Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
J Alzheimers Dis ; 99(2): 715-727, 2024.
Article in English | MEDLINE | ID: mdl-38728189

ABSTRACT

Background: There are various molecular hypotheses regarding Alzheimer's disease (AD) like amyloid deposition, tau propagation, neuroinflammation, and synaptic dysfunction. However, detailed molecular mechanism underlying AD remains elusive. In addition, genetic contribution of these molecular hypothesis is not yet established despite the high heritability of AD. Objective: The study aims to enable the discovery of functionally connected multi-omic features through novel integration of multi-omic data and prior functional interactions. Methods: We propose a new deep learning model MoFNet with improved interpretability to investigate the AD molecular mechanism and its upstream genetic contributors. MoFNet integrates multi-omic data with prior functional interactions between SNPs, genes, and proteins, and for the first time models the dynamic information flow from DNA to RNA and proteins. Results: When evaluated using the ROS/MAP cohort, MoFNet outperformed other competing methods in prediction performance. It identified SNPs, genes, and proteins with significantly more prior functional interactions, resulting in three multi-omic subnetworks. SNP-gene pairs identified by MoFNet were mostly eQTLs specific to frontal cortex tissue where gene/protein data was collected. These molecular subnetworks are enriched in innate immune system, clearance of misfolded proteins, and neurotransmitter release respectively. We validated most findings in an independent dataset. One multi-omic subnetwork consists exclusively of core members of SNARE complex, a key mediator of synaptic vesicle fusion and neurotransmitter transportation. Conclusions: Our results suggest that MoFNet is effective in improving classification accuracy and in identifying multi-omic markers for AD with improved interpretability. Multi-omic subnetworks identified by MoFNet provided insights of AD molecular mechanism with improved details.


Subject(s)
Alzheimer Disease , Deep Learning , Polymorphism, Single Nucleotide , Alzheimer Disease/genetics , Humans , Polymorphism, Single Nucleotide/genetics , Gene Regulatory Networks/genetics
2.
Res Sq ; 2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38405816

ABSTRACT

Alzheimer's disease (AD) is a highly heritable brain dementia, along with substantial failure of cognitive function. Large-scale genome-wide association studies (GWASs) have led to a significant set of SNPs associated with AD and related traits. GWAS hits usually emerge as clusters where a lead SNP with the highest significance is surrounded by other less significant neighboring SNPs. Although functionality is not guaranteed even with the strongest associations in GWASs, lead SNPs have historically been the focus of the field, with the remaining associations inferred to be redundant. Recent deep genome annotation tools enable the prediction of function from a segment of a DNA sequence with significantly improved precision, which allows in-silico mutagenesis to interrogate the functional effect of SNP alleles. In this project, we explored the impact of top AD GWAS hits on chromatin functions and whether it will be altered by the genetic context (i.e., alleles of neighboring SNPs). Our results showed that highly correlated SNPs in the same LD block could have distinct impacts on downstream functions. Although some GWAS lead SNPs showed dominant functional effects regardless of the neighborhood SNP alleles, several other SNPs did exhibit enhanced loss or gain of function under certain genetic contexts, suggesting potential additional information hidden in the LD blocks.

3.
medRxiv ; 2023 Oct 23.
Article in English | MEDLINE | ID: mdl-37961458

ABSTRACT

Alzheimer's disease (AD) is a highly heritable brain dementia, along with substantial failure of cognitive function. Large-scale genome-wide association studies (GWAS) have led to a significant set of SNPs associated with AD and related traits. GWAS hits usually emerge as clusters where a lead SNP with the highest significance is surrounded by other less significant neighboring SNPs. Although functionality is not guaranteed with even the strongest associations in the GWAS, the lead SNPs have been historically the focus of the field, with the remaining associations inferred as redundant. Recent deep genome annotation tools enable the prediction of function from a segment of DNA sequence with significantly improved precision, which allows in-silico mutagenesis to interrogate the functional effect of SNP alleles. In this project, we explored the impact of top AD GWAS hits on the chromatin functions, and whether it will be altered by the genomic context (i.e., alleles of neighborhood SNPs). Our results showed that highly correlated SNPs in the same LD block could have distinct impact on the downstream functions. Although some GWAS lead SNPs showed dominating functional effect regardless of the neighborhood SNP alleles, several other ones do get enhanced loss or gain of function under certain genomic context, suggesting potential extra information hidden in the LD blocks.

4.
Front Big Data ; 6: 1151893, 2023.
Article in English | MEDLINE | ID: mdl-37215688

ABSTRACT

Introduction: Brain imaging genetics aims to explore the genetic architecture underlying brain structure and functions. Recent studies showed that the incorporation of prior knowledge, such as subject diagnosis information and brain regional correlation, can help identify significantly stronger imaging genetic associations. However, sometimes such information may be incomplete or even unavailable. Methods: In this study, we explore a new data-driven prior knowledge that captures the subject-level similarity by fusing multi-modal similarity networks. It was incorporated into the sparse canonical correlation analysis (SCCA) model, which is aimed to identify a small set of brain imaging and genetic markers that explain the similarity matrix supported by both modalities. It was applied to amyloid and tau imaging data of the ADNI cohort, respectively. Results: Fused similarity matrix across imaging and genetic data was found to improve the association performance better or similarly well as diagnosis information, and therefore would be a potential substitute prior when the diagnosis information is not available (i.e., studies focused on healthy controls). Discussion: Our result confirmed the value of all types of prior knowledge in improving association identification. In addition, the fused network representing the subject relationship supported by multi-modal data showed consistently the best or equally best performance compared to the diagnosis network and the co-expression network.

5.
BMC Med Genomics ; 15(Suppl 2): 92, 2022 04 23.
Article in English | MEDLINE | ID: mdl-35461274

ABSTRACT

BACKGROUND: There is growing evidence indicating that a number of functional connectivity networks are disrupted at each stage of the full clinical Alzheimer's disease spectrum. Such differences are also detectable in cognitive normal (CN) carrying mutations of AD risk genes, suggesting a substantial relationship between genetics and AD-altered functional brain networks. However, direct genetic effect on functional connectivity networks has not been measured. METHODS: Leveraging existing AD functional connectivity studies collected in NeuroSynth, we performed a meta-analysis to identify two sets of brain regions: ones with altered functional connectivity in resting state network and ones without. Then with the brain-wide gene expression data in the Allen Human Brain Atlas, we applied a new biclustering method to identify a set of genes with differential co-expression patterns between these two set of brain regions. RESULTS: Differential co-expression analysis using biclustering method led to a subset of 38 genes which showed distinctive co-expression patterns between AD-related and non AD-related brain regions in default mode network. More specifically, we observed 4 sub-clusters with noticeable co-expression difference, where the difference in correlations is above 0.5 on average. CONCLUSIONS: This work applies a new biclustering method to search for a subset of genes with altered co-expression patterns in AD-related default mode network regions. Compared with traditional differential expression analysis, differential co-expression analysis yielded many more significant hits with extra insights into the wiring mechanism between genes. Particularly, the differential co-expression pattern was observed between two sets of genes, suggesting potential upstream genetic regulators in AD development.


Subject(s)
Alzheimer Disease , Connectome , Alzheimer Disease/genetics , Brain , Humans , Magnetic Resonance Imaging , Nerve Net
6.
Brief Bioinform ; 22(6)2021 11 05.
Article in English | MEDLINE | ID: mdl-33971669

ABSTRACT

A large number of genetic variations have been identified to be associated with Alzheimer's disease (AD) and related quantitative traits. However, majority of existing studies focused on single types of omics data, lacking the power of generating a community including multi-omic markers and their functional connections. Because of this, the immense value of multi-omics data on AD has attracted much attention. Leveraging genomic, transcriptomic and proteomic data, and their backbone network through functional relations, we proposed a modularity-constrained logistic regression model to mine the association between disease status and a group of functionally connected multi-omic features, i.e. single-nucleotide polymorphisms (SNPs), genes and proteins. This new model was applied to the real data collected from the frontal cortex tissue in the Religious Orders Study and Memory and Aging Project cohort. Compared with other state-of-art methods, it provided overall the best prediction performance during cross-validation. This new method helped identify a group of densely connected SNPs, genes and proteins predictive of AD status. These SNPs are mostly expression quantitative trait loci in the frontal region. Brain-wide gene expression profile of these genes and proteins were highly correlated with the brain activation map of 'vision', a brain function partly controlled by frontal cortex. These genes and proteins were also found to be associated with the amyloid deposition, cortical volume and average thickness of frontal regions. Taken together, these results suggested a potential pathway underlying the development of AD from SNPs to gene expression, protein expression and ultimately brain functional and structural changes.


Subject(s)
Alzheimer Disease/genetics , Databases, Nucleic Acid , Genomics , Polymorphism, Single Nucleotide , Transcriptome , Alzheimer Disease/metabolism , Genome-Wide Association Study , Humans
7.
Neuroimage ; 226: 117560, 2021 02 01.
Article in English | MEDLINE | ID: mdl-33189932

ABSTRACT

Progressive accumulation of tau neurofibrillary tangles in the brain is a defining pathologic feature of Alzheimer's disease (AD). Tau pathology exhibits a predictable spatiotemporal spreading pattern, but the underlying mechanisms of this spread are poorly understood. Although AD is conventionally considered a disease of the gray matter, it is also associated with pronounced and progressive deterioration of the white matter (WM). A link between abnormal tau and WM degeneration is suggested by findings from both animal and postmortem studies, but few studies demonstrated their interplay in vivo. Recent advances in diffusion magnetic resonance imaging and the availability of tau positron emission tomography (PET) have made it possible to evaluate the association of tau and WM degeneration (tau-WM) in vivo. In this study, we explored the spatial pattern of tau-WM associations across the whole brain to evaluate the hypothesis that tau deposition is associated with WM microstructural alterations not only in isolated tracts, but in continuous structural connections in a stereotypic pattern. Sixty-two participants, including 22 cognitively normal subjects, 22 individuals with subjective cognitive decline, and 18 with mild cognitive impairment were included in the study. WM characteristics were inferred by classic diffusion tensor imaging (DTI) and a complementary diffusion compartment model - neurite orientation dispersion and density imaging (NODDI) that provides a proxy for axonal density. A data-driven iterative searching (DDIS) approach, coupled with whole-brain graph theory analyses, was developed to continuously track tau-WM association patterns. Without applying prior knowledge of the tau spread, we observed a distinct spatial pattern that resembled the typical propagation of tau pathology in AD. Such association pattern was not observed between diffusion and amyloid-ß PET signal. Tau-related WM degeneration is characterized by an increase in the mean diffusivity (with a dominant change in the radial direction) and a decrease in the intra-axonal volume fraction. These findings suggest that cortical tau deposition (as measured in tau PET) is associated with a lower axonal packing density and greater diffusion freedom. In conclusion, our in vivo findings using a data-driven method on cross-sectional data underline the important role of WM alterations in the AD pathological cascade with an association pattern similar to the postmortem Braak staging of AD. Future studies will focus on longitudinal analyses to provide in vivo evidence of tau pathology spreads along neuroanatomically connected brain areas.


Subject(s)
Brain/diagnostic imaging , Cognitive Dysfunction/diagnostic imaging , Nerve Degeneration/diagnostic imaging , White Matter/diagnostic imaging , tau Proteins/metabolism , Aged , Aged, 80 and over , Brain/metabolism , Brain/pathology , Cognitive Dysfunction/metabolism , Cognitive Dysfunction/pathology , Cross-Sectional Studies , Diffusion Tensor Imaging , Female , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Nerve Degeneration/metabolism , Nerve Degeneration/pathology , Positron-Emission Tomography , White Matter/metabolism , White Matter/pathology
8.
Front Genet ; 12: 782953, 2021.
Article in English | MEDLINE | ID: mdl-35237294

ABSTRACT

Background: Human brain structural connectivity is an important imaging quantitative trait for brain development and aging. Mapping the network connectivity to the phenotypic variation provides fundamental insights in understanding the relationship between detailed brain topological architecture, function, and dysfunction. However, the underlying neurobiological mechanism from gene to brain connectome, and to phenotypic outcomes, and whether this mechanism changes over time, remain unclear. Methods: This study analyzes diffusion-weighted imaging data from two age-specific neuroimaging cohorts, extracts structural connectome topological network measures, performs genome-wide association studies of the measures, and examines the causality of genetic influences on phenotypic outcomes mediated via connectivity measures. Results: Our empirical study has yielded several significant findings: 1) It identified genetic makeup underlying structural connectivity changes in the human brain connectome for both age groups. Specifically, it revealed a novel association between the minor allele (G) of rs7937515 and the decreased network segregation measures of the left middle temporal gyrus across young and elderly adults, indicating a consistent genetic effect on brain connectivity across the lifespan. 2) It revealed rs7937515 as a genetic marker for body mass index in young adults but not in elderly adults. 3) It discovered brain network segregation alterations as a potential neuroimaging biomarker for obesity. 4) It demonstrated the hemispheric asymmetry of structural network organization in genetic association analyses and outcome-relevant studies. Discussion: These imaging genetic findings underlying brain connectome warrant further investigation for exploring their potential influences on brain-related complex diseases, given the significant involvement of altered connectivity in neurological, psychiatric and physical disorders.

9.
PLoS One ; 15(6): e0234748, 2020.
Article in English | MEDLINE | ID: mdl-32555747

ABSTRACT

Large-scale genome wide association studies (GWASs) have led to discovery of many genetic risk factors in Alzheimer's disease (AD), such as APOE, TOMM40 and CLU. Despite the significant progress, it remains a major challenge to functionally validate these genetic findings and translate them into targetable mechanisms. Integration of multiple types of molecular data is increasingly used to address this problem. In this paper, we proposed a modularity-constrained Lasso model to jointly analyze the genotype, gene expression and protein expression data for discovery of functionally connected multi-omic biomarkers in AD. With a prior network capturing the functional relationship between SNPs, genes and proteins, the newly introduced penalty term maximizes the global modularity of the subnetwork involving selected markers and encourages the selection of multi-omic markers with dense functional connectivity, instead of individual markers. We applied this new model to the real data collected in the ROS/MAP cohort where the cognitive performance was used as disease quantitative trait. A functionally connected subnetwork involving 276 multi-omic biomarkers, including SNPs, genes and proteins, were identified to bear predictive power. Within this subnetwork, multiple trans-omic paths from SNPs to genes and then proteins were observed. This suggests that cognitive performance deterioration in AD patients can be potentially a result of genetic variations due to their cascade effect on the downstream transcriptome and proteome level.


Subject(s)
Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Computational Biology/methods , Aged , Aged, 80 and over , Biomarkers/metabolism , Female , Genomics , Humans , Male , Polymorphism, Single Nucleotide
10.
BMC Med Genomics ; 13(Suppl 5): 53, 2020 04 03.
Article in English | MEDLINE | ID: mdl-32241275

ABSTRACT

BACKGROUND: Alzheimer's disease (AD) is one of the leading causes of death in the US and there is no validated drugs to stop, slow or prevent AD. Despite tremendous effort on biomarker discovery, existing findings are mostly individual biomarkers and provide limited insights into the transcriptomic decoupling underlying AD. We propose to explore the gene co-expression patterns in multiple AD stages, including cognitively normal (CN), early mild cognitive impairment (EMCI), late MCI and AD. METHODS: We modified traiditonal joint graphical lasso to model our asusmption that the co-expression networks in consecutive disease stages are largely similar with critical differences. In addition, we performed subsequent network comparison analysis for identification of stage specific transcriptomic decoupling. We focused our analysis on top AD-enriched pathways. RESULTS: We observed that 419 edges in CN, 420 edges in EMCI, 381 edges in LMCI and 250 edges in AD were frequently estimated with non zero weights. With modified JGL, the weight of all estimated edges in CN, EMCI and LMCI are zero. In AD group, 299 edges were occasionally estimated to be nonzero and the average correlation between genes was 0.0023. For co-expression change during AD progression, there are 66 pairs of genes that demonstrated a continuously decreasing or increasing co-expression from CN to EMCI, LMCI and AD.The network level clustering coefficient remains stable from CN to LMCI and then decreases significantly when progressing to AD. When evaluating edge level differences, we identified eight gene modules with continuously decreasing or increasing co-expression patterns during AD progression. Five of them shows significant changes from CN to EMCI and thus have the potential to serve system biomarkers for early screening of AD. CONCLUSION: We employed a modified joint graphical lasso for estimation of co-expression networks for multiple stages of AD. Comparing with graphical lasso, our modified joint graphical lasso model accounts for the similarity in consecutive disease stages. Our results on real data set revealed five gene clusters with obvious co-expression pattern change from CN to EMCI, which could be used as potential system-level biomarkers for early screening of AD.


Subject(s)
Algorithms , Alzheimer Disease/genetics , Alzheimer Disease/pathology , Biomarkers/analysis , Computational Biology/methods , Gene Regulatory Networks , Transcriptome , Disease Progression , High-Throughput Nucleotide Sequencing , Humans , Jagged-1 Protein
11.
J Exp Clin Cancer Res ; 38(1): 150, 2019 Apr 08.
Article in English | MEDLINE | ID: mdl-30961642

ABSTRACT

BACKGROUND: Deregulated ErbB signaling plays an important role in tumorigenesis of pancreatic cancer. However, patients with pancreatic cancer benefit little from current existed therapies targeting the ErbB signaling. Here, we explore the potential anti-tumor activity of Valproic acid against pancreatic cancer via targeting ErbB family members. METHODS: Cell viability assay and apoptosis evaluation were carried out to determine the efficacy of VPA on pancreatic cancer cells. Western blot analyses were performed to determine the expression and activation of proteins. Apoptosis enzyme-linked immunosorbent assay was used to quantify cytoplasmic histone associated DNA fragments. Lentiviral expression system was used to introduce overexpression of exogeneous genes or gene-targeting short hairpin RNAs (shRNAs). qRT-PCR was carried out to analyze the mRNAs and miRNAs expression levels. Tumor xenograft model was established to evaluate the in vivo anti-pancreatic cancer activity of VPA. RESULTS: VPA preferentially inhibited cell proliferation/survival of, and induced apoptosis in EGFR/ErbB2/ErbB3-coexpressing pancreatic cancer cells within its clinically achievable range [40~100 mg/L (0.24~0.6 mmol/L)]. Mechanistic investigations revealed that VPA treatment resulted in simultaneous significant down-regulation of EGFR, ErbB2, and ErbB3 in pancreatic cancer cells likely via induction of ErbB family members-targeting microRNAs. Moreover, the anti-pancreatic cancer activity of VPA was further validated in tumor xenograft model. CONCLUSIONS: Our data strongly suggest that VPA may be added to the treatment regimens for pancreatic cancer patients with co-overexpression of the ErbB family members.


Subject(s)
MicroRNAs/metabolism , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Receptor, ErbB-3/metabolism , Valproic Acid/therapeutic use , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Mice, Nude , Pancreatic Neoplasms/metabolism , Signal Transduction , Valproic Acid/pharmacology
12.
Connect Neuroimaging (2019) ; 11848: 42-53, 2019 Oct.
Article in English | MEDLINE | ID: mdl-34514470

ABSTRACT

The permutation test is an often used test procedure for determining statistical significance in brain network studies. Unfortunately, generating every possible permutation for large-scale brain imaging datasets such as HCP and ADNI with hundreds of subjects is not practical. Many previous attempts at speeding up the permutation test rely on various approximation strategies such as estimating the tail distribution with known parametric distributions. In this study, we propose the novel transposition test that exploits the underlying algebraic structure of the permutation group. The method is applied to a large number of diffusion tensor images in localizing the regions of the brain network differences.

13.
Connect Neuroimaging (2018) ; 11083: 58-66, 2018 Sep.
Article in English | MEDLINE | ID: mdl-30906933

ABSTRACT

Brain imaging genetics is an emerging research field to explore the underlying genetic architecture of brain structure and function measured by different imaging modalities. However, not all the changes in the brain are a consequential result of genetic effect and it is usually unknown which imaging phenotypes are promising for genetic analyses. In this paper, we focus on identifying highly heritable measures of structural brain networks derived from diffusion weighted imaging data. Using the twin data from the Human Connectome Project (HCP), we evaluated the reliability of fractional anisotropy measure, fiber length and fiber number of each edge in the structural connectome and seven network level measures using intraclass correlation coefficients. We then estimated the heritability of those reliable network measures using SOLAR-Eclipse software. Across all 64,620 network edges between 360 brain regions in the Glasser parcellation, we observed ~5% of them with significantly high heritability in fractional anisotropy, fiber length or fiber number. All the tested network level measures, capturing the network integrality, segregation or resilience, are highly heritable, with variance explained by the additive genetic effect ranging from 59% to 77%.

SELECTION OF CITATIONS
SEARCH DETAIL
...