Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
Nat Med ; 29(9): 2224-2232, 2023 09.
Article in English | MEDLINE | ID: mdl-37653343

ABSTRACT

Most complex human traits differ by sex, but we have limited insight into the underlying mechanisms. Here, we investigated the influence of biological sex on protein expression and its genetic regulation in 1,277 human brain proteomes. We found that 13.2% (1,354) of brain proteins had sex-differentiated abundance and 1.5% (150) of proteins had sex-biased protein quantitative trait loci (sb-pQTLs). Among genes with sex-biased expression, we found 67% concordance between sex-differentiated protein and transcript levels; however, sex effects on the genetic regulation of expression were more evident at the protein level. Considering 24 psychiatric, neurologic and brain morphologic traits, we found that an average of 25% of their putatively causal genes had sex-differentiated protein abundance and 12 putatively causal proteins had sb-pQTLs. Furthermore, integrating sex-specific pQTLs with sex-stratified genome-wide association studies of six psychiatric and neurologic conditions, we uncovered another 23 proteins contributing to these traits in one sex but not the other. Together, these findings begin to provide insights into mechanisms underlying sex differences in brain protein expression and disease.


Subject(s)
Genome-Wide Association Study , Sex Characteristics , Female , Male , Humans , Brain , Multifactorial Inheritance , Phenotype
2.
Neurobiol Dis ; 185: 106257, 2023 09.
Article in English | MEDLINE | ID: mdl-37562656

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disorder influenced by a complex interplay of environmental, epigenetic, and genetic factors. DNA methylation (5mC) and hydroxymethylation (5hmC) are DNA modifications that serve as tissue-specific and temporal regulators of gene expression. TET family enzymes dynamically regulate these epigenetic modifications in response to environmental conditions, connecting environmental factors with gene expression. Previous epigenetic studies have identified 5mC and 5hmC changes associated with AD. In this study, we performed targeted resequencing of TET1 on a cohort of early-onset AD (EOAD) and control samples. Through gene-wise burden analysis, we observed significant enrichment of rare TET1 variants associated with AD (p = 0.04). We also profiled 5hmC in human postmortem brain tissues from AD and control groups. Our analysis identified differentially hydroxymethylated regions (DhMRs) in key genes responsible for regulating the methylome: TET3, DNMT3L, DNMT3A, and MECP2. To further investigate the role of Tet1 in AD pathogenesis, we used the 5xFAD mouse model with a Tet1 KO allele to examine how Tet1 loss influences AD pathogenesis. We observed significant changes in neuropathology, 5hmC, and RNA expression associated with Tet1 loss, while the behavioral alterations were not significant. The loss of Tet1 significantly increased amyloid plaque burden in the 5xFAD mouse (p = 0.044) and lead to a non-significant trend towards exacerbated AD-associated stress response in 5xFAD mice. At the molecular level, we found significant DhMRs enriched in genes involved in pathways responsible for neuronal projection organization, dendritic spine development and organization, and myelin assembly. RNA-Seq analysis revealed a significant increase in the expression of AD-associated genes such as Mpeg1, Ctsd, and Trem2. In conclusion, our results suggest that TET enzymes, particularly TET1, which regulate the methylome, may contribute to AD pathogenesis, as the loss of TET function increases AD-associated pathology.


Subject(s)
Alzheimer Disease , Humans , Mice , Animals , Alzheimer Disease/metabolism , 5-Methylcytosine , Epigenesis, Genetic , DNA Methylation , Transcription Factors/metabolism , Mixed Function Oxygenases/genetics , Mixed Function Oxygenases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Immunologic/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism
3.
Nat Commun ; 13(1): 4314, 2022 07 26.
Article in English | MEDLINE | ID: mdl-35882878

ABSTRACT

Several common psychiatric and neurodegenerative diseases share epidemiologic risk; however, whether they share pathophysiology is unclear and is the focus of our investigation. Using 25 GWAS results and LD score regression, we find eight significant genetic correlations between psychiatric and neurodegenerative diseases. We integrate the GWAS results with human brain transcriptomes (n = 888) and proteomes (n = 722) to identify cis- and trans- transcripts and proteins that are consistent with a pleiotropic or causal role in each disease, referred to as causal proteins for brevity. Within each disease group, we find many distinct and shared causal proteins. Remarkably, 30% (13 of 42) of the neurodegenerative disease causal proteins are shared with psychiatric disorders. Furthermore, we find 2.6-fold more protein-protein interactions among the psychiatric and neurodegenerative causal proteins than expected by chance. Together, our findings suggest these psychiatric and neurodegenerative diseases have shared genetic and molecular pathophysiology, which has important ramifications for early treatment and therapeutic development.


Subject(s)
Mental Disorders , Neurodegenerative Diseases , Brain , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Humans , Mental Disorders/genetics , Neurodegenerative Diseases/genetics , Polymorphism, Single Nucleotide
4.
Article in English | MEDLINE | ID: mdl-35772923

ABSTRACT

OBJECTIVE: APOE is a strong risk factor for Alzheimer's disease (AD) and associated with higher low-density lipoprotein cholesterol (LDL-C) levels. Moreover, LDL-C is associated with the development of clinically ascertained AD; however, whether this association is present with the underlying neuropathological manifestations of AD or whether it is independent of the effect of APOE is unknown and is the focus of this paper. METHODS: Individuals in the Religious Orders Study/Memory and Ageing Project cohorts with longitudinal measures of blood lipids and detailed autopsies were studied. We modelled the relationship between blood lipids and 12 age-related brain pathologies using a linear mixed model adjusted for potential confounding factors and stratified by APOE genotype with overall significance determined by meta-analysis. Blood lipids considered were LDL-C, high-density lipoprotein cholesterol and triglycerides. Brain pathologies included AD pathology measured by silver staining (Braak stage, a modified Consortium to Establish a Registry for Alzheimer's Disease [CERAD] score and global AD pathology) and immunohistochemistry (beta-amyloid and neurofibrillary tangles) as well as cerebral microinfarct, cerebral macroinfarct, cerebral amyloid angiopathy, cerebral atherosclerosis, hippocampal sclerosis, TDP-43 cytoplasmic inclusions and Lewy bodies. RESULTS: 559 participants (69.1% female) had complete data for analysis. They were followed for a median of 7 years and a median of 3 years prior to dementia onset. LDL-C was associated with all measures of AD neuropathology (neurofibrillary tangles, beta-amyloid, Braak stage, modified CERAD score and global AD pathology) and cerebral amyloid angiopathy independent of APOE after adjusting for age, sex, cholesterol-lowering medication use, body mass index, smoking and education at false discovery rate (FDR) p-value <0.05. CONCLUSIONS: These findings implicate LDL-C in the pathophysiology of AD independent of APOE and suggest LDL-C is a modifiable risk factor for AD.

5.
Mol Psychiatry ; 27(7): 3075-3084, 2022 07.
Article in English | MEDLINE | ID: mdl-35449297

ABSTRACT

Genome-wide association studies (GWAS) have identified several risk loci for post-traumatic stress disorder (PTSD); however, how they confer PTSD risk remains unclear. We aimed to identify genes that confer PTSD risk through their effects on brain protein abundance to provide new insights into PTSD pathogenesis. To that end, we integrated human brain proteomes with PTSD GWAS results to perform a proteome-wide association study (PWAS) of PTSD, followed by Mendelian randomization, using a discovery and confirmatory study design. Brain proteomes (N = 525) were profiled from the dorsolateral prefrontal cortex using mass spectrometry. The Million Veteran Program (MVP) PTSD GWAS (n = 186,689) was used for the discovery PWAS, and the Psychiatric Genomics Consortium PTSD GWAS (n = 174,659) was used for the confirmatory PWAS. To understand whether genes identified at the protein-level were also evident at the transcript-level, we performed a transcriptome-wide association study (TWAS) using human brain transcriptomes (N = 888) and the MVP PTSD GWAS results. We identified 11 genes that contribute to PTSD pathogenesis via their respective cis-regulated brain protein abundance. Seven of 11 genes (64%) replicated in the confirmatory PWAS and 4 of 11 also had their cis-regulated brain mRNA levels associated with PTSD. High confidence level was assigned to 9 of 11 genes after considering evidence from the confirmatory PWAS and TWAS. Most of the identified genes are expressed in other PTSD-relevant brain regions and several are preferentially expressed in excitatory neurons, astrocytes, and oligodendrocyte precursor cells. These genes are novel, promising targets for mechanistic and therapeutic studies to find new treatments for PTSD.


Subject(s)
Stress Disorders, Post-Traumatic , Veterans , Brain , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study/methods , Humans , Polymorphism, Single Nucleotide/genetics , Proteome/genetics , Stress Disorders, Post-Traumatic/genetics , Stress Disorders, Post-Traumatic/psychology , Transcriptome , Veterans/psychology
6.
Genes (Basel) ; 12(6)2021 05 26.
Article in English | MEDLINE | ID: mdl-34073619

ABSTRACT

Cerebral atherosclerosis is a leading cause of stroke and an important contributor to dementia. Yet little is known about its genetic basis. To examine the association of common single nucleotide polymorphisms with cerebral atherosclerosis severity, we conducted a genomewide association study (GWAS) using data collected as part of two community-based cohort studies in the United States, the Religious Orders Study (ROS) and Rush Memory and Aging Project (MAP). Both studies enroll older individuals and exclude participants with signs of dementia at baseline. From our analysis of 1325 participants of European ancestry who had genotype and neuropathologically assessed cerebral atherosclerosis measures available, we found a novel locus for cerebral atherosclerosis in NTNG1. The locus comprises eight SNPs, including two independent significant SNPs: rs6664221 (ß = -0.27, 95% CI = (-0.35, -0.19), p = 1.29 × 10-10) and rs10881463 (ß = -0.20, 95% CI = (-0.27, -0.13), p = 3.40 × 10-8). We further found that the SNPs may influence cerebral atherosclerosis by regulating brain protein expression of CNOT3. CNOT3 is a subunit of CCR4-NOT, which has been shown to be a master regulator of mRNA stability and translation and an important complex for cholesterol homeostasis. In summary, we identify a novel genetic locus for cerebral atherosclerosis and a potential mechanism linking this variation to cerebral atherosclerosis progression. These findings offer insights into the genetic effects on cerebral atherosclerosis.


Subject(s)
Intracranial Arteriosclerosis/genetics , Netrins/genetics , Polymorphism, Single Nucleotide , Transcription Factors/genetics , Aged , Aged, 80 and over , Female , GPI-Linked Proteins/genetics , Humans , Male , Middle Aged
7.
Am J Geriatr Psychiatry ; 29(5): 488-498, 2021 05.
Article in English | MEDLINE | ID: mdl-33046355

ABSTRACT

OBJECTIVE: A wealth of evidence has linked purpose in life (PiL) to better mental and physical health and healthy aging. Here, the authors aimed to determine important correlates of PiL using a machine learning approach. METHODS: Participants were recruited from retirement communities by the Rush Memory and Aging Project and assessed for childhood experience, adulthood sociodemographic factors (e.g., education, income, marital status), lifestyle and health behavior (e.g., cognitively stimulating activities, exercise, social activities, social network size), psychological factors (e.g., depression, loneliness, perceived discrimination, perceived social support), personality traits (e.g., PiL, harm avoidance), and medical conditions. Elastic Net was implemented to identify important correlates of PiL. RESULTS: A total of 1,839 participants were included in our analysis. Among the 23 variables provided to Elastic Net, 10 were identified as important correlates of PiL. In order of decreasing effect size, factors associated with lower PiL were loneliness, harm avoidance, older age, and depressive symptoms, while those associated with greater PiL were perceived social support, more social activities, more years of education, higher income, intact late-life cognitive performance, and more middle-age cognitive activities. CONCLUSION: Our findings identify potentially important modifiable factors as targets for intervention strategies to enhance PiL.


Subject(s)
Healthy Aging , Loneliness , Adult , Aged , Aging , Humans , Machine Learning , Social Support
SELECTION OF CITATIONS
SEARCH DETAIL
...