Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Cell Mol Med ; 28(4): e18113, 2024 02.
Artigo em Inglês | MEDLINE | ID: mdl-38332530

RESUMO

The resistance to anoikis plays a critical role in the metastatic progression of various types of malignancies, including gastric cancer (GC). Nevertheless, the precise mechanism behind anoikis resistance is not fully understood. Here, our primary focus was to examine the function and underlying molecular mechanism of Integrin beta-like 1 (ITGBL1) in the modulation of anoikis resistance and metastasis in GC. The findings of our investigation have demonstrated that the overexpression of ITGBL1 significantly augmented the resistance of GC cells to anoikis and promoted their metastatic potential, while knockdown of ITGBL1 had a suppressive effect on both cellular processes in vitro and in vivo. Mechanistically, we proved that ITGBL1 has a role in enhancing the resistance of GC cells to anoikis and promoting metastasis through the AKT/Fibulin-2 (FBLN2) axis. The inhibition of AKT/FBLN2 signalling was able to reverse the impact of ITGBL1 on the resistance of GC cells to anoikis and their metastatic capability. Moreover, the expression levels of ITGBL1 were found to be significantly elevated in the cancerous tissues of patients diagnosed with GC, and there was a strong correlation observed between high expression levels of ITGBL1 and worse prognosis among individuals diagnosed with GC. Significantly, it was revealed that within our cohort of GC patients, individuals exhibiting elevated ITGBL1 expression and diminished FBLN2 expression experienced the worst prognosis. In conclusion, the findings of our study indicate that ITGBL1 may serve as a possible modulator of resistance to anoikis and the metastatic process in GC.


Assuntos
Anoikis , Proteínas de Ligação ao Cálcio , Neoplasias Gástricas , Humanos , Anoikis/genética , Neoplasias Gástricas/patologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas da Matriz Extracelular , Linhagem Celular Tumoral , Metástase Neoplásica , Integrina beta1/genética
2.
Cancer Sci ; 114(11): 4225-4236, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37661645

RESUMO

Ferroptosis, a newly discovered form of regulated cell death, has been reported to be associated with multiple cancers, including colorectal cancer (CRC). However, the underlying molecular mechanism is still unclear. In this study, we identified B7H3 as a potential regulator of ferroptosis resistance in CRC. B7H3 knockdown decreased but B7H3 overexpression increased the ferroptosis resistance of CRC cells, as evidenced by the expression of ferroptosis-associated genes (PTGS2, FTL, FTH, and GPX4) and the levels of important indicators of ferroptosis (malondialdehyde, iron load). Moreover, B7H3 promoted ferroptosis resistance by regulating sterol regulatory element binding protein 2 (SREBP2)-mediated cholesterol metabolism. Both exogenous cholesterol supplementation and treatment with the SREBP2 inhibitor betulin reversed the effect of B7H3 on ferroptosis in CRC cells. Furthermore, we verified that B7H3 downregulated SREBP2 expression by activating the AKT pathway. Additionally, multiplex immunohistochemistry was carried out to show the expression of B7H3, prostaglandin-endoperoxide synthase 2, and SREBP2 in CRC tumor tissues, which was associated with the prognosis of patients with CRC. In summary, our findings reveal a role for B7H3 in regulating ferroptosis by controlling cholesterol metabolism in CRC.


Assuntos
Neoplasias Colorretais , Ferroptose , Humanos , Colesterol/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Ciclo-Oxigenase 2 , Ferroptose/genética , Ferro/metabolismo
3.
J Transl Med ; 21(1): 163, 2023 03 02.
Artigo em Inglês | MEDLINE | ID: mdl-36864416

RESUMO

BACKGROUND: Gastric cancer (GC) is a major cancer burden throughout the world with a high mortality rate. The performance of current predictive and prognostic factors is still limited. Integrated analysis is required for accurate cancer progression predictive biomarker and prognostic biomarkers that help to guide therapy. METHODS: An AI-assisted bioinformatics method that combines transcriptomic data and microRNA regulations were used to identify a key miRNA-mediated network module in GC progression. To reveal the module's function, we performed the gene expression analysis in 20 clinical samples by qRT-PCR, prognosis analysis by multi-variable Cox regression model, progression prediction by support vector machine, and in vitro studies to elaborate the roles in GC cells migration and invasion. RESULTS: A robust microRNA regulated network module was identified to characterize GC progression, which consisted of seven miR-200/183 family members, five mRNAs and two long non-coding RNAs H19 and CLLU1. Their expression patterns and expression correlation patterns were consistent in public dataset and our cohort. Our findings suggest a two-fold biological potential of the module: GC patients with high-risk score exhibited a poor prognosis (p-value < 0.05) and the model achieved AUCs of 0.90 to predict GC progression in our cohort. In vitro cellular analyses shown that the module could influence the invasion and migration of GC cells. CONCLUSIONS: Our strategy which combines AI-assisted bioinformatics method with experimental and clinical validation suggested that the miR-200/183 family-mediated network module as a "pluripotent module", which could be potential marker for GC progression.


Assuntos
MicroRNAs , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/genética , MicroRNAs/genética , Biomarcadores Tumorais/genética , Biologia Computacional , Inteligência Artificial
4.
Cytokine ; 162: 156108, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36527892

RESUMO

Immunotherapy has been a promising, emerging treatment for various cancers. Gamma delta (γδ) T cells own a T cell receptor composed of γ- and δ- chain and act as crucial players in the anti-tumor immune effect. Currently, Vγ9Vδ2 T cells, the predominate γδ T cell subset in human peripheral blood, has been shown to exert multiple biological functions. In addition, a growing body of evidence notes that Vγ9Vδ2 T cells interact with tumor cells in many ways, such as TCR-mediated nonpeptidic-phosphorylated phosphoantigens (pAgs) recognization, NKG2D/NKG2D ligand (NKG2DL) pathway, Fas-FasL axis and antibody-dependent cellular cytotoxicity (ADCC) as well as exosome. More importantly, clinical studies with Vγ9Vδ2 T cells in cancers have propelled several clinical applications to investigate their safety and efficacy. Herein, this review summarized the underlying ways and mechanisms of interplay cancer cells and Vγ9Vδ2 T cells, which may help us to generate new strategies for tumor immunotherapy in the future.


Assuntos
Neoplasias , Receptores de Antígenos de Linfócitos T gama-delta , Humanos , Subfamília K de Receptores Semelhantes a Lectina de Células NK , Ativação Linfocitária , Neoplasias/terapia , Neoplasias/metabolismo , Imunoterapia , Subpopulações de Linfócitos T
5.
Cell Mol Life Sci ; 79(11): 563, 2022 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-36272033

RESUMO

Emerging evidence suggests that ferroptosis is involved in the pathogenesis of ulcerative colitis (UC). However, the key regulator of this process remains uncertain. In this study, we aimed to explore the roles of solute carrier (SLC) family 6 member 14 (SLC6A14) in regulating ferroptosis in UC. The expression of SLC6A14 was significantly increased and positively associated with that of prostaglandin-endoperoxide synthase 2 (PTGS2) in tissue samples from patients with UC. Moreover, a series of in vitro and in vivo experiments showed that SLC6A14 knockdown markedly suppressed ferroptosis. RNA sequencing revealed that SLC6A14 inhibited the expression of P21 (RAC1)-activated kinase 6 (PAK6) and that PAK6 knockdown abolished the effects of SLC6A14 on RAS-selective lethal 3 (RSL3)-induced ferroptosis in Caco-2 cells. Furthermore, chromatin immunoprecipitation (ChIP) and Western blot analysis demonstrated that SLC6A14 negatively regulated PAK6 expression in a CCAAT enhancer binding protein beta (C/EBPß)-dependent manner. Collectively, these findings indicate that SLC6A14 facilitates ferroptosis in UC by promoting C/EBPß expression and binding activity to inhibit PAK6 expression, suggesting that targeting SLC6A14-C/EBPß-PAK6 axis-mediated ferroptosis may be a promising therapeutic alternative for UC.


Assuntos
Colite Ulcerativa , Ferroptose , Humanos , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Colite Ulcerativa/genética , Ciclo-Oxigenase 2 , Células CACO-2 , Ferroptose/genética , Células Epiteliais/metabolismo , Quinases Ativadas por p21/genética , Quinases Ativadas por p21/metabolismo , Sistemas de Transporte de Aminoácidos
6.
Cancer Immunol Immunother ; 71(2): 311-325, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34159436

RESUMO

Recent studies have shown that tumor-derived exosomes participate in the communication between tumor cells and their microenvironment and mediate malignant biological behaviors including immune escape. In this study, we found that gastric cancer (GC) cell-derived exosomes could be effectively uptaken by Vγ9Vδ2 T cells, decrease the cell viability of Vγ9Vδ2 T cells, induce apoptosis, and reduce the production of cytotoxic cytokines IFN-γ and TNF-α. Furthermore, we demonstrated that exosomal miR-135b-5p was delivered into Vγ9Vδ2 T cells. Exosomal miR-135b-5p impaired the function of Vγ9Vδ2 T cells by targeting specificity protein 1 (SP1). More importantly, blocking the SP1 function by Plicamycin, an SP1 inhibitor, abolished the effect of stable miR-135b-5p knockdown GC cell-derived exosomes on Vγ9Vδ2 T cell function. Collectively, our results suggest that GC cell-derived exosomes impair the function of Vγ9Vδ2 T cells via miR-135b-5p/SP1 pathway, and targeting exosomal miR-135b-5p/SP1 axis may improve the efficiency of GC immunotherapy based on Vγ9Vδ2 T cells.


Assuntos
Exossomos/genética , MicroRNAs/genética , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Fator de Transcrição Sp1/antagonistas & inibidores , Neoplasias Gástricas/patologia , Linfócitos T/imunologia , Microambiente Tumoral , Apoptose , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/metabolismo , Células Tumorais Cultivadas
7.
Cancer Cell Int ; 22(1): 147, 2022 Apr 11.
Artigo em Inglês | MEDLINE | ID: mdl-35410218

RESUMO

INTRODUCTION: B7-H4 is overexpressed in colorectal cancer (CRC) and plays an important role in tumor growth and immunosuppression. However, the exact mechanism that regulates B7-H4 expression remains largely unknown. Here, we investigated whether protein kinase C δ (PKCδ) regulates the expression of B7-H4 in CRC. METHODS: By using immunohistochemical (IHC) and immunofluorescence (IF) staining, we analyzed the expression of B7-H4 and phospho-PKCδ (p-PKCδ) in 225 colorectal tumor samples and determined the clinical significance of the expression patterns. In vitro experiments were performed with the CRC cell lines HCT116 and SW620 to detect the effect of PKCδ activation on B7-H4 expression, and xenograft-bearing mice were treated with rottlerin to monitor the expression of B7-H4 and tumor metastasis. RESULTS: The B7-H4 expression level was significantly correlated with the p-PKCδ level (r = 0.378, P < 0.001) in tumor tissues. Coexpression of p-PKCδ and B7-H4 was significantly associated with moderate/poor differentiation (P = 0.024), lymph node metastasis (P = 0.001) and advanced Dukes' stage (P = 0.002). Western blot analysis showed that Phorbol-12-Myristate-13-Acetate (TPA) increased B7-H4 expression in a concentration-dependent manner and that rottlerin abrogated the TPA-induced increase in B7-H4 expression. The protein levels of B7-H4 and p-STAT3 were significantly reduced by a PKCδ-specific siRNA. Moreover, the STAT3 inhibitor cryptotanshinone significantly decreased the B7-H4 protein level in CRC cells. Knockdown of B7-H4 or PKCδ suppressed cell migration and motility. Rottlerin also inhibited B7-H4 expression and tumor metastasis in vivo. CONCLUSION: The B7-H4 expression level is significantly correlated with the p-PKCδ level and tumor metastasis in CRC samples. B7-H4 expression is upregulated by STAT3 activation via PKCδ and plays roles in PKCδ-induced cancer cell motility and metastasis, suggesting that the PKCδ/STAT3/B7-H4 axis may be a potential therapeutic target for CRC.

8.
Mol Cell Biochem ; 477(5): 1371-1380, 2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35142950

RESUMO

Increasing evidence indicates that anoikis resistance is a critical process for metastasis of cancer cells, making it the attractive therapeutic target for cancer benefit. Anoikis resistance is widely regulated by various factors, such as signaling pathways, integrins switch, and non-coding RNAs (ncRNAs). ncRNAs composed of microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), are frequently dysregulated in a variety of human malignancies and are closely related to anoikis resistance of cancer cells. Based on the available literature, we reviewed the molecular basis underlying ncRNAs modulating cancer cells anoikis resistance, which may contribute to a better understanding of cancer metastasis and provide new beneficial therapeutic strategies against cancer.


Assuntos
Neoplasias , RNA Longo não Codificante , Anoikis/genética , Humanos , Neoplasias/genética , RNA Circular/genética , RNA Longo não Codificante/genética , RNA Longo não Codificante/metabolismo , RNA não Traduzido/genética , RNA não Traduzido/metabolismo
9.
Cancer Sci ; 112(6): 2245-2259, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33756038

RESUMO

Long noncoding RNAs (lncRNAs) have been investigated in multiple human cancers including gastric cancer (GC). Our research aims to explore the role of H19 in aerobic glycolysis, proliferation, and immune escape of GC cells. The expression of H19 in GC samples was analyzed using Gene Expression Profiling Interactive Analysis, Gene Expression Omnibus data, and real-time quantitative PCR analysis. Relative quantification of glucose consumption and lactate production from cell supernatant were applied to assess the aerobic glycolysis of GC cells. Subcellular fractionation, luciferase reporter, and western blot assays certified the binding between genes. Cell Counting Kit-8 and colony formation assays were used to determine GC cell proliferation. Flow cytometry, ELISA, and real-time quantitative PCR assays were applied to analyze the immunosuppressive effect of H19. H19 was highly expressed in samples of patients with GC, and associated with tumor growth in vivo. H19 knockdown suppressed glucose consumption, lactate production, and proliferation of GC cells by regulating the microRNA (miR)-519d-3p/lactate dehydrogenase A (LDHA) axis. Both miR-519d-3p depletion and LDHA overexpression could reverse the H19 knockdown-induced decrease in aerobic glycolysis and proliferation. Moreover, conditioned medium from stable knockdown H19 GC cells modulated the activity of immune cells including γδT cells, Jurkat cells, and tumor-associated macrophages in a miR-519d-3p/LDHA/lactate axis-dependent manner. The H19/miR-519d-3p/LDHA axis mainly contributed to aerobic glycolysis, proliferation, and immune escape of GC cells.


Assuntos
Proliferação de Células , L-Lactato Desidrogenase/metabolismo , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Neoplasias Gástricas/patologia , Evasão Tumoral , Efeito Warburg em Oncologia , Animais , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Humanos , L-Lactato Desidrogenase/genética , Camundongos , MicroRNAs/genética , RNA Longo não Codificante/genética , Neoplasias Gástricas/genética , Neoplasias Gástricas/imunologia , Neoplasias Gástricas/metabolismo
10.
Cancer Immunol Immunother ; 70(5): 1213-1226, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33119798

RESUMO

Immunotherapy based on γδT cells has limited efficiency in solid tumors, including colon cancer (CC). The immune evasion of tumor cells may be the main cause of the difficulties of γδT cell-based treatment. In the present study, we explored whether and how B7-H3 regulates the resistance of CC cells to the cytotoxicity of Vγ9Vδ2 (Vδ2) T cells. We observed that B7-H3 overexpression promoted, while B7-H3 knockdown inhibited, CC cell resistance to the killing effect of Vδ2 T cells in vitro and in vivo. Mechanistically, we showed that B7-H3-mediated CC cell resistance to the cytotoxicity of Vδ2 T cells involved a molecular pathway comprising STAT3 activation and decreased ULBP2 expression. ULBP2 blockade or knockdown abolished the B7-H3 silencing-induced increase in the cytotoxicity of Vδ2 T cells to CC cells. Furthermore, cryptotanshinone, a STAT3 phosphorylation inhibitor, reversed the B7-H3 overexpression-induced decrease in ULBP2 expression and attenuated the killing effect of Vδ2 T cells on CC cells. Moreover, there was a negative correlation between the expression of B7-H3 and ULBP2 in the tumor tissues of CC patients. Our results suggest that the B7-H3-mediated STAT3/ULBP2 axis may be a potential candidate target for improving the efficiency of γδT cell-based immunotherapy in CC.


Assuntos
Antígenos B7/metabolismo , Vacinas Anticâncer/imunologia , Neoplasias do Colo/imunologia , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Fator de Transcrição STAT3/metabolismo , Linfócitos T/metabolismo , Animais , Antígenos B7/genética , Neoplasias do Colo/terapia , Citotoxicidade Imunológica , Feminino , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/metabolismo , Técnicas de Silenciamento de Genes , Células HCT116 , Xenoenxertos , Humanos , Imunoterapia Adotiva , Peptídeos e Proteínas de Sinalização Intercelular/genética , Camundongos , Camundongos SCID , Receptores de Antígenos de Linfócitos T gama-delta/genética , Receptores de Antígenos de Linfócitos T gama-delta/metabolismo , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/transplante , Evasão Tumoral
11.
FASEB J ; 34(3): 3743-3754, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31943384

RESUMO

Wear debris-induced osteolysis and ensuing aseptic loosening is the main cause of implant failure and revision surgery. Wear debris-induced inflammatory response plays key roles in peri-implant osteolysis. Recently, substantial of evidence suggests that hydrogen sulfide (H2 S), the third gasotransmitter, is a critical player regulating inflammation. However, the role and therapeutic potential of H2 S in wear debris-induced inflammation and osteolysis remains to be defined. In the present study, we investigated the effect of H2 S on wear debris-induced pro-inflammatory cytokines expression and osteolysis in vitro and in vivo. With a slow-releasing H2 S donor GYY4137, our study demonstrated that H2 S attenuated wear debris-induced osteolysis and osteoclastogenesis in murine calvaria resorption models. The expression of tumor necrosis factor-alpha (TNF-α), interleukin-1ß (IL-1ß), and interleukin-6 (IL-6) that stimulated by wear particles were significantly reduced by GYY4137. Further, the level of sirtuin 1 (SIRT1), which possesses anti-inflammation property, was examined in vivo and in macrophages. And we found that wear debris decreased the expression of SIRT1. Cotreated macrophages with GYY4137 in part reversed the decline of SIRT1. More importantly, with the SIRT1 recombinant lentivirus and small interfering RNAs (siRNA) against SIRT1, our data indicated that SIRT1 mediated the inhibitory effects of GYY4137 on wear debris-induced inflammation. Collectively, these results suggested that exogenous H2 S production (via H2 S donors) may represent a potential approach for the treatment of wear particle-induced osteolysis.


Assuntos
Sulfeto de Hidrogênio/uso terapêutico , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Osteólise/tratamento farmacológico , Osteólise/metabolismo , Sirtuína 1/metabolismo , Animais , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Camundongos , Células RAW 264.7 , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/efeitos dos fármacos , Microtomografia por Raio-X
12.
Exp Cell Res ; 386(1): 111719, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31726050

RESUMO

Gamma delta (γδ) T cell-based tumor immunotherapy has been one of the most promising cancer immunotherapeutic strategies. However, the key regulators of the Vγ9Vδ2 T cell-mediated antitumor response remain unclear. Recently, mounting reports have indicated that Tim-3 performs critical roles in the regulation of the activities of immune cells, including Vγ9Vδ2 T cells. However, the roles of Tim-3 in Vγ9Vδ2 T cell-mediated killing of colon cancer cells and the underlying mechanism remain largely unknown. Here, the proportion of Tim-3+ γδ T cells was significantly increased in both the peripheral blood and colon cancer tissue of patients and was significantly associated with TNM staging and tumor volume. Additionally, the activation of Tim-3 signaling significantly inhibited the killing efficiency of Vγ9Vδ2 T cells against colon cancer cells. In addition, Tim-3 signaling reduced the expression of perforin and granzyme B in Vγ9Vδ2 T cells. Blocking the perforin/granzyme B pathway also decreased the cytotoxicity of Vγ9Vδ2 T cells to colon cancer cells. Moreover, Tim-3 signaling reduced the perforin and granzyme B expression of Vγ9Vδ2 T cells in an ERK1/2 signaling pathway-dependent manner. This knowledge reveals that Tim-3 may be a promising therapeutic target to improve Vγ9Vδ2 T cell-based adoptive immunotherapy for colon cancer.


Assuntos
Neoplasias do Colo/imunologia , Granzimas/metabolismo , Receptor Celular 2 do Vírus da Hepatite A/metabolismo , Linfócitos Intraepiteliais/imunologia , Perforina/metabolismo , Idoso , Células Cultivadas , Neoplasias do Colo/terapia , Feminino , Granzimas/genética , Células HCT116 , Receptor Celular 2 do Vírus da Hepatite A/genética , Humanos , Imunoterapia/métodos , Sistema de Sinalização das MAP Quinases , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Perforina/genética
13.
Mol Ther ; 28(4): 1214-1228, 2020 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-32087149

RESUMO

Mesenchymal stem cells (MSCs) have shown great promise in inflammatory bowel disease (IBD) treatment, owing to their immunosuppressive capabilities, but their therapeutic effectiveness is sometimes thwarted by their low efficiency in entering the inflamed colon and variable immunomodulatory ability in vivo. Here, we demonstrated a new methodology to manipulate MSCs to express CX3C chemokine receptor 1 (CX3CR1) and interleukin-25 (IL-25) to promote their delivery to the inflamed colon and enhance their immunosuppressive capability. Compared to MSCs without treatment, MSCs infected with a lentivirus (LV) encoding CX3CR1 and IL-25 (CX3CR1&IL-25-LV-MSCs) exhibited enhanced targeting to the inflamed colon and could further move into extravascular space of the colon tissues via trans-endothelial migration in dextran sodium sulfate (DSS)-challenged mice after MSC intravenous injection. The administration of the CX3CR1&IL-25-LV-MSCs achieved a better therapeutic effect than that of the untreated MSCs, as indicated by pathological indices and inflammatory markers. Antibody-blocking studies indicated that the enhanced therapeutic effects of dual-functionalized MSCs were dependent on CX3CR1 and IL-25 function. Overall, this strategy, which is based on enhancing the homing and immunosuppressive abilities of MSCs, represents a promising therapeutic approach that may be valuable in IBD therapy.


Assuntos
Receptor 1 de Quimiocina CX3C/genética , Colite/terapia , Interleucinas/genética , Lentivirus/genética , Células-Tronco Mesenquimais/metabolismo , Animais , Receptor 1 de Quimiocina CX3C/metabolismo , Colite/induzido quimicamente , Colite/metabolismo , Modelos Animais de Doenças , Feminino , Interleucinas/metabolismo , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais/virologia , Camundongos , Ratos , Dodecilsulfato de Sódio/efeitos adversos , Resultado do Tratamento
14.
Biochem Biophys Res Commun ; 473(1): 133-139, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-26996127

RESUMO

Peri-prosthetic osteolysis and the consequent aseptic loosening constitute the most common reason for total joint arthroplasty failure and surgical revision. Although numerous studies suggest that pro-inflammatory cytokines induced by wear particles is involved in the pathological process of aseptic loosening, the underlying mechanism linking wear particles to pro-inflammatory cytokines remains to be illustrated. In the present study, we investigated the effect of autophagy on TNF-α secretion induced by TiAl6V4 particles (TiPs) in macrophages and in a calvarial resorption animal model. Our study demonstrated that TiPs activated autophage in macrophages and particle-induced osteolysis animal models as well as periprosthetic membranes of patients with aseptic loosening. The autophagy inhibitor 3-MA (3-methyladenine) could dramatically reduce TiPs-induced TNF-α expression both in macrophages and in membranes from animal models. Furthermore, inhibition of autophagy with 3-MA ameliorated the severity of osteolysis in PIO animal models. Collectively, these results suggest that autophagy plays a key role in TiPs-induced osteolysis by promoting TNF-α expression and that blocking autophagy may represent a potential therapeutic approach for treating particle-induced peri-implant osteolysis.


Assuntos
Autofagia , Macrófagos/metabolismo , Osteólise/induzido quimicamente , Titânio/química , Fator de Necrose Tumoral alfa/metabolismo , Adenina/análogos & derivados , Adenina/química , Idoso , Idoso de 80 Anos ou mais , Ligas , Animais , Modelos Animais de Doenças , Feminino , Humanos , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Falha de Prótese , Titânio/efeitos adversos , Regulação para Cima , Microtomografia por Raio-X , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Gut ; 63(8): 1247-57, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24000293

RESUMO

OBJECTIVE: Emerging evidence suggests that microRNA (miRNA)-mediated gene regulation influences a variety of autoimmune disease processes, including Crohn's disease (CD), but the biological function of miRNAs in CD remains unclear. We examine miRNA level in colon tissues and study the potential functions of miRNAs that regulate pathological genes during the inflammation process. DESIGN: miRNA levels were assayed in the inflamed colon of 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced and IL-10 knockout (KO) chronic colitis mice and CD patients by microarray or qRT-PCR. The influence of differently expressed miR-141 on its putative target genes, CXCL12ß, and leukocyte migration was investigated in colonic epithelia cells, colitis models and CD patients. The role of miR-141 was further studied in the experimental colitis mice by intracolonic administration of miR-141 precursors or inhibitors. RESULTS: An inverse correlation between miR-141 and CXCL12ß/total-CXCL12 was observed predominantly in the epithelial cells of the inflamed colons from colitic mice and CD patients. Further study demonstrated that miR-141 directly regulated CXCL12ß expression and CXCL12ß-mediated leukocyte migration. Upregulation or downregulation of miR-141 in the TNBS-induced or IL-10 KO colitic colon regulated leukocyte infiltration and alleviated or aggravated experimental colitis, respectively. Additionally, colonic overexpression of CXCL12ß abolished the therapeutic effect of miR-141 in TNBS-induced colitis. CONCLUSIONS: This study showed that the pathway of miR-141 targeting CXCL12ß is a possible mechanism underlying inflammatory cell trafficking during colonic inflammation process. Inhibiting colonic CXCL12ß expression and blocking colonic immune cell recruitment by using miRNA precursors represents a promising approach that may be valuable for CD treatment.


Assuntos
Quimiocina CXCL12/genética , Colite/metabolismo , Doença de Crohn/metabolismo , Regulação da Expressão Gênica , MicroRNAs/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Células Cultivadas , Colite/induzido quimicamente , Colite/prevenção & controle , Colo/metabolismo , Regulação para Baixo , Células Epiteliais/metabolismo , Feminino , Humanos , Interleucina-10/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , MicroRNAs/farmacologia , MicroRNAs/uso terapêutico , RNA Mensageiro/metabolismo , Ácido Trinitrobenzenossulfônico
16.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 40(5): 435-442, 2024 May.
Artigo em Zh | MEDLINE | ID: mdl-38790100

RESUMO

Objective To elucidate the expression characteristics and clinical significance of B7 homolog 3(B7-H3) in colorectal cancer (CRC) and to explore its associations with tumor glycolysis and immune cell infiltration in the tumor microenvironment. Methods The transcriptomic and clinicopathological data of CRC were obtained from the TCGA database and analyzed to determine the expression and clinical relevance of B7-H3. Correlations between glycolysis-related genes and B7-H3 expression were assessed based on TCGA data. The associations between B7-H3 expression and the infiltration of 22 types of immune cells were analyzed using the CIBERSORT algorithm. Immunohistochemical staining was used to verify the results of database analysis in surgical specimens and adjacent normal tissue specimens of 51 CRC patients.Results B7-H3 was significantly upregulated in CRC tissues and demonstrated strong correlations with tumor invasion depth, advanced TNM stage, and poor prognosis. Additionally, B7-H3 expression was closely associated with the upregulation of glycolysis-related genes in CRC. Immune cell infiltration analysis revealed that a total of 16 types of immune cells were significantly correlated with B7-H3 expression. Furthermore, an inverse relationship between B7-H3 expression and CD8+T cell infiltration was identified at the transcriptional level but not at the protein level. Conclusion B7-H3 is highly expressed in CRC tissues and is significantly correlated with disease progression and poor prognosis. Furthermore, the association of B7-H3 expression with glycolysis-related genes and immune cell infiltration suggests a pivotal role of B7-H3 in the regulation of tumor glycolysis and cancer immunity.


Assuntos
Antígenos B7 , Neoplasias Colorretais , Humanos , Antígenos B7/genética , Antígenos B7/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/metabolismo , Masculino , Feminino , Pessoa de Meia-Idade , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Prognóstico , Idoso , Regulação Neoplásica da Expressão Gênica , Glicólise/genética , Relevância Clínica
17.
Biochim Biophys Acta Rev Cancer ; 1879(1): 189031, 2024 01.
Artigo em Inglês | MEDLINE | ID: mdl-38036107

RESUMO

Colorectal cancer (CRC) is a common malignant tumor of the gastrointestinal tract and one of the most common causes of cancer-related deaths worldwide. Immune checkpoint inhibitors have become a milestone in many cancer treatments with significant curative effects. However, its therapeutic effect on colorectal cancer is still limited. B7-H3 is a novel immune checkpoint molecule of the B7/CD28 family and is overexpressed in a variety of solid tumors including colorectal cancer. B7-H3 was considered as a costimulatory molecule that promotes anti-tumor immunity. However, more and more studies support that B7-H3 is a co-inhibitory molecule and plays an important immunosuppressive role in colorectal cancer. Meanwhile, B7-H3 promoted metabolic reprogramming, invasion and metastasis, and chemoresistance in colorectal cancer. Therapies targeting B7-H3, including monoclonal antibodies, antibody drug conjugations, and chimeric antigen receptor T cells, have great potential to improve the prognosis of colorectal cancer patients.


Assuntos
Anticorpos Monoclonais , Neoplasias Colorretais , Humanos , Neoplasias Colorretais/patologia
18.
Cancer Lett ; 585: 216693, 2024 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-38301909

RESUMO

Necroptosis is a regulated necrotic cell death mechanism and plays a crucial role in the progression of cancers. However, the potential role and mechanism of necroptosis in colorectal cancer (CRC) has not been fully elucidated. In this study, we found that nuclear receptor subfamily 4 group A member 1 (NR4A1) was highly expressed in CRC cells treated with TNF-α, Smac mimetic, and z-VAD-FMK (TSZ). The depletion of NR4A1 significantly enhanced the sensitivity of CRC cells to TSZ-induced necroptosis, while NR4A1 overexpression suppressed these effects, as evidenced by the LDH assay, flow cytometry analysis of cell death, PI staining, and expression analysis of necrosome complexes (RIPK1, RIPK3, and MLKL). Moreover, NR4A1 deficiency made HT29 xenograft tumors sensitive to necroptotic cell death in vivo. Mechanistically, NR4A1 depletion promoted necroptosis activation in CRC through the RIG-I-like receptor pathway by interacting with DDX3. Importantly, the RIG-I pathway agonist poly(I:C) or inhibitor cFP abolished the effects of NR4A1 overexpression or suppression on necroptosis in CRC cells. Moreover, we observed that NR4A1 was highly expressed in CRC tissues and was associated with a poor prognosis. In conclusion, our results suggest that NR4A1 plays a critical role in modulating necroptosis in CRC cells and provide a new therapeutic target for CRC.


Assuntos
Neoplasias Colorretais , Proteínas Quinases , Humanos , Proteínas Quinases/metabolismo , Necroptose/fisiologia , Morte Celular , Necrose , Neoplasias Colorretais/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Proteína Serina-Treonina Quinases de Interação com Receptores/metabolismo , Apoptose , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/genética , Membro 1 do Grupo A da Subfamília 4 de Receptores Nucleares/metabolismo
19.
Cell Death Discov ; 10(1): 33, 2024 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-38228617

RESUMO

HOOK3, a member of the human hook microtubule-tethering protein family, has been implicated in the progression of cancer. However, the role of HOOK3 in the pathogenesis of gastric cancer (GC) remains incompletely understood. In this study, we investigated the expression of HOOK3 protein in GC tissues using immunohistochemistry (IHC). The findings of our study indicate that the expression levels of HOOK3 in GC tissues were relatively low. Furthermore, a significant negative association was seen between HOOK3 expression and the prognosis of patients with GC. The suppression of HOOK3 resulted in a notable increase in the proliferation, migration, invasion, and survival of GC cells. Conversely, the overexpression of HOOK3 had the opposite impact, reducing these cellular processes. Moreover, in vivo tests have shown evidence that the overexpression of HOOK3 significantly inhibited the formation of tumors and the spread of GC cells to the lungs. In a mechanistic manner, the analysis of RNA-seq data demonstrated that the knockdown of HOOK3 resulted in a notable increase in the expression of vascular endothelial growth factor A (VEGFA) in GC cells. Furthermore, the upregulation of VEGFA counteracted the impacts of HOOK3 upregulation on the proliferation, migration, invasion, and survival of GC cells. Furthermore, it was revealed that specificity protein 1 (SP1) exhibited the ability to bind to the promoter region of VEGFA. Moreover, the overexpression of SP1 successfully counteracted the inhibitory impact of HOOK3 overexpression on the expression of VEGFA in GC cells. In summary, the results of our study indicate that HOOK3 has a role in inhibiting the growth, migration, invasion, and survival of GC cells by modulating the SP1/VEGFA pathway. These findings contribute significant knowledge to our understanding of the underlying mechanisms involved in the development of GC.

20.
Life Sci ; : 122916, 2024 Jul 16.
Artigo em Inglês | MEDLINE | ID: mdl-39025206

RESUMO

AIMS: Gastric cancer (GC) is one of the most common malignant tumors of the digestive system. High expression of the mitotic kinase BUB1 has been shown to be associated with the development of many cancers, but the role of BUB1 in GC is still unclear. The current study aimed to investigate the role of BUB1 in GC. MATERIALS AND METHODS: BUB1 inhibitor, siRNA or BUB1 overexpression plasmid-mediated functional studies were performed in vitro and in vivo to explore the oncogenic role of BUB1 in GC. The expression of BUB1 and FGF18 in GC tumor samples was determined by IHC staining. RNA-seq, Western blot, MeRIP-qPCR and Co-IP assays were used to investigate the molecular mechanisms by which BUB1 regulates GC progression. KEY FINDINGS: Knockdown of BUB1 significantly inhibited the proliferation and metastasis of GC cells in vitro and in vivo. Moreover, overexpression of BUB1 significantly promoted the proliferation, migration and invasion of GC cells. High expression of BUB1 and FGF18 in GC tissues predicted poor prognosis in GC patients. Mechanistically, BUB1 interacted with METTL3 and induced m6A modification of TRAF6 mRNA, further activating the NF-κB/FGF18 axis in GC cells. SIGNIFICANCE: Our results confirmed that BUB1 acts as a positive regulator of GC cell proliferation and metastasis by activating the TRAF6/NF-κB/FGF18 pathway through METTL3-mediated m6A methylation. Targeting the BUB1/METTL3/TRAF6/NF-κB/FGF18 axis might be a novel diagnostic and therapeutic strategy in GC.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA