Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters










Publication year range
1.
Zhongguo Xiu Fu Chong Jian Wai Ke Za Zhi ; 38(4): 480-486, 2024 Apr 15.
Article in Chinese | MEDLINE | ID: mdl-38632070

ABSTRACT

Objective: To explore the therapeutic effect of basic fibroblast growth factor (bFGF) on spinal cord injury (SCI) in rats and the influence of Notch/signal transducer and activator of transcription 3 (STAT3) signaling pathway. Methods: A total of 40 10-week-old male Sprague Dawley (SD) rats were selected to establish T 10-segment SCI model by a free falling object. Among them, 32 successful models were randomly divided into model group and bFGF group, with 16 in each group. Another 16 SD rats were selected as sham-operation group, with only T 10 processes, dura mater, and spinal cord exposed. After modeling, the rats in bFGF group were intraperitoneally injected with 100 µg/kg bFGF (once a day for 28 days), and the rats in model group and sham-operation group were injected with normal saline in the same way. The survival of rats in each group were observed after modeling. Basso-Beattie-Bresnahan (BBB) scores were performed before modeling and at immediate, 14 days, and 28 days after modeling to evaluate the functional recovery of hind limbs. Then, the spinal cord tissue at the site of injury was taken at 28 days and stained with HE, Nissl, and propidium iodide (PI) to observe the pathological changes, neuronal survival (number of Nissl bodies) and apoptosis (number of PI red stained cells) of the spinal cord tissue; immunohistochemical staining and ELISA were used to detect the levels of astrocyte activation markers [glial fibrillary acidic protein (GFAP)] and inflammatory factors [interleukin 1ß (IL-1ß), tumor necrosis factor α (TNF-α), interferon γ (IFN-γ)] in tissues, respectively. Western blot was used to detect the expressions of Notch/STAT3 signaling pathway related proteins [Notch, STAT3, phosphoryl-STAT3 (p-STAT3), bone morphogenetic protein 2 (BMP-2)] in tissues. Results: All rats survived until the experiment was completed. At immediate after modeling, the BBB scores in model group and bFGF group significantly decreased when compared to sham-operation group ( P<0.05). At 14 and 28 days after modeling, the BBB scores in model group significantly decreased when compared to sham-operation group ( P<0.05); the bFGF group showed an increase compared to model group ( P<0.05). Compared with before modeling, the BBB scores of model group and bFGF group decreased at immediate after modeling, and gradually increased at 14 and 28 days, the differences between different time points were significant ( P<0.05). The structure of spinal cord tissue in sham-operation group was normal; in model group, there were more necrotic lesions in the spinal cord tissue and fewer Nissl bodies with normal structures; the number of necrotic lesions in the spinal cord tissue of the bFGF group significantly reduced compared to the model group, and some normally structured Nissl bodies were visible. Compared with sham-operation group, the number of Nissl bodies in spinal cord tissue significantly decreased, the number of PI red stained cells, GFAP, IL-1ß, TNF-α, IFN-γ, Notch, p-STAT3 /STAT3, BMP-2 protein expression levels significantly increased in model group ( P<0.05). The above indexes in bFGF group significantly improved when compared with model group ( P<0.05). Conclusion: bFGF can improve motor function and pathological injury repair of spinal cord tissue in SCI rats, improve neuronal survival, and inhibit neuronal apoptosis, excessive activation of astrocytes in spinal cord tissue and inflammatory response, the mechanism of which may be related to the decreased activity of Notch/STAT3 signaling pathway.


Subject(s)
Fibroblast Growth Factor 2 , Spinal Cord Injuries , Rats , Male , Animals , Rats, Sprague-Dawley , Fibroblast Growth Factor 2/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology , STAT3 Transcription Factor/therapeutic use , Tumor Necrosis Factor-alpha/metabolism , Spinal Cord Injuries/therapy , Spinal Cord/metabolism , Signal Transduction
2.
ACS Infect Dis ; 10(3): 988-999, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38317607

ABSTRACT

Escherichia coli continues to be the predominant Gram-negative pathogen causing neonatal meningitis worldwide. Inflammatory mediators have been implicated in the pathogenesis of meningitis and are key therapeutic targets. The role of interleukin-22 (IL-22) in various diseases is diverse, with both protective and pathogenic effects. However, little is understood about the mechanisms underlying the damaging effects of IL-22 on the blood-brain barrier (BBB) in E. coli meningitis. We observed that meningitic E. coli infection induced IL-22 expression in the serum and brain of mice. The tight junction proteins (TJPs) components ZO-1, Occludin, and Claudin-5 were degraded in the mouse brain and human brain microvascular endothelial cells (hBMEC) following IL-22 administration. Moreover, the meningitic E. coli-caused increase in BBB permeability in wild-type mice was restored by knocking out IL-22. Mechanistically, IL-22 activated the STAT3-VEGFA signaling cascade in E. coli meningitis, thus eliciting the degradation of TJPs to induce BBB disruption. Our data indicated that IL-22 is an essential host accomplice during E. coli-caused BBB disruption and could be targeted for the therapy of bacterial meningitis.


Subject(s)
Escherichia coli Infections , Meningitis, Bacterial , Meningitis, Escherichia coli , Humans , Mice , Animals , Blood-Brain Barrier , Meningitis, Escherichia coli/metabolism , Meningitis, Escherichia coli/microbiology , Meningitis, Escherichia coli/pathology , Escherichia coli/metabolism , Endothelial Cells , Interleukin-22 , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
3.
Toxicon ; 240: 107627, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38253207

ABSTRACT

BACKGROUND: Lung adenocarcinoma (LUAD) is the most popular type of lung cancer. Sulfotanshinone IIA sodium (STS IIA) has been proven to have an anticancer effect. However, its role in LUAD and its underlying mechanism remain unclear. OBJECTIVE: To investigate the role and mechanism of STS IIA in LUAD angiogenesis. METHODS: The mRNA levels of genes, including forkhead box O3 (FOXO3) and chemokine C-X-C motif ligand 1 (CXCL1), were detected by qRT-PCR. The levels of proteins, including FOXO3, CXCL1, and vascular endothelial growth factor (VEGF), were measured by Western blot. The proliferation and angiogenesis of human umbilical vein endothelial cells (HUVECs) were detected by the EdU assay and the tubule formation assay, respectively. The binding relationship between FOXO3 and CXCL1 was detected by dual-luciferase reporter assay. RESULTS: Our results illustrated that different concentrations of STS IIA inhibited the proliferation and angiogenesis of HUVECs. FOXO3 regulated the proliferation and angiogenesis of HUVECs inhibited by STS ⅡA via targeting CXCL1. Subsequently, we proved that exogenous CXCL1 alleviated the inhibition of proliferation and angiogenesis of HUVECs regulated by STS IIA via activating the STAT3/VEGF pathway. Finally, we found that STS IIA inhibited the angiogenesis of lung adenocarcinoma though FOXO3 to inhibit the CXCL1/STAT3/VEGF pathway. CONCLUSION: Our study finally elucidated the underlying molecular mechanism by which STS ⅡA inhibits LUAD angiogenesis.


Subject(s)
Adenocarcinoma of Lung , Vascular Endothelial Growth Factor A , Humans , Vascular Endothelial Growth Factor A/metabolism , Signal Transduction , Cell Proliferation , Angiogenesis , Human Umbilical Vein Endothelial Cells , Adenocarcinoma of Lung/metabolism , Neovascularization, Pathologic , Chemokine CXCL1/metabolism , Chemokine CXCL1/pharmacology , Forkhead Box Protein O3/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
5.
Clin Exp Hypertens ; 46(1): 2297642, 2024 Dec 31.
Article in English | MEDLINE | ID: mdl-38147409

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by progressive vascular remodeling caused by the excessive proliferation and survival of pulmonary artery smooth muscle cells (PASMCs). Dual-specificity tyrosine regulated kinase 1A (DYRK1A) is a pleiotropic kinase involved in the regulation of multiple biological functions, including cell proliferation and survival. However, the role and underlying mechanisms of DYRK1A in PAH pathogenesis remain unclear. We found that DYRK1A was upregulated in PASMCs in response to hypoxia, both in vivo and in vitro. Inhibition of DYRK1A by harmine significantly attenuated hypoxia-induced pulmonary hypertension and pulmonary artery remodeling. Mechanistically, we found that DYRK1A promoted pulmonary arterial remodeling by enhancing the proliferation and survival of PASMCs through activating the STAT3/Pim-1/NFAT pathway, because STAT3 gain-of-function via adeno-associated virus serotype 2 (AAV2) carrying the constitutively active form of STAT3 (STAT3C) nearly abolished the protective effect of harmine on PAH. Collectively, our results reveal a significant role for DYRK1A in pulmonary arterial remodeling and suggest it as a drug target with translational potential for the treatment of PAH.


Subject(s)
Hypertension, Pulmonary , Pulmonary Arterial Hypertension , Humans , Pulmonary Arterial Hypertension/metabolism , Vascular Remodeling , Harmine/adverse effects , Harmine/metabolism , Hypertension, Pulmonary/drug therapy , Hypertension, Pulmonary/metabolism , Pulmonary Artery , Hypoxia , Myocytes, Smooth Muscle/metabolism , Cell Proliferation , Cells, Cultured , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
6.
BMC Oral Health ; 23(1): 846, 2023 11 09.
Article in English | MEDLINE | ID: mdl-37946196

ABSTRACT

BACKGROUND: Toosendanin (TSN) exhibits potent antitumor activity against various tumor cell lines. However, its efficacy against oral squamous cell carcinoma (OSCC) remains unknown. Here, we investigated the effects of TSN on OSCC cells in vitro and verified them in vivo using a patient-derived xenograft (PDX) model. METHODS: The effect of TSN on OSCC cells was investigated by cytotoxicity assays and flow cytometry. The expression of proteins was detected by western blotting. An OSCC PDX model was constructed to further investigate the role of TSN in regulating the function of OSCC. RESULTS: The cell viability of CAL27 and HN6 cells decreased as the concentration of TSN increased within the experimental range. Compared with controls, TSN at lower doses inhibited cell proliferation and induced apoptosis through S-phase cell cycle arrest. TSN inhibited OSCC cell proliferation by downregulating the STAT3 pathway through the inhibition of STAT3 phosphorylation. After successful construction of the OSCC PDX model with high pathological homology to the primary tumor and treatment with an intraperitoneal injection of TSN, we showed that TSN significantly reduced the tumor size of the PDX model mice without obvious toxicity. CONCLUSIONS: Both in vitro and in vivo, TSN significantly inhibits the proliferation and promoted apoptosis of OSCC cells. Furthermore, TSN demonstrates potent inhibition of STAT3 phosphorylation, indicating its potential as a promising therapeutic agent for OSCC. Therefore, TSN holds great promise as a viable drug candidate for the treatment of OSCC.


Subject(s)
Carcinoma, Squamous Cell , Drugs, Chinese Herbal , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Animals , Mice , Carcinoma, Squamous Cell/pathology , Squamous Cell Carcinoma of Head and Neck , Mouth Neoplasms/pathology , Cell Proliferation , Drugs, Chinese Herbal/pharmacology , Drugs, Chinese Herbal/therapeutic use , Cell Line, Tumor , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology , STAT3 Transcription Factor/therapeutic use
7.
J Cosmet Dermatol ; 22(12): 3511-3519, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37563868

ABSTRACT

BACKGROUND: Hypertrophic scars (HS) often affect the normal function and appearance of the skin and bring adverse effects to the body and mind of patients, being a challenge in the fields of burns and plastic surgery as well as rehabilitation. Despite significant efficacy of centella triterpenes cream for treating HS clinically, its pharmacodynamics and molecular targets are still unclear. Therefore, the network pharmacology analysis combined with in vitro cell molecular biology experiments was used to explore the mechanism and targets of centella triterpenes cream treating HS in this study. METHODS: First, target genes of asiaticoside (AC) were obtained from the databases including the Comparative Toxicogenomics Database, similarity ensemble approach, SwissTargetPrediction and TargetNet, and HS targets were acquired from the databases like Disgenet, GeneCards, and Online Mendelian Inheritance in Man. The common targets of AC-HS were obtained through plotting a Venn diagram. Subsequently, STRING 11.0 was employed for analyzing the protein-protein interaction (PPI) network of the common targets, and cytoscape 3.9.0 for analyzing the connectivity of PPI and plotting the network diagram of "drug-component-target". Additionally, a modified tissue culture method was applied to separate primary normal fibroblasts (NFs) in human skin and hypertrophic scar fibroblasts (HSFs). HSFs after 24-h AC treatment were subjected to MTT assay to detect cell viability, scratch assay to assess cell migration ability, and western blot to test the protein expression levels of STAT3, p-STAT3, transforming growth factor-ß1 (TGF-ß1), collagen I (COL 1), fibronectin 1 (FN1), and alpha-smooth muscle actin (α-SMA). RESULTS: In network pharmacology analysis, 134 pharmacodynamic targets of AC and 2333 HS targets were obtained after retrieving the database, 50 AC-HS common targets were obtained by a Venn diagram, and a total of 178 edges and 13 core genes such as JUN and STAT3 were acquired by PPI analysis. In vitro experiments showed that the phosphorylation level of STAT3 (p-STAT3) was increased in HSFs. In addition to reducing p-STAT3 in HSFs, AC significantly inhibited the cell viability and migration of HSFs and downregulated the protein levels of TGF-ß1, COL 1, FN 1, and α-SMA. CONCLUSION: STAT3 can be activated in HS. AC may exert its pharmacological effects of inhibiting TGF-ß1 signal transduction and regulating extracellular matrix remodeling in HS by inhibiting STAT3 phosphorylation. However, the specific molecular mechanism of AC remains to be verified through further experiments.


Subject(s)
Centella , Cicatrix, Hypertrophic , Humans , Cicatrix, Hypertrophic/drug therapy , Transforming Growth Factor beta1/metabolism , Phosphorylation , Network Pharmacology , Fibroblasts , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
8.
Clinics (Sao Paulo) ; 78: 100268, 2023.
Article in English | MEDLINE | ID: mdl-37567042

ABSTRACT

OBJECTIVE: Trastuzumab is the preferred drug for the treatment of breast cancer. However, research on the cellular mechanisms of trastuzumab's potential cardiotoxicity is insufficient. The purpose of this study was to explore the toxic effects and potential mechanism of action of trastuzumab on cardiomyocytes. METHOD: Human Cardiomyocyte (HCM) viability was assessed using the MTT method. HCM apoptosis was detected using the Hoechst33342/PI Fluorescent staining. The LDH and CK activities of the cell were measured using commercially available LDH and CK assay kits. The expression levels of Notch2, JAK2, STAT3, cleaved caspase 3, bax, and bcl 2 in HCMs were detected using western blotting. RESULTS: The results showed that 250 mg/L trastuzumab induced cardiomyocyte injury and apoptosis, inhibited viability, activated the Notch2 receptor, and inhibited JAK2/STAT3 expression in HCM. Inhibition of Notch2 expression in HCM by targeted siNotch2 transfection reversed the trastuzumab-induced injury and apoptosis, and the expression of JAK2/STAT3 returned to normal levels. CONCLUSIONS: Trastuzumab induces Notch2 expression by inhibiting the JAK2/STAT3 pathway of HCMs, promotes cell apoptosis, and causes cardiomyocyteinjury. Notch2 may be a potential target of trastuzumab-inducedmyocardial injury. This experiment reveals the mechanism of trastuzumab-induced cardiotoxicity, providing a theoretical basis for the application of trastuzumab.


Subject(s)
Cardiotoxicity , Myocytes, Cardiac , Humans , Myocytes, Cardiac/metabolism , Trastuzumab/adverse effects , Trastuzumab/metabolism , Cardiotoxicity/etiology , Cardiotoxicity/metabolism , Receptor, Notch2/metabolism , Apoptosis , Janus Kinase 2/metabolism , Janus Kinase 2/pharmacology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
9.
Mol Biol (Mosk) ; 57(4): 706-708, 2023.
Article in Russian | MEDLINE | ID: mdl-37528791

ABSTRACT

Hepatocellular carcinoma (HCC) is the most frequently diagnosed primary liver tumor worldwide. Tumor-associated macrophages (TAMs) usually have a similar phenotype to M2-like macrophages and can participate in tumor progression by secreting cytokines to suppress the immune response and activity of tumor-infiltrating lymphocytes. We investigated the role of M2 macrophages in HCC progression and explored the effects of vascular endothelial growth factor receptor 2 inhibitor-apatinib. As a cellular model of HCC, Hepb3 cell line was used. M2 macrophages were obtained by differentiation of THP-1 cells. The Transwell chamber was used to co-culture M2 macrophages and Hepb3 cells. CCK-8 and EdU assays were conducted to measure cell viability and proliferation capacity. Transwell migration assay was performed to estimate cellular metastatic potential. Cytokine expression levels were assessed by ELISA. Western blotting was used to characterize activation of the VEGFR2/STAT3/PD-L1 axis. It has been shown that co-culture with M2 macrophages increased viability, cytokine production, promoted proliferation, invasion, and migration of Hepb3 cells. The secretion of TGF-ß1, IL-6, MMP-9, and VEGF was significantly increased after co-culture. In contrast apatinib suppressed M2 macrophage-induced proliferation, cell viability, invasion, and migration of Hepb3 cells. Moreover, apatinib markedly decreased expression levels of p-VEGFR2, p-STAT3, and PD-L1 in Hepb3 cells under the co-culture conditions. In conclusion, apatinib treatment can suppress TAMs-mediated malignant behavior of HCC cells via modulation of the VEGFR2/STAT3/PD-L1 signaling pathway.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , B7-H1 Antigen/genetics , Vascular Endothelial Growth Factor A/genetics , Cell Line , Signal Transduction , Macrophages/metabolism , Macrophages/pathology , Cytokines/metabolism , Cell Line, Tumor , Cell Proliferation , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
10.
Curr Drug Targets ; 24(11): 919-928, 2023.
Article in English | MEDLINE | ID: mdl-37534791

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is associated with a high mortality rate due to early recurrence and its metastasis features. To this day, effective treatment options for metastatic HCC remain a major challenge to patient treatment. Flavokawain B (FKB) is a naturally occurring chalcone molecule capable of providing effective therapy against this life-threatening disease. OBJECTIVE: This study investigated the anti-metastatic effects of FKB on the growth and development of metastatic HCC. METHODS: HepG2 cells were used in this study and a neutral red assay was performed to determine the IC50 value of FKB. Cell scratch and exclusion zone assays were performed to assess the rate of cell migration and invasion. Relative mRNA levels of UCK2, STAT3, VEGF and HIF-1α genes were quantified using RT-qPCR. RESULTS: FKB inhibited the proliferation of HepG2 cells at an IC50 value of 28 µM after 72 h of incubation. Its cytotoxic effect was confirmed to induce apoptosis through the phase-contrast inverted microscope. Cell migration and invasion were significantly inhibited at 7, 14, and 28 µM of FKB as compared to untreated cells. The inhibition in the cell migration significantly increased with the increasing concentrations of the bioactive compound. The relative expression levels of the UCK2 gene and its downstream genes, STAT3, VEGF and HIF-1α, were significantly downregulated after 72 h exposure to FKB treatment. CONCLUSION: Our data suggest that FKB inhibited HepG2 proliferation and further suppressed its metastasis partly by regulating the STAT3/Hif-1α/VEGF signalling pathway. FKB could be a potential alternative and viable strategy against HCC.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Vascular Endothelial Growth Factor A/genetics , Liver Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cell Proliferation , Cell Line, Tumor , Uridine Kinase , STAT3 Transcription Factor/pharmacology
11.
Clinics (Sao Paulo) ; 78: 100265, 2023.
Article in English | MEDLINE | ID: mdl-37562217

ABSTRACT

OBJECTIVES: The authors determined the level of Expression of Leptin (LEP) in Polycystic Ovary Syndrome (PCOS) patients with or without obesity and in GCs treated with insulin. METHODS: LEP expression was first assessed in ovary cortex specimens collected from women with PCOS with or without obesity as well as from healthy controls. Ovarian Granulosa Cells (OGCs) induced by insulin extracted from a mouse model were used in further functional research. RESULTS: Real-time PCR and western blotting indicated that LEP expression was upregulated in GCs induced by insulin, in comparison with that in GCs not induced by insulin. Furthermore, the knockdown of LEP resulted in a reduction in growth and multiplication and an increase in apoptosis and inflammation in GCs induced by insulin. Next, the authors evaluated the effect of LEP on three key pathways of inflammation (MAPK, NF-kB, and JAK1/STAT3); results showed that the JAK1/STAT3 pathway was induced by LEP knockdown, as evidenced by the upregulation of phosphor-JAK1, phosphor-STAT3, and nuclear STAT3 expression. Administration of curcumin, a specific inhibitor of STAT3, counteracted the effect of LEP knockdown on cell inflammation and apoptosis. CONCLUSION: The present data suggest that upregulation of LEP expression in the PCOS granulosa cell model is essential for reducing apoptosis and inflammation by modulating the JAK1/STAT3 pathway axis.


Subject(s)
Polycystic Ovary Syndrome , Humans , Mice , Animals , Female , Polycystic Ovary Syndrome/metabolism , Leptin/adverse effects , Leptin/metabolism , Granulosa Cells/metabolism , Insulin , Obesity , Apoptosis , Janus Kinase 1/metabolism , Janus Kinase 1/pharmacology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
12.
J Eur Acad Dermatol Venereol ; 37(9): 1920-1927, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37262304

ABSTRACT

BACKGROUND: Pemphigus vulgaris (PV) is a potentially fatal autoimmune bullous disease primarily caused by acantholysis of keratinocytes attributed to pathogenic desmoglein-3 (Dsg3) autoantibodies. Interleukin-37 (IL-37) reportedly plays important roles in a variety of autoimmune diseases, but its role in PV is not clear. OBJECTIVES: To investigate whether IL-37 plays a role in the occurrence and progression of PV. METHODS: HaCaT keratinocytes were stimulated with anti-Dsg3 antibody to establish an in vitro PV model, which was defined as anti-Dsg3 group. Cells incubated with medium without anti-Dsg3 treatment were used as control. IL-37 was cultured with these cells infected with or without lentiviral vector shRNA-Caveolin-1 (sh-Cav-1-LV). Cell dissociation assay and immunocytofluorescence were performed to assess keratinocyte dissociation, keratin retraction and Dsg3 endocytosis. Real-time PCR was used to detect the mRNA level of Cav-1, and western blot was used to determine the protein expression of Cav-1, Dsg3, STAT3 and phosphorylated-STAT3 (p-STAT3). RESULTS: The anti-Dsg3 group showed more cell debris, increased keratin retraction, increased Dsg3 endocytosis, reduced Cav-1 expression and co-localization than the control group, while IL-37 treatment neutralized all of these changes. Interestingly, Cav-1 knockdown supressed the inhibitory effect of IL-37 on keratinocyte dissociation and Dsg3 internalization. The protein expression of p-STAT3 was increased in keratinocytes of the PV model but decreased by IL-37. Re-activation of the STAT3 pathway by colivelin supressed the inhibitory effect of IL-37 on keratinocyte dissociation and Dsg3 internalization, along with upregulation of Cav-1 and Dsg3. CONCLUSIONS: IL-37 inhibited keratinocyte dissociation and Dsg3 endocytosis in an in vitro PV model through the upregulating Cav-1 and inhibiting STAT3 pathway.


Subject(s)
Caveolin 1 , Interleukins , Humans , Autoantibodies , Caveolin 1/metabolism , Caveolin 1/pharmacology , Desmoglein 3 , Endocytosis , Interleukins/metabolism , Keratinocytes/metabolism , Keratins/metabolism , Pemphigus/pathology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology , Up-Regulation
13.
Clin Breast Cancer ; 23(4): 388-396, 2023 06.
Article in English | MEDLINE | ID: mdl-36872108

ABSTRACT

BACKGROUND: This study was to investigate the functional role and mechanism of receptor activator of nuclear factor-kappa B ligand (RANKL) associated autophagy and chemoresistance in breast cancer. MATERIALS AND METHODS: Cell Counting Kit-8 (CCK-8) assay was used to detect the cell viability. Real-time polymerase chain reaction (PCR) was used for determining the relative mRNA levels of key genes and protein expression was assessed by Western blotting. Immunofluorescence was performed to evaluate the changes in the autophagy flux. Short hairpin (shRNA) was used to knockdown the expression of the target genes in breast cancer cells. Based on The Cancer Genome Atlas (TCGA) database, we explored the expression of receptor activator of nuclear factor-kappa B (RANK), autophagy and signal transducer and activator of transcription 3 (STAT3) signaling associated genes and analyzed their correlation with the prognosis of breast cancer patients. RESULTS: The findings showed that receptor activator of nuclear factor-kappa B ligand (RANKL), the ligand of RANK, could effectively enhance the chemoresistance potential of breast cancer cells. Our results showed that RANKL induced autophagy and enhanced the expression of autophagy associated genes in breast cancer cells. The knockdown of RANK suppressed RANKL mediated autophagy induction in these cells. Furthermore, the inhibition of autophagy suppressed RANKL mediated chemoresistance in breast cancer cells. We found STAT3 signaling pathway was involved in RANKL-induced autophagy. Analysis of the expression of RANK, and autophagy and STAT3 signaling associated genes in breast cancer tissues showed that the expression of autophagy and STAT3 signaling associated genes was correlated with the prognosis of breast cancer patients. CONCLUSION: The present study suggests that the RANKL/RANK axis may potentially mediate chemoresistance in breast cancer cells by inducing autophagy through the STAT3 signaling pathway.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , RANK Ligand/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology , Signal Transduction , Autophagy
14.
Physiol Genomics ; 55(5): 213-221, 2023 05 01.
Article in English | MEDLINE | ID: mdl-36939206

ABSTRACT

Current research has shown that inhibiting deoxythymidylate kinase (DTYMK) can significantly reduce development of lung cancer without liver kinase B1. However, its underlying regulatory mechanism is still unclear. We therefore aimed to investigate whether DTYMK inhibitors could suppress lung adenocarcinoma (LUAD) progression. In this study, human tissues, A549 cells, and xenograft tumors were used to explore the regulation and mechanism of DTYMK on LUAD cell proliferation and migration. Meanwhile, YMU1 (a DTYMK inhibitor) was applied to A549 cells and xenograft tumors to investigate its potential as a drug for LUAD. DTYMK was overexpressed in LUAD tissues and correlated with tumor stage. Knockdown of DTYMK suppressed cell viability, migration, and invasion. In addition, the activation of signal transducers and activators of transcription 3 (STAT3) was repressed upon DTYMK inhibition. YMU1 showed the same effect as DTYMK knockdown in vivo and in vitro. DTYMK plays an important role in progression of LUAD through the STAT3 signaling pathway. YMU1 may have the potential to inhibit the development of LUAD.NEW & NOTEWORTHY DTYMK plays an important role in progression of LUAD through the STAT3 signaling pathway. YMU1 may serve as a novel drug to suppress the development of LUAD.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Humans , Thymidine Monophosphate/pharmacology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Signal Transduction , Lung/pathology , Cell Proliferation , Cell Movement , Gene Expression Regulation, Neoplastic , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
15.
Poult Sci ; 102(3): 102441, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36599221

ABSTRACT

This study aimed to evaluate the effect of isoleucine (Ile) on growth performance, meat quality and lipid metabolism of broilers fed a low-protein diet (LPD). The 396 one-day-old male Cobb broilers were allocated to 4 treatment groups as follows: control diet (CON), LPD, LPD + 0.13% Ile (LPD-LI) and LPD + 0.26% Ile (LPD-HI), with nine replicates of 11 broilers each for 42 d. The Ile increased average daily gain, average daily feed intake, fiber density and the mRNA level of myosin heavy chain (MyHC)-I in breast muscle, and decreased feed to gain ratio, shear force, fiber diameter and the mRNA level of MyHC-IIb in breast muscle, which were impaired by the LPD. Compared to the LPD group, broilers in LPD-LI and LPD-HI groups had lower serum lipid levels, liver fat content, abdominal adipose percentage and mRNA levels of peroxisome proliferator-activated receptor-γ, CCAAT/enhancer binding protein-α, ki-67, topoisomerase II alpha (TOP2A) and thioredoxin-dependent peroxidase 2 in abdominal adipose and liver X receptors-α, sterol regulatory element binding protein 1 (SREBP1), acetyl-CoA carboxylase (ACC) and fatty acid synthase (FAS) in liver, and higher mRNA levels of peroxisome proliferator activated receptor-α, carnitine palmitoyl-transferase 1 (CPT-1), and acyl-CoA oxidase 1 (ACOX1) in liver, which were equal to the CON levels. A LPD supplemented with Ile decreased enzyme activities of ACC and FAS in liver and glycerol-3-phosphate dehydrogenase and TOP2A in abdominal adipose, and increased enzyme activities of CPT-1 and ACOX1 in liver. Furthermore, Ile supplementation enhanced the mRNA level of leptin receptor and protein levels of phospho-5' adenosine monophosphate-activated protein kinase (AMPK), mechanistic target of rapamycin, ribosomal protein 70 S6 kinase, janus kinase 2 (JAK2), and signal transducer and activator of transcription 3 (STAT3), and decreased the protein level of SREBP1 in the liver of broilers in LPD group. In conclusion, dietary supplementation with Ile to 0.83% could improve growth performance and meat quality and alleviate lipid deposition of broilers fed a LPD through activating AMPK and JAK2/STAT3 signaling pathways.


Subject(s)
Chickens , Isoleucine , Male , Animals , Isoleucine/metabolism , Chickens/physiology , Diet, Protein-Restricted/veterinary , AMP-Activated Protein Kinases/metabolism , Janus Kinase 2/metabolism , Janus Kinase 2/pharmacology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology , Dietary Supplements , Diet/veterinary , Liver/metabolism , Signal Transduction , RNA, Messenger/metabolism , Lipids , Lipid Metabolism
16.
Eur J Haematol ; 110(4): 435-443, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36576398

ABSTRACT

BACKGROUND: It is known that the microenvironmental cytokine interferon gamma (IFN-γ) provides a survival advantage for chronic lymphocytic leukemia (CLL) cells. However, the mechanisms involved in this effect have not been properly investigated. METHODS: Herein, we conducted a comprehensive screening of the effects of IFN-γ on signaling pathways and gene expression profiles in CLL cells by using western blotting, real-time quantitative reverse transcription (RT-qPCR) and high-throughput RNA sequencing (RNA-seq). RESULTS: We found that IFN-γ not only activated the pro-survival signal transducer and activator of transcription 3 (STAT3), but also activated the protein kinase B and extracellular signal-regulated kinase signaling pathways. RNA-seq analysis showed that IFN-γ stimulation changed the expression profiles of more than 500 genes, with 391 being up-regulated and 123 down-regulated. These genes are involved in numerous biological processes, including anti-apoptosis, cell migration, and proliferation. IFN-γ significantly up-regulated the expression of CD38, BCL6, CXCL9, BCL2A1, SCOS3, IL-10, HGF, EGFR, THBS-1, FN1, and MUC1, which encode proteins potentially associated with disease progression, worse prognosis or poor response to treatment. Blocking janus kinases1/2 (JAK1/2) or STAT3 signal by specific inhibitors affected the expression of most genes, suggesting a pivotal role of the JAK1/2-STAT3 pathway in IFN-γ pro-survival effects in CLL. CONCLUSIONS: Our data demonstrate that IFN-γ regulates a complex pro-survival signal network in CLL through JAK1/2-STAT3, which provides a rational explanation for IFN-γ promoting CLL cells survival and drug resistance.


Subject(s)
Interferon-gamma , Leukemia, Lymphocytic, Chronic, B-Cell , Humans , Cytokines/metabolism , Interferon-gamma/immunology , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Signal Transduction , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
17.
Eur J Pharmacol ; 938: 175434, 2023 Jan 05.
Article in English | MEDLINE | ID: mdl-36462735

ABSTRACT

Benign prostatic hyperplasia (BPH) is a chronic proliferative non-tumorous disease that mainly bothers males older than 50 and significantly disturbs the quality of life. Cryptotanshinone (CTS), a herbal extract, has been proven with therapeutic effects on various diseases. However, the effects and possible mechanisms of CTS in BPH have not yet been elucidated. This study aims to investigate the efficacy of CTS on the BPH-associated pathological processes and the possible mechanisms underlying it. Herein, CTS was intragastrically administrated to estradiol/testosterone (E2/T) (1:100)-induced BPH rats, and finasteride (Fi) was used as the positive control. Human benign prostatic hyperplasia epithelial cells (BPH-1) and normal human prostate stromal cells (WPMY-1) were used for the in vitro experiments. Results indicated that E2/T injection was able to induce BPH manifestation, featured with increased prostate index. Furthermore, it accelerated proliferation, epithelial-mesenchymal transition (EMT), stromal collagen deposition, and inhibited apoptosis of rat prostate. However, the administration of CTS partially reversed the changes mentioned above. The therapeutic effects of CTS on BPH were also confirmed by in vitro experiments. The efficacy of CTS on these processes might be attributed to the suppression of AR and EGFR/STAT3 axis activity. In conclusion, CTS might suppress BPH progression by modulating proliferation, apoptosis, EMT, and stromal collagen deposition via suppressing AR and EGFR/STAT3 axis.


Subject(s)
Prostatic Hyperplasia , Male , Rats , Humans , Animals , Prostatic Hyperplasia/chemically induced , Quality of Life , Apoptosis , Fibrosis , Cell Proliferation , Collagen/adverse effects , ErbB Receptors , STAT3 Transcription Factor/pharmacology
18.
Burns ; 49(4): 889-900, 2023 06.
Article in English | MEDLINE | ID: mdl-35850880

ABSTRACT

OBJECTIVE: Curcumin, a natural extract from the rhizomes of Curcuma longa, is also known as a curcuminoid. Curcumin has been studied as a therapeutic drug for wound healing because of its anti-inflammatory, anti-oxidant, and anti-bacterial activities. However, the detailed mechanism of curcumin in wound healing is not clear. It is well-known that the skin is the largest organ in humans and prevents tissues from damage, including infection, radiation, and mechanical damage. Wound healing of the skin is a complex physiological regulation process requiring various cell types and cytokines; hence, wound healing, including surgery and care, incurs a huge expenditure each year. Transient receptor potential cation channel subfamily M member 7 (TRPM7) regulates multiple physiological and pharmacological processes through its channel and kinase activities. In addition, TRPM7 regulates cell adhesion, migration, and anti-oxidative activity, thereby playing a regulatory role in the wound healing process. This study aimed to explore the function of curcumin in the wound healing process. METHODS: We first established TRPM7 overexpression and knockdown models in fibroblasts using lentivirus. CCK-8 and wound healing assays were used to clarify whether overexpression of TRPM7 promoted proliferation and migration in fibroblasts. Expression of target genes and proteins was detected using qPCR and western blotting. Concentrations of migration-related cytokines were measured using ELISA. RESULTS: Proliferation and migration of fibroblasts increased after curcumin treatment and was further enhanced after overexpression of TRPM7. In addition, expression of proliferation-related genes and proteins was elevated after TRPM7 overexpression. Further, the secretion of migration-related cytokines was elevated after TRPM7 overexpression. CONCLUSION: Curcumin treatment promoted proliferation and migration of fibroblasts, and these effects were mediated by the signal transducer and activator of transcription 3 (STAT3)/SMAD family member 3/hypoxia-inducible factor 1 subunit alpha signaling pathway. Thus, we conclude that overexpression of TRPM7 might contribute to wound healing.


Subject(s)
Burns , Curcumin , TRPM Cation Channels , Humans , Burns/metabolism , Curcumin/metabolism , Curcumin/pharmacology , Cytokines/metabolism , Fibroblasts/metabolism , Protein Serine-Threonine Kinases , Signal Transduction , Smad3 Protein/metabolism , Smad3 Protein/pharmacology , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology , TRPM Cation Channels/metabolism , Wound Healing
19.
J Thromb Thrombolysis ; 55(1): 116-125, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36396837

ABSTRACT

The morbidity and mortality rates of cardiovascular disease are markedly higher in patients with diabetes than in non-diabetic patients, including patients with ischemia-reperfusion injury (IRI). However, the cardiovascular protective effects of Empagliflozin (EMPA) on IRI in diabetes mellitus have rarely been studied. In this study, we established a cardiomyocyte hypoxia/reoxygenation (H/R) injury model to mimic myocardial I/R injuries that occur in vivo. H9C2 cells were subjected to high glucose (HG) treatment plus H/R injury to mimic myocardial I/R injuries that occur in diabetes mellitus. Next, different concentrations of EMPA were added to the H9C2 cells and its protective effect was detected. STAT3 knockdown with recombinant plasmids was used to determine its roles. Our results showed that H/R injury-induced cell apoptosis, necroptosis, oxidative stress, and endoplasmic reticulum stress were further promoted by HG conditions, and HG treatment plus an H/R injury inhibited the activation of JAK2/STAT3 signaling. EMPA was found to protect against H/R-induced cardiomyocyte injury under HG conditions and activate JAK2/STAT3 signaling, while down-regulation of STAT3 reversed the protective effect of EMPA. When taken together, these findings indicate that EMPA protects against I/R-induced cardiomyocyte injury by activating JAK2/STAT3 signaling under HG conditions. Our results clarified the mechanisms that underlie the cardiovascular protective effects of EMPA in diabetes mellitus and provide new therapeutic targets for IRI in diabetes mellitus.


Subject(s)
Hypoxia , Myocytes, Cardiac , Humans , Myocytes, Cardiac/metabolism , Cell Line , Apoptosis , Glucose/pharmacology , Janus Kinase 2 , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
20.
Chemosphere ; 312(Pt 1): 136545, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36155021

ABSTRACT

OBJECTIVE: It has been suggested that Bisphenol A (BPA), a high-production-volume industrial chemical, can accelerate the development of various type of cancers. However, the effect of BPA on osteosarcoma and the underlying mechanisms are yet to be established. Therefore, in this study we tried to explore the carcinogenic effects of BPA on osteosarcoma and the underlying mechanism. METHODS: SaOs-2 cancer cell line was used to treat with BPA at the doses of 0.1, 1, 10 µM DGLAP5 knockdown and overexpression methods were constructed by using adenovirus mediated transfection, and the functional analysis of DGLAP5 was investigated to evaluate the carcinogenic effect of BPA on osteosarcoma through DLGAP5. Xenograft and metastatic mouse model were used to evaluate in vivo experiments. RESULTS: In this study, BPA at 10 µM promoted the proliferation, migration and invasion in vitro, and accelerate the progression and metastasis in vivo. Also, exposure to BPA was associated with poor survival of osteosarcoma in mice. In addition, we observed that BPA at 10 µM significantly increased the expression of DGLAP5 in osteosarcoma. Silencing DGLAP5 could reverse the effect of BPA on proliferation, migration and invasion. Mechanically, BPA promoted IL-6, JAK2, and STAT3 expression and promoted tumor progression in an IL-6-dependent manner through up-regulation of DLGAP5. CONCLUSION: Our findings demonstrated that BPA could promote the proliferation, migration, invasion of osteosarcoma cells and related to poor survival in a mouse model. DLGAP5 is one of the most critical targets of BPA to act as a carcinogen through IL-6/JAK2/STAT3 signaling pathway.


Subject(s)
Bone Neoplasms , Osteosarcoma , Humans , Mice , Animals , Interleukin-6/genetics , Interleukin-6/metabolism , Cell Movement , Osteosarcoma/chemically induced , Osteosarcoma/genetics , Signal Transduction , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Bone Neoplasms/chemically induced , Bone Neoplasms/genetics , Cell Proliferation , Cell Line, Tumor , Neoplasm Proteins/metabolism , Neoplasm Proteins/pharmacology , Neoplasm Proteins/therapeutic use , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Janus Kinase 2/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...