Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
1.
J Biol Chem ; 294(11): 3881-3898, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30655286

ABSTRACT

Coronary artery disease (CAD) is the leading cause of death worldwide. Long noncoding RNAs (lncRNAs) are a class of noncoding transcripts of > 200 nucleotides and are increasingly recognized as playing functional roles in physiology and disease. ANRIL is an lncRNA gene mapped to the chromosome 9p21 genetic locus for CAD identified by the first series of genome-wide association studies (GWAS). However, ANRIL's role in CAD and the underlying molecular mechanism are unknown. Here, we show that the major ANRIL transcript in endothelial cells (ECs) is DQ485454 with a much higher expression level in ECs than in THP-1 monocytes. Of note, DQ485454 expression was down-regulated in CAD coronary arteries compared with non-CAD arteries. DQ485454 overexpression significantly reduced monocyte adhesion to ECs, transendothelial monocyte migration (TEM), and EC migration, which are critical cellular processes involved in CAD initiation, whereas siRNA-mediated ANRIL knockdown (KD) had the opposite effect. Microarray and follow-up quantitative RT-PCR analyses revealed that the ANRIL KD down-regulated expression of AHNAK2, CLIP1, CXCL11, ENC1, EZR, LYVE1, WASL, and TNFSF10 genes and up-regulated TMEM100 and TMEM106B genes. Mechanistic studies disclosed that overexpression of CLIP1, EZR, and LYVE1 reversed the effects of ANRIL KD on monocyte adhesion to ECs, TEM, and EC migration. These findings indicate that ANRIL regulates EC functions directly related to CAD, supporting the hypothesis that ANRIL is involved in CAD pathogenesis at the 9p21 genetic locus and identifying a molecular mechanism underlying lncRNA-mediated regulation of EC function and CAD development.


Subject(s)
Coronary Artery Disease/metabolism , Coronary Artery Disease/pathology , Cytoskeletal Proteins/metabolism , Endothelial Cells/metabolism , Microtubule-Associated Proteins/metabolism , Neoplasm Proteins/metabolism , RNA, Long Noncoding/metabolism , Up-Regulation , Vesicular Transport Proteins/metabolism , Cell Movement , Cells, Cultured , Cytoskeletal Proteins/genetics , Humans , Microtubule-Associated Proteins/genetics , Middle Aged , Neoplasm Proteins/genetics , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Vesicular Transport Proteins/genetics
2.
RNA Biol ; 17(10): 1391-1401, 2020 10.
Article in English | MEDLINE | ID: mdl-32602777

ABSTRACT

Each gene typically has multiple alternatively spliced transcripts. Different transcripts are assumed to play a similar biological role; however, some transcripts may simply lose their function due to loss of important functional domains. Here, we show that two different transcripts of lncRNA gene ANRIL associated with coronary artery disease (CAD) play antagonizing roles against each other. We previously reported that DQ485454, the short transcript, is downregulated in coronary arteries from CAD patients, and reduces monocyte adhesion to endothelial cells (ECs) and transendothelial monocyte migration (TEM). Interestingly, the longest transcript NR_003529 is significantly upregulated in coronary arteries from CAD patients. Overexpression of ANRIL transcript NR_003529 increases monocyte adhesion to ECs and TEM, whereas knockdown of NR_003529 expression reduces monocyte adhesion to ECs and TEM. Much more dramatic effects were observed for the combination of overexpression of NR_003529 and knockdown of DQ485454 or the combination of knockdown of NR_003529 and overexpression of DQ485454. The antagonizing effects of ANRIL transcripts NR_003529 and DQ485454 were associated with their opposite effects on expression of downstream target genes EZR, CXCL11 or TMEM106B. Our results demonstrate that different transcripts of lncRNA can exert antagonizing effects on biological functions, thereby providing important insights into the biology of lncRNA. The data further support the hypothesis that ANRIL is the causative gene at the 9p21 CAD susceptibility locus.


Subject(s)
Alternative Splicing , Coronary Artery Disease/genetics , Endothelial Cells/metabolism , Gene Expression Regulation , RNA, Long Noncoding/genetics , Biomarkers , Cell Adhesion/genetics , Coronary Artery Disease/diagnosis , Coronary Artery Disease/metabolism , Coronary Vessels/metabolism , Coronary Vessels/pathology , Disease Susceptibility , Gene Knockdown Techniques , Humans , Monocytes/metabolism , Monocytes/pathology , RNA Isoforms , Transendothelial and Transepithelial Migration/genetics
3.
Ann Neurol ; 82(3): 466-478, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28856709

ABSTRACT

OBJECTIVE: Rett syndrome (RTT) and epileptic encephalopathy (EE) are devastating neurodevelopmental disorders with distinct diagnostic criteria. However, highly heterogeneous and overlapping clinical features often allocate patients into the boundary of the two conditions, complicating accurate diagnosis and appropriate medical interventions. Therefore, we investigated the specific molecular mechanism that allows an understanding of the pathogenesis and relationship of these two conditions. METHODS: We screened novel genetic factors from 34 RTT-like patients without MECP2 mutations, which account for ∼90% of RTT cases, by whole-exome sequencing. The biological function of the discovered variants was assessed in cell culture and Xenopus tropicalis models. RESULTS: We identified a recurring de novo variant in GABAB receptor R2 (GABBR2) that reduces the receptor function, whereas different GABBR2 variants in EE patients possess a more profound effect in reducing receptor activity and are more responsive to agonist rescue in an animal model. INTERPRETATION: GABBR2 is a genetic factor that determines RTT- or EE-like phenotype expression depending on the variant positions. GABBR2-mediated γ-aminobutyric acid signaling is a crucial factor in determining the severity and nature of neurodevelopmental phenotypes. Ann Neurol 2017;82:466-478.


Subject(s)
Mutation , Receptors, GABA-B/genetics , Rett Syndrome/genetics , Spasms, Infantile/genetics , Exome , Genotype , HEK293 Cells , Humans , Methyl-CpG-Binding Protein 2/genetics , Phenotype , Signal Transduction/genetics
5.
Sci Transl Med ; 14(648): eabf3136, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35675436

ABSTRACT

Brugada syndrome (BrS) is a fatal arrhythmia that causes an estimated 4% of all sudden death in high-incidence areas. SCN5A encodes cardiac sodium channel NaV1.5 and causes 25 to 30% of BrS cases. Here, we report generation of a knock-in (KI) mouse model of BrS (Scn5aG1746R/+). Heterozygous KI mice recapitulated some of the clinical features of BrS, including an ST segment abnormality (a prominent J wave) on electrocardiograms and development of spontaneous ventricular tachyarrhythmias (VTs), seizures, and sudden death. VTs were caused by shortened cardiac action potential duration and late phase 3 early afterdepolarizations associated with reduced sodium current density (INa) and increased Kcnd3 and Cacna1c expression. We developed a gene therapy using adeno-associated virus serotype 9 (AAV9) vector-mediated MOG1 delivery for up-regulation of MOG1, a chaperone that binds to NaV1.5 and traffics it to the cell surface. MOG1 was chosen for gene therapy because the large size of the SCN5A coding sequence (6048 base pairs) exceeds the packaging capacity of AAV vectors. AAV9-MOG1 gene therapy increased cell surface expression of NaV1.5 and ventricular INa, reversed up-regulation of Kcnd3 and Cacna1c expression, normalized cardiac action potential abnormalities, abolished J waves, and blocked VT in Scn5aG1746R/+ mice. Gene therapy also rescued the phenotypes of cardiac arrhythmias and contractile dysfunction in heterozygous humanized KI mice with SCN5A mutation p.D1275N. Using a small chaperone protein may have broad implications for targeting disease-causing genes exceeding the size capacity of AAV vectors.


Subject(s)
Brugada Syndrome , Cardiomyopathies , Animals , Arrhythmias, Cardiac/therapy , Brugada Syndrome/genetics , Brugada Syndrome/metabolism , Brugada Syndrome/therapy , Cardiomyopathies/genetics , Cardiomyopathies/therapy , Death, Sudden , Disease Models, Animal , Genetic Therapy , Mice , Mutation/genetics , NAV1.5 Voltage-Gated Sodium Channel/genetics , NAV1.5 Voltage-Gated Sodium Channel/metabolism , Protein Transport
6.
J Am Heart Assoc ; 9(7): e014146, 2020 04 07.
Article in English | MEDLINE | ID: mdl-32237974

ABSTRACT

Background Epistasis describes how gene-gene interactions affect phenotypes, and could have a profound impact on human diseases such as coronary artery disease (CAD). The goal of this study was to identify gene-gene interactions in CAD using an easily generalizable multi-stage approach. Methods and Results Our forward genetic approach consists of multiple steps that combine statistical and functional approaches, and analyze information from global gene expression profiling, functional interactions, and genetic interactions to robustly identify gene-gene interactions. Global gene expression profiling shows that knockdown of ANRIL (DQ485454) at 9p21.3 GWAS (genome-wide association studies) CAD locus upregulates TMEM100 and TMEM106B. Functional studies indicate that the increased monocyte adhesion to endothelial cells and transendothelial migration of monocytes, 2 critical processes in the initiation of CAD, by ANRIL knockdown are reversed by knockdown of TMEM106B, but not of TMEM100. Furthermore, the decreased monocyte adhesion to endothelial cells and transendothelial migration of monocytes induced by ANRIL overexpression was reversed by overexpressing TMEM106B. TMEM106B expression was upregulated by >2-fold in CAD coronary arteries. A significant association was found between variants in TMEM106B (but not in TMEM100) and CAD (P=1.9×10-8). Significant gene-gene interaction was detected between ANRIL variant rs2383207 and TMEM106B variant rs3807865 (P=0.009). A similar approach also identifies significant interaction between rs6903956 in ADTRP and rs17465637 in MIA3 (P=0.005). Conclusions We demonstrate 2 pairs of epistatic interactions between GWAS loci for CAD and offer important insights into the genetic architecture and molecular mechanisms for the pathogenesis of CAD. Our strategy has broad applicability to the identification of epistasis in other human diseases.


Subject(s)
Cardiovascular Diseases/genetics , Endothelial Cells/metabolism , Epistasis, Genetic , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/metabolism , Case-Control Studies , Cells, Cultured , Data Interpretation, Statistical , Gene Regulatory Networks , Genetic Predisposition to Disease , Genome-Wide Association Study , Heart Disease Risk Factors , Humans , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Middle Aged , Models, Statistical , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Phenotype , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Risk Assessment , Transcriptome
7.
Front Immunol ; 9: 1984, 2018.
Article in English | MEDLINE | ID: mdl-30279688

ABSTRACT

Bile acids (BAs) control metabolism and inflammation by interacting with several receptors. Here, we report that intravenous infusion of taurodeoxycholate (TDCA) decreases serum pro-inflammatory cytokines, normalizes hypotension, protects against renal injury, and prolongs mouse survival during sepsis. TDCA increases the number of granulocytic myeloid-derived suppressor cells (MDSCLT) distinctive from MDSCs obtained without TDCA treatment (MDSCL) in the spleen of septic mice. FACS-sorted MDSCLT cells suppress T-cell proliferation and confer protection against sepsis when adoptively transferred better than MDSCL. Proteogenomic analysis indicated that TDCA controls chromatin silencing, alternative splicing, and translation of the immune proteome of MDSCLT, which increases the expression of anti-inflammatory molecules such as oncostatin, lactoferrin and CD244. TDCA also decreases the expression of pro-inflammatory molecules such as neutrophil elastase. These findings suggest that TDCA globally edits the proteome to increase the number of MDSCLT cells and affect their immune-regulatory functions to resolve systemic inflammation during sepsis.


Subject(s)
Myeloid-Derived Suppressor Cells/immunology , Sepsis/immunology , T-Lymphocytes/immunology , Taurodeoxycholic Acid/metabolism , Animals , Cell Count , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Gene Expression Regulation , Humans , Immune Tolerance , Leukocyte Elastase/genetics , Leukocyte Elastase/metabolism , Lipopolysaccharides/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oncostatin M/genetics , Oncostatin M/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL