Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 77
Filter
1.
J Cardiovasc Med (Hagerstown) ; 25(8): 569-586, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38916232

ABSTRACT

BACKGROUND: Spontaneous coronary artery dissection (SCAD) is a multifactorial process that involves predisposing factors and precipitating stressors. Genetic abnormality has been implicated to play a mechanistic role in the development of SCAD. This systematic review aims to summarize the current evidence concerning the link between SCAD and genetic abnormalities. METHODS: We reviewed original studies published until May 2023 that reported SCAD patients with a genetic mutation by searching PubMed, Embase Ovid, and Google Scholar. Registries, cohort studies, and case reports were included if a definitive SCAD diagnosis was reported, and the genetic analysis was performed. Exclusion criteria included editorials, reviews, letters or commentaries, animal studies, meeting papers, and studies from which we were unable to extract data. Data were extracted from published reports. RESULTS: A total of 595 studies were screened and 55 studies were identified. Among 116 SCAD patients with genetic abnormalities, 20% had mutations in the COL gene, 13.70% TLN1 gene, and 8.42% TSR1 gene. Mutations affecting the genes encoding COL and TLN1 were most frequently reported (20 and 13.7%, respectively). Interestingly, 15 genes of this collection were also reported in patients with thoracic aortic diseases as well. The genetic commonality between fibromuscular dysplasia (FMD) and SCAD was also included. CONCLUSION: In this review, the inherited conditions and reported genes of undetermined significance from case reports associated with SCAD are collected. A brief description of the encoded protein and the clinical features associated with pathologic genes is provided. Current data suggested that the diagnostic yield of genetic studies for patients with SCAD would be low and routine genetic screening of such patients with no clinical features indicative of associated disorders remains debatable. This review can be used as a guide for clinicians to recognize inherited syndromic and nonsyndromic disorders associated with SCAD.


Subject(s)
Coronary Vessel Anomalies , Genetic Predisposition to Disease , Mutation , Vascular Diseases , Humans , Coronary Vessel Anomalies/genetics , Coronary Vessel Anomalies/diagnosis , Vascular Diseases/genetics , Vascular Diseases/congenital , Vascular Diseases/diagnosis , Risk Factors , Phenotype , Female , Fibromuscular Dysplasia/genetics , Fibromuscular Dysplasia/diagnosis , Fibromuscular Dysplasia/complications , Male
2.
Stroke ; 55(6): 1699-1706, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38690664

ABSTRACT

Systemic vasculopathy has occasionally been reported in cases of moyamoya disease (MMD). Since the pathological relationship between moyamoya vasculopathy (MMV) and moyamoya-related systemic vasculopathy (MMRSV) remains unclear, it was examined herein by a review of histopathologic studies in consideration of clinicopathological and genetic viewpoints. Although luminal stenosis was a common finding in MMV and MMRSV, histopathologic findings of vascular remodeling markedly differed. MMV showed intimal hyperplasia, marked medial atrophy, and redundant tortuosity of the internal elastic lamina, with outer diameter narrowing called negative remodeling. MMRSV showed hyperplasia, mainly in the intima and sometimes in the media, with disrupted stratification of the internal elastic lamina. Systemic vasculopathy has also been observed in patients with non-MMD carrying the RNF213 (ring finger protein 213) mutation, leading to the concept of RNF213 vasculopathy. RNF213 vasculopathy in patients with non-MMD was histopathologically similar to MMRSV. Cases of MMRSV have sometimes been diagnosed with fibromuscular dysplasia. Fibromuscular dysplasia is similar to MMD not only in the histopathologic findings of MMRSV but also from clinicopathological and genetic viewpoints. The significant histopathologic difference between MMV and MMRSV may be attributed to a difference in the original vascular wall structure and its resistance to pathological stress between the intracranial and systemic arteries. To understand the pathogeneses of MMD and MMRSV, a broader perspective that includes RNF213 vasculopathy and fibromuscular dysplasia as well as an examination of the 2- or multiple-hit theory consisting of genetic factors, vascular structural conditions, and vascular environmental factors, such as blood immune cells and hemodynamics, are needed.


Subject(s)
Moyamoya Disease , Ubiquitin-Protein Ligases , Moyamoya Disease/genetics , Moyamoya Disease/pathology , Humans , Ubiquitin-Protein Ligases/genetics , Adenosine Triphosphatases/genetics , Mutation , Fibromuscular Dysplasia/genetics , Fibromuscular Dysplasia/pathology , Fibromuscular Dysplasia/complications
6.
Circ Genom Precis Med ; 15(6): e003496, 2022 12.
Article in English | MEDLINE | ID: mdl-36374587

ABSTRACT

BACKGROUND: The risk of arterial diseases may be elevated among family members of individuals having multifocal fibromuscular dysplasia (FMD). We sought to investigate the risk of arterial diseases in families of individuals with FMD. METHODS: Family histories for 73 probands with FMD were obtained, which included an analysis of 463 total first-degree relatives focusing on FMD and related arterial disorders. A polygenic risk score for FMD (PRSFMD) was constructed from prior genome-wide association findings of 584 FMD cases and 7139 controls and evaluated for association with an abdominal aortic aneurysm (AAA) in a cohort of 9693 AAA cases and 294 049 controls. A previously published PRSAAA was also assessed among the FMD cases and controls. RESULTS: Of all first degree relatives of probands, 9.3% were diagnosed with FMD, aneurysms, and dissections. Aneurysmal disease occurred in 60.5% of affected relatives and 5.6% of all relatives. Among 227 female first-degree relatives of probands, 4.8% (11) had FMD, representing a relative risk (RR)FMD of 1.5 ([95% CI, 0.75-2.8]; P=0.19) compared with the estimated population prevalence of 3.3%, though not of statistical significance. Of all fathers of FMD probands, 11% had AAAs resulting in a RRAAA of 2.3 ([95% CI, 1.12-4.6]; P=0.014) compared with population estimates. The PRSFMD was found to be associated with an AAA (odds ratio, 1.03 [95% CI, 1.01-1.05]; P=2.6×10-3), and the PRSAAA was found to be associated with FMD (odds ratio, 1.53 [95% CI, 1.2-1.9]; P=9.0×10-5) as well. CONCLUSIONS: FMD and AAAs seem to be sex-dimorphic manifestations of a heritable arterial disease with a partially shared complex genetic architecture. Excess risk of having an AAA according to a family history of FMD may justify screening in family members of individuals having FMD.


Subject(s)
Aortic Aneurysm, Abdominal , Fibromuscular Dysplasia , Male , Humans , Female , Fibromuscular Dysplasia/epidemiology , Fibromuscular Dysplasia/genetics , Fibromuscular Dysplasia/complications , Genome-Wide Association Study , Aortic Aneurysm, Abdominal/epidemiology , Aortic Aneurysm, Abdominal/genetics , Arteries , Risk Factors
8.
Clin Sci (Lond) ; 136(16): 1241-1255, 2022 08 31.
Article in English | MEDLINE | ID: mdl-36043395

ABSTRACT

Artery stenosis is a common cause of hypertension and stroke and can be due to atherosclerosis accumulation in the majority of cases and in a small fraction of patients to arterial fibromuscular dysplasia (FMD). Artery stenosis due to atherosclerosis is widely studied with known risk factors (e.g. increasing age, male gender, and dyslipidemia) to influence its etiology, including genetic factors. However, the causes of noninflammatory and nonatherosclerotic stenosis in FMD are less understood. FMD occurs predominantly in early middle-age women, a fraction of the population where cardiovascular risk is different and understudied. FMD arteriopathies are often diagnosed in the context of hypertension and stroke and co-occur mainly with spontaneous coronary artery dissection, an atypical cause of acute myocardial infarction. In this review, we provide a comprehensive overview of the recent advances in the understanding of molecular origins of FMD. Data were obtained from genetic studies using complementary methodological approaches applied to familial, syndromic, and sporadic forms of this intriguing arteriopathy. Rare variation analyses point toward mechanisms related to impaired prostacyclin signaling and defaults in fibrillar collagens. The study of common variation, mainly through a recent genome-wide association study, describes a shared genetic link with blood pressure, in addition to point at potential risk genes involved in actin cytoskeleton and intracellular calcium homeostasis supporting impaired vascular contraction as a key mechanism. We conclude this review with future strategies and approaches needed to fully understand the genetic and molecular mechanisms related to FMD.


Subject(s)
Atherosclerosis , Cardiovascular Diseases , Fibromuscular Dysplasia , Hypertension , Stroke , Atherosclerosis/complications , Cardiovascular Diseases/complications , Cardiovascular Diseases/genetics , Constriction, Pathologic/complications , Female , Fibromuscular Dysplasia/complications , Fibromuscular Dysplasia/diagnosis , Fibromuscular Dysplasia/genetics , Genome-Wide Association Study , Humans , Hypertension/complications , Male , Middle Aged , Risk Factors , Stroke/complications
10.
Arterioscler Thromb Vasc Biol ; 42(5): 597-609, 2022 05.
Article in English | MEDLINE | ID: mdl-35387477

ABSTRACT

BACKGROUND: Genome-wide association studies have revealed robust associations of common genetic polymorphisms in an intron of the PHACTR-1 (phosphatase and actin regulator 1) gene (chr6p24), with cervical artery dissection, spontaneous coronary artery dissection, and fibromuscular dysplasia. The aim was to assess its role in the pathogenesis of cervical artery dissection or fibromuscular dysplasia. METHODS: Using various tissue-specific Cre-driver mouse lines, Phactr1 was deleted either in endothelial cells using 2 tissue-specific Cre-driver (PDGFB [platelet-derived growth factor B]-CreERT2 mice and Tie2 [tyrosine kinase with immunoglobulin and EGF homology domains]-Cre) and smooth muscle cells (smooth muscle actin-CreERT2) with a third tissue-specific Cre-driver. RESULTS: To test the efficacy of the Phactr1 deletion after cre-induction, we confirmed first, a decrease in Phactr1 transcription and Phactr1 expression in endothelial cell and smooth muscle cell isolated from Phactr1iPDGFB and Phactr1iSMA mice. Irrespective to the tissue or the duration of the deletion, mice did not spontaneously display pathological phenotype or vascular impairment: mouse survival, growth, blood pressure, large vessel morphology, or actin organization were not different in knockout mice than their comparatives littermates. Challenging vascular function and repair either by angiotensin II-induced hypertension or limb ischemia did not lead to vascular morphology or function impairment in Phactr1-deleted mice. Similarly, there were no more consequences of Phactr1 deletion during embryogenesis in endothelial cells. CONCLUSIONS: Loss of PHACTR-1 function in the cells involved in vascular physiology does not appear to induce a pathological vascular phenotype. The in vivo effect of the intronic variation described in genome-wide association studies is unlikely to involve downregulation in PHACTR-1 expression.


Subject(s)
Actins , Arterial Occlusive Diseases/metabolism , Fibromuscular Dysplasia , Microfilament Proteins/metabolism , Actins/metabolism , Animals , Endothelial Cells/metabolism , Fibromuscular Dysplasia/genetics , Genome-Wide Association Study , Mice , Mice, Knockout , Mice, Transgenic , Microfilament Proteins/genetics , Myocytes, Smooth Muscle/metabolism , Phosphoric Monoester Hydrolases/metabolism
12.
Cardiovasc Res ; 118(1): 65-83, 2022 01 07.
Article in English | MEDLINE | ID: mdl-33739371

ABSTRACT

Fibromuscular dysplasia (FMD) is a non-atherosclerotic vascular disease that may involve medium-sized muscular arteries throughout the body. The majority of FMD patients are women. Although a variety of genetic, mechanical, and hormonal factors play a role in the pathogenesis of FMD, overall, its cause remains poorly understood. It is probable that the pathogenesis of FMD is linked to a combination of genetic and environmental factors. Extensive studies have correlated the arterial lesions of FMD to histopathological findings of arterial fibrosis, cellular hyperplasia, and distortion of the abnormal architecture of the arterial wall. More recently, the vascular phenotype of lesions associated with FMD has been expanded to include arterial aneurysms, dissections, and tortuosity. However, in the absence of a string-of-beads or focal stenosis, these lesions do not suffice to establish the diagnosis. While FMD most commonly involves renal and cerebrovascular arteries, involvement of most arteries throughout the body has been reported. Increasing evidence highlights that FMD is a systemic arterial disease and that subclinical alterations can be found in non-affected arterial segments. Recent significant progress in FMD-related research has led to improve our understanding of the disease's clinical manifestations, natural history, epidemiology, and genetics. Ongoing work continues to focus on FMD genetics and proteomics, physiological effects of FMD on cardiovascular structure and function, and novel imaging modalities and blood-based biomarkers that can be used to identify subclinical FMD. It is also hoped that the next decade will bring the development of multi-centred and potentially international clinical trials to provide comparative effectiveness data to inform the optimal management of patients with FMD.


Subject(s)
Arteries , Biomedical Research/trends , Fibromuscular Dysplasia , Molecular Diagnostic Techniques/trends , Animals , Arteries/metabolism , Arteries/pathology , Arteries/physiopathology , Fibromuscular Dysplasia/diagnosis , Fibromuscular Dysplasia/genetics , Fibromuscular Dysplasia/metabolism , Fibromuscular Dysplasia/physiopathology , Gene Expression Profiling/trends , Genetic Predisposition to Disease , Hemodynamics , Humans , Phenotype , Predictive Value of Tests , Prognosis , Proteomics/trends , Risk Assessment , Risk Factors , Vascular Remodeling
13.
Nat Commun ; 12(1): 6031, 2021 10 15.
Article in English | MEDLINE | ID: mdl-34654805

ABSTRACT

Fibromuscular dysplasia (FMD) is an arteriopathy associated with hypertension, stroke and myocardial infarction, affecting mostly women. We report results from the first genome-wide association meta-analysis of six studies including 1556 FMD cases and 7100 controls. We find an estimate of SNP-based heritability compatible with FMD having a polygenic basis, and report four robustly associated loci (PHACTR1, LRP1, ATP2B1, and LIMA1). Transcriptome-wide association analysis in arteries identifies one additional locus (SLC24A3). We characterize open chromatin in arterial primary cells and find that FMD associated variants are located in arterial-specific regulatory elements. Target genes are broadly involved in mechanisms related to actin cytoskeleton and intracellular calcium homeostasis, central to vascular contraction. We find significant genetic overlap between FMD and more common cardiovascular diseases and traits including blood pressure, migraine, intracranial aneurysm, and coronary artery disease.


Subject(s)
Cardiovascular Diseases/complications , Cardiovascular Diseases/genetics , Fibromuscular Dysplasia/complications , Fibromuscular Dysplasia/genetics , Genome-Wide Association Study , Adult , Arteries , Cytoskeletal Proteins/genetics , Female , Fibroblasts , Gene Expression Regulation , Humans , Intracranial Aneurysm , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Male , Microfilament Proteins/genetics , Middle Aged , Plasma Membrane Calcium-Transporting ATPases/genetics , Sodium-Calcium Exchanger/genetics , Transcriptome
14.
Circ Res ; 128(12): 1958-1972, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34110898

ABSTRACT

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.


Subject(s)
Coronary Vessel Anomalies , Fibromuscular Dysplasia , Sex Factors , Vascular Diseases/congenital , Aneurysm/etiology , Aortic Dissection/etiology , Angiography , Constriction, Pathologic/etiology , Coronary Vessel Anomalies/diagnostic imaging , Coronary Vessel Anomalies/epidemiology , Coronary Vessel Anomalies/genetics , Coronary Vessel Anomalies/pathology , Female , Fibromuscular Dysplasia/diagnostic imaging , Fibromuscular Dysplasia/epidemiology , Fibromuscular Dysplasia/genetics , Fibromuscular Dysplasia/pathology , Genetic Predisposition to Disease , Genetic Variation , Humans , Hypertension/etiology , Male , Myocardial Infarction/etiology , Stroke/etiology , Vascular Diseases/diagnostic imaging , Vascular Diseases/epidemiology , Vascular Diseases/genetics , Vascular Diseases/pathology
16.
EuroIntervention ; 17(6): 508-515, 2021 Aug 27.
Article in English | MEDLINE | ID: mdl-33319763

ABSTRACT

BACKGROUND: Spontaneous coronary artery dissection (SCAD) is an increasingly reported but poorly understood condition. Few European data are available. AIMS: The aims of this study were to obtain European data on SCAD, determine the prevalence of fibromuscular dysplasia (FMD) and enable genetic analyses in this population. METHODS: Data from a national French registry of SCAD cases were analysed prospectively and retrospectively. Clinical and angiographic data and management strategy were collected. Major adverse cardiovascular events (MACE) were analysed after one year of follow-up. Subjects were screened for FMD and blood was collected for DNA extraction. RESULTS: From June 2016 to August 2018, 373 SCAD cases were confirmed by the core lab. Mean age was 51.5 years. Patients were mostly women (90.6%) and 54.7% of cases had less than two cardiovascular risk factors. At one year, 295 patients (79.1%) were treated conservatively, the MACE rate was 12.3%, and there were no cases of mortality. The recurrence rate of SCAD was 3.3%. FMD was found at ≥1 arterial site in 45.0% of cases. We also confirmed the genetic association between the PHACTR1 locus and SCAD (odds ratio=1.66, p=7.08×10-8). CONCLUSIONS: Here we describe the DISCO registry, the largest European SCAD cohort where FMD was found in 45% of cases and the genetic association with PHACTR1 was confirmed. This nationwide cohort is a valuable resource for future clinical and genetic investigation to understand SCAD aetiology.


Subject(s)
Coronary Vessel Anomalies , Fibromuscular Dysplasia , Vascular Diseases , Coronary Angiography , Coronary Vessel Anomalies/diagnostic imaging , Coronary Vessel Anomalies/epidemiology , Coronary Vessel Anomalies/genetics , Coronary Vessels , Dissection , Female , Fibromuscular Dysplasia/epidemiology , Fibromuscular Dysplasia/genetics , Humans , Middle Aged , Prevalence , Registries , Retrospective Studies , Vascular Diseases/diagnostic imaging , Vascular Diseases/epidemiology , Vascular Diseases/genetics
17.
Cardiovasc Res ; 117(4): 1154-1165, 2021 03 21.
Article in English | MEDLINE | ID: mdl-32531060

ABSTRACT

AIMS: Fibromuscular dysplasia (FMD) and spontaneous coronary artery dissection (SCAD) are related, non-atherosclerotic arterial diseases mainly affecting middle-aged women. Little is known about their physiopathological mechanisms. We aimed to identify rare genetic causes to elucidate molecular mechanisms implicated in FMD and SCAD. METHODS AND RESULTS: We analysed 29 exomes that included familial and sporadic FMD. We identified one rare loss-of-function variant (LoF) (frequencygnomAD = 0.000075) shared by two FMD sisters in the prostaglandin I2 receptor gene (PTGIR), a key player in vascular remodelling. Follow-up was conducted by targeted or Sanger sequencing (1071 FMD and 363 SCAD patients) or lookups in exome (264 FMD) or genome sequences (480 SCAD), all independent and unrelated. It revealed four additional LoF allele carriers, in addition to several rare missense variants, among FMD patients, and two LoF allele carriers among SCAD patients, including one carrying a rare splicing mutation (c.768 + 1C>G). We used burden test to test for enrichment in patients compared to gnomAD controls, which detected a putative enrichment in FMD (PTRAPD = 8 × 10-4), but not a significant enrichment (PTRAPD = 0.12) in SCAD. The biological effects of variants on human prostaclycin receptor (hIP) signalling and protein expression were characterized using transient overexpression in human cells. We confirmed the LoFs (Q163X and P17RfsX6) and one missense (L67P), identified in one FMD and one SCAD patient, to severely impair hIP function in vitro. CONCLUSIONS: Our study shows that rare genetic mutations in PTGIR are enriched among FMD patients and found in SCAD patients, suggesting a role for prostacyclin signalling in non-atherosclerotic stenosis and dissection.


Subject(s)
Coronary Vessel Anomalies/genetics , Fibromuscular Dysplasia/genetics , Loss of Function Mutation , Mutation, Missense , Receptors, Epoprostenol/genetics , Vascular Diseases/congenital , Adult , Aged , Australia , Coronary Vessel Anomalies/diagnosis , Coronary Vessel Anomalies/metabolism , DNA Mutational Analysis , Databases, Genetic , Europe , Female , Fibromuscular Dysplasia/diagnosis , Fibromuscular Dysplasia/metabolism , Genetic Predisposition to Disease , HEK293 Cells , Humans , Male , Middle Aged , Phenotype , Predictive Value of Tests , Receptors, Epoprostenol/metabolism , Risk Assessment , Risk Factors , United States , Vascular Diseases/diagnosis , Vascular Diseases/genetics , Vascular Diseases/metabolism
19.
Arterioscler Thromb Vasc Biol ; 40(11): 2686-2699, 2020 11.
Article in English | MEDLINE | ID: mdl-32938213

ABSTRACT

OBJECTIVE: While rare variants in the COL5A1 gene have been associated with classical Ehlers-Danlos syndrome and rarely with arterial dissections, recurrent variants in COL5A1 underlying a systemic arteriopathy have not been described. Monogenic forms of multifocal fibromuscular dysplasia (mFMD) have not been previously defined. Approach and Results: We studied 4 independent probands with the COL5A1 pathogenic variant c.1540G>A, p.(Gly514Ser) who presented with arterial aneurysms, dissections, tortuosity, and mFMD affecting multiple arteries. Arterial medial fibroplasia and smooth muscle cell disorganization were confirmed histologically. The COL5A1 c.1540G>A variant is predicted to be pathogenic in silico and absent in gnomAD. The c.1540G>A variant is on a shared 160.1 kb haplotype with 0.4% frequency in Europeans. Furthermore, exome sequencing data from a cohort of 264 individuals with mFMD were examined for COL5A1 variants. In this mFMD cohort, COL5A1 c.1540G>A and 6 additional relatively rare COL5A1 variants predicted to be deleterious in silico were identified and were associated with arterial dissections (P=0.005). CONCLUSIONS: COL5A1 c.1540G>A is the first recurring variant recognized to be associated with arterial dissections and mFMD. This variant presents with a phenotype reminiscent of vascular Ehlers-Danlos syndrome. A shared haplotype among probands supports the existence of a common founder. Relatively rare COL5A1 genetic variants predicted to be deleterious by in silico analysis were identified in ≈2.7% of mFMD cases, and as they were enriched in patients with arterial dissections, may act as disease modifiers. Molecular testing for COL5A1 should be considered in patients with a phenotype overlapping with vascular Ehlers-Danlos syndrome and mFMD.


Subject(s)
Aortic Dissection/genetics , Arteries/pathology , Collagen Type V/genetics , Ehlers-Danlos Syndrome/genetics , Fibromuscular Dysplasia/genetics , Polymorphism, Single Nucleotide , Adult , Aortic Dissection/diagnostic imaging , Aortic Dissection/pathology , Arteries/diagnostic imaging , Ehlers-Danlos Syndrome/diagnostic imaging , Ehlers-Danlos Syndrome/pathology , Female , Fibromuscular Dysplasia/diagnostic imaging , Fibromuscular Dysplasia/pathology , Genetic Predisposition to Disease , Haplotypes , Humans , Male , Middle Aged , Phenotype , Young Adult
20.
Nat Commun ; 11(1): 4432, 2020 09 04.
Article in English | MEDLINE | ID: mdl-32887874

ABSTRACT

Spontaneous coronary artery dissection (SCAD) is a non-atherosclerotic cause of myocardial infarction (MI), typically in young women. We undertook a genome-wide association study of SCAD (Ncases = 270/Ncontrols = 5,263) and identified and replicated an association of rs12740679 at chromosome 1q21.2 (Pdiscovery+replication = 2.19 × 10-12, OR = 1.8) influencing ADAMTSL4 expression. Meta-analysis of discovery and replication samples identified associations with P < 5 × 10-8 at chromosome 6p24.1 in PHACTR1, chromosome 12q13.3 in LRP1, and in females-only, at chromosome 21q22.11 near LINC00310. A polygenic risk score for SCAD was associated with (1) higher risk of SCAD in individuals with fibromuscular dysplasia (P = 0.021, OR = 1.82 [95% CI: 1.09-3.02]) and (2) lower risk of atherosclerotic coronary artery disease and MI in the UK Biobank (P = 1.28 × 10-17, HR = 0.91 [95% CI :0.89-0.93], for MI) and Million Veteran Program (P = 9.33 × 10-36, OR = 0.95 [95% CI: 0.94-0.96], for CAD; P = 3.35 × 10-6, OR = 0.96 [95% CI: 0.95-0.98] for MI). Here we report that SCAD-related MI and atherosclerotic MI exist at opposite ends of a genetic risk spectrum, inciting MI with disparate underlying vascular biology.


Subject(s)
Coronary Vessel Anomalies/genetics , Genes, Neoplasm , Myocardial Infarction/genetics , Vascular Diseases/congenital , ADAMTS Proteins/genetics , Carotid Artery Diseases/complications , Carotid Artery Diseases/genetics , Chromosomes/genetics , Cohort Studies , Coronary Artery Disease/genetics , Female , Fibromuscular Dysplasia/complications , Fibromuscular Dysplasia/genetics , Genome-Wide Association Study , Humans , Low Density Lipoprotein Receptor-Related Protein-1/genetics , Male , Meta-Analysis as Topic , Microfilament Proteins/genetics , Risk Factors , Vascular Diseases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...