Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Ann Rheum Dis ; 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38479789

ABSTRACT

OBJECTIVES: Osteoarthritis is a complex disease with a huge public health burden. Genome-wide association studies (GWAS) have identified hundreds of osteoarthritis-associated sequence variants, but the effector genes underpinning these signals remain largely elusive. Understanding chromosome organisation in three-dimensional (3D) space is essential for identifying long-range contacts between distant genomic features (e.g., between genes and regulatory elements), in a tissue-specific manner. Here, we generate the first whole genome chromosome conformation analysis (Hi-C) map of primary osteoarthritis chondrocytes and identify novel candidate effector genes for the disease. METHODS: Primary chondrocytes collected from 8 patients with knee osteoarthritis underwent Hi-C analysis to link chromosomal structure to genomic sequence. The identified loops were then combined with osteoarthritis GWAS results and epigenomic data from primary knee osteoarthritis chondrocytes to identify variants involved in gene regulation via enhancer-promoter interactions. RESULTS: We identified 345 genetic variants residing within chromatin loop anchors that are associated with 77 osteoarthritis GWAS signals. Ten of these variants reside directly in enhancer regions of 10 newly described active enhancer-promoter loops, identified with multiomics analysis of publicly available chromatin immunoprecipitation sequencing (ChIP-seq) and assay for transposase-accessible chromatin using sequencing (ATAC-seq) data from primary knee chondrocyte cells, pointing to two new candidate effector genes SPRY4 and PAPPA (pregnancy-associated plasma protein A) as well as further support for the gene SLC44A2 known to be involved in osteoarthritis. For example, PAPPA is directly associated with the turnover of insulin-like growth factor 1 (IGF-1) proteins, and IGF-1 is an important factor in the repair of damaged chondrocytes. CONCLUSIONS: We have constructed the first Hi-C map of primary human chondrocytes and have made it available as a resource for the scientific community. By integrating 3D genomics with large-scale genetic association and epigenetic data, we identify novel candidate effector genes for osteoarthritis, which enhance our understanding of disease and can serve as putative high-value novel drug targets.

2.
EMBO Rep ; 17(11): 1641-1656, 2016 11.
Article in English | MEDLINE | ID: mdl-27702986

ABSTRACT

Reprogramming of somatic cells to induced pluripotent stem cells rewrites the code of cell fate at the chromatin level. Yet, little is known about this process physically. Here, we describe a fluorescence recovery after photobleaching method to assess the dynamics of heterochromatin/euchromatin and show significant heterochromatin loosening at the initial stage of reprogramming. We identify growth arrest and DNA damage-inducible protein a (Gadd45a) as a chromatin relaxer in mouse embryonic fibroblasts, which also enhances somatic cell reprogramming efficiency. We show that residue glycine 39 (G39) in Gadd45a is essential for interacting with core histones, opening chromatin and enhancing reprogramming. We further demonstrate that Gadd45a destabilizes histone-DNA interactions and facilitates the binding of Yamanaka factors to their targets for activation. Our study provides a method to screen factors that impact on chromatin structure in live cells, and identifies Gadd45a as a chromatin relaxer.


Subject(s)
Cell Cycle Proteins/genetics , Cellular Reprogramming , Heterochromatin/metabolism , Induced Pluripotent Stem Cells/physiology , Nuclear Proteins/genetics , Animals , Cell Cycle Proteins/metabolism , Cell Differentiation/genetics , Cellular Reprogramming/genetics , DNA/genetics , DNA/metabolism , Embryonic Stem Cells/metabolism , Fibroblasts/metabolism , Glycine/metabolism , Heterochromatin/genetics , Histones/genetics , Histones/metabolism , Induced Pluripotent Stem Cells/metabolism , Mice , Nuclear Proteins/metabolism , Photobleaching
3.
Stem Cells ; 34(1): 83-92, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26388522

ABSTRACT

Somatic cell reprogramming is accompanied by changes in lipid metabolism. While attempting to dissect the molecular mechanisms of the lipid metabolic switch during reprogramming, we found that overexpression of sterol regulatory element binding protein-1 (Srebp-1), a transcriptional factor required for lipid homeostasis, enhances reprogramming efficiency, while knockdown or pharmaceutical inhibition of Srebp-1 is inhibitory. Srebp-1 overexpression blocks the formation of partially reprogrammed cells, and functions in the early phase of reprogramming. Furthermore, Srebp-1 functions in nucleus and depends on its transcriptional activity but not its ability to bind the E-box motif and regulation of canonical targets. Mechanistically, Srebp-1 interacts with c-Myc, facilitates its binding to downstream pluripotent targets, strengthens the function of c-Myc in enhancing other Yamanaka factors' binding, and thereby promotes the expression of pluripotent genes. These results elucidate a novel role for Srebp-1 in somatic cell reprogramming and provide insights into understanding the metabolic switch during reprogramming.


Subject(s)
Cellular Reprogramming , Proto-Oncogene Proteins c-myc/metabolism , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Cellular Reprogramming/genetics , Gene Expression Regulation , Mice , Pluripotent Stem Cells/metabolism , Protein Binding
4.
Biophys J ; 109(5): 892-9, 2015 Sep 01.
Article in English | MEDLINE | ID: mdl-26331247

ABSTRACT

Mitochondria are highly dynamic cell organelles. Continual cycles of fusion and fission play an important role in mitochondrial metabolism and cellular signaling. Previously, a novel mitochondrial morphology, the donut, was reported in cells after hypoxia-reoxygenation or osmotic pressure changes. However, the mechanism of donut formation remained elusive. Here, we obtained the distribution of donut diameters (D = 2R) and found that 95% are >0.8 µm. We also performed highly precise measurements of the mitochondrial tubule diameters using superresolution and electron microscopy. Then, we set up a model by calculating the mitochondrial bending energy and osmotic potential during donut formation. It shows that the bending energy is increased as the radius of curvature, R, gets smaller in the process of donut formation, especially for radii <0.4 µm, creating a barrier to donut formation. The calculations also show that osmotic potential energy release can balance the rising bending energy through volume expansion. Finally, we revealed the donut formation process in a Gibbs free-energy-dependent model combining calculations and measurements.


Subject(s)
Mitochondria/metabolism , Models, Biological , Animals , Biomechanical Phenomena , Cell Hypoxia , Cell Line , Mice , Microscopy , Osmotic Pressure , Oxygen/metabolism , Thermodynamics
5.
Biochem Biophys Res Commun ; 444(3): 411-4, 2014 Feb 14.
Article in English | MEDLINE | ID: mdl-24472544

ABSTRACT

Tbx3, a member of T-box gene family, has been reported to play critical roles in embryonic development and cell fate determination. In mammalian tissues, Tbx3 is expressed as two isoforms called Tbx3 and Tbx3+2a. However, the differences between the two isoforms in pluripotency maintaining remain obscure. Here we show that both Tbx3 and Tbx3+2a are highly expressed in mouse embryonic stem cells (mESCs) or induced pluripotent stem cells (iPSCs). Overexpression of either Tbx3 or Tbx3+2a could induce the differentiation of mESCs. Mechanistic studies suggest both Tbx3 and Tbx3+2a inhibit the transcriptional activity of pluripotency related transcription factor Nanog. Moreover, Tbx3+2a could directly interact with Nanog while Tbx3 couldn't, indicating the requirement of the 2a domain in Nanog binding. In summary, our results not only reveal the essential roles of Tbx3 and Tbx3+2a in pluripotency maintaining, but also point out the different mechanistic modes between these two isoforms.


Subject(s)
Embryonic Stem Cells/cytology , Homeodomain Proteins/physiology , Pluripotent Stem Cells/cytology , Protein Isoforms/physiology , T-Box Domain Proteins/physiology , Transcription, Genetic/physiology , Animals , Base Sequence , Cells, Cultured , DNA Primers , Embryonic Stem Cells/metabolism , HEK293 Cells , Humans , Mice , Nanog Homeobox Protein , Pluripotent Stem Cells/metabolism , Polymerase Chain Reaction , Protein Binding
6.
Biol Reprod ; 90(6): 119, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24648398

ABSTRACT

Mammalian spermatogenesis is regulated by coordinated gene expression in a spatiotemporal manner. The spatiotemporal regulation of major sperm proteins plays important roles during normal development of the male gamete, of which the underlying molecular mechanisms are poorly understood. A-kinase anchoring protein 3 (AKAP3) is one of the major components of the fibrous sheath of the sperm tail that is formed during spermiogenesis. In the present study, we analyzed the expression of sperm-specific Akap3 and the potential regulatory factors of its protein synthesis during mouse spermiogenesis. Results showed that the transcription of Akap3 precedes its protein synthesis by about 2 wk. Nascent AKAP3 was found to form protein complex with PKA and RNA binding proteins (RBPs), including PIWIL1, PABPC1, and NONO, as revealed by coimmunoprecipitation and protein mass spectrometry. RNA electrophoretic gel mobility shift assay showed that these RBPs bind sperm-specific mRNAs, of which proteins are synthesized during the elongating stage of spermiogenesis. Biochemical and cell biological experiments demonstrated that PIWIL1, PABPC1, and NONO interact with each other and colocalize in spermatids' RNA granule, the chromatoid body. In addition, NONO was found in extracytoplasmic granules in round spermatids, whereas PIWIL1 and PABPC1 were diffusely localized in cytoplasm of elongating spermatids, indicating their participation at different steps of mRNA metabolism during spermatogenesis. Interestingly, type I PKA subunits colocalize with PIWIL1 and PABPC1 in the cytoplasm of elongating spermatids and cosediment with the RBPs in polysomal fractions on sucrose gradients. Further biochemical analyses revealed that activation of PKA positively regulates AKAP3 protein synthesis without changing its mRNA level in elongating spermatids. Taken together, these results indicate that PKA signaling directly participates in the regulation of protein translation in postmeiotic male germ cells, underscoring molecular mechanisms that regulate protein synthesis during mouse spermiogenesis.


Subject(s)
A Kinase Anchor Proteins/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , RNA-Binding Proteins/metabolism , Signal Transduction/physiology , Spermatids/physiology , Spermatogenesis/physiology , A Kinase Anchor Proteins/metabolism , Animals , Argonaute Proteins/metabolism , Cells, Cultured , Cytoplasm/metabolism , DNA-Binding Proteins/metabolism , Male , Mice, Inbred Strains , Poly(A)-Binding Proteins/metabolism , Protein Biosynthesis/physiology , RNA Processing, Post-Transcriptional/physiology , RNA, Messenger/metabolism , Sperm Tail/physiology , Spermatids/cytology
7.
Hepatology ; 57(5): 1838-46, 2013 May.
Article in English | MEDLINE | ID: mdl-23299860

ABSTRACT

UNLABELLED: Hepatocellular carcinoma (HCC) is the fifth most common malignancy and is the third leading cause of cancer death worldwide. Recently, the multitargeted kinase inhibitor sorafenib was shown to be the first systemic agent to improve survival in advanced HCC. Unlike other malignancies such as breast cancer, in which molecular subtypes have been clearly defined (i.e., luminal, HER2 amplified, basal, etc.) and tied to effective molecular therapeutics (hormone blockade and trastuzumab, respectively), in HCC this translational link does not exist. Molecular profiling studies of human HCC have identified unique molecular subtypes of the disease. We hypothesized that a panel of human HCC cell lines would maintain molecular characteristics of the clinical disease and could then be used as a model for novel therapeutics. Twenty human HCC cell lines were collected and RNA was analyzed using the Agilent microarray platform. Profiles from the cell lines in vitro recapitulate previously described subgroups from clinical material. Next, we evaluated whether molecular subgroup would have predictive value for response to the Src/Abl inhibitor dasatinib. The results demonstrate that sensitivity to dasatinib was associated with a progenitor subtype. Dasatinib was effective at inducing cell cycle arrest and apoptosis in "progenitor-like" cell lines but not in resistant lines. CONCLUSION: These findings suggest that cell line models maintain the molecular background of HCC and that subtype may be important for selecting patients for response to novel therapies. In addition, it highlights a potential role for Src family signaling in this progenitor subtype of HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Protein Kinase Inhibitors/therapeutic use , Pyrimidines/therapeutic use , Thiazoles/therapeutic use , src-Family Kinases/antagonists & inhibitors , Apoptosis/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Dasatinib , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , In Vitro Techniques , Male , Pharmacogenetics , Protein Kinase Inhibitors/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/pharmacology , Thiazoles/pharmacology , src-Family Kinases/drug effects , src-Family Kinases/genetics
8.
Cell Death Differ ; 29(11): 2316-2331, 2022 11.
Article in English | MEDLINE | ID: mdl-35614132

ABSTRACT

Metabolic switch is critical for cell fate determination through metabolic functions, epigenetic modifications, and gene expression. However, the mechanisms underlying these alterations and their functional roles remain unclear. Here, we show that Plin2-mediated moderate lipid hydrolysis is critical for pluripotency of embryonic stem cells (ESCs). Upon exit from pluripotency, lipid droplet (LD)-associated protein Plin2 is recognized by Hsc70 and degraded via chaperone-mediated autophagy to facilitate LD mobilization. Enhancing lipid hydrolysis by Plin2 knockout promotes pluripotency exit, which is recovered by ATGL inhibition. Mechanistically, excessive lipid hydrolysis induces a dramatic lipidomic remodeling characterized by decreased cardiolipin and phosphatidylethanolamine, which triggers defects in mitochondrial cristae and fatty acid oxidation, resulting in reduced acetyl-CoA and histone acetylation. Our results reveal how LD mobilization is regulated and its critical role in ESC pluripotency, and indicate the mechanism linking LD homeostasis to mitochondrial remodeling and epigenetic regulation, which might shed light on development and diseases.


Subject(s)
Histones , Lipid Droplets , Lipid Droplets/metabolism , Acetylation , Histones/metabolism , Epigenesis, Genetic , Lipidomics , Perilipin-2/genetics , Perilipin-2/metabolism , Lipids
9.
Cell Death Differ ; 29(5): 1042-1054, 2022 05.
Article in English | MEDLINE | ID: mdl-34815549

ABSTRACT

Somatic cell reprogramming is an ideal model for studying epigenetic regulation as it undergoes dramatic chromatin remodeling. However, a role for phosphorylation signaling in chromatin protein modifications for reprogramming remains unclear. Here, we identified mitogen-activated protein kinase kinase 6 (Mkk6) as a chromatin relaxer and found that it could significantly enhance reprogramming. The function of Mkk6 in heterochromatin loosening and reprogramming requires its kinase activity but does not depend on its best-known target, P38. We identified Gatad2b as a novel target of Mkk6 phosphorylation that acts downstream to elevate histone acetylation levels and loosen heterochromatin. As a result, Mkk6 over-expression facilitates binding of Sox2 and Klf4 to their targets and promotes pluripotency gene expression during reprogramming. Our studies not only reveal an Mkk phosphorylation mediated modulation of chromatin status in reprogramming, but also provide new rationales to further investigate and improve the cell fate determination processes.


Subject(s)
Chromatin , Heterochromatin , Cellular Reprogramming , Epigenesis, Genetic , MAP Kinase Kinase 6/genetics , MAP Kinase Kinase 6/metabolism , Phosphorylation
10.
Hepatology ; 51(4): 1363-72, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20091883

ABSTRACT

The T cell immunoglobulin and mucin domain-containing molecules (TIM) protein family, which is expressed by T cells, plays a crucial role in regulating host adaptive immunity and tolerance. However, its role in local inflammation, such as innate immunity-dominated organ ischemia-reperfusion injury (IRI), remains unknown. Liver IRI occurs frequently after major hepatic resection or liver transplantation. Using an antagonistic anti-TIM-1 antibody (Ab), we studied the role of TIM-1 signaling in the model of partial warm liver ischemia followed by reperfusion. Anti-TIM-1 Ab monotherapy ameliorated the hepatocellular damage and improved liver function due to IR, as compared with controls. Histological examination has revealed that anti-TIM-1 Ab treatment decreased local neutrophil infiltration, inhibited sequestration of T lymphocytes, macrophages, TIM-1 ligand-expressing TIM-4(+) cells, and reduced liver cell apoptosis. Intrahepatic neutrophil activity and induction of proinflammatory cytokines/chemokines were also reduced in the treatment group. In parallel in vitro studies, anti-TIM-1 Ab suppressed interferon-gamma (IFN-gamma) production in concanavalin A (conA)-stimulated spleen T cells, and diminished tumor necrosis factor alpha (TNF-alpha)/interleukin (IL)-6 expression in a macrophage/spleen T cell coculture system. This is the first study to provide evidence for the novel role of TIM-1 signaling in the mechanism of liver IRI. TIM-1 regulates not only T for the role of cell activation but may also affect macrophage function in the local inflammation response. These results provide compelling data for further investigation of TIM-1 pathway in the mechanism of IRI, to improve liver function, expand the organ donor pool, and improve the overall success of liver transplantation.


Subject(s)
Ischemia/etiology , Liver/blood supply , Membrane Proteins/physiology , Reperfusion Injury/etiology , Animals , Apoptosis , Cell Communication , Cytokines/biosynthesis , Hepatitis A Virus Cellular Receptor 1 , Liver/pathology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Neutrophil Infiltration , Signal Transduction , T-Lymphocytes/immunology
11.
Liver Transpl ; 16(5): 600-10, 2010 May.
Article in English | MEDLINE | ID: mdl-20440769

ABSTRACT

Janus kinase/signal transducers and activators of transcription (JAK/STAT) signaling is one of the major pathways for cytokine signal transduction. However, the role of the JAK/STAT pathway in liver ischemia/reperfusion is not clear. This study focuses on Janus kinase-2 (JAK2), which functions upstream of signal transducer and activator of transcription 1 (STAT1) in JAK/STAT, and its role in the mechanism of liver ischemia/reperfusion injury (IRI). Partial warm ischemia was produced in the hepatic lobes of C57BL/6 mice for 90 minutes, and this was followed by 6 hours of reperfusion. Mice were treated with a JAK2 inhibitor (tyrphostin AG490; 40 mg/kg intraperitoneally) or vehicle 60 minutes prior to ischemic insult. JAK2 blockade resulted in a significant reduction of hepatocyte apoptosis and liver injury. Macrophage and neutrophil infiltration, as assessed by immunohistochemistry, was markedly decreased in AG490-treated livers in comparison with controls. The expression of pro-inflammatory cytokines [tumor necrosis factor alpha, interleukin 6 (IL-6), and IL-1beta] and chemokines [chemokine (C-X-C motif) ligand 10 (CXCL-10) and CXCL-2] was also significantly reduced in the AG490-treated group in comparison with controls. AG490-treated livers showed fewer cells positive for terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling and reduced cleaved caspase-3 protein expression in parallel with increased B-cell lymphoma extra large expression. We employed AG490 (75 mM) in primary bone marrow-derived macrophage (BMM) and hepatoma cell (CRL1830) cultures, which were both stimulated with lipopolysaccharide (LPS; 10 ng/mL). In BMM cultures, AG490 depressed otherwise LPS-induced pro-inflammatory gene expression programs (IL-6, IL-12p40, IL-1beta, CXCL-10, and inducible nitric oxide synthase). In hepatoma cells, AG490 reduced cleaved caspase-3 expression. Moreover, JAK2 blockade inhibited STAT1 and STAT3 phosphorylation. This is the first report documenting that JAK2 signaling is essential in the pathophysiology of liver IRI, as its selective blockage ameliorated the disease process and protected livers from inflammation and apoptosis.


Subject(s)
Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Liver Diseases/metabolism , Reperfusion Injury/metabolism , Signal Transduction/physiology , Tyrphostins/pharmacology , Animals , Apoptosis/physiology , Cell Movement/immunology , Cells, Cultured , Cytokines/genetics , Enzyme Inhibitors/pharmacology , Gene Expression/immunology , Hepatocytes/drug effects , Hepatocytes/metabolism , Hepatocytes/pathology , Lipopolysaccharides/pharmacology , Liver Diseases/drug therapy , Liver Diseases/immunology , Macrophages/cytology , Macrophages/immunology , Male , Mice , Mice, Inbred C57BL , Neutrophils/cytology , Neutrophils/immunology , Phosphorylation/drug effects , Phosphorylation/physiology , Reperfusion Injury/drug therapy , Reperfusion Injury/immunology , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , T-Lymphocytes/cytology , T-Lymphocytes/immunology
12.
Nat Metab ; 2(9): 882-892, 2020 09.
Article in English | MEDLINE | ID: mdl-32839595

ABSTRACT

Somatic cell reprogramming provides insight into basic principles of cell fate determination, which remain poorly understood. Here we show that the transcription factor Glis1 induces multi-level epigenetic and metabolic remodelling in stem cells that facilitates the induction of pluripotency. We find that Glis1 enables reprogramming of senescent cells into pluripotent cells and improves genome stability. During early phases of reprogramming, Glis1 directly binds to and opens chromatin at glycolytic genes, whereas it closes chromatin at somatic genes to upregulate glycolysis. Subsequently, higher glycolytic flux enhances cellular acetyl-CoA and lactate levels, thereby enhancing acetylation (H3K27Ac) and lactylation (H3K18la) at so-called 'second-wave' and pluripotency gene loci, opening them up to facilitate cellular reprogramming. Our work highlights Glis1 as a powerful reprogramming factor, and reveals an epigenome-metabolome-epigenome signalling cascade that involves the glycolysis-driven coordination of histone acetylation and lactylation in the context of cell fate determination.


Subject(s)
DNA-Binding Proteins/metabolism , Epigenome , Induced Pluripotent Stem Cells , Metabolome , Signal Transduction/genetics , Signal Transduction/physiology , Transcription Factors/metabolism , Acetyl Coenzyme A/metabolism , Animals , Cellular Reprogramming , Cellular Senescence , Chromatin Immunoprecipitation , Glucose/metabolism , Lactic Acid/metabolism , Male , Mice , Plasmids/genetics
14.
Liver Transpl ; 15(8): 939-47, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19642132

ABSTRACT

Neutrophils are considered crucial effector cells in the pathophysiology of organ ischemia/reperfusion injury (IRI). Although neutrophil elastase (NE) accounts for a substantial portion of the neutrophil activity, the function of NE in liver IRI remains unclear. This study focuses on the role of NE in the mechanism of liver IRI. Partial warm ischemia was produced in the left and middle hepatic lobes of C57BL/6 mice for 90 minutes, and this was followed by 6 to 24 hours of reperfusion. Mice were treated with neutrophil elastase inhibitor (NEI; 2 mg/kg per os) at 60 minutes prior to the ischemia insult. NEI treatment significantly reduced serum alanine aminotransferase levels in comparison with controls. Histological examination of liver sections revealed that unlike in controls, NEI treatment ameliorated hepatocellular damage and decreased local neutrophil infiltration, as assessed by myeloperoxidase assay, naphthol AS-D chloroacetate esterase stains, and immunohistochemistry (anti-Ly-6G). The expression of pro-inflammatory cytokines (tumor necrosis factor alpha and interleukin 6) and chemokines [chemokine (C-X-C motif) ligand 1 (CXCL-1), CXCL-2, and CXCL-10] was significantly reduced in the NEI treatment group, along with diminished apoptosis, according to terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining and caspase-3 activity. In addition, toll-like receptor 4 (TLR4) expression was diminished in NEI-pretreated livers, and this implies a putative role of NE in the TLR4 signal transduction pathway. Thus, targeting NE represents a useful approach for preventing liver IRI and hence expanding the organ donor pool and improving the overall success of liver transplantation. Liver Transpl 15:939-947, 2009. (c) 2009 AASLD.


Subject(s)
Enzyme Inhibitors/pharmacology , Leukocyte Elastase/antagonists & inhibitors , Liver Transplantation/methods , Liver/pathology , Reperfusion Injury/prevention & control , Animals , Apoptosis , Carboxylic Ester Hydrolases/metabolism , Humans , Immunohistochemistry/methods , Liver Transplantation/adverse effects , Male , Mice , Mice, Inbred C57BL , Naphthols/metabolism , Peroxidase/metabolism , Reperfusion Injury/drug therapy , Treatment Outcome
15.
Zhonghua Yi Xue Za Zhi ; 88(26): 1837-40, 2008 Jul 08.
Article in Zh | MEDLINE | ID: mdl-19040020

ABSTRACT

OBJECTIVE: To investigate the effect of chronic restraint stress (CRS) on the esophageal epithelial barrier function and discuss the role of emotional factors in the pathogenesis of gastroesophageal reflux. METHODS: Twenty-four male SD rats were randomly divided into 4 equal groups: CRS group submitted to partial restraint stress 2 h per day for 7 days, CRS + acid/pepsin perfusion group submitted to partial restraint stress 2 h per day for 7 days and then anesthetization and perfusion of hydrochloric acid (pH=2) and pepsin (1 g/L) into the distal esophagus at the speed of 4 ml/h for 60 min, acid/pepsin perfusion group submitted to acid/pepsin perfusion into the esophagus, and control group. Then the rats were killed to collect the specimens of distal esophagus to undergo HE staining and light microscopy. Histological scoring was conducted. The diameter of intercellular spaces of esophageal epithelium was assessed. RESULTS: The amounts of food intake and body weight increment of the CRS and CRS + acid/pepsin perfusion groups were significantly lower than those of the control and acid/pepsin perfusion groups (all P < 0.01). The histological score of the CRS + acid/pepsin perfusion group was the highest. The diameter of intercellular spaces of esophagus of the CRS + acid/pepsin perfusion group was (0.516 +/- 0.010) microm, significantly greater than those of the CRS, acid/pepsin perfusion, and control groups [(0.457 +/- 0.031) microm, (0.441 +/- 0.021) microm, and (0.235 +/- 0.027) microm respectively, P < 0.01 or P < 0.05]. The histological score of the CRS + acid/pepsin perfusion group was 2.33 +/- 0.33, significantly higher than those of the CRS, acid/ pepsin perfusion, and control groups (1.17 +/- 0.31, 1.33 +/- 0.33; and 0.33 +/- 0.21 respectively, P < 0.05 or P < 0.01). CONCLUSION: CRS enlarges significantly the esophageal DIS and impairs the epithelial barrier function of the esophagus. Psychological factors induce or enhance the reflux symptom in the GERD patients, especially in non-erosive reflux disease and functional heartburn patients.


Subject(s)
Esophagus/physiopathology , Gastroesophageal Reflux/physiopathology , Stress, Psychological/physiopathology , Animals , Epithelial Cells/metabolism , Epithelial Cells/pathology , Esophagus/pathology , Extracellular Space/metabolism , Gastroesophageal Reflux/metabolism , Gastroesophageal Reflux/pathology , Male , Mucous Membrane/metabolism , Mucous Membrane/pathology , Rats , Rats, Sprague-Dawley , Restraint, Physical
16.
Cell Death Dis ; 8(10): e3107, 2017 10 12.
Article in English | MEDLINE | ID: mdl-29022923

ABSTRACT

MicroRNAs (miRNAs) play crucial roles in the establishment of pluripotent state by controlling pluripotent network. However, the molecular mechanisms controlling miRNAs during somatic cell reprogramming remain obscure. In this study, we show Gadd45a (growth arrest and DNA-damage-inducible protein 45a) enhances reprogramming by activating miR-295. Furthermore, we show that Gadd45a binds the promoter regions of miR-295. Nuclease accessibility assay indicates that Gadd45a opens the promoter regions of miR-295. Levels of H3K9Ac and H3K27Ac on the promoter regions of miR-295 were also increased. In conclusion, our results indicate that Gadd45a relaxes the promoter regions of miR-295 and promotes the expression of miR-295 during reprogramming, implying a concise mechanism of Gadd45a and miR-290 cluster cooperation in cell-fate determination.


Subject(s)
Cell Cycle Proteins/metabolism , Cellular Reprogramming/genetics , MicroRNAs/genetics , Nuclear Proteins/metabolism , Pluripotent Stem Cells/cytology , Animals , Cell Differentiation/genetics , Cell Proliferation/genetics , DNA Damage/genetics , DNA Repair/genetics , DNA-Binding Proteins/metabolism , Mice , Pluripotent Stem Cells/virology , Promoter Regions, Genetic/genetics
18.
J Alzheimers Dis ; 21(3): 823-31, 2010.
Article in English | MEDLINE | ID: mdl-20634586

ABSTRACT

Amyloid-ß (Aß) peptide-induced impairment of hippocampal synaptic plasticity is considered an underlying mechanism for memory loss in the early stages of Alzheimer's disease and its animal models. We previously reported inhibition of long-term potentiation (LTP) and miniature excitatory postsynaptic currents by oligomeric Aß(1-42) at hippocampal synapses. While multiple cellular mechanisms could be involved in Aß-induced synaptic dysfunction, blockade of activity-dependent autophosphorylation of Ca2+ and calmodulin-dependent protein kinase II (CaMKII) appeared to be a major component of Aß action in our studies. The present study further tested this hypothesis and examined the therapeutic potential of trkB receptor-acting neurotrophins in rescuing Aß-induced synaptic and signaling impairments. As expected, treatment of rat hippocampal slices with Aß(1-42) significantly reduced LTP in the Schaffer collateral-CA1 pathway and dentate medial perforant path. LTP-associated CaMKII activation and AMPA receptor phosphorylation were blocked by Aß(1-42) at the same concentration that inhibited LTP. Aß-induced LTP impairment, however, was prevented when slices were co-treated with neurotrophin 4 (NT4). Western blotting and immunohistochemical analyses confirmed that treatment with NT4 or brain-derived neurotrophic factor, another trkB-acting neurotrophin, could oppose Aß action, enhancing autophosphorylation of CaMKII, and AMPA receptor phosphorylation at a CaMKII-dependent site. These findings support the view that CaMKII is a key synaptic target of Aß toxicity as well as a potential therapeutic site of neurotrophins for Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/pharmacology , Brain-Derived Neurotrophic Factor/pharmacology , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Hippocampus/physiology , Long-Term Potentiation/drug effects , Nerve Growth Factors/pharmacology , Peptide Fragments/pharmacology , Amyloid beta-Peptides/metabolism , Analysis of Variance , Animals , Blotting, Western , Brain-Derived Neurotrophic Factor/metabolism , Calcium/metabolism , Electrophysiology , Hippocampus/drug effects , Immunohistochemistry , Long-Term Potentiation/physiology , Male , Nerve Growth Factors/metabolism , Peptide Fragments/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Receptors, AMPA/metabolism , Synapses/drug effects , Synapses/physiology
19.
Transplantation ; 89(9): 1050-6, 2010 May 15.
Article in English | MEDLINE | ID: mdl-20160675

ABSTRACT

BACKGROUND.: A neutrophil elastase (NE) inhibitor, Sivelestat, has been approved for the treatment of acute lung injury associated with systemic inflammation in humans. Some reports have also shown its protective effects in liver inflammatory states. We have recently documented the importance of NE in the pathophysiology of liver ischemia/reperfusion injury, a local Ag-independent inflammation response. This study was designed to explore putative cytoprotective functions of clinically available Sivelestat in liver ischemia/reperfusion injury. METHODS.: Partial warm ischemia was produced in the left and middle hepatic lobes of C57BL/6 mice for 90 min, followed by 6 or 24 hr of reperfusion. The mice were given Sivelestat (100 mg/kg, subcutaneous) at 10 min before ischemia, 10 min before reperfusion, and at 1 and 3 hr of reperfusion thereafter. RESULTS.: Sivelestat treatment significantly reduced serum alanine aminotransferase levels and NE activity, when compared with controls. Histological liver examination has revealed that unlike in controls, Sivelestat ameliorated the hepatocellular damage and decreased local neutrophil activity and infiltration. The expression of proinflammatory cytokines (tumor necrosis factor-alpha and interleukin-6), chemokines (CXCL-1, CXCL-2, and CXCL-10), and toll-like receptor 4 was significantly reduced in the treatment group, along with diminished apoptosis through caspase-3 pathway. Moreover, in vitro studies confirmed downregulation of proinflammatory cytokine and chemokine programs in mouse macrophage cell cultures, along with depression of innate toll-like receptor 4 signaling. CONCLUSION.: Sivelestat-mediated NE inhibition may represent an effective therapeutic option in liver transplantation and other inflammation disease states.


Subject(s)
Glycine/analogs & derivatives , Liver/pathology , Proteinase Inhibitory Proteins, Secretory/therapeutic use , Reperfusion Injury/prevention & control , Sulfonamides/therapeutic use , Animals , Chemokine CXCL1/genetics , DNA Primers , Glycine/therapeutic use , Humans , Immunohistochemistry , Inflammation/prevention & control , Interleukin-6/genetics , Leukocyte Elastase/metabolism , Liver/drug effects , Male , Mice , Mice, Inbred C57BL , Neutrophils/enzymology , Peroxidase/blood , Polymerase Chain Reaction , RNA/genetics , RNA/isolation & purification , Tumor Necrosis Factor-alpha/genetics
20.
J Neurophysiol ; 92(5): 2853-8, 2004 Nov.
Article in English | MEDLINE | ID: mdl-15212428

ABSTRACT

Accumulation of amyloid beta-peptides (Abeta) in the brain has been linked with memory loss in Alzheimer's disease and its animal models. However, the synaptic mechanism by which Abeta causes memory deficits remains unclear. We previously showed that acute application of Abeta inhibited long-term potentiation (LTP) in the hippocampal perforant path via activation of calcineurin, a Ca2+ -dependent protein phosphatase. This study examined whether Abeta could also inhibit Ca2+/calmodulin dependent protein kinase II (CaMKII), further disrupting the dynamic balance between protein kinase and phosphatase during synaptic plasticity. Immunoblot analysis was conducted to measure autophosphorylation of CaMKII at Thr286 and phosphorylation of the GluR1 subunit of AMPA receptors in single rat hippocampal slices. A high-frequency tetanus applied to the perforant path significantly increased CaMKII autophosphorylation and subsequent phosphorylation of GluR1 at Ser831, a CaMKII-dependent site, in the dentate area. Acute application of Abeta1-42 inhibited dentate LTP and associated phosphorylation processes, but was without effect on phosphorylation of GluR1 at Ser845, a protein kinase A-dependent site. These results suggest that activity-dependent CaMKII autophosphorylation and AMPA receptor phosphorylation are essential for dentate LTP. Disruption of such mechanisms could directly contribute to Abeta-induced deficits in hippocampal synaptic plasticity and memory.


Subject(s)
Amyloid beta-Peptides/pharmacology , Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Hippocampus/physiology , Long-Term Potentiation/physiology , Receptors, AMPA/metabolism , Animals , Calcium-Calmodulin-Dependent Protein Kinase Type 2 , Hippocampus/drug effects , In Vitro Techniques , Long-Term Potentiation/drug effects , Male , Models, Neurological , Peptide Fragments/pharmacology , Phosphorylation , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL