Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
1.
Nat Commun ; 15(1): 4359, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38777835

ABSTRACT

Cystine-knot peptides (CKPs) are naturally occurring peptides that exhibit exceptional chemical and proteolytic stability. We leveraged the CKP carboxypeptidase A1 inhibitor as a scaffold to construct phage-displayed CKP libraries and subsequently screened these collections against HTRA1, a trimeric serine protease implicated in age-related macular degeneration and osteoarthritis. The initial hits were optimized by using affinity maturation strategies to yield highly selective and potent picomolar inhibitors of HTRA1. Crystal structures, coupled with biochemical studies, reveal that the CKPs do not interact in a substrate-like manner but bind to a cryptic pocket at the S1' site region of HTRA1 and abolish catalysis by stabilizing a non-competent active site conformation. The opening and closing of this cryptic pocket is controlled by the gatekeeper residue V221, and its movement is facilitated by the absence of a constraining disulfide bond that is typically present in trypsin fold serine proteases, thereby explaining the remarkable selectivity of the CKPs. Our findings reveal an intriguing mechanism for modulating the activity of HTRA1, and highlight the utility of CKP-based phage display platforms in uncovering potent and selective inhibitors against challenging therapeutic targets.


Subject(s)
Catalytic Domain , High-Temperature Requirement A Serine Peptidase 1 , Peptides , High-Temperature Requirement A Serine Peptidase 1/metabolism , High-Temperature Requirement A Serine Peptidase 1/genetics , Humans , Peptides/chemistry , Peptides/metabolism , Peptides/pharmacology , Peptide Library , Crystallography, X-Ray , Protein Binding , Cystine/chemistry , Cystine/metabolism , Models, Molecular
2.
J Clin Invest ; 134(10)2024 May 15.
Article in English | MEDLINE | ID: mdl-38747292

ABSTRACT

Cerebral small vessel disease (cSVD) encompasses a heterogeneous group of age-related small vessel pathologies that affect multiple regions. Disease manifestations range from lesions incidentally detected on neuroimaging (white matter hyperintensities, small deep infarcts, microbleeds, or enlarged perivascular spaces) to severe disability and cognitive impairment. cSVD accounts for approximately 25% of ischemic strokes and the vast majority of spontaneous intracerebral hemorrhage and is also the most important vascular contributor to dementia. Despite its high prevalence and potentially long therapeutic window, there are still no mechanism-based treatments. Here, we provide an overview of the recent advances in this field. We summarize recent data highlighting the remarkable continuum between monogenic and multifactorial cSVDs involving NOTCH3, HTRA1, and COL4A1/A2 genes. Taking a vessel-centric view, we discuss possible cause-and-effect relationships between risk factors, structural and functional vessel changes, and disease manifestations, underscoring some major knowledge gaps. Although endothelial dysfunction is rightly considered a central feature of cSVD, the contributions of smooth muscle cells, pericytes, and other perivascular cells warrant continued investigation.


Subject(s)
Cerebral Small Vessel Diseases , Collagen Type IV , Receptor, Notch3 , Humans , Cerebral Small Vessel Diseases/genetics , Cerebral Small Vessel Diseases/physiopathology , Cerebral Small Vessel Diseases/pathology , Receptor, Notch3/genetics , Receptor, Notch3/metabolism , Collagen Type IV/genetics , Collagen Type IV/metabolism , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , Animals
3.
Invest Ophthalmol Vis Sci ; 65(4): 34, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38648039

ABSTRACT

Purpose: The purpose of this study was to determine if levels of the HtrA1 protein in serum or vitreous humor are influenced by genetic risk for age-related macular degeneration (AMD) at the 10q26 locus, age, sex, AMD status, and/or AMD disease severity, and, therefore, to determine the contribution of systemic and ocular HtrA1 to the AMD disease process. Methods: A custom-made sandwich ELISA assay (SCTM ELISA) for detection of the HtrA1 protein was designed and compared with three commercial assays (R&D Systems, MyBiosource 1 and MyBiosource 2) using 65 serum samples. Concentrations of HtrA1 were thereafter determined in serum and vitreous samples collected from 248 individuals and 145 human donor eyes, respectively. Results: The SCTM ELISA demonstrated high specificity, good recovery, and parallelism within its linear detection range and performed comparably to the R&D Systems assay. In contrast, we were unable to demonstrate the specificity of the two assays from MyBioSource using either recombinant or native HtrA1. Analyses of concentrations obtained using the validated SCTM assay revealed that genetic risk at the 10q26 locus, age, sex, or AMD status are not significantly associated with altered levels of the HtrA1 protein in serum or in vitreous humor (P > 0.05). Conclusions: HtrA1 levels in serum and vitreous do not reflect the risk for AMD associated with the 10q26 locus or disease status. Localized alteration in HTRA1 expression in the retinal pigment epithelium, rather than systemic changes in HtrA1, is the most likely driver of elevated risk for developing AMD among individuals with risk variants at the 10q26 locus.


Subject(s)
High-Temperature Requirement A Serine Peptidase 1 , Macular Degeneration , Serine Endopeptidases , Vitreous Body , Aged , Female , Humans , Male , Chromosomes, Human, Pair 10/genetics , Enzyme-Linked Immunosorbent Assay/methods , Genetic Predisposition to Disease , High-Temperature Requirement A Serine Peptidase 1/blood , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , Macular Degeneration/genetics , Macular Degeneration/metabolism , Macular Degeneration/diagnosis , Risk Factors , Sensitivity and Specificity , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Vitreous Body/metabolism
4.
Front Biosci (Schol Ed) ; 16(1): 3, 2024 Feb 29.
Article in English | MEDLINE | ID: mdl-38538345

ABSTRACT

Age-related macular degeneration (AMD) is a multifactorial genetic disease, with at least 52 identifiable associated gene variants at 34 loci, including variants in complement factor H (CFH) and age-related maculopathy susceptibility 2/high-temperature requirement A serine peptidase-1 (ARMS2/HTRA1). Genetic factors account for up to 70% of disease variability. However, population-based genetic risk scores are generally more helpful for clinical trial design and stratification of risk groups than for individual patient counseling. There is some evidence of pharmacogenetic influences on various treatment modalities used in AMD patients, including Age-Related Eye Disease Study (AREDS) supplements, photodynamic therapy (PDT), and anti-vascular endothelial growth factor (anti-VEGF) agents. However, there is currently no convincing evidence that genetic information plays a role in routine clinical care.


Subject(s)
Macular Degeneration , Proteins , Humans , Macular Degeneration/drug therapy , Macular Degeneration/genetics , Dietary Supplements , High-Temperature Requirement A Serine Peptidase 1/genetics , Vascular Endothelial Growth Factors/genetics , Vascular Endothelial Growth Factors/therapeutic use , Polymorphism, Single Nucleotide , Risk Factors
5.
Nat Commun ; 15(1): 2436, 2024 Mar 18.
Article in English | MEDLINE | ID: mdl-38499535

ABSTRACT

Parkinson's disease (PD) is closely linked to α-synuclein (α-syn) misfolding and accumulation in Lewy bodies. The PDZ serine protease HTRA1 degrades fibrillar tau, which is associated with Alzheimer's disease, and inactivating mutations to mitochondrial HTRA2 are implicated in PD. Here, we report that HTRA1 inhibits aggregation of α-syn as well as FUS and TDP-43, which are implicated in amyotrophic lateral sclerosis (ALS) and frontotemporal dementia. The protease domain of HTRA1 is necessary and sufficient for inhibiting aggregation, yet this activity is proteolytically-independent. Further, HTRA1 disaggregates preformed α-syn fibrils, rendering them incapable of seeding aggregation of endogenous α-syn, while reducing HTRA1 expression promotes α-syn seeding. HTRA1 remodels α-syn fibrils by targeting the NAC domain, the key domain catalyzing α-syn amyloidogenesis. Finally, HTRA1 detoxifies α-syn fibrils and prevents formation of hyperphosphorylated α-syn accumulations in primary neurons. Our findings suggest that HTRA1 may be a therapeutic target for a range of neurodegenerative disorders.


Subject(s)
Parkinson Disease , alpha-Synuclein , Humans , alpha-Synuclein/genetics , alpha-Synuclein/metabolism , Amyloid/metabolism , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , Parkinson Disease/genetics , Parkinson Disease/metabolism , Lewy Bodies/metabolism
6.
Int J Mol Sci ; 25(6)2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38542204

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of blindness in the elderly worldwide. The prevalence and phenotypes of AMD differ among populations, including between people in Taiwan and other regions. We performed a genome-wide association study to identify genetic variants and to develop genetic models to predict the risk of AMD development and progression in the Taiwanese population. In total, 4039 patients with AMD and 16,488 non-AMD controls (aged ≥ 65 years) were included. We identified 31 AMD-associated variants (p < 5 × 10-8) on chromosome 10q26, surrounding PLEKHA1-ARMS2-HTRA1. Two genetic models were constructed using the clump and threshold method. Model 1 included the single nucleotide polymorphism rs11200630 and showed a 1.31-fold increase in the risk of AMD per risk allele (95% confidence interval (CI) = 1.20-1.43, p < 0.001). In model 2, 1412 single-nucleotide polymorphisms were selected to construct a polygenic risk score (PRS). Individuals with the top 5% PRS had a 1.40-fold higher AMD risk compared with that of individuals with a PRS in the bottom quartile (95% CI = 1.04-1.89, p = 0.025). Moreover, the PRS in the upper quartile was related to a decreased age at AMD diagnosis by 0.62 years (95% CI = -1.15, -0.09, p = 0.023). Both genetic models provide useful predictive power for populations at high risk of AMD, affording a basis for identifying patients requiring close follow-up and early intervention.


Subject(s)
Macular Degeneration , Proteins , Aged , Humans , Proteins/genetics , Genome-Wide Association Study , Macular Degeneration/diagnosis , Macular Degeneration/epidemiology , Macular Degeneration/genetics , High-Temperature Requirement A Serine Peptidase 1/genetics , Polymorphism, Single Nucleotide , Early Diagnosis , Genetic Predisposition to Disease , Risk Factors , Genotype
7.
JAMA Neurol ; 81(5): 551-552, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38466301

ABSTRACT

This case report describes the evaluation of a 44-year-old man with a history of headache, dizziness, and imbalance and imaging that showed lacunar infarctions and bilateral white matter hyperintensities.


Subject(s)
Cerebral Small Vessel Diseases , High-Temperature Requirement A Serine Peptidase 1 , Humans , High-Temperature Requirement A Serine Peptidase 1/genetics , Cerebral Small Vessel Diseases/genetics , Cerebral Small Vessel Diseases/diagnostic imaging , Cerebral Hemorrhage/genetics , Cerebral Hemorrhage/diagnostic imaging , Male , Heterozygote , Female , Middle Aged
8.
Tissue Cell ; 87: 102329, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38367326

ABSTRACT

High-temperature requirement A1 (HtrA1), a multidomain serine protease acting on Extracellular matrix (ECM) rearrangement, is also secreted by osteoblasts and osteoclasts. Recent and conflicting literature highlights HtrA1's role as a controller of bone remodeling, proposing it as a possible target for pathologies with unbalanced bone resorption, like Osteoporosis (OP). To add knowledge on this molecule function in bone physiopathology, here we compared HtrA1 distribution in the ECM of healthy (H) and OP bone tissue, also examining its localization in the sites of new bone formation. HtrA1 was homogeneously expressed in the mature bone ECM of H tissue showing a 55.6 ± 16.4% of the stained area, with a significant (p=0.0001) decrease in OP percentage stained area (21.1 ± 13.1). Moreover, HtrA1 was present in the endosteum and cells involved in osteogenesis, mainly in those "entrapped" in woven bone, whereas osteocytes in mature lamellar bone were negative. Based on our previous observation in OP tissue of a significantly increased expression of Decorin and Osteocalcin, both involved in bone mineralization and remodeling and equally substrates for HtrA1, we speculate that HtrA1 by controlling the proper amount of Decorin and Osteocalcin favors normal bone maturation and mineralization. Besides, we suggest that late-osteoblasts and pre-osteocytes secrete HtrA1 in the adjacent matrix whilst proceeding with their maturation and that HtrA1 expression is further modified during the remodeling from woven to the lamellar bone. Overall, our data suggest HtrA1 as a positive regulator of bone matrix formation and maturation: its reduced expression in mature OP bone, affecting protein content and distribution, could hamper correct bone remodeling and mineralization.


Subject(s)
Osteoporosis , Serine Proteases , Humans , Osteocalcin/metabolism , Serine Proteases/metabolism , Bone Matrix/metabolism , Decorin/metabolism , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Bone and Bones/metabolism , Extracellular Matrix/metabolism , Osteoporosis/genetics
9.
Adv Exp Med Biol ; 1415: 27-36, 2023.
Article in English | MEDLINE | ID: mdl-37440010

ABSTRACT

Age-related macular degeneration (AMD) is the leading cause of blindness in the global aging population. Familial aggregation and genome-wide association (GWA) studies have identified gene variants associated with AMD, implying a strong genetic contribution to AMD development. Two loci, on human Chr 1q31 and 10q26, respectively, represent the most influential of all genetic factors. While the role of CFH at Chr 1q31 is well established, uncertainty remains about the genes ARMS2 and HTRA1, at the Chr 10q26 locus. Since both genes are in strong linkage disequilibrium, assigning individual gene effects is difficult. In this chapter, we review current literature about ARMS2 and HTRA1 and their relevance to AMD risk. Future studies will be necessary to unravel the mechanisms by which they contribute to AMD.


Subject(s)
Macular Degeneration , Proteins , Humans , Aged , Proteins/genetics , Serine Endopeptidases/genetics , Genome-Wide Association Study , High-Temperature Requirement A Serine Peptidase 1/genetics , Macular Degeneration/genetics , Linkage Disequilibrium , Polymorphism, Single Nucleotide , Complement Factor H/genetics , Genotype
10.
J Stroke Cerebrovasc Dis ; 32(8): 107225, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37348440

ABSTRACT

Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy (CARASIL) is an extremely rare hereditary cerebral small vessel disease caused by homozygous or compound heterozygous mutations in the gene coding for high-temperature requirement A serine peptidase 1 (HtrA1). Given the rare nature of the disease, delays in diagnosis and misdiagnosis are not uncommon. In this article, we reported the first case of CARASIL from Saudi Arabia with a novel homozygous variant c.1156C>T in exon 7 of the HTRA1 gene. The patient was initially misdiagnosed with primary progressive multiple sclerosis and treated with rituximab. CARASIL should be considered in the differential diagnosis of patients with suspected atypical progressive multiple sclerosis who have additional signs such as premature scalp alopecia and low back pain with diffuse white matter lesions in brain MRI. Genetic testing is important to confirm the diagnosis.


Subject(s)
Cerebral Arterial Diseases , Cerebrovascular Disorders , Leukoencephalopathies , Multiple Sclerosis , Humans , Cerebral Infarction/diagnostic imaging , Cerebral Infarction/genetics , Cerebral Infarction/pathology , Leukoencephalopathies/diagnostic imaging , Leukoencephalopathies/genetics , Cerebrovascular Disorders/genetics , Alopecia/diagnosis , Alopecia/genetics , Mutation , High-Temperature Requirement A Serine Peptidase 1/genetics
11.
Proc Natl Acad Sci U S A ; 120(19): e2215005120, 2023 05 09.
Article in English | MEDLINE | ID: mdl-37126685

ABSTRACT

Genome-wide association studies (GWAS) have identified genetic risk loci for age-related macular degeneration (AMD) on the chromosome 10q26 (Chr10) locus and are tightly linked: the A69S (G>T) rs10490924 single-nucleotide variant (SNV) and the AATAA-rich insertion-deletion (indel, del443/ins54), which are found in the age-related maculopathy susceptibility 2 (ARMS2) gene, and the G512A (G>A) rs11200638 SNV, which is found in the high-temperature requirement A serine peptidase 1 (HTRA1) promoter. The fourth variant is Y402H complement factor H (CFH), which directs CFH signaling. CRISPR manipulation of retinal pigment epithelium (RPE) cells may allow one to isolate the effects of the individual SNV and thus identify SNV-specific effects on cell phenotype. Clustered regularly interspaced short palindromic repeats (CRISPR) editing demonstrates that rs10490924 raised oxidative stress in induced pluripotent stem cell (iPSC)-derived retinal cells from patients with AMD. Sodium phenylbutyrate preferentially reverses the cell death caused by ARMS2 rs10490924 but not HTRA1 rs11200638. This study serves as a proof of concept for the use of patient-specific iPSCs for functional annotation of tightly linked GWAS to study the etiology of a late-onset disease phenotype. More importantly, we demonstrate that antioxidant administration may be useful for reducing reactive oxidative stress in AMD, a prevalent late-onset neurodegenerative disorder.


Subject(s)
Induced Pluripotent Stem Cells , Macular Degeneration , Humans , High-Temperature Requirement A Serine Peptidase 1/genetics , Induced Pluripotent Stem Cells/metabolism , Clustered Regularly Interspaced Short Palindromic Repeats , Proteins/metabolism , Serine Endopeptidases/genetics , Genome-Wide Association Study , Macular Degeneration/genetics , Oxidative Stress , Polymorphism, Single Nucleotide , Complement Factor H/genetics , Genotype
12.
In Vitro Cell Dev Biol Anim ; 59(3): 166-178, 2023 Mar.
Article in English | MEDLINE | ID: mdl-37017808

ABSTRACT

The present study identified a novel upstream long chain non-coding (lncRNA) NEAT1/miR-141-3p/HTRA1 axis that regulated the activation of NLR family pyrin domain containing 3 (NLRP3) inflammasome to modulate endometriosis (EM) development. Specifically, clinical data suggested that the expression of NLRP3 and apoptosis-associated speck-like protein containing CARD (ASC), the cleavage of caspase-1 and gasdermin D (GSDMD), and the production of inflammatory cytokines (interleukin (IL)-1ß, IL-6, tumor necrosis factor (TNF)-α, and IL-18) were all significantly increased in the ectopic endometrium (EE) tissues, compared to the normal endometrium (NE) tissues. Then, through analyzing the datasets from GEO database (GSE2339, GSE58178, and GSE7305) using the GEO2R bioinformatics tools, we verified that HtrA Serine Peptidase 1 (HTRA1) was especially enriched in the EE tissues compared to the NE tissues. To further confirm the biological functions of HTRA1, HTRA1 was overexpressed or downregulated in primary human endometrial stromal cells (hESCs) isolated from NE tissues or EE tissues, respectively. The results showed that upregulation of HTRA1 activated NLRP3 inflammasome-mediated pyroptotic cell death and cellular inflammation in NE-derived hESCs, whereas silencing of HTRA1 played an opposite role in EE-derived hESCs. In addition, the lncRNA NEAT1/miR-141-3p axis was screened as the upstream regulator of HTRA1. Mechanistically, lncRNA NEAT1 sponged miR-141-3p to positively regulate HTRA1 in a competing endogenous RNA (ceRNA) mechanisms-dependent manner. The recovery experiments in hESCs from NE and EE tissues confirmed that lncRNA NEAT1 overexpression promoted NLRP3 inflammasome-mediated pyroptotic cell death through regulating the miR-141-3p/HTRA1 axis. Taken together, this study firstly uncovered the underlying mechanisms by which a novel lncRNA NEAT1/miR-141-3p/HTRA1-NLRP3 pathway contributed to the development of EM, which provided novel diagnostic and therapeutic biomarkers for this disease.


Subject(s)
Endometriosis , MicroRNAs , RNA, Long Noncoding , Animals , Female , Humans , Endometriosis/genetics , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , Inflammasomes/genetics , Inflammasomes/metabolism , Inflammation/genetics , Inflammation/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Pyroptosis , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/genetics
15.
J Appl Toxicol ; 43(8): 1214-1224, 2023 08.
Article in English | MEDLINE | ID: mdl-36861143

ABSTRACT

Inorganic arsenic (iAs) has been a human health concern and is associated with intestinal malignancies. However, the molecular mechanisms of the iAs-induced oncogenic process in intestine epithelial cells remain elusive, partly because of the known hormesis effect of arsenic. Here, we established that six-month exposure to iAs at a concentration similar to those found in contaminated drinking water could promote malignant characteristics, including enhanced proliferation and migration, resistance to apoptosis, and mesenchymal-like transition in Caco-2 cells. Transcriptome analysis and mechanism study revealed that key genes and pathways involved in cell adhesion, inflammation and oncogenic regulation were altered during chronic iAs exposure. Specifically, we uncovered that down-regulation of HTRA1 was essential for the iAs-induced acquisition of the cancer hallmarks. Further, we evidenced that the loss of HTRA1 during iAs-exposure could be restored by HDAC6 inhibition. Caco-2 cells with chronic exposure to iAs exhibited enhanced sensitivity to WT-161, a specific inhibitor of HDAC6, when used alone than in combination with a chemotherapeutic agent. These findings provide valuable information for understanding the mechanisms of arsenic-induced carcinogenesis and facilitating the health management of populations in arsenic-polluted areas.


Subject(s)
Arsenic , High-Temperature Requirement A Serine Peptidase 1 , Histone Deacetylase 6 , Humans , Arsenic/analysis , Caco-2 Cells , Carcinogenesis , Down-Regulation , Drinking Water/analysis , Histone Deacetylase 6/genetics , Histone Deacetylase 6/metabolism , High-Temperature Requirement A Serine Peptidase 1/genetics
16.
Molecules ; 28(5)2023 Feb 28.
Article in English | MEDLINE | ID: mdl-36903482

ABSTRACT

Inflammation and elevated expression of high temperature requirement A serine peptidase 1 (HTRA1) are known high risk factors for age-related macular degeneration (AMD). However, the specific mechanism that HTRA1 causes AMD and the relationship between HTRA1 and inflammation remains unclear. We found that lipopolysaccharide (LPS) induced inflammation enhanced the expression of HTRA1, NF-κB, and p-p65 in ARPE-19 cells. Overexpression of HTRA1 up-regulated NF-κB expression, and on the other hand knockdown of HTRA1 down-regulated the expression of NF-κB. Moreover, NF-κB siRNA has no significant effect on the expression of HTRA1, suggesting HTRA1 works upstream of NF-κB. These results demonstrated that HTRA1 plays a pivotal role in inflammation, explaining possible mechanism of overexpressed HTRA1-induced AMD. Celastrol, a very common anti-inflammatory and antioxidant drug, was found to suppress inflammation by inhibiting phosphorylation of p65 protein efficaciously in RPE cells, which may be applied to the therapy of age-related macular degeneration.


Subject(s)
Macular Degeneration , NF-kappa B , Humans , NF-kappa B/metabolism , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , High-Temperature Requirement A Serine Peptidase 1/pharmacology , Lipopolysaccharides/pharmacology , Retinal Pigment Epithelium/metabolism , Macular Degeneration/metabolism , Inflammation/drug therapy , Epithelial Cells/metabolism , Retinal Pigments/metabolism
17.
J Biomed Inform ; 141: 104345, 2023 05.
Article in English | MEDLINE | ID: mdl-36958462

ABSTRACT

Stroke is the second largest cause of mortality in the world. Genome-wide association studies (GWAS) have identified some genetic variants associated with stroke risk, but their putative functional causal genes are unknown. Hence, we aimed to identify putative functional causal gene biomarkers of stroke risk. We used a summary-based Mendelian randomisation (SMR) approach to identify the pleiotropic associations of genetically regulated traits (i.e., gene expression and DNA methylation) with stroke risk. Using SMR approach, we integrated cis-expression quantitative loci (cis-eQTLs) and cis-methylation quantitative loci (cis-mQTLs) data with GWAS summary statistics of stroke. We also utilised heterogeneity in dependent instruments (HEIDI) test to distinguish pleiotropy from linkage from the observed associations identified through SMR analysis. Our integrative SMR analyses and HEIDI test revealed 45 candidate biomarker genes (FDR < 0.05; PHEIDI > 0.01) that were pleiotropically or potentially causally associated with stroke risk. Of those candidate biomarker genes, 10 genes (HTRA1, PMF1, FBN2, C9orf84, COL4A1, BAG4, NEK6, SH2B3, SH3PXD2A, ACAD10) were differentially expressed in genome-wide blood transcriptomics data from stroke and healthy individuals (FDR < 0.05). Functional enrichment analysis of the identified candidate biomarker genes revealed gene ontologies and pathways involved in stroke, including "cell aging", "metal ion binding" and "oxidative damage". Based on the evidence of genetically regulated expression of genes through SMR and directly measured expression of genes in blood, our integrative analysis suggests ten genes as blood biomarkers of stroke risk. Furthermore, our study provides a better understanding of the influence of DNA methylation on the expression of genes linked to stroke risk.


Subject(s)
Stroke , Systems Biology , Humans , Genome-Wide Association Study , Phenotype , Stroke/diagnosis , Stroke/genetics , Genetic Markers , Genetic Predisposition to Disease , Polymorphism, Single Nucleotide , NIMA-Related Kinases/genetics , High-Temperature Requirement A Serine Peptidase 1/genetics , Acyl-CoA Dehydrogenase/genetics
18.
J Hypertens ; 41(4): 587-596, 2023 04 01.
Article in English | MEDLINE | ID: mdl-36651169

ABSTRACT

BACKGROUND: It has been reported that the alteration of circular RNAs (circRNAs) during preeclampsia (PE) can be associated with the pathogenesis of this disease. Herein, this work investigated the potential functions and mechanism of circ_0001326 in PE process. METHODS: The levels of genes and proteins were evaluated by quantitative real-time PCR (qRT-PCR) and western blotting. The functional experiments were conducted using cell counting kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), flow cytometry, transwell, and wound healing assays, respectively. The binding between miR-188-3p and circ_0001326 or HtrA serine peptidase 1 (HTRA1) was verified by bioinformatics analysis and dual-luciferase reporter assays. RESULTS: Circ_0001326 and HTRA1 expression was increased, while miR-188-3p expression was decreased in the placental tissues of preeclamptic singleton pregnant women compared with the normal pregnant women. Functionally, up-regulation of circ_0001326 or HTRA1, or down-regulation of miR-188-3p led to the arrest of cell growth, invasion, migration and epithelial-mesenchymal transition (EMT) process in trophoblast cells. Mechanistically, circ_0001326 acted as a sponge for miR-188-3p, which directly targeted HTRA1. Moreover, circ_0001326 could regulate HTRA1 through sequestering miR-188-3p. A series of rescue experiments showed that miR-188-3p reversed the inhibitory effects of circ_0001326 knockdown on above behaviors of trophoblast cells. Besides that, HTRA1 silencing attenuated the action of miR-188-3p inhibitor on trophoblast cell phenotype alteration. CONCLUSION: Our study demonstrated that circ_0001326 could promote trophoblast cell proliferation, invasion, migration and EMT in PE by miR-188-3p/HTRA1 axis, indicating a novel insight into the pathogenesis of PE.


Subject(s)
MicroRNAs , Pre-Eclampsia , Female , Humans , Pregnancy , Trophoblasts , Epithelial-Mesenchymal Transition/genetics , Placenta , Pre-Eclampsia/genetics , Cell Proliferation/genetics , Serine , MicroRNAs/genetics , Cell Movement/genetics , High-Temperature Requirement A Serine Peptidase 1/genetics
19.
JCI Insight ; 8(4)2023 02 22.
Article in English | MEDLINE | ID: mdl-36656640

ABSTRACT

Heart failure (HF) is characterized by global alterations in myocardial DNA methylation, yet little is known about the epigenetic regulation of the noncoding genome and potential reversibility of DNA methylation with left ventricular assist device (LVAD) therapy. Genome-wide mapping of myocardial DNA methylation in 36 patients with HF at LVAD implantation, 8 patients at LVAD explantation, and 7 nonfailing (NF) donors using a high-density bead array platform identified 2,079 differentially methylated positions (DMPs) in ischemic cardiomyopathy (ICM) and 261 DMPs in nonischemic cardiomyopathy (NICM). LVAD support resulted in normalization of 3.2% of HF-associated DMPs. Methylation-expression correlation analysis yielded several protein-coding genes that are hypomethylated and upregulated (HTRA1, FBXO16, EFCAB13, and AKAP13) or hypermethylated and downregulated (TBX3) in HF. A potentially novel cardiac-specific super-enhancer long noncoding RNA (lncRNA) (LINC00881) is hypermethylated and downregulated in human HF. LINC00881 is an upstream regulator of sarcomere and calcium channel gene expression including MYH6, CACNA1C, and RYR2. LINC00881 knockdown reduces peak calcium amplitude in the beating human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). These data suggest that HF-associated changes in myocardial DNA methylation within coding and noncoding genomes are minimally reversible with mechanical unloading. Epigenetic reprogramming strategies may be necessary to achieve sustained clinical recovery from heart failure.


Subject(s)
Cardiomyopathies , F-Box Proteins , Heart Failure , Induced Pluripotent Stem Cells , Humans , Epigenesis, Genetic , DNA Methylation , Induced Pluripotent Stem Cells/metabolism , Heart Failure/genetics , Myocytes, Cardiac/metabolism , Cardiomyopathies/metabolism , F-Box Proteins/metabolism , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism
20.
J Cell Physiol ; 238(3): 566-581, 2023 03.
Article in English | MEDLINE | ID: mdl-36715607

ABSTRACT

Nuclear protein 1 (NUPR1) is a stress-induced protein activated by various stresses, such as inflammation and oxidative stress. We previously reported that Nupr1 deficiency increased bone volume by enhancing bone formation in 11-week-old mice. Analysis of differentially expressed genes between wild-type (WT) and Nupr1-knockout (Nupr1-KO) osteocytes revealed that high temperature requirement A 1 (HTRA1), a serine protease implicated in osteogenesis and transforming growth factor-ß signaling was markedly downregulated in Nupr1-KO osteocytes. Nupr1 deficiency also markedly reduced HtrA1 expression, but enhanced SMAD1 signaling in in vitro-cultured primary osteoblasts. In contrast, Nupr1 overexpression enhanced HtrA1 expression in osteoblasts, suggesting that Nupr1 regulates HtrA1 expression, thereby suppressing osteoblastogenesis. Since HtrA1 is also involved in cellular senescence and age-related diseases, we analyzed aging-related bone loss in Nupr1-KO mice. Significant spine trabecular bone loss was noted in WT male and female mice during 6-19 months of age, whereas aging-related trabecular bone loss was attenuated, especially in Nupr1-KO male mice. Moreover, cellular senescence-related markers were upregulated in the osteocytes of 6-19-month-old WT male mice but markedly downregulated in the osteocytes of 19-month-old Nupr1-KO male mice. Oxidative stress-induced cellular senescence stimulated Nupr1 and HtrA1 expression in in vitro-cultured primary osteoblasts, and Nupr1 overexpression enhanced p16ink4a expression in osteoblasts. Finally, NUPR1 expression in osteocytes isolated from the bones of patients with osteoarthritis was correlated with age. Collectively, these results indicate that Nupr1 regulates HtrA1-mediated osteoblast differentiation and senescence. Our findings unveil a novel Nupr1/HtrA1 axis, which may play pivotal roles in bone formation and age-related bone loss.


Subject(s)
Bone and Bones , Down-Regulation , High-Temperature Requirement A Serine Peptidase 1 , Osteoporosis , Signal Transduction , Smad1 Protein , Animals , Female , Male , Mice , Bone and Bones/metabolism , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 1/metabolism , Mice, Knockout , Osteoblasts/metabolism , Osteocytes/metabolism , Osteogenesis , Osteoporosis/metabolism , Osteoporosis/prevention & control , Smad1 Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...