Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Eur J Pharmacol ; 960: 176122, 2023 Dec 05.
Article in English | MEDLINE | ID: mdl-37863414

ABSTRACT

Ulcerative colitis is an inflammatory bowel disease with increasing prevalence and incidence. Current treatments for ulcerative colitis are not generally applicative and are often accompanied by side effects. IGF2 is an endogenous protein that plays roles in anti-inflammation and stemness maintenance, but little is known about its mechanism and function in the progression of ulcerative colitis. In this study, mouse recombinant IGF2 was used in a mouse model of ulcerative colitis established by DSS. IGF2 expression was reduced in colon tissues but not plasma of DSS-induced colitis mice. IGF2R expression was also decreased in colitis colons, which was then elevated by recombinant IGF2. Recombinant IGF2 alleviated colon injury in colitis, which was evaluated by colon shortening, body weight loss and DAI score. IGF2 treatment also relieved the inflammatory response in colitis, which was assessed by the spleen weight index, MPO activity and proinflammatory cytokine expression and was also detected in LPS-stimulated RAW264.7 cells in vitro. Moreover, IGF2R was predicted and further verified to interact with the Sting protein, and the cGAS-Sting pathway as a key pathway for stemness regulation, was upregulated in colonic colons, which was blocked by IGF2 treatment. Additionally, IGF2 treatment can maintain colonic stemness and further repair colonic tight junction function in DSS-induced colitis. In conclusion, IGF2/IGF2R downregulated the cGAS-Sting pathway to sustain colonic stemness and barrier integrity to protect against ulcerative colitis induced by DSS.


Subject(s)
Colitis, Ulcerative , Colitis , Mice , Animals , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/metabolism , Colon , Colitis/chemically induced , Colitis/drug therapy , Colitis/metabolism , Signal Transduction , Nucleotidyltransferases/metabolism , Dextran Sulfate/adverse effects , Disease Models, Animal , Mice, Inbred C57BL
2.
Article in English | MEDLINE | ID: mdl-31678514

ABSTRACT

Atherosclerosis (AS) is characterized by lipids metabolism disorder and inflammatory response. Accumulating evidence has demonstrated that Wingless type 5a (Wnt5a) is implicated in cardiovascular diseases through non-canonical Wnt cascades. However, its precise role during the pathogenesis of AS is still unclear. Therefore, the present study aims to investigate the role and the underlying mechanism of Wnt5a/receptor tyrosine kinase-like orphan receptor 2 (Ror2) pathways in the promotion of AS process through affecting lipid accumulation and inflammation. In atherosclerotic clinical samples, Wnt5a levels were measured by using enzyme-linked immunosorbent assay (ELISA) assay. In vivo experiments were conducted by using apolipoprotein E knockout (apoE-/-) mice model. Vascular smooth muscle cells (VSMCs) were applied for in vitro studies. Wnt5a was highly expressed in both of atherosclerotic clinical samples and apoE-/- mice. The knockdown of Wnt5a significantly inhibited cholesterol accumulation and inflammatory response. Additionally, the lipopolysaccharide (LPS)-induced inflammation aggravated the cholesterol accumulation and decreased adenosine triphosphate (ATP)-binding cassette transporter A1 (ABCA1) expression in VSMCs. Depletion of intracellular cholesterol by ß-cyclodextrin (ß-CD) led to the upregulation of ABCA1 and the inhibition of inflammation. Conversely, the overexpression of Wnt5a inhibited ABCA1 expression, facilitated cholesterol accumulation, impared cholesterol efflux, promoted NF-κB nuclear translocation and the inflammatory cytokines secretion. Moreover, the knockdown of Ror2 increased ABCA1 expression and reduced Wnt5a-induced cholesterol accumulation and inflammatory responses. Furthermore, the knockdown of ABCA1 enhanced cholesterol accumulation and inflammatory response. Therefore, Wnt5a/Ror2 pathway was critical in regulating cholesterol homeostasis and inflammatory response, which might be a promising therapeutic target for AS therapy.


Subject(s)
Atherosclerosis/metabolism , Cholesterol/metabolism , Inflammation/metabolism , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt-5a Protein/metabolism , ATP Binding Cassette Transporter 1/genetics , ATP Binding Cassette Transporter 1/metabolism , Animals , Atherosclerosis/blood , Atherosclerosis/immunology , Case-Control Studies , Disease Models, Animal , Gene Knockdown Techniques , Humans , Inflammation/blood , Inflammation/immunology , Male , Mice , Mice, Knockout, ApoE , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle , Receptor Tyrosine Kinase-like Orphan Receptors/blood , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Signal Transduction/immunology , Wnt-5a Protein/blood , Wnt-5a Protein/genetics
3.
J Breast Cancer ; 22(1): 15-28, 2019 03.
Article in English | MEDLINE | ID: mdl-30941230

ABSTRACT

PURPOSE: Pokemon, also known as ZBTB7A, belongs to the POZ and Krüppel (POK) family of transcription repressors and is implicated in tumor progression as a key proto-oncogene. This present study aimed at determining the mechanism by which Pokemon inhibits transforming growth factor ß (TGFß)-Smad4 pathway-dependent proliferation arrest of breast cancer cells via specificity protein 1 (SP1). METHODS: Over-expressing plasmid or small interfering RNA (siRNA) transfection was used to regulate Pokemon levels. The EdU incorporation assay, MTS assay, and clone formation were used to identify the inhibitory effect of Pokemon siRNA on cell proliferation. Quantitative real-time polymerase chain reaction assay confirmed that Pokemon deletion inhibited the expression of proliferation-associated genes. The dual-luciferase reporter assay, electrophoretic mobility shift assay, and co-immunoprecipitation assay were used to analyze binding between Pokemon, Smad4, and SP1. RESULTS: Pokemon deletion induced proliferation arrest of breast cancer cells and inhibited the expression of proliferation-associated genes, especially Smad4. Pokemon bound with SP1 to interdict Smad4 promoter activity. Information on clinical samples was obtained from The Cancer Genome Atlas data, in which the Pokemon mRNA levels showed a negative correlation with Smad4 levels in different subtypes of breast cancer in two independent datasets. CONCLUSION: We demonstrated that Pokemon binds to SP1 to down-regulate Smad4 expression, thereby promoting proliferation of breast cancer cells. This suggests that Pokemon is a potential TGFß-signaling participant in breast cancer progression.

4.
Oncol Rep ; 38(2): 1303-1311, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28677794

ABSTRACT

Our previous study reported several alternative splicing variants of arginine N-methyltransferase 2 (PRMT2), which lose different exons in the C-terminals of the wild-type PRMT2 gene. Particularly, due to frame-shifting, PRMT2ß encodes a novel amino acid sequence at the C-terminus of the protein, the function of which is not understood. In the present study, we determined the role of PRMT2ß in breast cancer cell proliferation, apoptosis and its effect on the Akt signaling pathway. Stable breast cancer MCF7 cell line with lentivirus-mediated PRMT2ß overexpression was obtained after selection by puromycin for 2 weeks. The effect of lentivirus-mediated PRMT2ß overexpression on breast cancer cellular oncogenic properties was evaluated by MTT, colony formation, cell cycle analysis and apoptosis assays in MCF7 cells. Luciferase activity assay and western blot analysis were performed to characterize the effects of PRMT2ß on cyclin D1 promoter activities and the Akt signaling pathway. Tissue microarray was performed to investigate the association of PRMT2ß with breast cancer progression. Lentivirus-mediated PRMT2ß overexpression suppressed the cell proliferation and colony formation of breast cancer MCF7 cells. PRMT2ß overexpression induced cell cycle arrest and apoptosis of MCF7 cells. Furthermore, PRMT2ß was revealed to suppress the transcription activity of the cyclin D1 promoter, and PRMT2ß was also found to inhibit cyclin D1 expression via the suppression of Akt/GSK-3ß signaling in breast cancer cells. Clinically, it was revealed that PRMT2ß expression was negatively correlated with human epidermal growth factor receptor 2 (HER2) (p=0.033) in breast tumors. Our results revealed that PRMT2ß, a novel splice variant of PRMT2, plays potential antitumor effect by suppressing cyclin D1 expression and inhibiting Akt signaling activity. This also opens a new avenue for treating breast cancer.


Subject(s)
Alternative Splicing , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic , Intracellular Signaling Peptides and Proteins/genetics , Protein-Arginine N-Methyltransferases/genetics , Apoptosis , Breast Neoplasms/metabolism , Case-Control Studies , Cell Proliferation , Cyclin D1/genetics , Cyclin D1/metabolism , Female , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Prognosis , Protein Isoforms , Protein-Arginine N-Methyltransferases/metabolism , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Tumor Cells, Cultured
5.
Int J Oncol ; 50(5): 1567-1578, 2017 May.
Article in English | MEDLINE | ID: mdl-28393241

ABSTRACT

The role of transforming growth factor-ß1 (TGF-ß1) is complicated and plays a different role in the development of cancer. High mobility group A (HMGA1) participates in multiple cellular biology processes, and exerts important roles in the epithelial-mesenchymal transition (EMT). However, the correlation of TGF-ß1 and HMGA1 in cancer cells is not yet fully understood. In this study, we determined the effects of TGF-ß1 on HMGA1 expression in thyroid cancer cells and examined the role of HMGA1 in thyroid cancer progression. With real-time PCR and immunofluorescence staining, our study demonstrated that TGF-ß1 induced the expression of HMGA1 through phosphoinositide 3-kinase (PI3K) and the extracellular signal-related kinase (ERK) signaling in thyroid cancer cells. With luciferase reported assay, the HMGA1 promoter activity was activated by TGF-ß1 in the SW579 cells. Furthermore, lentivirus-mediated HMGA1 knockdown inhibits cellular oncogenic properties of thyroid cancer cells. Clinically, tissue microarray revealed that HMGA1 was expressed in thyroid carcinoma more than that in normal thyroid tissues (P<0.001); expression of HMGA1 and MMP-2 was identified to be positively correlated (P=0.017). The present study established the first link between HMGA1 and TGF-ß1 in the regulation of thyroid cancer proliferation and invasion, and provided evidence for the pivotal role of HMGA1 in the progression of thyroid cancer, indicating HMGA1 to be potential biological marker for the diagnosis of thyroid cancer.


Subject(s)
HMGA1a Protein/genetics , Matrix Metalloproteinase 2/genetics , Thyroid Neoplasms/genetics , Transforming Growth Factor beta1/genetics , Adult , Aged , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Female , Gene Expression Regulation, Neoplastic , HMGA1a Protein/biosynthesis , Humans , MAP Kinase Signaling System , Male , Matrix Metalloproteinase 2/biosynthesis , Middle Aged , Neoplasm Invasiveness/genetics , Thyroid Neoplasms/pathology
6.
J Transl Med ; 14: 80, 2016 Mar 23.
Article in English | MEDLINE | ID: mdl-27008379

ABSTRACT

BACKGROUND: S100A13 and high mobility group A (HMGA1) are known to play essential roles in the carcinogenesis and progression of cancer. However, the correlation between S100A13 and HMGA1 during cancer progression is not yet well understood. In this study, we determined the effects of S100A13 on HMGA1 expression in thyroid cancer cells and examined the role of HMGA1 in thyroid cancer progression. METHODS: Stable ectopic S100A13 expression TT cellular proliferation was evaluated by nude mice xenografts assays. The effect of lentivirus-mediated S100A13 knockdown on thyroid cancer cellular oncogenic properties were evaluated by MTT, colony formation assays and transwell assays in TPC1 and SW579 cells. The effect of siRNA-mediated HMGA1 knockdown on thyroid cancer cellular proliferation and invasion were evaluated by MTT, colony formation assays and transwell assays. The tissue microarray was performed to investigate the correlation between S100A13 and HMGA1 expression in tumor tissues. RESULTS: The ectopic expression of S100A13 could increase tumor growth in a TT cell xenograft mouse model. Moreover, lentivirus-mediated S100A13 knockdown led to the inhibition of cellular oncogenic properties in thyroid cancer cells, and HMGA1 was found to be involved in the effect of S100A13 on thyroid cancer growth and invasion. Furthermore, siRNA-mediated HMGA1 knockdown was proved to inhibit the growth of TPC1 cells and invasive abilities of SW579 cells. Clinically, it was revealed that both S100A13 and HMGA1 showed a higher expression levels in thyroid cancer cases compared with those in matched normal thyroid cases (P = 0.007 and P = 0.000); S100A13 and HMGA1 expressions were identified to be positively correlated (P = 0.004, R = 0.316) when analyzed regardless of thyroid cancer types. CONCLUSIONS: This is the first report for the association between HMGA1 and S100A13 expression in the modulation of thyroid cancer growth and invasion. Those results would provide an essential insight into the effect of S100A13 on carcinogenesis of thyroid tumor, rending S100A13 to be potential biological marker for the diagnosis of thyroid cancer.


Subject(s)
HMGA1a Protein/metabolism , S100 Proteins/metabolism , Thyroid Neoplasms/metabolism , Thyroid Neoplasms/pathology , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Female , Gene Knockdown Techniques , Humans , Lentivirus/metabolism , Male , Mice, Inbred BALB C , Mice, Nude , Microscopy, Fluorescence , Neoplasm Invasiveness , RNA, Small Interfering/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , Xenograft Model Antitumor Assays
7.
Asian Pac J Cancer Prev ; 13(10): 5201-5, 2012.
Article in English | MEDLINE | ID: mdl-23244135

ABSTRACT

BACKGROUND: To evaluate the role of the X-ray repair cross complementing group 3 (XRCC3) T241M polymorphism in head and neck cancer susceptibility. MATERIALS AND METHODS: We performed a meta-analysis of all available studies, which included 3,191 cases and 5,090 controls. RESULTS: Overall, a significant risk effect of the T241M polymorphism was not found under homologous contrast (MM vs TT: OR=1.293, 95% CI=0.926-1.805; TM vs TT: OR=1.148 95% CI=0.930-1.418) and recessive models (MM vs TT+TM): OR=1.170, 95% CI=0.905- 1.512, but a significantly increased risk was observed under a dominant model (MM+TM vs TT): OR=1.243, 95% CI=1.001-1.544. In stratified analyses, there were no significant associations for Asians or Caucasians. CONCLUSION: Our meta-analysis suggested the XRCC3 241M allele (MM+TM) might act as a head and neck cancer risk factor among all subjects, and the effect of T241M polymorphism on head and neck susceptibility should be studied with a larger, stratified population.


Subject(s)
DNA-Binding Proteins/genetics , Genetic Predisposition to Disease , Head and Neck Neoplasms/etiology , Polymorphism, Genetic/genetics , Case-Control Studies , Humans , Prognosis , Risk Factors
8.
Biochem Biophys Res Commun ; 420(1): 17-23, 2012 Mar 30.
Article in English | MEDLINE | ID: mdl-22390933

ABSTRACT

It is well-known that sphingosine-1-phosphate (S1P), the phospholipid content of HDL, binding to S1P receptors can raise COX-2 expression and PGI(2) release through p38MAPK/CREB pathway. In the present study we assess the action of SR-B1 initiated PI3K-Akt-eNOS signaling in the regulation of COX-2 expression and PGI(2) production in response to HDL. We found that apoA1 could increase PGI(2) release and COX-2 expression in ECV 304 endothelial cells. Furthermore, SR-B1 was found to be involved in HDL induced up-regulation of COX-2 and PGI(2). Over-expressed SR-B1 did not significantly increase the expression of COX-2 and the PGI(2) levels, but knock-down of SR-B1 by siRNA could significantly attenuate COX-2 expression and PGI(2) release together with p38MAPK and CREB phosphorylation. Consistently, the declines of p-p38MAPK, p-CREB, COX-2 and PGI(2) were also observed after incubation with LY294002 (25µmol/L; PI3K special inhibitor) or L-NAME (50µmol/L; eNOS special inhibitor). In addition, we demonstrated the increases of PGI(2) release, COX-2 expression and p38MAPK phosphorylation, when nitric oxide level was raised through the incubation of L-arginine (10 or 20nmol/L) in endothelial cells. Taking together, our data support that SR-B1 mediated PI3K-Akt-eNOS signaling was involved in HDL-induced COX-2 expression and PGI(2) release in endothelial cells.


Subject(s)
Endothelial Cells/metabolism , Epoprostenol/biosynthesis , Lipoproteins, HDL/metabolism , Scavenger Receptors, Class B/metabolism , Apolipoprotein A-I/metabolism , Apolipoprotein A-I/pharmacology , Cell Line , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclooxygenase 2/biosynthesis , Endothelial Cells/drug effects , Humans , Lipoproteins, HDL/pharmacology , Nitric Oxide Synthase Type III/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Scavenger Receptors, Class B/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Recent Pat Anticancer Drug Discov ; 7(2): 154-67, 2012 May 01.
Article in English | MEDLINE | ID: mdl-22339355

ABSTRACT

ATP citrate lyase (ACL or ACLY) is an extra-mitochondrial enzyme widely distributed in various human and animal tissues. ACL links glucose and lipid metabolism by catalyzing the formation of acetyl-CoA and oxaloacetate from citrate produced by glycolysis in the presence of ATP and CoA. ACL is aberrantly expressed in many immortalized cells and tumors, such as breast, liver, colon, lung and prostate cancers, and is correlated reversely with tumor stage and differentiation, serving as a negative prognostic marker. ACL is an upstream enzyme of the long chain fatty acid synthesis, providing acetyl-CoA as an essential component of the fatty acid synthesis. Therefore, ACL is a key enzyme of cellular lipogenesis and potent target for cancer therapy. As a hypolipidemic strategy of metabolic syndrome and cancer treatment, many small chemicals targeting ACL have been designed and developed. This review article provides an update for the research and development of ACL inhibitors with a focus on their patent status, offering a new insight into their potential application.


Subject(s)
ATP Citrate (pro-S)-Lyase/antagonists & inhibitors , Antineoplastic Agents/chemistry , Drug Design , Enzyme Inhibitors/chemistry , Lipogenesis/drug effects , Neoplasms/drug therapy , Neoplasms/enzymology , ATP Citrate (pro-S)-Lyase/chemistry , ATP Citrate (pro-S)-Lyase/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Citric Acid/analogs & derivatives , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Humans , Mice , Patents as Topic , Protein Processing, Post-Translational
10.
Eur J Pharmacol ; 676(1-3): 51-6, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-22173123

ABSTRACT

Numerous studies revealed that spinal inflammation and immune response play an important role in neuropathic pain. In this study, we investigated the effects of intrathecal injection of a Toll-like receptor (TLR4) inhibitor epigallocatechin gallate (EGCG) on neuropathic pain induced by chronic constriction injury of the sciatic nerve (CCI). A total of 120 rats were randomly assigned into 4 groups: sham-operated group, CCI group, CCI plus normal saline group and CCI plus EGCG group. CCI and sham surgeries were performed and both thermal hyperalgesia and mechanical allodynia were tested. Lumbar spinal cord was sampled and the mRNA and protein expressions of TLR4 and High Mobility Group 1 protein (HMGB1) were detected, the contents of tumor necrosis factor alpha (TNF-α), interleukin-1 beta (IL-1ß) and interleukin-10 (IL-10) were measured by ELISA, and immunohistochemistry for nuclear factor kappa B (NF-κB) was also carried out. When compared with the sham group, both mechanical and heat pain thresholds were significantly decreased, and the mRNA and protein expressions of TLR4 and HMGB1, the contents of TNF-α, IL-1ß and IL-10 in the spinal cords and NF-κB expression in the spinal dorsal horn were markedly increased in CCI rats (P<0.05). After intrathecal injection of EGCG (1mg/kg) once daily from 1day before to 3days after CCI surgery, the expressions of TLR4, NF-κB, HMGB1, TNF-α and IL-1ß were markedly decreased while the content of IL-10 in the spinal cord increased significantly accompanied by dramatical improvement of pain behaviors in CCI rats (P<0.05). These results show that the TLR4 signaling pathway plays an important role in the occurrence and development of neuropathic pain, and the therapy targeting TLR4 might be a novel strategy in the treatment of neuropathic pain.


Subject(s)
Catechin/analogs & derivatives , Injections, Spinal , Neuralgia/drug therapy , Toll-Like Receptor 4/antagonists & inhibitors , Animals , Catechin/administration & dosage , Catechin/pharmacology , Catechin/therapeutic use , Chronic Disease , Constriction , Gene Expression Regulation/drug effects , HMGB1 Protein/genetics , HMGB1 Protein/metabolism , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Male , NF-kappa B/metabolism , Neuralgia/genetics , Neuralgia/metabolism , Neuralgia/prevention & control , Pain Threshold/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Sciatic Nerve/drug effects , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/physiopathology , Spinal Cord/drug effects , Spinal Cord/metabolism , Spinal Cord/physiopathology , Toll-Like Receptor 4/genetics , Toll-Like Receptor 4/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL