Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell ; 174(3): 576-589.e18, 2018 07 26.
Article in English | MEDLINE | ID: mdl-30033361

ABSTRACT

Genome-wide association studies (GWAS) have identified rs11672691 at 19q13 associated with aggressive prostate cancer (PCa). Here, we independently confirmed the finding in a cohort of 2,738 PCa patients and discovered the biological mechanism underlying this association. We found an association of the aggressive PCa-associated allele G of rs11672691 with elevated transcript levels of two biologically plausible candidate genes, PCAT19 and CEACAM21, implicated in PCa cell growth and tumor progression. Mechanistically, rs11672691 resides in an enhancer element and alters the binding site of HOXA2, a novel oncogenic transcription factor with prognostic potential in PCa. Remarkably, CRISPR/Cas9-mediated single-nucleotide editing showed the direct effect of rs11672691 on PCAT19 and CEACAM21 expression and PCa cellular aggressive phenotype. Clinical data demonstrated synergistic effects of rs11672691 genotype and PCAT19/CEACAM21 gene expression on PCa prognosis. These results provide a plausible mechanism for rs11672691 associated with aggressive PCa and thus lay the ground work for translating this finding to the clinic.


Subject(s)
Prostatic Neoplasms/genetics , RNA, Long Noncoding/genetics , RNA, Untranslated/genetics , Adult , Alleles , Cell Line, Tumor , Chromosomes, Human, Pair 19/genetics , Cohort Studies , Gene Expression Regulation, Neoplastic/genetics , Gene Frequency/genetics , Genetic Predisposition to Disease/genetics , Genome-Wide Association Study , Genotype , Homeodomain Proteins , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Prognosis
2.
Genet Res (Camb) ; 2022: 9792913, 2022.
Article in English | MEDLINE | ID: mdl-35645614

ABSTRACT

Background: We aimed to construct the lncRNA-associated competing endogenous RNA (ceRNA) network and distinguish feature lncRNAs associated with tendinopathy. Methods: We downloaded the gene profile of GSE26051 from the Gene Expression Omnibus (GEO), including 23 normal samples and 23 diseased tendons. Differentially expressed mRNAs (DEmRNAs) and differentially expressed lncRNAs (DElncRNAs) were identified, and functional and pathway enrichment analyses were performed. Protein-protein interaction (PPI) network was constructed and further analyzed by module mining. Moreover, a ceRNA regulatory network was constructed based on the identified lncRNA-mRNA coexpression relationship pairs and miRNA-mRNA regulation pairs. Results: We identified 1117 DEmRNAs and 57 DElncRNAs from the GEO data. The downregulated DEmRNAs were particularly associated with muscle contraction and muscle filament, while the upregulated ones were linked to extracellular matrix organization and cell adhesion. From the PPI network, 11 modules were extracted. Genes in MCODE 2 (such as TPM4) were significantly involved in cardiomyopathy, and genes in MCODE 4 (such as COL4A3 and COL4A4) were involved in focal adhesion, ECM-receptor interaction, and PI3K-Akt signaling pathway. The ceRNA network contained 7 lncRNAs (MIR133A1HG, LINC01405, PRKCQ-AS1, C10orf71-AS1, MBNL1-AS1, HOTAIRM1, and DNM3OS), 63 mRNAs, and 41 miRNAs. Downregulated lncRNA MIR133A1HG could competitively bind with hsa-miR-659-3p and hsa-miR-218-1-3p to regulate the TPM3. Meanwhile, MIR133A1HG could competitively bind with hsa-miR-1179 to regulate the COL4A3. Downregulated C10orf71-AS1 could competitively bind with hsa-miR-130a-5p to regulate the COL4A4. Conclusions: Seven important lncRNAs, particularly MIR133A1HG and C10orf71-AS1, were found associated with tendinopathy according to the lncRNA-associated ceRNA network.


Subject(s)
MicroRNAs , RNA, Long Noncoding , Tendinopathy , Gene Regulatory Networks/genetics , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Phosphatidylinositol 3-Kinases/genetics , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Tendinopathy/genetics
3.
FASEB J ; 34(6): 8416-8427, 2020 06.
Article in English | MEDLINE | ID: mdl-32350948

ABSTRACT

During human erythroid maturation, Hsp70 translocates into the nucleus and protects GATA-1 from caspase-3 cleavage. Failure of Hsp70 to localize to the nucleus was found in Myelodysplastic syndrome (MDS) erythroblasts and can induce dyserythropoiesis, with arrest of maturation and death of erythroblasts. However, the mechanism of the nuclear trafficking of Hsp70 in erythroblasts remains unknown. Here, we found the hematopoietic transcriptional regulator, EDAG, to be a novel binding partner of Hsp70 that forms a protein complex with Hsp70 and GATA-1 during human normal erythroid differentiation. EDAG overexpression blocked the cytoplasmic translocation of Hsp70 induced by EPO deprivation, inhibited GATA-1 degradation, thereby promoting erythroid maturation in an Hsp70-dependent manner. Furthermore, in myelodysplastic syndrome (MDS) patients with dyserythropoiesis, EDAG is dramatically down-regulated, and forced expression of EDAG has been found to restore the localization of Hsp70 in the nucleus and elevate the protein level of GATA-1 to a significant extent. In addition, EDAG rescued the dyserythropoiesis of MDS patients by increasing erythroid differentiation and decreasing cell apoptosis. This study demonstrates the molecular mechanism of Hsp70 nuclear sustaining during erythroid maturation and establishes that EDAG might be a suitable therapeutic target for dyserythropoiesis in MDS patients.


Subject(s)
Cell Nucleus/metabolism , Erythroblasts/metabolism , Erythropoiesis/physiology , HSP70 Heat-Shock Proteins/metabolism , Myelodysplastic Syndromes/metabolism , Nuclear Proteins/metabolism , Apoptosis/physiology , Caspase 3/metabolism , Cell Differentiation/physiology , Cells, Cultured , Cytoplasm/metabolism , Gene Expression Regulation/physiology , Hematologic Diseases/metabolism , Humans
4.
Stem Cells ; 32(8): 2278-89, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24740910

ABSTRACT

Erythroid differentiation-associated gene (EDAG) has been considered to be a transcriptional regulator that controls hematopoietic cell differentiation, proliferation, and apoptosis. The role of EDAG in erythroid differentiation of primary erythroid progenitor cells and in vivo remains unknown. In this study, we found that EDAG is highly expressed in CMPs and MEPs and upregulated during the erythroid differentiation of CD34(+) cells following erythropoietin (EPO) treatment. Overexpression of EDAG induced erythroid differentiation of CD34(+) cells in vitro and in vivo using immunodeficient mice. Conversely, EDAG knockdown reduced erythroid differentiation in EPO-treated CD34(+) cells. Detailed mechanistic analysis suggested that EDAG forms complex with GATA1 and p300 and increases GATA1 acetylation and transcriptional activity by facilitating the interaction between GATA1 and p300. EDAG deletion mutants lacking the binding domain with GATA1 or p300 failed to enhance erythroid differentiation, suggesting that EDAG regulates erythroid differentiation partly through forming EDAG/GATA1/p300 complex. In the presence of the specific inhibitor of p300 acetyltransferase activity, C646, EDAG was unable to accelerate erythroid differentiation, indicating an involvement of p300 acetyltransferase activity in EDAG-induced erythroid differentiation. ChIP-PCR experiments confirmed that GATA1 and EDAG co-occupy GATA1-targeted genes in primary erythroid cells and in vivo. ChIP-seq was further performed to examine the global occupancy of EDAG during erythroid differentiation and a total of 7,133 enrichment peaks corresponding to 3,847 genes were identified. Merging EDAG ChIP-Seq and GATA1 ChIP-Seq datasets revealed that 782 genes overlapped. Microarray analysis suggested that EDAG knockdown selectively inhibits GATA1-activated target genes. These data provide novel insights into EDAG in regulation of erythroid differentiation.


Subject(s)
Cell Differentiation/physiology , E1A-Associated p300 Protein/metabolism , GATA1 Transcription Factor/metabolism , Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Nuclear Proteins/metabolism , Acetylation , Animals , Blotting, Western , Cell Separation , Erythroid Cells/cytology , Erythroid Cells/metabolism , Female , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Heterografts , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Oligonucleotide Array Sequence Analysis , Transcriptome
5.
Int Immunopharmacol ; 123: 110747, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37586299

ABSTRACT

Diabetic cardiomyopathy (DCM) is a prevalent cardiovascular complication of diabetes mellitus, characterized by high morbidity and mortality rates worldwide. However, treatment options for DCM remain limited. For decades, a substantial body of evidence has suggested that the inflammatory response plays a pivotal role in the development and progression of DCM. Notably, DCM is closely associated with alterations in inflammatory cells, exerting direct effects on major resident cells such as cardiomyocytes, vascular endothelial cells, and fibroblasts. These cellular changes subsequently contribute to the development of DCM. This article comprehensively analyzes cellular, animal, and human studies to summarize the latest insights into the impact of inflammation on DCM. Furthermore, the potential therapeutic effects of current anti-inflammatory drugs in the management of DCM are also taken into consideration. The ultimate goal of this work is to consolidate the existing literature on the inflammatory processes underlying DCM, providing clinicians with the necessary knowledge and tools to adopt a more efficient and evidence-based approach to managing this condition.


Subject(s)
Diabetes Mellitus , Diabetic Cardiomyopathies , Animals , Humans , Diabetic Cardiomyopathies/drug therapy , Diabetic Cardiomyopathies/etiology , Endothelial Cells , Inflammation/drug therapy , Inflammation/complications , Myocytes, Cardiac , Anti-Inflammatory Agents/therapeutic use , Anti-Inflammatory Agents/pharmacology , Diabetes Mellitus/drug therapy
6.
Oncogene ; 41(21): 2945-2957, 2022 05.
Article in English | MEDLINE | ID: mdl-35437307

ABSTRACT

PLK1 and Smad4 are two important factors in prostate cancer initiation and progression. They have been reported to play the opposite role in Pten-deleted mice, one is an oncogene, the other is a tumor suppressor. Moreover, they could reversely regulate the PI3K/AKT/mTOR pathway and the activation of MYC. However, the connections between PLK1 and Smad4 have never been studied. Here, we showed that PLK1 could interact with Smad4 and promote the ubiquitination and degradation of Smad4 in PCa cells. PLK1 and PELO could bind to different domains of Smad4 and formed a protein complex. PELO facilitated the degradation of Smad4 through cooperating with PLK1, thereby resulting in proliferation and metastasis of prostate cancer cell. Changes in protein levels of Smad4 led to the alteration of biological function that caused by PLK1 in prostate cancer cells. Further studies showed that PELO upregulation was positively associated with high grade PCa and knockdown of PELO expression significantly decreased PCa cell proliferation and metastasis in vitro and vivo. PELO knockdown in PCa cells could enhance the tumor suppressive role of PLK1 inhibitor. In addition, blocking the interaction between PELO and Smad4 by using specific peptide could effectively inhibit PCa cell metastasis ability in vitro and vivo. Overall, these findings identified a novel regulatory relationship among PLK1, Smad4 and PELO, and provided a potential therapeutic strategy for advanced PCa therapy by co-targeting PLK1 and PELO.


Subject(s)
Cell Cycle Proteins , Endonucleases , Phosphatidylinositol 3-Kinases , Prostatic Neoplasms , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation/genetics , Endonucleases/genetics , Endonucleases/metabolism , Humans , Male , Mice , Phosphatidylinositol 3-Kinases/metabolism , Prostate/pathology , Prostatic Neoplasms/pathology , Smad4 Protein/genetics , Smad4 Protein/metabolism , Ubiquitination
7.
Adv Sci (Weinh) ; 9(5): e2103838, 2022 02.
Article in English | MEDLINE | ID: mdl-34923767

ABSTRACT

Hematopoietic stem and progenitor cells (HSPCs) possess the remarkable ability to regenerate the whole blood system in response to ablated stress demands. Delineating the mechanisms that maintain HSPCs during regenerative stresses is increasingly important. Here, it is shown that Hemgn is significantly induced by hematopoietic stresses including irradiation and bone marrow transplantation (BMT). Hemgn deficiency does not disturb steady-state hematopoiesis in young mice. Hemgn-/- HSPCs display defective engraftment activity during BMT with reduced homing and survival and increased apoptosis. Transcriptome profiling analysis reveals that upregulated genes in transplanted Hemgn-/- HSPCs are enriched for gene sets related to interferon gamma (IFN-γ) signaling. Hemgn-/- HSPCs show enhanced responses to IFN-γ treatment and increased aging over time. Blocking IFN-γ signaling in irradiated recipients either pharmacologically or genetically rescues Hemgn-/- HSPCs engraftment defect. Mechanistical studies reveal that Hemgn deficiency sustain nuclear Stat1 tyrosine phosphorylation via suppressing T-cell protein tyrosine phosphatase TC45 activity. Spermidine, a selective activator of TC45, rescues exacerbated phenotype of HSPCs in IFN-γ-treated Hemgn-/- mice. Collectively, these results identify that Hemgn is a critical regulator for successful engraftment and reconstitution of HSPCs in mice through negatively regulating IFN-γ signaling. Targeted Hemgn may be used to improve conditioning regimens and engraftment during HSPCs transplantation.


Subject(s)
Hematopoietic Stem Cell Transplantation , Interferon-gamma , Animals , Hematopoiesis , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/metabolism , Interferon-gamma/metabolism , Mice , Transplantation Conditioning
8.
Int J Cancer ; 124(3): 739-44, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19006069

ABSTRACT

The HSulf-1 gene is an important factor that modulates the sulfation status of heparan sulfate proteoglycans (HSPGs) in the extracellular matrix, resulting in disturbance of HSPG-related signal transduction pathways. Recently, HSulf-1 has been reported to be down-regulated in several human cancers. In this study, we first cloned and characterized the 5' promoter region of the HSulf-1 gene (around 400 bp) that contained high basal promoter activity. We also found that this functional promoter region was hypermethylated in a number of human cancer cell lines. Furthermore, we found that hypermethylation in this promoter region correlated with the down-regulation of the HSulf-1 expression in human breast and gastric cancer cell lines and tissue samples. These results suggest that the promoter hypermethylation may be one of the mechanisms of the HSulf-1 gene silencing in human breast and gastric cancers. Finally, we demonstrated that the HSulf-1 promoter was more frequently (p<0.05) methylated in cell-free DNA extracted from serum samples of human breast and gastric cancer patients than that of healthy people (76.2%, 55.0% and 19.0%, respectively), indicating that detection of the HSulf-1 promoter methylation in serum samples may have clinical implications in early detection and diagnosis of human breast and gastric cancers.


Subject(s)
Breast Neoplasms/genetics , DNA Methylation/genetics , Gene Silencing , Promoter Regions, Genetic , Stomach Neoplasms/genetics , Sulfotransferases/genetics , Adult , Aged , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Reverse Transcriptase Polymerase Chain Reaction , Transfection
9.
Zhongguo Gu Shang ; 32(6): 504-507, 2019 Jun 25.
Article in Zh | MEDLINE | ID: mdl-31277531

ABSTRACT

OBJECTIVE: To explore the method and effect of ultrasound-guided suprascapular nerve block combined with acupuncture in the treatment of calcified tendinitis of rotator cuff. METHODS: From January 2015 to December 2017, total 30 patients with calcified tendinitis, including 23 cases of supraspinatus tendon, 5 cases of infraspinatus tendon and 2 cases of subscapular tendon, were treated with ultrasound-guided suprascapular nerve block combined with acupuncture. There were 7 males and 23 females, ranging in age from 36 to 71 years old, with an average of 51.6 years old. There were 17 cases on the right and 13 cases on the left. VAS pain score, Constant-murley score, UCLA score and X-ray examination were used to evaluate the clinical results before and after surgery. RESULTS: The mean follow-up was 14.3 months (6 to 30 months). The preoperative VAS score was 3.82±1.13, Constant-Murley score was 36.91±7.95 and UCLA score was 11.35±2.17. The final follow-up scores were 1.32±1.06, 90.61±2.89 and 33.22±1.51, respectively. The final follow-up scores were improved significantly(P<0.05). CONCLUSIONS: Conservative treatment of calcified rotator cuff tendinitis is ineffective. Suprascapular nerve block guided by ultrasound combined with acupuncture has a good therapeutic effect. It is a minimally invasive, economic, safe and effective method, which is worth promoting.


Subject(s)
Acupuncture Therapy , Nerve Block , Rotator Cuff Injuries , Tendinopathy , Adult , Aged , Arthroscopy , Female , Humans , Male , Middle Aged , Rotator Cuff , Tendinopathy/therapy , Treatment Outcome
10.
PLoS One ; 13(1): e0190794, 2018.
Article in English | MEDLINE | ID: mdl-29324880

ABSTRACT

EDAG is multifunctional transcriptional regulator primarily expressed in the linloc-kit+Sca-1+ hematopoietic stem cells (HSC) and CD34+ progenitor cells. Previous studies indicate that EDAG is required for maintaining hematopoietic lineage commitment balance. Here using ex vivo culture and HSC transplantation models, we report that EDAG enhances the proliferative potential of human cord blood CD34+ cells, increases survival, prevents cell apoptosis and promotes their repopulating capacity. Moreover, EDAG overexpression induces rapid entry of CD34+ cells into the cell cycle. Gene expression profile analysis indicate that EDAG knockdown leads to down-regulation of various positive cell cycle regulators including cyclin A, B, D, and E. Together these data provides novel insights into EDAG in regulation of expansion and survival of human hematopoietic stem/progenitor cells.


Subject(s)
Antigens, CD34/metabolism , Cell Proliferation/physiology , Cell Survival/physiology , Fetal Blood/metabolism , Hematopoietic Stem Cells/metabolism , Nuclear Proteins/metabolism , Animals , Apoptosis/physiology , Cell Cycle/physiology , Cells, Cultured , Cord Blood Stem Cell Transplantation , Cyclins/metabolism , Female , Fetal Blood/cytology , Hematopoietic Stem Cells/chemistry , Humans , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Nuclear Proteins/genetics
11.
Oncotarget ; 8(57): 96774-96790, 2017 Nov 14.
Article in English | MEDLINE | ID: mdl-29228570

ABSTRACT

Keratin 8 (CK8) is the major component of the intermediate filaments of simple or single-layered epithelia. Gene targeting mice model suggest that CK8 is involved in colonic active ion transport, colorectal hyperplasia and inflammation. In the present study, we found that CK8 is downregulated in the colon during DSS-induced colitis and AOM/DSS-induced colitis-associated colorectal cancer (CAC) development. In human patients with colon cancer, CK8 is downregulated. Using CK8 heterozygous knockout mice (CK8+/-), we found that CK8+/- mice are highly susceptible to DSS-induced colitis and more prone to AOM/DSS-induced CAC than wild type (WT) mice. The colonic permeability is increased with DSS or AOM/DSS treatment, leading to alteration of gut microbiota in CK8+/- mice with CAC. Metagenomic analysis of fecal microbiota suggests Firmicutes and Proteobacteria are increased in CK8+/- mice with CAC, while Bacteroidetes and Verrucomicrobia are decreased. Antibiotic treatment decreases the incidence of colorectal cancer tumorigenesis and TLR4 inhibitor attenuates the susceptibility of CK8+/- mice to DSS-induced colitis. These data suggest CK8 protects mice from colitis and colitis-associated colorectal cancer by modulating colonic permeability and gut microbiota composition homeostasis.

13.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 24(4): 1173-8, 2016 Aug.
Article in Zh | MEDLINE | ID: mdl-27531795

ABSTRACT

OBJECTIVE: To construct the ovexpression lentivirus vector of PPP2Cß, the catalytic subunit of protein phosphatase 2A, so as to obtain high-titer packaged lentivirus particles, and to examine the effect of PPP2Cß on the erythroid differentiation Methods: The CDS of PPP2Cß was cloned into the second generation of lentivirus vector FUGW, which should be used to co-transfect HEK 293T cells with the lentiviral expression vector and packaging vectors including pMD2G and pSPAX2. Lentiviruses were harvested at 36 and 48 hours after transfection. Titers of viral stock were determined by using flow cytometric analysis. The Western blot was performed to detect the expression level of PPP2Cß in K562 cells transinfected with the lentiviruses. Benzidine staining and real-time PCR analysis were used to assess the erythroid differentiation of K562 cells. RESULTS: The PPP2Cß overexpressing lentivirus vectors were constructed, the high-titer lentiviral particles were obtained, and then the PPP2Cß overexpression K562 cell line was established and promote erythroid differentiation of K562 cells. CONCLUSION: This study suggests that overexpression PPP2Cß can promote K562 cell erythroid differentiation.


Subject(s)
Cell Differentiation , Erythroid Cells , Genetic Vectors , Humans , K562 Cells , Lentivirus , Protein Phosphatase 2 , Real-Time Polymerase Chain Reaction , Transfection
14.
Sci Rep ; 6: 32710, 2016 09 02.
Article in English | MEDLINE | ID: mdl-27586056

ABSTRACT

Toll-like receptors (TLRs) have critical roles in innate immunity and inflammation and the detailed mechanisms by which TLR signaling is fine tuned remain unclear. Keratin 8 (CK8) belongs to the type II keratin family and is the major compontent of the intermediate filaments of simple or single-layered epithelia. Here we report that down-regulation of CK8 in mice enhanced TLR-mediated responses, rendering mice more susceptible to lipopolysaccharide (LPS)-induced endotoxin shock and Escherichia coli-caused septic peritonitis with reduced survival, elevated levels of inflammation cytokines and more severe tissue damage. We found that CK8 suppressed TLR-induced nuclear factor (NF)-κB activation and interacted with the adaptor tumor necrosis factor (TNF) receptor-associated factor 6 (TRAF6) to prevent its polyubiquitination. Our findings demonstrate a novel role of CK8 in negative regulation of TLR/NF-κB signaling and highlight a previously unidentified nonclassical function for CK8 in limiting inflammatory responses.


Subject(s)
Inflammation/pathology , Keratin-8/metabolism , Shock, Septic/pathology , TNF Receptor-Associated Factor 6/metabolism , Toll-Like Receptors/metabolism , Ubiquitination , Animals , Cytokines/blood , Disease Models, Animal , Endotoxins/toxicity , Escherichia coli Infections/pathology , Mice , NF-kappa B/metabolism , Peritonitis/pathology , Survival Analysis
15.
Oxid Med Cell Longev ; 2015: 959253, 2015.
Article in English | MEDLINE | ID: mdl-25874026

ABSTRACT

Nrf2 is the key transcription factor regulating the antioxidant response which is crucial for cytoprotection against extracellular stresses. Numerous in vivo studies indicate that Nrf2 plays a protective role in anti-inflammatory response. 3-(3-Pyridylmethylidene)-2-indolinone (PMID) is a synthesized derivative of 2-indolinone compounds. Our previous study suggested that PMID induces the activation of Nrf2/ARE pathway, then protecting against oxidative stress-mediated cell death. However, little is known regarding the anti-inflammatory properties of PMID in severe inflammatory phenotypes. In the present study we determined if PMID treatment protects mice from dextran sodium sulphate- (DSS-) induced colitis. The result suggests that treatment with PMID prior to colitis induction significantly reduced body weight loss, shortened colon length, and decreased disease activity index compared to control mice. Histopathological analysis of the colon revealed attenuated inflammation in PMID pretreated animals. The levels of inflammatory markers in colon tissue and serum were reduced associated with inhibition of NF-κB activation. The expression levels of Nrf2-dependent genes such as HO-1, NQO1, and Nrf2 were increased in PMID pretreated mice. However, PMID pretreatment did not prevent DSS-induced colitis in Nrf2 knockout mice. These data indicate that PMID pretreatment in mice confers protection against DSS-induced colitis in Nrf2-dependent manner, suggesting a potential role of PMID in anti-inflammatory response.


Subject(s)
Anti-Inflammatory Agents/therapeutic use , Colitis/drug therapy , Indoles/therapeutic use , Pyridines/therapeutic use , Animals , Anti-Inflammatory Agents/chemical synthesis , Anti-Inflammatory Agents/pharmacology , Colitis/chemically induced , Colitis/pathology , Colon/drug effects , Colon/pathology , Colon/physiology , Cytokines/analysis , Cytokines/genetics , Dextran Sulfate/toxicity , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Indoles/chemical synthesis , Indoles/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Knockout , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , NF-E2-Related Factor 2/deficiency , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , NF-kappa B/metabolism , Pyridines/chemical synthesis , Pyridines/pharmacology , Severity of Illness Index
16.
World J Gastroenterol ; 21(36): 10375-84, 2015 Sep 28.
Article in English | MEDLINE | ID: mdl-26420964

ABSTRACT

AIM: To investigate the effects of Recql5 deficiency on liver injury induced by lipopolysaccharide/D-galactosamine (LPS/D-Gal). METHODS: Liver injury was induced in wild type (WT) or Recql5-deficient mice using LPS/D-Gal, and assessed by histological, serum transaminases, and mortality analyses. Hepatocellular apoptosis was quantified by transferase dUTP nick end labeling assay and Western blot analysis of cleaved caspase-3. Liver inflammatory chemokine and cytochrome P450 expression was analyzed by quantitative reverse transcription-PCR. Neutrophil infiltration was evaluated by myeloperoxidase activity. Expression and phosphorylation of ERK, JNK, p65, and H2A.X was determined by Western blot. Oxidative stress was evaluated by measuring malondialdehyde production and nitric oxide synthase, superoxide dismutase, glutathione peroxidase, catalase, and glutathione reductase activity. RESULTS: Following LPS/D-Gal exposure, Recql5-deficient mice exhibited enhanced liver injury, as evidenced by more severe hepatic hemorrhage, higher serum aspartate transaminase and alanine transaminase levels, and lower survival rate. As compared to WT mice, Recql5-deficient mice showed an increased number of apoptotic hepatocytes and higher cleaved caspase-3 levels. Recql5-deficient mice exhibited increased DNA damage, as evidenced by increased γ-H2A.X levels. Inflammatory cytokine levels, neutrophil infiltration, and ERK phosphorylation were also significantly increased in the knockout mice. Additionally, Recql5-deficient mice exhibited increased malondialdehyde production and elevated inducible nitric oxide synthase, superoxide dismutase, glutathione peroxidase, catalase, and glutathione reductase activity, indicative of enhanced oxidative stress. Moreover, CYP450 expression was significantly downregulated in Recql5-deficient mice after LPS/D-Gal treatment. CONCLUSION: Recql5 protects the liver against LPS/D-Gal-induced injury through suppression of hepatocyte apoptosis and oxidative stress and modulation of CYP450 expression.


Subject(s)
Chemical and Drug Induced Liver Injury/prevention & control , Galactosamine , Hepatocytes/enzymology , Lipopolysaccharides , Liver/enzymology , RecQ Helicases/metabolism , Animals , Apoptosis , Caspase 3/metabolism , Chemical and Drug Induced Liver Injury/enzymology , Chemical and Drug Induced Liver Injury/etiology , Chemical and Drug Induced Liver Injury/genetics , Chemical and Drug Induced Liver Injury/pathology , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Cytokines/genetics , Cytokines/metabolism , DNA Damage , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Hepatocytes/pathology , Inflammation Mediators/metabolism , Liver/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Infiltration , Oxidative Stress , Phosphorylation , RecQ Helicases/deficiency , RecQ Helicases/genetics , Signal Transduction/drug effects , Time Factors
17.
PLoS One ; 9(3): e91557, 2014.
Article in English | MEDLINE | ID: mdl-24637716

ABSTRACT

Hematopoiesis is a complex process regulated by sets of transcription factors in a stage-specific and context-dependent manner. THAP11 is a transcription factor involved in cell growth, ES cell pluripotency, and embryogenesis. Here we showed that THAP11 was down-regulated during erythroid differentiation but up-regulated during megakaryocytic differentiation of cord blood CD34+ cells. Overexpression of THAP11 in K562 cells inhibited the erythroid differentiation induced by hemin with decreased numbers of benzidine-positive cells and decreased mRNA levels of α-globin (HBA) and glycophorin A (GPA), and knockdown of THAP11 enhanced the erythroid differentiation. Conversely, THAP11 overexpression accelerated the megakaryocytic differentiation induced by phorbol myristate acetate (PMA) with increased percentage of CD41+ cells, increased numbers of 4N cells, and elevated CD61 mRNA levels, and THAP11 knockdown attenuated the megakaryocytic differentiation. The expression levels of transcription factors such as c-Myc, c-Myb, GATA-2, and Fli1 were changed by THAP11 overexpression. In this way, our results suggested that THAP11 reversibly regulated erythroid and megakaryocytic differentiation.


Subject(s)
Cell Differentiation/genetics , Erythroid Cells/cytology , Erythroid Cells/metabolism , Hematopoiesis/genetics , Megakaryocytes/cytology , Megakaryocytes/metabolism , Repressor Proteins/genetics , Antigens, CD34/metabolism , Cell Differentiation/drug effects , Fetal Blood/cytology , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Hemin/pharmacology , Humans , K562 Cells , RNA, Messenger/genetics , RNA, Messenger/metabolism , Repressor Proteins/metabolism , Transcription Factors/metabolism
18.
Cell Signal ; 26(5): 1089-97, 2014 May.
Article in English | MEDLINE | ID: mdl-24509415

ABSTRACT

GATA-2, a member of zinc finger GATA transcription factor family, plays key role in the hematopoietic stem cells self-renewal and differentiation. The transforming growth factor-ß (TGFß) signaling pathway is a major signaling network that controls cell proliferation, differentiation and tumor suppression. Here we found that GATA-2 negatively regulated TGF-ß signaling pathway in Smad4-dependent manner. GATA-2 specifically interacts with Smad4 with its N-terminal while the zinc finger domain of GATA-2 is essential for negative regulation of TGFß. Although GATA-2 did not affect the phosphorylation of Smad2/3 and the complex Smad2/3/4 formation in response to TGFß, the DNA binding activity of Smad4 was decreased significantly by GATA-2 overexpression. Overexpression of GATA-2 in K562 cells led to reduced TGFß-induced erythroid differentiation while knockdown of GATA-2 enhanced TGFß-induced erythroid differentiation. All these results suggest that GATA-2 is a novel negative regulator of TGFß signal pathway.


Subject(s)
GATA2 Transcription Factor/metabolism , Smad4 Protein/metabolism , Transforming Growth Factor beta/metabolism , Activins/pharmacology , Cell Differentiation/drug effects , Cell Line , DNA/metabolism , GATA2 Transcription Factor/antagonists & inhibitors , GATA2 Transcription Factor/genetics , HEK293 Cells , Hep G2 Cells , Histone Deacetylases/metabolism , Humans , K562 Cells , Phosphorylation/drug effects , Protein Binding , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta1/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL