Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
BMC Med ; 21(1): 253, 2023 07 13.
Article in English | MEDLINE | ID: mdl-37442994

ABSTRACT

BACKGROUND: PARP inhibitor (PARPi), as a kind of DNA damage repair inhibitor, has been shown to be effective in various solid tumors and hematologic malignancies. Natural killer/T cell lymphoma (NKTCL) is a highly aggressive malignancy, the treatment of which has long been a major challenge in the clinic. Here, we investigated the efficacy and mechanism of PARPi, and the therapeutic value of PARPi combined with cisplatin in NKTCL. METHODS: The cell proliferation, cell apoptosis, and cell cycle of NKTCL cells were detected respectively by CCK-8 and flow cytometry. The changes of mRNA expression and protein level were measured respectively by mRNA-sequencing, quantitative real-time PCR, western blotting, and immunofluorescence. LMO2 expression was detected by immunohistochemistry and western blotting. Targeted knockdown of LMO2 was conducted by short hairpin RNA. The tumor xenograft models were established to evaluate the efficacy of drugs in vivo. RESULTS: PARPi inhibited cell proliferation, promoted cell apoptosis, and induced S-phase cell cycle arrest in NKTCL cells. PARPi led to the accumulation of DNA damage by blocking DNA repair and DNA replication. Additionally, LMO2 deficiency reduced the sensitivity of NKTCL cells to PARPi. Finally, the combination of PARPi and cisplatin exhibited significant synergistic effects both in vitro and in vivo. CONCLUSIONS: In summary, we found that PARPi exerted an anti-tumor effect via LMO2 and synergized with cisplatin in NKTCL, which provides the theoretical basis for the clinical application of PARPi.


Subject(s)
Antineoplastic Agents , Lymphoma, T-Cell , Lymphoma , Humans , Cisplatin/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Cell Line, Tumor , Antineoplastic Agents/pharmacology , Killer Cells, Natural , RNA, Messenger , Proto-Oncogene Proteins/pharmacology , Adaptor Proteins, Signal Transducing/pharmacology , LIM Domain Proteins/pharmacology
2.
Cell Biochem Funct ; 41(5): 542-552, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37170668

ABSTRACT

PDZ-LIM domain-containing Protein 2 (PDLIM2) has been reported to be downregulated in ovarian cancer. However, its exact function and mechanism in regulating ovarian cancer progression have not been elucidated. This work researched the exert effect and mechanism of PDLIM2 on ovarian cancer progression. Briefly, PDLIM2 expression in clinical tissues of ovarian cancer patients and cells was investigated by qRT-PCR and Western blot. The function of PDLIM2 on the proliferation, colony formation, migration and invasion of ovarian cancer cells was explored via cell counting kit-8, colony formation and Transwell assays. To verify whether PDLIM2 regulates ovarian cancer progression via regulating the transforming growth factor-ß (TGF-ß)/Smad pathway, exogenous TGF-ß (10 ng/mL) treatment was performed on the PDLIM2-overexpressed ovarian cancer cells. PDLIM2 effect on the in vivo growth of ovarian cancer cells was researched by establishing a xenograft tumor model. Immunohistochemistry and Western blot were performed to protein expression in cells and tissues. As a result, PDLIM2 was low-expressed in ovarian cancer tissues/cells. PDLIM2 upregulation attenuated the proliferation, colony formation, migration, invasion and epithelial-mesenchymal transition (EMT) of ovarian cancer cells, and inactivated the TGF-ß/Smad pathway. The opposite results were found in the PDLIM2-silenced ovarian cancer cells. Exogenous TGF-ß treatment abrogated the inhibition of PDLIM2 on the malignant behavior of ovarian cancer cells. PDLIM2 upregulation attenuated the in vivo growth and EMT of ovarian cancer cells. Thus, PDLIM2 attenuates the proliferation, migration, invasion and EMT of ovarian cancer cells via inactivating the TGF-ß/Smad pathway. PDLIM2 may be a usefully target for ovarian cancer treatment.


Subject(s)
Ovarian Neoplasms , Transforming Growth Factor beta , Humans , Female , Transforming Growth Factor beta/metabolism , Cell Movement , Smad Proteins/metabolism , Cell Line, Tumor , Signal Transduction , Epithelial-Mesenchymal Transition , Transforming Growth Factor beta1/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microfilament Proteins/pharmacology , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/pharmacology
3.
Reprod Biol Endocrinol ; 20(1): 64, 2022 Apr 04.
Article in English | MEDLINE | ID: mdl-35379225

ABSTRACT

BACKGROUND: Our previous two-dimensional electrophoresis experiment showed that the expression of LASP1 in patients with endometriosis was significantly higher than that of control endometrium. However, the molecular mechanism by which LASP1 is regulated in endometriosis/adenomyosis is unknown. METHODS: Herein, qPCR was performed to analyze the expression levels of LASP1 and miR-218-5p between endometriosis (Ems) cells and control cells. Fluorescence in situ hybridization was carried out to measure the expression level of miR-218-5p in ectopic endometrium versus normal endometrium. After miR-218-5p mimic or inhibitor were transfected, the transwell experiment was carried out to see the effect of miR-218-5p on the migration of endometrial stromal cells (ESCs). EdU was used to measure cell proliferation rate. Dual-luciferase reporter assay was used to verify the binding of hsa-miR-218-5p to the 3'UTR of LASP1. Western blot and immunofluorescence analysis were carried out to identify the protein expression pattern of LASP1 and EMT markers in endometrial tissue. RESULTS: The miR-218-5p is mainly secreted from blood vessels and expressed in the muscle layer around the endometrium, which inhibits the expression level of LASP1 by binding the 3'UTR region of LASP1 in normal ESCs. Overexpression of miR-218-5p impedes the epithelial-to-mesenchymal transition (EMT) and prevents the migration of ESCs and the expression of Vimentin in Ems. CONCLUSIONS: Our findings revealed that miR-218-5p in endometrial microenvironment prevents the migration of ectopic endometrial stromal cells by inhibiting LASP1.


Subject(s)
MicroRNAs , Adaptor Proteins, Signal Transducing/genetics , Cell Movement/genetics , Cytoskeletal Proteins/metabolism , Cytoskeletal Proteins/pharmacology , Endometrium/metabolism , Female , Humans , In Situ Hybridization, Fluorescence , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/pharmacology , MicroRNAs/genetics , MicroRNAs/metabolism , Stromal Cells/metabolism
4.
Neurobiol Dis ; 155: 105397, 2021 07.
Article in English | MEDLINE | ID: mdl-34015491

ABSTRACT

Subanesthetic doses of ketamine induce schizophrenia-like behaviors in mice including hyperlocomotion and deficits in working memory and sensorimotor gating. Here, we examined the effect of in vivo ketamine administration on neuronal properties and endocannabinoid (eCB)-dependent modulation of synaptic transmission onto layer 2/3 pyramidal neurons in brain slices of the prefrontal cortex, a region tied to the schizophrenia-like behavioral phenotypes of ketamine. Since deficits in working memory and sensorimotor gating are tied to activation of the tyrosine phosphatase PTP1B in glutamatergic neurons, we asked whether PTP1B contributes to these effects of ketamine. Ketamine increased membrane resistance and excitability of pyramidal neurons. Systemic pharmacological inhibition of PTP1B by Trodusquemine restored these neuronal properties and prevented each of the three main ketamine-induced behavior deficits. Ketamine also reduced mobilization of eCB by pyramidal neurons, while unexpectedly reducing their inhibitory inputs, and these effects of ketamine were blocked or occluded by PTP1B ablation in glutamatergic neurons. While ablation of PTP1B in glutamatergic neurons prevented ketamine-induced deficits in memory and sensorimotor gating, it failed to prevent hyperlocomotion (a psychosis-like phenotype). Taken together, these results suggest that PTP1B in glutamatergic neurons mediates ketamine-induced deficits in eCB mobilization, memory and sensorimotor gating whereas PTP1B in other cell types contributes to hyperlocomotion. Our study suggests that the PTP1B inhibitor Trodusquemine may represent a new class of fast-acting antipsychotic drugs to treat schizophrenia-like symptoms.


Subject(s)
Cholestanes/pharmacology , Ketamine/toxicity , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Schizophrenia/chemically induced , Schizophrenia/prevention & control , Spermine/analogs & derivatives , Adaptor Proteins, Signal Transducing/pharmacology , Anesthetics, Dissociative/toxicity , Animals , Cholestanes/therapeutic use , Dose-Response Relationship, Drug , LIM Domain Proteins/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Schizophrenia/metabolism , Spermine/pharmacology , Spermine/therapeutic use
5.
Biochem Biophys Res Commun ; 449(4): 386-91, 2014 Jul 11.
Article in English | MEDLINE | ID: mdl-24866244

ABSTRACT

HCA587, also known as MAGE-C2, belonging to the MAGE gene family which is characterized by a conserved MAGE Homology Domain, is active in various types of tumors and silent in normal tissues except in male germ-line cells. The biological function of HCA587 is largely unknown. To analyze it, we attempted to identify protein partners of HCA587. We immunopurified HCA587-containing complex from HEK293 cells and identified BS69, a potential tumor suppressor, as an associated protein by mass spectrometry, and the following Immunoprecipitation and GST pull-down assays confirmed HCA587 interaction with BS69. Interestingly, overexpression of HCA587 promoted ubiquitination and the proteasomal degradation of BS69 whereas knockdown of endogenous HCA587 increased the protein level of BS69. Consistent with a functional role for BS69 in negatively regulating LMP1-induced NF-κB activation, overexpression of HCA587 resulted in a significant enhancement of LMP1-induced IL-6 production. These data indicate that HCA587 is a new negative regulator of BS69.


Subject(s)
Antigens, Neoplasm/metabolism , Carrier Proteins/metabolism , Neoplasm Proteins/metabolism , Proteasome Endopeptidase Complex/metabolism , Adaptor Proteins, Signal Transducing/pharmacology , Antigens, Neoplasm/biosynthesis , Antigens, Neoplasm/isolation & purification , Carrier Proteins/biosynthesis , Cell Cycle Proteins , Co-Repressor Proteins , Cytoskeletal Proteins/pharmacology , DNA-Binding Proteins , HEK293 Cells , HeLa Cells , Humans , Interleukin-6/biosynthesis , LIM Domain Proteins/pharmacology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/isolation & purification , Ubiquitination
6.
Arthritis Rheumatol ; 75(3): 438-448, 2023 03.
Article in English | MEDLINE | ID: mdl-36103378

ABSTRACT

OBJECTIVE: Interstitial lung disease (ILD) is a serious complication and leading cause of mortality in patients with systemic sclerosis (SSc). In this study, we explored the role of LIM and cysteine-rich domains protein 1 (LMCD1) as a novel factor in the pathogenesis of SSc-related ILD (SSc-ILD). METHODS: The expression and effects of LMCD1 were studied in lung tissue samples and fibroblasts from SSc-ILD patients and control subjects as well as in lung tissue samples from animal models. RESULTS: LMCD1 was consistently elevated in lung tissue samples and in fibroblasts isolated from SSc-ILD patients as compared to controls. Additionally, LMCD1 was found to be highly expressed in the lung in the fibroblast-specific protein (FSP)-driven, constitutively active transforming growth factor ß receptor type I (TGFßR1) transgenic mouse model of ILD and the bleomycin-induced mouse model of ILD. In lung fibroblasts from SSc-ILD patients, LMCD1 is an essential factor for the TGFß-induced generation of type I collagen, fibronectin, and α-smooth muscle actin (α-SMA). Depletion of LMCD1 by small interfering RNA reduced the expression of extracellular matrix proteins and lowered transcriptional activity and expression of α-SMA, as well as decreased the proliferation and contractile activity of SSc-ILD lung fibroblasts. In dense fibrotic areas of affected lung tissue, lung LMCD1 colocalized with α-SMA. In cultured scleroderma lung fibroblasts, LMCD1 colocalized and interacted with serum response factor which mediates LMCD1-induced contractile activity of lung fibroblasts. CONCLUSION: Our study identifies LMCD1 as a profibrotic molecule contributing to the activation of myofibroblasts and the persistent fibroproliferation observed in SSc-ILD. Thus, LMCD1 may be a potential novel therapeutic target for patients with SSc-ILD.


Subject(s)
Lung Diseases, Interstitial , Pulmonary Fibrosis , Scleroderma, Localized , Scleroderma, Systemic , Animals , Mice , Pulmonary Fibrosis/complications , Myofibroblasts/metabolism , Scleroderma, Systemic/pathology , Lung Diseases, Interstitial/etiology , Lung/pathology , Fibroblasts/metabolism , Scleroderma, Localized/complications , Co-Repressor Proteins/metabolism , Co-Repressor Proteins/pharmacology , LIM Domain Proteins/metabolism , LIM Domain Proteins/pharmacology
7.
Eur J Pharm Sci ; 182: 106372, 2023 Mar 01.
Article in English | MEDLINE | ID: mdl-36621614

ABSTRACT

Epidermal growth factor receptor tyrosine kinase inhibitor (EGFR TKIs) was one of the main drugs in the treatment of non-small cell lung cancer (NSCLC). Previous studies had demonstrated that PDZ and LIM Domain 5 (PDLIM5) played an important role in EGFR TKIs resistance. However, there was no feasible method to eliminate EGFR TKIs resistance by suppressing this gene. Here, we formulated a novel mesoporous silica-loaded PDLIM5 siRNA (Small interfering RNA) nanoplatforms. The results have shown that PDLIM5 siRNA could be effectively bound to the nanoplatforms and had good biocompatibility. Further exploration suggested that the nano-platform combined with ultrasonic irradiation could be very effective for siRNA delivery and ultrasound imaging. Moreover, Epithelial-mesenchymal transformation (EMT) changes occurred in PC-9 Gefitinib resistance (PC-9/GR) cells during the development of drug resistance. When PDLIM5 siRNA entered PC-9/GR cells, the sensitivity of drug-resistant cells to gefitinib could be restored through the transforming growth factor-ß (TGF-ß)/EMT pathway. Therefore, PDLIM5 may be an important reason for the resistance of NSCLC cells to gefitinib, and this nanoplatform may become a novel treatment for EGFR TKIs resistance in NSCLC patients.


Subject(s)
Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/metabolism , Gefitinib/pharmacology , Gefitinib/therapeutic use , RNA, Small Interfering/genetics , Lung Neoplasms/metabolism , Epithelial-Mesenchymal Transition , ErbB Receptors , Quinazolines , Drug Resistance, Neoplasm , Ultrasonography , Cell Line, Tumor , Protein Kinase Inhibitors/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Adaptor Proteins, Signal Transducing/metabolism , Adaptor Proteins, Signal Transducing/pharmacology , Adaptor Proteins, Signal Transducing/therapeutic use , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , LIM Domain Proteins/pharmacology
8.
Psychoneuroendocrinology ; 115: 104629, 2020 05.
Article in English | MEDLINE | ID: mdl-32171900

ABSTRACT

OBJECTIVE: Prenatal stress (PS) contributes to depression-like behavior in the offspring. PDLIM5 is involved in the onset of mental disorders. This study is to investigate the role and mechanism of PDLIM5 in depression-like behavior of PS offspring rats. METHODS: PS model was used to analyze the effects of different treatments to PS offspring rats with different sex, including PDLIM5, PDLIM5 shRNA and 5-aza-2' -deoxycytidine (5-azaD). The depression-like behavior was assessed by the sucrose preference test (SPT) and forced swimming test (FST). The mRNA and protein expression levels of PDLIM5 in the hippocampus of PS offspring rats were detected by qRT-PCR and western blot, respectively. The methylation of PDLIM5 promoter were analyzed by bisulfite sequencing. RESULTS: Our data revealed that PS offspring rats showed a significant decrease in sucrose preference and a prolonged immobility time. Injection of PDLIM5 significantly improved the depression-like behavior in PS offspring rats, whereas administration of PDLIM5 shRNA aggravated it. In addition, PDLIM5 expression was decreased at the mRNA and protein levels, and the methylation level of PDLIM5 promoter was increased in hippocampus of PS male but not female offspring rats. Furthermore, microinjection of 5-azaD improved the PS induced depression-like behavior in offspring rats. Moreover, in male PS offspring rats, microinjection of 5-azaD reversed the effect of PS on PDLIM5 expression and promoter methylation. CONCLUSION: PDLIM5 can significantly improve the depression-like behavior of both male and female PS offspring rats, while the PDLIM5 promoter methylation is only observed in male PS offspring rats. Our study may provide new mechanism for the pathogenesis of depression and experimental evidence for sex-based precise treatment.


Subject(s)
Adaptor Proteins, Signal Transducing/pharmacology , Behavior, Animal/drug effects , Depression/drug therapy , Hippocampus/drug effects , LIM Domain Proteins/pharmacology , Pregnancy Complications , Prenatal Exposure Delayed Effects/drug therapy , Sex Characteristics , Stress, Psychological , Adaptor Proteins, Signal Transducing/administration & dosage , Animals , DNA Methylation/genetics , Disease Models, Animal , Epigenesis, Genetic/genetics , Female , LIM Domain Proteins/administration & dosage , Male , Pregnancy , Promoter Regions, Genetic/genetics , RNA, Small Interfering , Rats , Rats, Sprague-Dawley , Recombinant Proteins
9.
PLoS One ; 10(4): e0122773, 2015.
Article in English | MEDLINE | ID: mdl-25835392

ABSTRACT

Hydrogen peroxide-inducible clone-5 (Hic-5) is a transforming growth factor (TGF)-ß1-inducible focal adhesion protein. We previously demonstrated that Hic-5 was localized in mesangial cells and its expression was associated with glomerular cell proliferation and matrix expansion in human and rat glomerulonephritis (GN). In the present study, we first assessed the role of Hic-5 in mesangioproliferative GN by injecting Habu venom into heminephrectomized wild type (Hic-5+/+) and Hic-5-deficient (Hic-5-/-) mice. Hic-5+/+ GN mice exhibited glomerular cell proliferation on day 7. Surprisingly, glomerular cell number and Ki-67-positive cells in Hic-5-/- GN mice were significantly greater than those in Hic-5+/+ GN mice on day 7, although the number of glomerular apoptotic cells and the expression of growth factors (platelet-derived growth factor-BB and TGF-ß1) and their receptors were similarly increased in both Hic-5+/+ and Hic-5-/- GN mice. In culture experiments, proliferation assays showed that platelet-derived growth factor-BB and TGF-ß1 enhanced the proliferation of Hic-5-/- mesangial cells compared with Hic-5+/+ mesangial cells. In addition, mitogenic regulation by Hic-5 was associated with altered and coordinated expression of cell cycle-related proteins including cyclin D1 and p21. The present results suggest that Hic-5 might regulate mesangial cell proliferation in proliferative GN in mice. In conclusion, modulation of Hic-5 expression might have a potential to prevent mesangial cell proliferation in the acute mitogenic phase of glomerulonephritis.


Subject(s)
Cell Proliferation/drug effects , Cytoskeletal Proteins/metabolism , DNA-Binding Proteins/metabolism , Glomerular Mesangium/metabolism , Glomerulonephritis/genetics , LIM Domain Proteins/metabolism , Mesangial Cells/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Becaplermin , Crotalid Venoms/toxicity , Cyclin D1/genetics , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cytoskeletal Proteins/genetics , Cytoskeletal Proteins/pharmacology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/pharmacology , Female , Gene Expression Regulation , Glomerular Mesangium/drug effects , Glomerular Mesangium/pathology , Glomerulonephritis/chemically induced , Glomerulonephritis/metabolism , Glomerulonephritis/pathology , Humans , Ki-67 Antigen/genetics , Ki-67 Antigen/metabolism , LIM Domain Proteins/genetics , LIM Domain Proteins/pharmacology , Male , Mesangial Cells/drug effects , Mesangial Cells/pathology , Mice , Mice, Knockout , Nephrectomy , Primary Cell Culture , Proto-Oncogene Proteins c-sis/genetics , Proto-Oncogene Proteins c-sis/metabolism , Proto-Oncogene Proteins c-sis/pharmacology , Signal Transduction , Transforming Growth Factor beta1/genetics , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/pharmacology , Trimeresurus/metabolism
10.
Shock ; 39(2): 189-96, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23324889

ABSTRACT

Acute lung injury (ALI) is a clinical syndrome characterized by hypoxia, which is caused by the breakdown of the alveolar capillary barrier. Interleukin 1ß (IL-1ß), a cytokine released within the airspace in ALI, downregulates the α subunit of the epithelial sodium channel (αENaC) transcription and protein expression via p38 MAP kinase-dependent signaling. Although induction of the heat shock response can restore alveolar fluid clearance compromised by IL-1ß following the onset of severe hemorrhagic shock in rats, the mechanisms are not fully understood. In this study, we report that the induction of the heat shock response prevents IL-1ß-dependent inhibition of αENaC mRNA expression and subsequent channel function. Heat shock results in IRAK1 detergent insolubility and a disruption of Hsp90 binding to IRAK1. Likewise, TAK1, another client protein of Hsp90 and signaling component of the IL-1ß pathway, is also detergent insoluble after heat shock. Twenty-four hours after heat shock, both IRAK1 and TAK1 are again detergent soluble, which correlates with the IL-1ß-dependent p38 activation. Remarkably, IL-1ß-dependent p38 activation 24 h after heat shock did not result in an inhibition of αENaC mRNA expression and channel function. Further analysis demonstrates prolonged preservation of αENaC expression by the activation of the heat shock response that involves inducible Hsp70. Inhibition of Hsp70 at 24 h after heat shock results in p38-dependent IL-1ß inhibition of αENaC mRNA expression, whereas overexpression of Hsp70 attenuates the p38-dependent IL-1ß inhibition of αENaC mRNA expression. These studies demonstrate new mechanisms by which the induction of the heat shock response protects the barrier function of the alveolar epithelium in ALI.


Subject(s)
Acute Lung Injury/prevention & control , Amiloride/pharmacology , Epithelial Sodium Channel Blockers/pharmacology , Heat-Shock Response/physiology , Interleukin-1beta/physiology , Pulmonary Alveoli/metabolism , Animals , Benzoquinones/pharmacology , Cytoskeletal Proteins/pharmacology , DNA-Binding Proteins/pharmacology , Epithelial Sodium Channels/drug effects , HSP70 Heat-Shock Proteins/metabolism , Interleukin-1 Receptor-Associated Kinases/metabolism , LIM Domain Proteins/pharmacology , Lactams, Macrocyclic/pharmacology , MAP Kinase Kinase Kinases/metabolism , MAP Kinase Signaling System/physiology , Male , RNA, Messenger/metabolism , Rats , Respiratory Mucosa/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL