI) and arteriosclerotic occlusion (ASO) of the lower extremities. METHODS: Genomic DNA was extracted from the peripheral blood of patients with lower-extremity vascular diseases, including 125 cases of DVT, 125 cases of CVI and 125 cases of ASO. DNA samples extracted from 197 healthy individuals were used as control samples. PCR-RFLP was used to investigate the polymorphisms of MTR in these subjects. RESULTS: The frequency of the G allele in MTR was 6.8%, 6.1% and 12.8% for the DVT group, CVI group and ASO group, respectively (p = 0.003). The frequency of the GG allele was 13.6%, 12.2% and 22.4% for the DVT group, CVI group and ASO group, respectively (p = 0.014). Only the allele frequency of GG in the ASO group was higher than that in the control group, and the disease risk was also 1.3 times higher than that in the control group (OR = 1.299, 95% CI = 1.025 ∼ 2.575). CONCLUSION: Patients with the G allele in MTR have a high risk for ASO, and the GG allele is a risk gene for ASO.


Asunto(s)
Enfermedades Vasculares , Humanos , Alelos , Genotipo , Estudios de Casos y Controles , Factores de Riesgo , Enfermedades Vasculares/genética , Extremidad Inferior , Homocisteína , 5-Metiltetrahidrofolato-Homocisteína S-Metiltransferasa/genética
18.
J Cell Physiol ; 236(4): 2333-2351, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-32875580

RESUMEN

Proprotein convertase subtilisin/kexin type-9 (PCSK9), a member of the proprotein convertase family, is an important drug target because of its crucial role in lipid metabolism. Emerging evidence suggests a direct role of localized PCSK9 in the pathogenesis of vascular diseases. With this in our consideration, we reviewed PCSK9 physiology with respect to recent development and major studies (clinical and experimental) on PCSK9 functionality in vascular disease. PCSK9 upregulates low-density lipoprotein (LDL)-cholesterol levels by binding to the LDL-receptor (LDLR) and facilitating its lysosomal degradation. PCSK9 gain-of-function mutations have been confirmed as a novel genetic mechanism for familial hypercholesterolemia. Elevated serum PCSK9 levels in patients with vascular diseases may contribute to coronary artery disease, atherosclerosis, cerebrovascular diseases, vasculitis, aortic diseases, and arterial aging pathogenesis. Experimental models of atherosclerosis, arterial aneurysm, and coronary or carotid artery ligation also support PCSK9 contribution to inflammatory response and disease progression, through LDLR-dependent or -independent mechanisms. More recently, several clinical trials have confirmed that anti-PCSK9 monoclonal antibodies can reduce systemic LDL levels, total nonfatal cardiovascular events, and all-cause mortality. Interaction of PCSK9 with other receptor proteins (LDLR-related proteins, cluster of differentiation family members, epithelial Na+ channels, and sortilin) may underlie its roles in vascular disease. Improved understanding of PCSK9 roles and molecular mechanisms in various vascular diseases will facilitate advances in lipid-lowering therapy and disease prevention.


Asunto(s)
Arterias/enzimología , Hipercolesterolemia/enzimología , Proproteína Convertasa 9/metabolismo , Enfermedades Vasculares/enzimología , Animales , Anticolesterolemiantes/uso terapéutico , Arterias/efectos de los fármacos , Arterias/patología , Regulación Enzimológica de la Expresión Génica , Humanos , Hipercolesterolemia/tratamiento farmacológico , Hipercolesterolemia/genética , Hipercolesterolemia/patología , Mutación , Inhibidores de PCSK9 , Proproteína Convertasa 9/genética , Inhibidores de Serina Proteinasa/uso terapéutico , Transducción de Señal , Enfermedades Vasculares/tratamiento farmacológico , Enfermedades Vasculares/genética , Enfermedades Vasculares/patología
19.
Hum Mol Genet ; 28(24): 4197-4207, 2019 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-31630189

RESUMEN

Raised albumin-creatinine ratio (ACR) is an indicator of microvascular damage and renal disease. We aimed to identify genetic variants associated with raised ACR and study the implications of carrying multiple ACR-raising alleles with metabolic and vascular-related disease. We performed a genome-wide association study of ACR using 437 027 individuals from the UK Biobank in the discovery phase, 54 527 more than previous studies, and followed up our findings in independent studies. We identified 62 independent associations with ACR across 56 loci (P < 5 × 10-8), of which 20 were not previously reported. Pathway analyses and the identification of 20 of the 62 variants (at r2 > 0.8) coinciding with signals for at least 16 related metabolic and vascular traits, suggested multiple pathways leading to raised ACR levels. After excluding variants at the CUBN locus, known to alter ACR via effects on renal absorption, an ACR genetic risk score was associated with a higher risk of hypertension, and less strongly, type 2 diabetes and stroke. For some rare genotype combinations at the CUBN locus, most individuals had ACR levels above the microalbuminuria clinical threshold. Contrary to our hypothesis, individuals carrying more CUBN ACR-raising alleles, and above the clinical threshold, had a higher frequency of vascular disease. The CUBN allele effects on ACR were twice as strong in people with diabetes-a result robust to an optimization-algorithm approach to simulating interactions, validating previously reported gene-diabetes interactions (P ≤ 4 × 10-5). In conclusion, a variety of genetic mechanisms and traits contribute to variation in ACR.


Asunto(s)
Albuminuria/genética , Creatinina/metabolismo , Enfermedades Renales/genética , Albúminas/metabolismo , Alelos , Creatinina/análisis , Diabetes Mellitus Tipo 2/genética , Femenino , Frecuencia de los Genes/genética , Predisposición Genética a la Enfermedad/genética , Estudio de Asociación del Genoma Completo , Genotipo , Humanos , Hipertensión/genética , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Reino Unido , Enfermedades Vasculares/genética , Enfermedades Vasculares/metabolismo
20.
J Hepatol ; 75(4): 924-934, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34052252

RESUMEN

BACKGROUND & AIMS: Porto-sinusoidal vascular disease (PSVD) is a rare vascular liver disease of unknown etiology that causes portal hypertension. It usually affects young individuals and shortens live expectancy. The deregulated pathways involved in PSVD development are unknown and therefore we lack curative treatments. The purpose of this study was to integrate transcriptomic and clinical data by comprehensive network-based modeling in order to uncover altered biological processes in patients with PSVD. METHODS: We obtained liver tissue samples from 20 consecutive patients with PSVD and 21 sex- and age-matched patients with cirrhosis and 13 histologically normal livers (HNL) (initial cohort) and performed transcriptomic analysis. Microarray data were analyzed using weighted gene correlation network analysis to identify clusters of highly correlated genes differently expressed in patients with PSVD. We next evaluated the molecular pathways enriched in patients with PSVD and the core-related genes from the most significantly enriched pathways in patients with PSVD. Our main findings were validated using RNA sequencing in a different cohort of PSVD, cirrhosis and HNL (n = 8 for each group). RESULTS: Patients with PSVD have a distinctive genetic profile enriched mainly in canonical pathways involving hemostasis and coagulation but also lipid metabolism and oxidative phosphorylation. Serpin family (SERPINC1), the apolipoproteins (APOA, APOB, APOC), ATP synthases (ATP5G1, ATP5B), fibrinogen genes (FGB, FGA) and alpha-2-macroglobulin were identified as highly connective genes that may have an important role in PSVD pathogenesis. CONCLUSION: PSVD has a unique transcriptomic profile and we have identified deregulation of pathways involved in vascular homeostasis as the main pathogenic event of disease development. LAY SUMMARY: Porto-sinusoidal vascular disease is a rare but life-shortening disease that affects mainly young people. Knowledge of the disrupted pathways involved in its development will help to identify novel therapeutic targets and new treatments. Using a systems biology approach, we identify that pathways regulating endothelial function and tone may act as drivers of porto-sinusoidal vascular disease.


Asunto(s)
Expresión Génica/genética , Redes Reguladoras de Genes/genética , Enfermedades Vasculares/genética , Adulto , Femenino , Expresión Génica/inmunología , Redes Reguladoras de Genes/inmunología , Humanos , Masculino , Persona de Mediana Edad , Enfermedades Vasculares/fisiopatología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA