Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell Immunol ; 391-392: 104743, 2023.
Article in English | MEDLINE | ID: mdl-37451918

ABSTRACT

The significance of peripheral immunity in the pathogenesis and progression of Alzheimer's diseases (AD) has been recognized. Brain-infiltrated peripheral immune components transporting across the blood-brain barrier (BBB) may reshape the central immune environment. However, mechanisms of how these components open the BBB for AD occurrence and development and correlations between peripheral and central immunity have not been fully explored. Herein, we formulate a hypothesis whereby peripheral immunity as a critical factor allows AD to progress. Peripheral central immune cell crosstalk is associated with early AD pathology and related risk factors. The damaged BBB permits peripheral immune cells to enter the central immune system to deprive its immune privilege promoting the progression toward developing AD. This review summarizes the influences of risk factors on peripheral immunity, alongside their functions, highlighting the concept of peripheral and central immunity as an integrated system in AD pathogenesis, which has received scant attention before.


Subject(s)
Alzheimer Disease , Humans , Central Nervous System , Brain , Blood-Brain Barrier/pathology , Risk Factors
2.
Hum Reprod ; 38(11): 2247-2258, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37713654

ABSTRACT

STUDY QUESTION: Can potential mechanisms involved in the likely concurrence of diminished ovarian reserve (DOR) and miscarriage be identified using genetic data? SUMMARY ANSWER: Concurrence between ovarian reserve and spontaneous miscarriage was observed, and may be attributed to shared genetic risk loci enriched in antigen processing and presentation and autoimmune disease pathways. WHAT IS KNOWN ALREADY: Previous studies have shown that lower serum anti-Müllerian hormone (AMH) levels are associated with increased risk of embryo aneuploidy and spontaneous miscarriage, although findings have not been consistent across all studies. A recent meta-analysis suggested that the association between DOR and miscarriage may not be causal, but rather a result of shared underlying causes such as clinical conditions or past exposure. Motivated by this hypothesis, we conducted the present analysis to explore the concurrence between DOR and miscarriage, and to investigate potential mechanisms using genetic data. STUDY DESIGN, SIZE, DURATION: Three data sources were used in the study: the clinical IVF data were retrospectively collected from an academically affiliated Reproductive Medicine Center (17 786 cycles included); the epidemiological data from the UK Biobank (UKB), which is a large-scale, population-based, prospective cohort study (35 316 white women included), were analyzed; and individual-level genotype data from the UKB were extracted for further analysis. PARTICIPANTS/MATERIALS, SETTING, METHODS: There were three modules of analysis. First, clinical IVF data were used to test the association between ovarian reserve biomarkers and the subsequent early spontaneous miscarriage risk. Second, the UKB data were used to test the association of spontaneous miscarriage history and early menopause. Third, individual-level genotype data from the UKB were analyzed to identify specific pleiotropic genes which affect the development of miscarriage and menopause. MAIN RESULTS AND THE ROLE OF CHANCE: In the analysis of clinical IVF data, the risk of early spontaneous miscarriage was 1.57 times higher in the group with AMH < 1.1 ng/ml group (P < 0.001), 1.62 times for antral follicular count <5 (P < 0.001), and 1.39 times for FSH ≥10 mIU/ml (P < 0.001) in comparison with normal ovarian reserve groups. In the analysis of UKB data, participants with a history of three or more miscarriages had a one-third higher risk of experiencing early menopause (odds ratio: 1.30, 95% CI 1.13-1.49, P < 0.001), compared with participants without spontaneous miscarriage history. We identified 158 shared genetic risk loci that affect both miscarriage and menopause, which enrichment analysis showed were involved in antigen processing and presentation and autoimmune disease pathways. LIMITATIONS, REASONS FOR CAUTION: The analyses of the UKB data were restricted to participants of European ancestry, as 94.6% of the cohort were of white ethnicity. Further studies are needed in non-white populations. Additionally, maternal age at the time of spontaneous miscarriage was not available in the UKB cohort, therefore we adjusted for age at baseline assessment in the models instead. It is known that miscarriage rate in IVF is higher compared to natural conception, highlighting a need for caution when generalizing our findings from the IVF cohort to the general population. WIDER IMPLICATIONS OF THE FINDINGS: Our findings have implications for IVF clinicians in terms of patient counseling on the prognosis of IVF treatment, as well as for genetic counseling regarding miscarriage. Our results highlight the importance of further research on the shared genetic architecture and common pathophysiological basis of DOR and miscarriage, which may lead to new therapeutic opportunities. STUDY FUNDING/COMPETING INTEREST(S): This work was supported by the Hunan Youth Science and Technology Innovation Talent Project (2020RC3060), the International Postdoctoral Exchange Fellowship Program (Talent-Introduction Program, YJ20220220), the fellowship of China Postdoctoral Science Foundation (2022M723564), and the Natural Science Foundation of Hunan Province, China (2023JJ41016). This work has been accepted for poster presentation at the 39th Annual Meeting of ESHRE, Copenhagen, Denmark, 25-28 June 2023 (Poster number: P-477). The authors declare no conflict of interest. TRIAL REGISTRATION NUMBER: N/A.


Subject(s)
Abortion, Spontaneous , Autoimmune Diseases , Menopause, Premature , Ovarian Diseases , Ovarian Reserve , Pregnancy , Humans , Female , Adolescent , Abortion, Spontaneous/epidemiology , Retrospective Studies , Prospective Studies , Anti-Mullerian Hormone , Fertilization in Vitro/methods
3.
Hered Cancer Clin Pract ; 21(1): 1, 2023 Jan 28.
Article in English | MEDLINE | ID: mdl-36707860

ABSTRACT

The current understanding of the inherited risk of colorectal cancer (CRC) started with an observational clinical era in the late 19th century, which was followed by a genetic era starting in the late 20th century. Genome-wide linkage analysis allowed mapping several high-risk genes, which marked the beginning of the genetic era. The current high-throughput genomic phase includes genome-wide association study (GWAS) and genome-wide sequencing approaches which have revolutionized the conception of the inherited risk of CRC. On the one hand, GWAS has allowed the identification of multiple low risk loci correlated with CRC. On the other, genome-wide sequencing has led to the discovery of a second batch of high-to-moderate-risk genes that correlate to atypical familial CRC and polyposis syndromes. In contrast to other common cancers, which are usually dominated by a polygenic background, CRC risk is believed to be equally explained by monogenic and polygenic architectures, which jointly contribute to a quarter of familial clustering. Despite the fact that genome-wide approaches have allowed the identification of a continuum of responsible high-to-moderate-to-low-risk variants, much of the predisposition and familial clustering of CRC has not yet been explained. Other genetic, epigenetic and environmental factors might be playing important roles as well. In this review we aim to provide insights on the complementary roles played by different genomic approaches in allowing the current understanding of the genetic architecture of inherited CRC.

4.
Am J Hum Genet ; 102(6): 1169-1184, 2018 06 07.
Article in English | MEDLINE | ID: mdl-29805045

ABSTRACT

Causal genes and variants within genome-wide association study (GWAS) loci can be identified by integrating GWAS statistics with expression quantitative trait loci (eQTL) and determining which variants underlie both GWAS and eQTL signals. Most analyses, however, consider only the marginal eQTL signal, rather than dissect this signal into multiple conditionally independent signals for each gene. Here we show that analyzing conditional eQTL signatures, which could be important under specific cellular or temporal contexts, leads to improved fine mapping of GWAS associations. Using genotypes and gene expression levels from post-mortem human brain samples (n = 467) reported by the CommonMind Consortium (CMC), we find that conditional eQTL are widespread; 63% of genes with primary eQTL also have conditional eQTL. In addition, genomic features associated with conditional eQTL are consistent with context-specific (e.g., tissue-, cell type-, or developmental time point-specific) regulation of gene expression. Integrating the 2014 Psychiatric Genomics Consortium schizophrenia (SCZ) GWAS and CMC primary and conditional eQTL data reveals 40 loci with strong evidence for co-localization (posterior probability > 0.8), including six loci with co-localization of conditional eQTL. Our co-localization analyses support previously reported genes, identify novel genes associated with schizophrenia risk, and provide specific hypotheses for their functional follow-up.


Subject(s)
Genome-Wide Association Study , Prefrontal Cortex/pathology , Quantitative Trait Loci/genetics , Schizophrenia/genetics , Cells, Cultured , Epigenesis, Genetic , Genome, Human , Humans
5.
Immunol Cell Biol ; 99(5): 496-508, 2021 05.
Article in English | MEDLINE | ID: mdl-33483996

ABSTRACT

The conceptual basis for a genetic predisposition underlying the risk for developing type 1 diabetes (T1D) predates modern human molecular genetics. Over half of the genetic risk has been attributed to the human leukocyte antigen (HLA) class II gene region and to the insulin (INS) gene locus - both thought to confer direction of autoreactivity and tissue specificity. Notwithstanding, questions still remain regarding the functional contributions of a vast array of minor polygenic risk variants scattered throughout the genome that likely influence disease heterogeneity and clinical outcomes. Herein, we summarize the available literature related to the T1D-associated coding variants defined at the time of this review, for the genes PTPN22, IFIH1, SH2B3, CD226, TYK2, FUT2, SIRPG, CTLA4, CTSH and UBASH3A. Data from genotype-selected human cohorts are summarized, and studies from the non-obese diabetic (NOD) mouse are presented to describe the functional impact of these variants in relation to innate and adaptive immunity as well as to ß-cell fragility, with expression profiles in tissues and peripheral blood highlighted. The contribution of each variant to progression through T1D staging, including environmental interactions, are discussed with consideration of how their respective protein products may serve as attractive targets for precision medicine-based therapeutics to prevent or suspend the development of T1D.


Subject(s)
Diabetes Mellitus, Type 1 , Animals , Diabetes Mellitus, Type 1/genetics , Genetic Predisposition to Disease , Genotype , Mice , Mice, Inbred NOD , Polymorphism, Single Nucleotide
6.
Proc Natl Acad Sci U S A ; 115(45): 11567-11572, 2018 11 06.
Article in English | MEDLINE | ID: mdl-30348779

ABSTRACT

Whole-exome sequencing has been successful in identifying genetic factors contributing to familial or sporadic Parkinson's disease (PD). However, this approach has not been applied to explore the impact of de novo mutations on PD pathogenesis. Here, we sequenced the exomes of 39 early onset patients, their parents, and 20 unaffected siblings to investigate the effects of de novo mutations on PD. We identified 12 genes with de novo mutations (MAD1L1, NUP98, PPP2CB, PKMYT1, TRIM24, CEP131, CTTNBP2, NUS1, SMPD3, MGRN1, IFI35, and RUSC2), which could be functionally relevant to PD pathogenesis. Further analyses of two independent case-control cohorts (1,852 patients and 1,565 controls in one cohort and 3,237 patients and 2,858 controls in the other) revealed that NUS1 harbors significantly more rare nonsynonymous variants (P = 1.01E-5, odds ratio = 11.3) in PD patients than in controls. Functional studies in Drosophila demonstrated that the loss of NUS1 could reduce the climbing ability, dopamine level, and number of dopaminergic neurons in 30-day-old flies and could induce apoptosis in fly brain. Together, our data suggest that de novo mutations could contribute to early onset PD pathogenesis and identify NUS1 as a candidate gene for PD.


Subject(s)
Brain/metabolism , Dopaminergic Neurons/metabolism , Mutation , Nerve Tissue Proteins/genetics , Parkinson Disease/genetics , Receptors, Cell Surface/genetics , Adult , Age of Onset , Animals , Apoptosis/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/antagonists & inhibitors , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Aryl Hydrocarbon Receptor Nuclear Translocator/metabolism , Base Sequence , Brain/pathology , Case-Control Studies , Cohort Studies , Disease Models, Animal , Dopamine/metabolism , Dopaminergic Neurons/pathology , Drosophila Proteins/antagonists & inhibitors , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Early Diagnosis , Female , Gene Expression , Gene Regulatory Networks , Humans , Male , Nerve Tissue Proteins/metabolism , Parents , Parkinson Disease/diagnosis , Parkinson Disease/metabolism , Parkinson Disease/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Receptors, Cell Surface/metabolism , Siblings
7.
Am J Med Genet B Neuropsychiatr Genet ; 186(1): 28-39, 2021 01.
Article in English | MEDLINE | ID: mdl-33522098

ABSTRACT

Schizophrenia is a severe mental disease characterized with positive symptoms, negative symptoms, and cognitive impairments. Although recent genome-wide association studies (GWASs) have identified over 145 risk loci for schizophrenia, pinpointing the causal variants and genes at the reported loci and elucidating their roles in schizophrenia remain major challenges. Here we identify a functional single-nucleotide polymorphism (SNP; rs213237) in ZNF323 promoter by using functional fine-mapping. We found that allelic differences at rs213237 affected the ZNF323 promoter activity significantly. Consistently, expression quantitative trait loci (eQTL) analysis showed that rs213237 was significantly associated with ZNF323 expression in diverse human brain tissues, suggesting that rs213237 may contribute to schizophrenia risk through regulating ZNF323 expression. Interestingly, we found that ZNF323 protein was localized in the nucleus and knockdown of ZNF323 in macaque neural stem cells (mNSCs) significantly impaired proliferation and survival of mNSCs. We further showed that stable knockdown of ZNF323 in SH-SY5Y cells resulted in significant decrease of the tyrosine hydroxylase (TH) protein expression. Finally, transcriptome analysis revealed that ZNF323 may regulate pivotal schizophrenia risk genes (including VIPR2 and NPY) and schizophrenia-associated pathways (including PI3K-AKT and NOTCH signaling pathways), suggesting that ZNF323 may be a major regulator of schizophrenia risk genes. Our study reveals how a genetic variant in ZNF323 promoter contributes to schizophrenia risk through regulating ZNF323 expression. More importantly, our findings demonstrate that ZNF323 may have a pivotal role in schizophrenia pathogenesis through regulating schizophrenia risk genes and schizophrenia-associated biological processes (including neurodevelopment, PI3K-AKT, and NOTCH signaling pathways).


Subject(s)
DNA-Binding Proteins/metabolism , Genome-Wide Association Study , Neural Stem Cells/pathology , Neuroblastoma/pathology , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Schizophrenia/pathology , Transcription Factors/metabolism , Animals , DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Humans , Macaca , Neural Stem Cells/metabolism , Neuroblastoma/genetics , Neuroblastoma/metabolism , Phenotype , Schizophrenia/genetics , Schizophrenia/metabolism , Transcription Factors/genetics , Tumor Cells, Cultured
8.
J Cell Mol Med ; 24(2): 1245-1255, 2020 01.
Article in English | MEDLINE | ID: mdl-31782902

ABSTRACT

Nowadays, gene expression profiling has been widely used in screening out prognostic biomarkers in numerous kinds of carcinoma. Our studies attempt to construct a clinical nomogram which combines risk gene signature and clinical features for individual recurrent risk assessment and offer personalized managements for clear cell renal cell carcinoma. A total of 580 differentially expressed genes (DEGs) were identified via microarray. Functional analysis revealed that DEGs are of fundamental importance in ccRCC progression and metastasis. In our study, 338 ccRCC patients were retrospectively analysed and a risk gene signature which composed of 5 genes was obtained from a LASSO Cox regression model. Further analysis revealed that identified risk gene signature could usefully distinguish the patients with poor prognosis in training cohort (hazard ratio [HR] = 3.554, 95% confidence interval [CI] 2.261-7.472, P < .0001, n = 107). Moreover, the prognostic value of this gene-signature was independent of clinical features (P = .002). The efficacy of risk gene signature was verified in both internal and external cohorts. The area under receiver operating characteristic curve of this signature was 0.770, 0.765 and 0.774 in the training, testing and external validation cohorts, respectively. Finally, a nomogram was developed for clinicians and did well in the calibration plots. This nomogram based on risk gene signature and clinical features might provide a practical way for recurrence prediction and facilitating personalized managements of ccRCC patients after surgery.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Renal Cell/mortality , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/mortality , Neoplasm Recurrence, Local/mortality , Nomograms , Transcriptome , Adult , Aged , Aged, 80 and over , Carcinoma, Renal Cell/genetics , Carcinoma, Renal Cell/pathology , Female , Follow-Up Studies , Humans , Kidney Neoplasms/genetics , Kidney Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Prognosis , Retrospective Studies , Survival Rate
9.
Mol Biol Rep ; 47(7): 5127-5136, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32583282

ABSTRACT

Common sequence variations in the VRK2 gene contribute to genetic risk for various psychiatric diseases including schizophrenia and major depressive disorder. Despite the clear importance of studying the regulation and function of VRK2 for understanding the causes of these diseases, the organisation and expression of the gene remain poorly characterised. Using reverse-transcriptase-PCR, we have amplifed exons of Vrk2 mRNA from regions of mouse brain, and from different cell classes comprising neurones, astrocytes and microglial cells. We find that Vrk2 mRNA is expressed in all cell types, and that the splicing of the mouse Vrk2 gene is much more complex than previously appreciated. In addition to the predicted alternative splicing (absence/presence) of the penultimate 3 prime exon, we also detected a variety of 5 prime structures, including two novel exons spanning the first characterised exon (exon 1), which we term exons 1a and 1b. While expressed in neurones and astrocytes, exon 1b was not expressed in microglial cells. Expression of transcripts containing exon 1a in microglia was increased by immune stimulation. An additional truncated transcript lacking 7 central exons was also identified. As with the human gene, the results confirm complex patterns of alternative splicing which are likely to be relevant for understanding the physiological and pathological function of the gene in the CNS.


Subject(s)
Alternative Splicing , Brain/metabolism , Protein Serine-Threonine Kinases/genetics , Animals , Brain/cytology , Cells, Cultured , Mice , Mice, Inbred C57BL , Microglia/metabolism , Neurons/metabolism , Protein Serine-Threonine Kinases/metabolism
10.
Eur Arch Psychiatry Clin Neurosci ; 270(2): 169-181, 2020 Mar.
Article in English | MEDLINE | ID: mdl-30929061

ABSTRACT

Preliminary evidence suggests that BDNF (brain derived neurotrophic factor) rs6265 genetic polymorphism, BDNF gene promotor methylation and BDNF serum levels might play an important role in the pathogenesis of affective disorders. As studies testing the BDNF system across molecular levels are sparse, this study aimed at investigating the BDNF val66met genotype, BDNF DNA methylation changes and peripheral BDNF serum levels in acute and remitted phases of MDD (major depressive disorder) and BD (bipolar disorder) and healthy controls. We found a significant difference of methylation levels at CpG site 1-1-1 and 3-1-1 between MDD and healthy controls (p < 0.003) with MDD patients showing significantly higher methylation levels. CpG 5-2-1 revealed a statistically significant difference between MDD and healthy controls and MDD and BD (p = 0.00003). Similar to the results of the methylation analysis a significant difference between MDD and healthy controls was found in BDNF serum levels (p = 0.002) with significantly lower BDNF serum levels in MDD compared to healthy controls. A difference between the samples from admission and discharge from hospital of both BDNF gene methylation and serum levels could not be detected in the present study and no influence of the BDNF val66met genotype on neither methylation nor BDNF serum level.


Subject(s)
Bipolar Disorder/blood , Brain-Derived Neurotrophic Factor/blood , DNA Methylation , Depressive Disorder, Major/blood , Adult , Biomarkers/blood , Bipolar Disorder/genetics , Bipolar Disorder/therapy , Brain-Derived Neurotrophic Factor/genetics , CpG Islands , Depressive Disorder, Major/genetics , Depressive Disorder, Major/therapy , Female , Humans , Longitudinal Studies , Male , Middle Aged , Patient Admission , Patient Discharge , Promoter Regions, Genetic , Remission Induction
11.
Eur Heart J ; 40(41): 3385-3392, 2019 11 01.
Article in English | MEDLINE | ID: mdl-31228190

ABSTRACT

AIMS: Efficacy of aspirin in primary prevention of cardiovascular disease (CVD) may be influenced by a common allele in guanylate cyclase GUCY1A3, which has been shown to modify platelet function and increase CVD risk. METHODS AND RESULTS: We investigated whether homozygotes of the GUCY1A3 rs7692387 risk (G) allele benefited from aspirin in two long-term, randomized placebo-controlled trials of aspirin in primary CVD prevention: the Women's Genome Health Study (WGHS, N = 23 294) and a myocardial infarction (MI, N = 550) and stroke (N = 382) case-control set from the Physician's Health Study (PHS, N = 22 071). Bleeding risk was evaluated in the WGHS. In the placebo group of the WGHS, the GUCY1A3 risk (G) allele was confirmed to increase CVD risk [hazard ratio 1.38; 95% confidence interval (CI) 1.08-1.78; P = 0.01]. Random-effects meta-analysis of the WGHS and PHS revealed that aspirin reduced CVD events among risk allele homozygotes [G/G: odds ratio (OR) 0.79; 95% CI 0.65-0.97; P = 0.03] but increased CVD events among non-risk allele carriers (e.g. G/A: OR 1.39; 95% CI 1.03-1.87; P = 0.03) thus implying an interaction between genotype stratum and aspirin intake (Pinteraction = 0.01). Bleeding associated with aspirin increased in all genotype groups, with higher risks in heterozygotes. CONCLUSION: In two randomized placebo-controlled trials in the setting of primary prevention, aspirin reduced the incidence of CVD events in individuals homozygous for the GUCY1A3 risk (G) allele, whereas heterozygote individuals had more events when taking aspirin.


Subject(s)
Aspirin , Cardiovascular Diseases , Coronary Artery Disease , Soluble Guanylyl Cyclase/genetics , Adult , Aged , Aged, 80 and over , Aspirin/adverse effects , Aspirin/therapeutic use , Cardiovascular Diseases/drug therapy , Cardiovascular Diseases/epidemiology , Cardiovascular Diseases/genetics , Cardiovascular Diseases/prevention & control , Coronary Artery Disease/epidemiology , Coronary Artery Disease/genetics , Female , Genetic Predisposition to Disease/epidemiology , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Primary Prevention
12.
Genomics ; 111(4): 590-597, 2019 07.
Article in English | MEDLINE | ID: mdl-29627504

ABSTRACT

Complex diseases, such as obesity, type II diabetes and chronic obstructive pulmonary disease (COPD) as metabolic disorder-related diseases are major concern for worldwide public health in the 21st century. The identification of these disease risk genes has attracted increasing interest in computational systems biology. In this paper, a novel method was proposed to prioritize disease risk genes (PDRG) by integrating functional annotations, protein interactions and gene expression information to assess similarity between genes in a disease-related metabolic network. The gene prioritization method was successfully carried out for obesity and COPD, the effectiveness of which was superior to those of ToppGene and ToppNet in both literature validation and recall rate by LOOCV. Our method could be applied broadly to other metabolism-related diseases, helping to prioritize novel disease risk genes, and could shed light on diagnosis and effective therapies.


Subject(s)
Diabetes Mellitus/genetics , Genome-Wide Association Study/methods , Metabolic Syndrome/genetics , Multifactorial Inheritance , Obesity/genetics , Pulmonary Disease, Chronic Obstructive/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study/standards , Humans
13.
Int J Cancer ; 145(10): 2692-2700, 2019 11 15.
Article in English | MEDLINE | ID: mdl-30927251

ABSTRACT

Mutations in BRCA1 and BRCA2 genes predispose to breast and ovarian cancer (BC/OC) with a high lifetime risk, whereas mutations in PALB2, CHEK2, ATM, FANCM, RAD51C and RAD51D genes cause a moderately elevated risk. In the Finnish population, recurrent mutations have been identified in all of these genes, the latest being CHEK2 c.319+2T>A and c.444+1G>A. By genotyping 3,156 cases and 2,089 controls, we estimated the frequencies of CHEK2 c.319+2T>A and c.444+1G>A in Finnish BC patients. CHEK2 c.319+2T>A was detected in 0.7% of the patients, and it was associated with a high risk of BC in the unselected patient group (OR = 5.40 [95% CI 1.58-18.45], p = 0.007) and similarly in the familial patient group. CHEK2 c.444+1G>A was identified in 0.1% of all patients. Additionally, we evaluated the combined prevalence of recurrent moderate-risk gene mutations in 2,487 BC patients, 556 OC patients and 261 BRCA1/2 carriers from 109 families. The overall frequency of the mutations was 13.3% in 1,141 BRCA1/2-negative familial BC patients, 7.5% in 1,727 unselected BC patients and 7.2% in 556 unselected OC patients. At least one moderate-risk gene mutation was found in 12.5% of BRCA1 families and 7.1% of BRCA1 index patients, as well as in 17.0% of BRCA2 families and 11.3% of BRCA2 index patients, and the mutations were associated with an additional risk in the BRCA1/2 index patients (OR = 2.63 [1.15-5.48], p = 0.011). These results support gene panel testing of even multiple members of BC families where several mutations may segregate in different individuals.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Genetic Predisposition to Disease , Ovarian Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Breast Neoplasms/epidemiology , Case-Control Studies , Cohort Studies , Datasets as Topic , Female , Finland/epidemiology , Genetic Testing/methods , Heterozygote , Humans , Medical History Taking , Middle Aged , Mutation , Ovarian Neoplasms/epidemiology , Prevalence , Risk Assessment/methods , Young Adult
14.
J Cell Sci ; 129(10): 2003-15, 2016 05 15.
Article in English | MEDLINE | ID: mdl-27044754

ABSTRACT

One of the defining pathological features of Alzheimer's disease is the intraneuronal accumulation of tau (also known as MAPT) protein. Tau is also secreted from neurons in response to various stimuli and accumulates in the cerebrospinal fluid of Alzheimer's disease patients. Tau pathology might spread from cell to cell through a mechanism involving secretion and uptake. Here, we developed an assay to follow cellular release and uptake of tau dimers. Individual silencing of ten common late-onset Alzheimer's disease risk genes in HEK293T cells expressing the tau reporters suggested that FRMD4A is functionally linked to tau secretion. FRMD4A depletion by using RNA interference (RNAi) reduced and overexpression increased tau secretion. The activity of cytohesins, interactors of FRMD4A and guanine-nucleotide-exchange factors of Arf6, was necessary for FRMD4A-induced tau secretion. Increased Arf6 and cell polarity signaling through Par6 and atypical protein kinase Cζ (aPKCζ) stimulated tau secretion. In mature cortical neurons, FRMD4A RNAi or inhibition of cytohesins strongly upregulated secretion of endogenous tau. These results suggest that FRMD4A, a genetic risk factor for late-onset Alzheimer's disease, regulates tau secretion by activating cytohesin-Arf6 signaling. We conclude that genetic risk factors of Alzheimer's disease might modulate disease progression by altering tau secretion.


Subject(s)
ADP-Ribosylation Factors/genetics , Adaptor Proteins, Signal Transducing/genetics , Alzheimer Disease/genetics , Guanine Nucleotide Exchange Factors/genetics , tau Proteins/genetics , ADP-Ribosylation Factor 6 , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Gene Expression Regulation , HEK293 Cells , Humans , Neurogenesis/genetics , Neurons/metabolism , Neurons/pathology , Phosphorylation , Protein Aggregation, Pathological/genetics , Signal Transduction , tau Proteins/metabolism
15.
BMC Cancer ; 18(1): 165, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29422015

ABSTRACT

BACKGROUND: Breast cancer risk for BRCA1 and BRCA2 pathogenic mutation carriers is modified by risk factors that cluster in families, including genetic modifiers of risk. We considered genetic modifiers of risk for carriers of high-risk mutations in other breast cancer susceptibility genes. METHODS: In a family known to carry the high-risk mutation PALB2:c.3113G>A (p.Trp1038*), whole-exome sequencing was performed on germline DNA from four affected women, three of whom were mutation carriers. RESULTS: RNASEL:p.Glu265* was identified in one of the PALB2 carriers who had two primary invasive breast cancer diagnoses before 50 years. Gene-panel testing of BRCA1, BRCA2, PALB2 and RNASEL in the Australian Breast Cancer Family Registry identified five carriers of RNASEL:p.Glu265* in 591 early onset breast cancer cases. Three of the five women (60%) carrying RNASEL:p.Glu265* also carried a pathogenic mutation in a breast cancer susceptibility gene compared with 30 carriers of pathogenic mutations in the 586 non-carriers of RNASEL:p.Glu265* (5%) (p < 0.002). Taqman genotyping demonstrated that the allele frequency of RNASEL:p.Glu265* was similar in affected and unaffected Australian women, consistent with other populations. CONCLUSION: Our study suggests that RNASEL:p.Glu265* may be a genetic modifier of risk for early-onset breast cancer predisposition in carriers of high-risk mutations. Much larger case-case and case-control studies are warranted to test the association observed in this report.


Subject(s)
Breast Neoplasms/genetics , Endoribonucleases/genetics , Genetic Predisposition to Disease/genetics , Adult , Age of Onset , Australia , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Female , Heterozygote , Humans , Middle Aged , Mutation , Pedigree , Young Adult
16.
Genet Epidemiol ; 39(6): 489-97, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26202568

ABSTRACT

Chronic psychosocial stress adversely affects health and is associated with the development of disease [Williams, 2008]. Systematic epidemiological and genetic studies are needed to uncover genetic variants that interact with stress to modify metabolic responses across the life cycle that are the proximal contributors to the development of cardiovascular disease and precipitation of acute clinical events. Among the central challenges in the field are to perform and replicate gene-by-environment (G × E) studies. The challenge of measurement of individual experience of psychosocial stress is magnified in this context. Although many research datasets exist that contain genotyping and disease-related data, measures of psychosocial stress are often either absent or vary substantially across studies. In this paper, we provide an algorithm to create a synthetic measure of chronic psychosocial stress across multiple datasets, applying a consistent criterion that uses proxy indicators of stress components. We validated the computed scores of chronic psychosocial stress by observing moderately strong and significant correlations with the self-rated chronic psychosocial stress in the Multi-Ethnic Study of Atherosclerosis Cohort (Rho = 0.23, P < 0.0001) and with the measures of depressive symptoms in five datasets (Rho = 0.15-0.42, Ps = 0.005 to <0.0001) and by comparing the distributions of the self-rated and computed measures. Finally, we demonstrate the utility of this computed chronic psychosocial stress variable by providing three additional replications of our previous finding of gene-by-stress interaction with central obesity traits [Singh et al., 2015].


Subject(s)
Gene-Environment Interaction , Stress, Psychological , Trans-Activators/genetics , Body Mass Index , Cardiovascular Diseases/etiology , Cardiovascular Diseases/genetics , Databases, Factual , Genotype , Humans , Phenotype , Polymorphism, Single Nucleotide
17.
Clin Immunol ; 163: 96-107, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26762769

ABSTRACT

Multiple Sclerosis (MS) is an autoimmune disease treated by therapies targeting peripheral blood cells. We previously identified that expression of two MS-risk genes, the transcription factors EOMES and TBX21 (ET), was low in blood from MS and stable over time. Here we replicated the low ET expression in a new MS cohort (p<0.0007 for EOMES, p<0.028 for TBX21) and demonstrate longitudinal stability (p<10(-4)) and high heritability (h(2)=0.48 for EOMES) for this molecular phenotype. Genes whose expression correlated with ET, especially those controlling cell migration, further defined the phenotype. CD56+ cells and other subsets expressed lower levels of Eomes or T-bet protein and/or were under-represented in MS. EOMES and TBX21 risk SNP genotypes, and serum EBNA-1 titres were not correlated with ET expression, but HLA-DRB1*1501 genotype was. ET expression was normalised to healthy control levels with natalizumab, and was highly variable for glatiramer acetate, fingolimod, interferon-beta, dimethyl fumarate.


Subject(s)
Multiple Sclerosis/genetics , T-Box Domain Proteins/genetics , Adult , Aged , CD56 Antigen , Case-Control Studies , Cell Movement , Dimethyl Fumarate/therapeutic use , Epstein-Barr Virus Nuclear Antigens/blood , Female , Fingolimod Hydrochloride/therapeutic use , Gene Expression Regulation , Genetic Predisposition to Disease , Glatiramer Acetate/therapeutic use , HLA-DRB1 Chains/genetics , Humans , Immunologic Factors/therapeutic use , Immunosuppressive Agents/therapeutic use , Interferon-beta/therapeutic use , Longitudinal Studies , Male , Middle Aged , Multiple Sclerosis/drug therapy , Natalizumab/therapeutic use , Polymorphism, Single Nucleotide , Young Adult
18.
Tumour Biol ; 37(2): 1845-51, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26318431

ABSTRACT

The oral squamous cell carcinoma (OSCC) is one of the most common malignant epithelial neoplasms and considered to be caused by the genetic damage. In addition, smoking habit and excessive alcohol consumption have been estimated to be the main risk factors. Although the association between OSCC and genetic susceptibility loci has been observed in many different populations, most of these studies simply focused on the single nucleotide polymorphism. Therefore, we made a contrast analysis between the 112 OSCC patients from the GEO database and 245 normal samples from the HapMap project. First, we performed a genome-wide haplotype association study by comparing the frequency of the haplotypes in the case-control experiment. Then, we mapped the haplotypes to the corresponding genes, screened the risk genes according to significant haplotypes (P < 1E-04), and prioritized the OSCC genes based on their similarity to the known OSCC susceptibility genes. We filtered four OSCC genes including SERPINB9, SERPINE2, GAK, and HSP90B1 through the gene global prioritization score (P < 0.005). SERPINB9 ranked first in the candidate gene list and contained a significant haplotype TAGGA (P value = 3.12E-11). The second risk gene was SERPINE2 with the haplotype GGGCCCTTT, which was closely similar to the SERPINB9.


Subject(s)
Carcinoma, Squamous Cell/genetics , Genetic Predisposition to Disease/genetics , Intracellular Signaling Peptides and Proteins/genetics , Membrane Glycoproteins/genetics , Mouth Neoplasms/genetics , Protein Serine-Threonine Kinases/genetics , Serpin E2/genetics , Serpins/genetics , Alcohol Drinking/adverse effects , Carcinoma, Squamous Cell/etiology , Case-Control Studies , Genome-Wide Association Study/methods , Haplotypes/genetics , Humans , Mouth Neoplasms/etiology , Polymorphism, Single Nucleotide/genetics , Risk Factors , Smoking/adverse effects , Smoking/genetics
19.
Tumour Biol ; 36(10): 7457-63, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25908172

ABSTRACT

This study was aimed to identify the prognostic risk markers for thyroid papillary carcinoma (TPC) by bioinformatics. The clinical data of TPC and their microRNAs (miRNAs) and genes expression profile data were downloaded from The Cancer Genome Atlas. Elastic net-Cox's proportional regression hazards model (EN-COX) was used to identify the prognostic associated factors. The receiver operating characteristic (ROC) curve and Kaplan-Meier (KM) curve were used to screen the significant prognostic risk miRNA and genes. Then, the target genes of the obtained miRNAs were predicted followed by function prediction. Finally, the significant risk genes were performed literature mining and function analysis. Total 1046 miRNAs and 20531 genes in 484 cases samples were identified after data preprocessing. From the EN-COX model, 30 prognostic risk factors were obtained. Based on the 30 risk factors, 3 miRNAs and 11 genes were identified from the ROC and KM curves. The target genes of miRNA-342 such as B-cell CLL/lymphoma 2 (BCL2) were mainly enriched in the biological process related to cellular metabolic process and Disease Ontology terms of lymphoma. The target genes of miRNA-93 were mainly enriched in the pathway of G1 phase. Among the 11 prognostic risk genes, v-maf avian musculoaponeurotic fibrosarcoma oncogene homologue F (MAFF), SRY (sex-determining region Y)-box 4 (SOX4), and retinoic acid receptor, alpha (RARA) encoded transcription factors. Besides, RARA was enriched in four pathways. These prognostic markers such as miRNA-93, miRNA-342, RARA, MAFF, SOX4, and BCL2 may be used as targets for TPC chemoprevention.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Papillary/genetics , Computational Biology/methods , MicroRNAs/genetics , Thyroid Gland/metabolism , Thyroid Neoplasms/genetics , Carcinoma, Papillary/mortality , Carcinoma, Papillary/pathology , Case-Control Studies , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Staging , Prognosis , Proportional Hazards Models , ROC Curve , Survival Rate , Thyroid Neoplasms/mortality , Thyroid Neoplasms/pathology
20.
Neurosci Bull ; 40(8): 1160-1172, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38206551

ABSTRACT

With the advancements in gene sequencing technologies, including genome-wide association studies, polygenetic risk scores, and high-throughput sequencing, there has been a tremendous advantage in mapping a detailed blueprint for the genetic model of bipolar disorder (BD). To date, intriguing genetic clues have been identified to explain the development of BD, as well as the genetic association that might be applied for the development of susceptibility prediction and pharmacogenetic intervention. Risk genes of BD, such as CACNA1C, ANK3, TRANK1, and CLOCK, have been found to be involved in various pathophysiological processes correlated with BD. Although the specific roles of these genes have yet to be determined, genetic research on BD will help improve the prevention, therapeutics, and prognosis in clinical practice. The latest preclinical and clinical studies, and reviews of the genetics of BD, are analyzed in this review, aiming to summarize the progress in this intriguing field and to provide perspectives for individualized, precise, and effective clinical practice.


Subject(s)
Bipolar Disorder , Genetic Predisposition to Disease , Genome-Wide Association Study , Bipolar Disorder/genetics , Humans , Genetic Predisposition to Disease/genetics , Animals
SELECTION OF CITATIONS
SEARCH DETAIL