Gene-mapping study of extremes of cerebral small vessel disease reveals TRIM47 as a strong candidate

Abstract Cerebral small vessel disease is a leading cause of stroke and a major contributor to cognitive decline and dementia, but our understanding of specific genes underlying the cause of sporadic cerebral small vessel disease is limited. We report a genome-wide association study and a whole-exome association study on a composite extreme phenotype of cerebral small vessel disease derived from its most common MRI features: white matter hyperintensities and lacunes. Seventeen population-based cohorts of older persons with MRI measurements and genome-wide genotyping (n = 41 326), whole-exome sequencing (n = 15 965), or exome chip (n = 5249) data contributed 13 776 and 7079 extreme small vessel disease samples for the genome-wide association study and whole-exome association study, respectively. The genome-wide association study identified significant association of common variants in 11 loci with extreme small vessel disease, of which the chr12q24.11 locus was not previously reported to be associated with any MRI marker of cerebral small vessel disease. The whole-exome association study identified significant associations of extreme small vessel disease with common variants in the 5′ UTR region of EFEMP1 (chr2p16.1) and one probably damaging common missense variant in TRIM47 (chr17q25.1). Mendelian randomization supports the causal association of extensive small vessel disease severity with increased risk of stroke and Alzheimer’s disease. Combined evidence from summary-based Mendelian randomization studies and profiling of human loss-of-function allele carriers showed an inverse relation between TRIM47 expression in the brain and blood vessels and extensive small vessel disease severity. We observed significant enrichment of Trim47 in isolated brain vessel preparations compared to total brain fraction in mice, in line with the literature showing Trim47 enrichment in brain endothelial cells at single cell level. Functional evaluation of TRIM47 by small interfering RNAs-mediated knockdown in human brain endothelial cells showed increased endothelial permeability, an important hallmark of cerebral small vessel disease pathology. Overall, our comprehensive gene-mapping study and preliminary functional evaluation suggests a putative role of TRIM47 in the pathophysiology of cerebral small vessel disease, making it an important candidate for extensive in vivo explorations and future translational work.

[1]  V. Salomaa,et al.  Genetic basis of lacunar stroke: a pooled analysis of individual patient data and genome-wide association studies , 2021, The Lancet Neurology.

[2]  N. Eriksson,et al.  Cerebral small vessel disease genomics and its implications across the lifespan , 2020, Nature Communications.

[3]  C. Lewis,et al.  Genome-wide association study of MRI markers of cerebral small vessel disease in 42,310 participants , 2020, Nature Communications.

[4]  Ryan L. Collins,et al.  The mutational constraint spectrum quantified from variation in 141,456 humans , 2020, Nature.

[5]  S. Robertson,et al.  Biallelic variants in EFEMP1 in a man with a pronounced connective tissue phenotype , 2019, European Journal of Human Genetics.

[6]  Selective RNA interference and gene silencing using reactive oxygen species-responsive lipid nanoparticles. , 2019, Chemical communications.

[7]  M. Dichgans,et al.  Small vessel disease: mechanisms and clinical implications , 2019, The Lancet Neurology.

[8]  Tanya M. Teslovich,et al.  Exome sequencing of 20,791 cases of type 2 diabetes and 24,440 controls , 2019, Nature.

[9]  Richard Frayne,et al.  Quantifying blood-brain barrier leakage in small vessel disease: Review and consensus recommendations , 2019, Alzheimer's & Dementia.

[10]  L. Beckett,et al.  White matter hyperintensities in vascular contributions to cognitive impairment and dementia (VCID): Knowledge gaps and opportunities , 2019, Alzheimer's & dementia.

[11]  M. Fornage,et al.  Association of variants in HTRA1 and NOTCH3 with MRI-defined extremes of cerebral small vessel disease in older subjects , 2019, Brain : a journal of neurology.

[12]  C. Sudlow,et al.  Genetic variation in PLEKHG1 is associated with white matter hyperintensities (n = 11,226) , 2019, Neurology.

[13]  C. Sudlow,et al.  Genetic and lifestyle risk factors for MRI-defined brain infarcts in a population-based setting , 2019, Neurology.

[14]  P. Navas,et al.  The mitochondrial phosphatase PPTC7 orchestrates mitochondrial metabolism regulating coenzyme Q10 biosynthesis. , 2018, Biochimica et biophysica acta. Bioenergetics.

[15]  Yeting Zhang,et al.  Functional equivalence of genome sequencing analysis pipelines enables harmonized variant calling across human genetics projects , 2018, Nature Communications.

[16]  Koji Ando,et al.  A molecular atlas of cell types and zonation in the brain vasculature , 2018, Nature.

[17]  Bernard Mazoyer,et al.  Burden of Dilated Perivascular Spaces, an Emerging Marker of Cerebral Small Vessel Disease, Is Highly Heritable , 2018, Stroke.

[18]  Andrew D. Johnson,et al.  Multiancestry genome-wide association study of 520,000 subjects identifies 32 loci associated with stroke and stroke subtypes , 2018, Nature Genetics.

[19]  Feng Li,et al.  The deubiquitinating enzyme cylindromatosis mitigates nonalcoholic steatohepatitis , 2018, Nature Medicine.

[20]  N. Plesnila,et al.  Cylindromatosis mediates neuronal cell death in vitro and in vivo , 2018, Cell Death & Differentiation.

[21]  Evan Z. Macosko,et al.  Heritability enrichment of specifically expressed genes identifies disease-relevant tissues and cell types , 2017, Nature Genetics.

[22]  A. Mishra,et al.  A Novel Approach for Pathway Analysis of GWAS Data Highlights Role of BMP Signaling and Muscle Cell Differentiation in Colorectal Cancer Susceptibility , 2017, Twin Research and Human Genetics.

[23]  J. van der Grond,et al.  Archetypal NOTCH3 mutations frequent in public exome: implications for CADASIL , 2016, Annals of clinical and translational neurology.

[24]  P. Visscher,et al.  Integration of summary data from GWAS and eQTL studies predicts complex trait gene targets , 2016, Nature Genetics.

[25]  Anders Albrechtsen,et al.  Weighting sequence variants based on their annotation increases power of whole-genome association studies , 2016, Nature Genetics.

[26]  Xiaowei Zhan,et al.  RVTESTS: an efficient and comprehensive tool for rare variant association analysis using sequence data , 2016, Bioinform..

[27]  Kaitlin M. Fitzpatrick,et al.  Genome-wide meta-analysis of cerebral white matter hyperintensities in patients with stroke , 2016, Neurology.

[28]  M. Cohen-Salmon,et al.  Purification of Mouse Brain Vessels. , 2015, Journal of visualized experiments : JoVE.

[29]  Yakir A Reshef,et al.  Partitioning heritability by functional annotation using genome-wide association summary statistics , 2015, Nature Genetics.

[30]  Lorna M. Lopez,et al.  Multiethnic Genome-Wide Association Study of Cerebral White Matter Hyperintensities on MRI , 2015, Circulation. Cardiovascular genetics.

[31]  M. Daly,et al.  LD Score regression distinguishes confounding from polygenicity in genome-wide association studies , 2014, Nature Genetics.

[32]  S. MacGregor,et al.  VEGAS2: Software for More Flexible Gene-Based Testing , 2014, Twin Research and Human Genetics.

[33]  Zoltán Kutalik,et al.  Quality control and conduct of genome-wide association meta-analyses , 2014, Nature Protocols.

[34]  Lisa J. Martin,et al.  Meta-analysis of genome-wide association studies identifies 1q22 as a susceptibility locus for intracerebral hemorrhage. , 2014, American journal of human genetics.

[35]  Nick C Fox,et al.  Neuroimaging standards for research into small vessel disease and its contribution to ageing and neurodegeneration , 2013, The Lancet Neurology.

[36]  Xihong Lin,et al.  Optimal tests for rare variant effects in sequencing association studies. , 2012, Biostatistics.

[37]  P. Visscher,et al.  Conditional and joint multiple-SNP analysis of GWAS summary statistics identifies additional variants influencing complex traits , 2012, Nature Genetics.

[38]  Boer,et al.  Genome-wide association studies of cerebral white matter lesion burden: the CHARGE Consortium , 2011 .

[39]  Yun Li,et al.  METAL: fast and efficient meta-analysis of genomewide association scans , 2010, Bioinform..

[40]  P. Bork,et al.  A method and server for predicting damaging missense mutations , 2010, Nature Methods.

[41]  M. Fornage,et al.  Genome-Wide Association Studies of MRI-Defined Brain Infarcts: Meta-Analysis From the CHARGE Consortium , 2010, Stroke.

[42]  Renata C. Geer,et al.  The NCBI BioSystems database , 2009, Nucleic Acids Res..

[43]  Z. Wszolek,et al.  VASCULAR RISK FACTORS AND DEMENTIA: HOW TO MOVE FORWARD? , 2009, Neurology.

[44]  K. Lunetta,et al.  Methods in Genetics and Clinical Interpretation Cohorts for Heart and Aging Research in Genomic Epidemiology (CHARGE) Consortium Design of Prospective Meta-Analyses of Genome-Wide Association Studies From 5 Cohorts , 2010 .

[45]  Takako Sasaki,et al.  Lack of fibulin-3 causes early aging and herniation, but not macular degeneration in mice. , 2007, Human molecular genetics.

[46]  S. Greenberg,et al.  Small vessels, big problems. , 2006, The New England journal of medicine.

[47]  Charles DeCarli,et al.  Genetic Variation in White Matter Hyperintensity Volume in the Framingham Study , 2004, Stroke.

[48]  R. Holman,et al.  Vascular Factors and Risk of Dementia: Design of the Three-City Study and Baseline Characteristics of the Study Population , 2003, Neuroepidemiology.

[49]  A. Rao Faculty Opinions recommendation of CYLD is a deubiquitinating enzyme that negatively regulates NF-kappaB activation by TNFR family members. , 2003 .

[50]  G. Courtois,et al.  The tumour suppressor CYLD negatively regulates NF-κB signalling by deubiquitination , 2003, Nature.

[51]  S. Ebrahim,et al.  'Mendelian randomization': can genetic epidemiology contribute to understanding environmental determinants of disease? , 2003, International journal of epidemiology.

[52]  F. Baas,et al.  GOA, a novel gene encoding a ring finger B-box coiled-coil protein, is overexpressed in astrocytoma. , 2001, Biochemical and biophysical research communications.

[53]  K. Luo,et al.  Negative feedback regulation of TGF-beta signaling by the SnoN oncoprotein. , 1999, Science.