Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Biochim Biophys Acta Mol Basis Dis ; 1869(8): 166821, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37516255

RESUMO

Development of colorectal cancer (CRC) accompanied with genomic instability. Genomic instability was promoted by microRNAs (miRNAs) inhibiting key genes in DNA damage repair and spindle assembly processes. Whether miR-653-3p affects genomic instability is unknown. The aim of this study is to explore the effect of miR-653-3p on genomic instability in CRC cells. Based on RT-qPCR analysis, miR-653-3p was highly expressed in CRC cells. Through single-cell electrophoresis assay and chromosome karyotype analysis, we determined ectopic expression of miR-653-3p induced increased DNA damage but inhibited apoptosis by promoting chromosomal instability. Mechanistically, luciferase assay identified the direct interaction of miR-653-3p with the 3' UTR of SIRT1, and western blot analysis indicated miR-653-3p inhibited SIRT1 and then promoted STAT3 phosphorylation and TWIST1 expression. The results of karyotype analysis showed that the upregulation of SIRT1 and the downregulation of TWIST1 caused by the downregulation of miR-653-3p suppressed chromosomal instability. Additionally, our evidence showed that miR-653-3p promoted CRC cell proliferation, migration, and 5-FU resistance, and miR-653-3p induced the development of CRC in the xenograft mice model. Altogether, our evidence suggests that miR-653-3p regulates SIRT1/TWIST1 signaling pathway and plays an important role in promoting genomic instability, proliferation, migration, and chemoresistance of CRC cells, which may serve as a promising therapeutic target for CRC therapy.


Assuntos
Neoplasias Colorretais , MicroRNAs , Humanos , Animais , Camundongos , Sirtuína 1/genética , Sirtuína 1/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Transdução de Sinais , Instabilidade Genômica , Instabilidade Cromossômica , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada a Twist/genética
2.
Cell Signal ; 101: 110517, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36332797

RESUMO

Colorectal cancer (CRC) continues to represent one of the major causes of cancer-related mortality and morbidity. MicroRNAs (miRNAs) are confirmed to be involved in modulating substential biological processes by affecting the expression of targeted genes, including carcinogenesis. In the present study, the expression pattern and functional roles of microRNA-15a-5p (miR-15a-5p) in CRC cells were investigated. The data from TCGA database indicated that miR-15a-5p is highly expressed in CRC tissues. Moreover, ectopic expression of miR-15a-5p facilitated the proliferation, migration, and invasion of CRC cells. Furthermore, bioinformatic analysis combinating with dual-luciferase assay revealed that SIRT4 acts as a crucial target of miR-15a-5p. Accordingly, overexpression of SIRT4 suppresses the miR-15a-5p-mediated enhancement in the proliferation, migration, and invasion of CRC cells, while the opposite phenotypes were observed after inhibition of SIRT4. Moreover, we further revealed that miR-15a-5p restrained the expression of SIRT4 to exacerbate the malignant phenotypes by modulating STAT3/TWIST1 and PETN/AKT signaling in CRC cells. Alternatively, inhibition of the miR-15a-5p/SIRT4 axis enhanced the chemosensitivity of 5-fluorouracil- and oxaliplatin-resistant HCT116 cells. Altogether, our evidence suggests that miR-15a-5p plays an essential role in promoting the proliferation, migration, and chemoresistance of CRC cells via targeting SIRT4 to modulate STAT3/TWIST1 and PETN/AKT signaling, which may serve as a promising therapeutic target for CRC therapy.


Assuntos
Neoplasias Colorretais , MicroRNAs , Humanos , Linhagem Celular Tumoral , Proliferação de Células/genética , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , MicroRNAs/metabolismo , Proteínas Nucleares/metabolismo , Fenótipo , Proteínas Proto-Oncogênicas c-akt/metabolismo , PTEN Fosfo-Hidrolase/genética , PTEN Fosfo-Hidrolase/metabolismo , Transdução de Sinais , Fator de Transcrição STAT3/metabolismo , Proteína 1 Relacionada a Twist/genética , Proteína 1 Relacionada a Twist/metabolismo
3.
Mol Biol Rep ; 49(9): 8337-8347, 2022 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-35690960

RESUMO

BACKGROUND: The immunomodulatory function of mesenchymal stem cells (MSCs) has been considered to be vital for MSC-based therapies. Many works have been devoted to excavate effective strategies for enhancing the immunomodulation effect of MSCs. Nonetheless, canine MSC-mediated immunomodulation is still poorly understood. METHODS AND RESULTS: The inflammatory microenvironment was simulated through the employment of interferon-γ (IFN-γ) in a culture system. Compared with unstimulated cBMSCs, IFN-γ stimulation increased the mRNA levels of Toll-like receptor 3 (TLR3) and indoleamine 2, 3-dioxygenase 1 (IDO-1), and simultaneously enhanced the secretion of immunosuppressive molecules, including interleukin (IL)-10, hepatocyte growth factor (HGF), and kynurenine in cBMSCs. IFN-γ stimulation significantly enhanced the ability of cBMSCs and their supernatant to suppress the proliferation of murine spleen lymphocytes. Lymphocyte subtyping evaluation revealed that cBMSCs and their supernatant diminished the percentage of CD3+CD4+ and CD3+CD8+ lymphocytes compared with the control group, with a decreasing CD4+/CD8+ ratio. Notably, exposure to IFN-γ decreased the CD4+/CD8+ ratio more effectively than unstimulated cells or supernatant. Additionally, IFN-γ-stimulation increased the mRNA levels of the Th1 cytokines TNF-α, and remarkably decreased the mRNA level of the Th2 cytokine IL-4 and IL-10. CONCLUSION: Our findings substantiate that IFN-γ stimulation can enhance the immunomodulatory properties of cBMSCs by promoting TLR3-dependent activation of the IDO/kynurenine pathway, increasing the secretion of immunoregulatory molecules and strengthening interactions with T lymphocytes, which may provide a meaningful strategy for the clinical application of cBMSCs in immune-related diseases.


Assuntos
Terapia de Imunossupressão , Indolamina-Pirrol 2,3,-Dioxigenase , Interferon gama , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Receptor 3 Toll-Like , Animais , Proliferação de Células , Células Cultivadas , Cães , Terapia de Imunossupressão/métodos , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Interferon gama/farmacologia , Cinurenina/metabolismo , Cinurenina/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/imunologia , Camundongos , RNA Mensageiro/metabolismo , Receptor 3 Toll-Like/metabolismo
4.
Exp Cell Res ; 418(1): 113265, 2022 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-35716785

RESUMO

Understanding the mechanisms of colorectal cancer (CRC) progression is critical for developing innovative treatment strategies. As an endoplasmic reticulum-located protein, B cell receptor-associated protein 31 (BCAP31) has been identified to be highly expressed in multiple cancers. However, its function and molecular mechanism in CRC remain not fully understood. In the present study, BCAP31 expression and its correlation with the clinical stage were analyzed based on TCGA database. We demonstrated that loss of BCAP31 suppressed CRC cell proliferation in vitro and tumor growth in vivo. Mechanistically, we demonstrated that Emerin was an interaction partner and downstream molecule of BCAP31. Knockdown of BCAP31 promoted the nuclear envelope localization of Emerin, leading to a reduction of ß-catenin accumulation in the nucleus, which resulted in downregulation of Wnt/ß-catenin downstream target genes, including c-Myc, cyclin D1, Survivin, and Mcl-1. Moreover, downregulation of Emerin partially restored the BCAP31 depletion-mediated ß-catenin protein level and tumor suppressive effects in CRC cells.Our data highlights the pivotal role of BCAP31 depletion in inhibiting cell proliferation in CRC cells, and mechanistically via Emerin/ß-catenin signaling, which may serve as a promising target for CRC treatment.


Assuntos
Neoplasias Colorretais , beta Catenina , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/patologia , Regulação Neoplásica da Expressão Gênica , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas Nucleares , Receptores de Antígenos de Linfócitos B/genética , Via de Sinalização Wnt , beta Catenina/genética , beta Catenina/metabolismo
5.
Biochem Biophys Res Commun ; 615: 116-122, 2022 07 30.
Artigo em Inglês | MEDLINE | ID: mdl-35609416

RESUMO

MicroRNAs (miRNAs) play a crucial role in cancer progression due to their capability to modulate the expression of various target genes. However, given the heterogeneity of tumor cells, miRNAs have been confirmed to exert different regulatory effects. Here, bioinformatic analysis results indicated that expression of miR-330-5p is decreased in colorectal cancer (CRC) tissues and inversely correlated with SND1 expression. Notably, ectopic expression of miR-330-5p restrained tumor cell proliferation, migration, and enhance the sensitivity of CRC cells to 5-FU. Moreover, similar phenotypes were substantiated after inhibition of SND1 expression using RNA interference. Conversely, overexpression of SND1 facilitated the malignant phenotypes of CRC cells and restored miR-330-5p-mediated tumor-suppressive activities in CRC cells. Mechanistically, miR-330-5p directly binds to SND1-3'-untranslated region (3'-UTR), thus involving in inhibiting CRC cells proliferation and invasion and promoting apoptosis. Taken together, miR-330-5p may act as a tumor suppressor by targeting the expression of SND1, suggesting that the miR-330-5p/SND1 axis may be a meaningful regulator for CRC intervention.


Assuntos
Neoplasias Colorretais , MicroRNAs , Regiões 3' não Traduzidas , Apoptose/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Neoplasias Colorretais/patologia , Endonucleases/genética , Endonucleases/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , MicroRNAs/metabolismo
6.
Nutrients ; 14(4)2022 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-35215376

RESUMO

Inflammatory activation and intestinal flora imbalance play an essential role in the development and progression of colorectal cancer (CRC). Berberine (BBR) has attracted great attention in recent years due to its heath-related benefits in inflammatory disorders and tumors, but the intricate mechanisms have not been fully elucidated. In this study, the effects and the mechanism of BBR on colon cancer were investigated in an azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced colitis-associated carcinogenesis mice model. Our results showed that pre-administration of BBR showed a decrease in weight loss, disease activity index (DAI) score, and the number of colon tumors in mice, compared with the model group. The evidence from pathological examination indicated that the malignancy of intestinal tumors was ameliorated after pre-administration of BBR. Additionally, pre-administration with BBR suppressed the expression of pro-inflammatory factors (interleukin (IL)-6, IL-1ß, cyclooxygenase (COX)-2 and tumor necrosis factor (TNF)-α) and the cell-proliferation marker Ki67, while expression of the tight junction proteins (ZO-1 and occludin) were increased in colon tissue. Moreover, the levels of critical pathway proteins involved in the inflammatory process (p-STAT3 and p-JNK) and cell cycle regulation molecules (ß-catenin, c-Myc and CylinD1) exhibited lower expression levels. Besides, 16S rRNA sequence analysis indicated that pre-administration of BBR increased the ratio of Firmicutes/Bacteroidetes (F:M) and the relative abundance of potentially beneficial bacteria, while the abundance of cancer-related bacteria was decreased. Gavage with Lactobacillus rhamnosus can improve the anti-tumor effect of BBR. Overall, pre-administration of BBR exerts preventive effects in colon carcinogenesis, and the mechanisms underlying these effects are correlated with the inhibition of inflammation and tumor proliferation and the maintenance of intestinal homeostasis.


Assuntos
Berberina , Colite , Microbioma Gastrointestinal , Animais , Azoximetano/toxicidade , Carcinogênese/metabolismo , Colite/induzido quimicamente , Colite/tratamento farmacológico , Colite/metabolismo , Colo/metabolismo , Sulfato de Dextrana/toxicidade , Modelos Animais de Doenças , Inflamação/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , RNA Ribossômico 16S/metabolismo
7.
Int J Mol Sci ; 22(21)2021 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-34768788

RESUMO

Senescence in mesenchymal stem cells (MSCs) not only hinders the application of MSCs in regenerative medicine but is also closely correlated with biological aging and the development of degenerative diseases. In this study, we investigated the anti-aging effects of curcumin (Cur) on canine bone marrow-derived MSCs (cBMSCs), and further elucidated the potential mechanism of action based on the modulation of autophagy. cBMSCs were expanded in vitro with standard procedures to construct a cell model of premature senescence. Our evidence indicates that compared with the third passage of cBMSCs, many typical senescence-associated phenotypes were observed in the sixth passage of cBMSCs. Cur treatment can improve cBMSC survival and retard cBMSC senescence according to observations that Cur (1 µM) treatment can improve the colony-forming unit-fibroblasts (CFU-Fs) efficiency and upregulated the mRNA expression of pluripotent transcription factors (SOX-2 and Nanog), as well as inhibiting the senescence-associated beta-galactosidase (SA-ß-gal) activities and mRNA expression of the senescence-related markers (p16 and p21) and pro-inflammatory molecules (tumor necrosis factor-α (TNF-α) and interleukin-6 (IL-6)). Furthermore, Cur (0.1 µM~10 µM) was observed to increase autophagic activity, as identified by upregulation of microtubule-associated protein 1 light chain 3 (LC3), unc51-like autophagy-activating kinase-1 (ULK1), autophagy-related gene (Atg) 7 and Atg12, and the generation of type II of light chain 3 (LC3-II), thereby increasing autophagic vacuoles and acidic vesicular organelles, as well as causing a significant decrease in the p62 protein level. Moreover, the autophagy activator rapamycin (RAP) and Cur were found to partially ameliorate the senescent features of cBMSCs, while the autophagy inhibitor 3-methyladenine (3-MA) was shown to aggravate cBMSCs senescence and Cur treatment was able to restore the suppressed autophagy and counteract 3-MA-induced cBMSC senescence. Hence, our study highlights the important role of Cur-induced autophagy and its effects for ameliorating cBMSC senescence and provides new insight for delaying senescence and improving the therapeutic potential of MSCs.


Assuntos
Senescência Celular/efeitos dos fármacos , Curcumina/farmacologia , Células-Tronco Mesenquimais/metabolismo , Animais , Autofagossomos/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Autofagia/fisiologia , Senescência Celular/fisiologia , China , Curcumina/metabolismo , Cães , Feminino , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos
8.
In Vitro Cell Dev Biol Anim ; 57(7): 685-694, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-34518994

RESUMO

The destruction of biological activity such as senescence and apoptosis caused by oxidative stress could play a pivotal role in the poor therapeutic efficiency of bone marrow mesenchymal stem cells (BMSCs) transplantation. Mitoquinone (MitoQ) has a highly effective mitochondrial antioxidant effect, and has been widely used in many oxidative damage models. This study aimed to investigate the protective effect of MitoQ on the oxidative stress-mediated senescence of canine BMSCs and the underlying mechanism. The senescence of BMSCs was determined by senescence-associated ß-galactosidase staining and quantitative real-time PCR. The expression of p-Nrf2 protein was detected by Western blotting. The results demonstrated that, as BMSCs were expanded in vitro, the senescent phenotype appeared. And the senescence of BMSCs may be caused by oxidative stress, manifested by increasing the level of ROS and decreasing the activity of antioxidant enzymes. Treatment of MitoQ down-regulated the mRNA levels of senescence-related and apoptosis-related genes, but up-regulated the mRNA levels of proliferation-related genes. Meanwhile, ROS generation and senescent activity were reduced in MitoQ-treated BMSCs. Further mechanism studies showed that MitoQ obviously promoted Nrf2 phosphorylation, and also facilitated the translocation of Nrf2 into the nucleus. Moreover, treatment of MitoQ increased the mRNA levels of downstream antioxidant genes and enhanced the activities of superoxide dismutase, catalase, and glutathione peroxidase. Thus, our study revealed that MitoQ, via the Nrf2/ARE signaling pathway, exerts an antioxidant effect as well as potentially delays OS-mediated senescence during BMSCs that were expanded in vitro, which may serve as a novel strategy to optimize the clinical application of BMSCs.


Assuntos
Elementos de Resposta Antioxidante/fisiologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Fator 2 Relacionado a NF-E2/metabolismo , Compostos Organofosforados/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Ubiquinona/análogos & derivados , Animais , Antígenos CD/metabolismo , Elementos de Resposta Antioxidante/efeitos dos fármacos , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/genética , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Senescência Celular/efeitos dos fármacos , Cães , Enzimas/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Estresse Oxidativo/fisiologia , Substâncias Protetoras/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ubiquinona/farmacologia
9.
Mol Cell Biochem ; 476(2): 1135-1149, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33196943

RESUMO

Mesenchymal stem cells (MSCs) are considered to be a promising therapeutic material due to their capacities for self-renewal, multilineage differentiation, and immunomodulation and have attracted great attention in regenerative medicine. However, MSCs may lose their biological functions because of donor age or disease and environmental pressure before and after transplantation, which hinders the application of MSC-based therapy. As a major intracellular lysosome-dependent degradative process, autophagy plays a pivotal role in maintaining cellular homeostasis and withstanding environmental pressure and may become a potential therapeutic target for improving MSC functions. Recent studies have demonstrated that the regulation of autophagy is a promising approach for improving the biological properties of MSCs. More in-depth investigations about the role of autophagy in MSC biology are required to contribute to the clinical application of MSCs. In this review, we focus on the role of autophagy regulation by various physical and chemical factors on the biological functions of MSCs in vitro and in vivo, and provide some strategies for enhancing the therapeutic efficacy of MSCs.


Assuntos
Autofagia , Homeostase , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/citologia , Medicina Regenerativa , Animais , Diferenciação Celular , Humanos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA