Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
1.
J Cell Sci ; 137(8)2024 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-38563084

RESUMO

Angiogenesis is a tightly controlled dynamic process demanding a delicate equilibrium between pro-angiogenic signals and factors that promote vascular stability. The spatiotemporal activation of the transcriptional co-factors YAP (herein referring to YAP1) and TAZ (also known WWTR1), collectively denoted YAP/TAZ, is crucial to allow for efficient collective endothelial migration in angiogenesis. The focal adhesion protein deleted-in-liver-cancer-1 (DLC1) was recently described as a transcriptional downstream target of YAP/TAZ in endothelial cells. In this study, we uncover a negative feedback loop between DLC1 expression and YAP activity during collective migration and sprouting angiogenesis. In particular, our study demonstrates that signaling via the RhoGAP domain of DLC1 reduces nuclear localization of YAP and its transcriptional activity. Moreover, the RhoGAP activity of DLC1 is essential for YAP-mediated cellular processes, including the regulation of focal adhesion turnover, traction forces, and sprouting angiogenesis. We show that DLC1 restricts intracellular cytoskeletal tension by inhibiting Rho signaling at the basal adhesion plane, consequently reducing nuclear YAP localization. Collectively, these findings underscore the significance of DLC1 expression levels and its function in mitigating intracellular tension as a pivotal mechanotransductive feedback mechanism that finely tunes YAP activity throughout the process of sprouting angiogenesis.


Assuntos
Adesões Focais , Proteínas Ativadoras de GTPase , Mecanotransdução Celular , Proteínas Supressoras de Tumor , Proteínas de Sinalização YAP , Animais , Humanos , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/genética , Movimento Celular , Retroalimentação Fisiológica , Adesões Focais/metabolismo , Adesões Focais/genética , Proteínas Ativadoras de GTPase/metabolismo , Proteínas Ativadoras de GTPase/genética , Células Endoteliais da Veia Umbilical Humana/metabolismo , Mecanotransdução Celular/genética , Neovascularização Fisiológica , Proteínas Supressoras de Tumor/metabolismo , Proteínas Supressoras de Tumor/genética , Proteínas de Sinalização YAP/metabolismo
2.
Genes Dev ; 31(17): 1754-1769, 2017 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-28982759

RESUMO

The Bcl-2 family protein Bim triggers mitochondrial apoptosis. Bim is expressed in nonapoptotic cells at the mitochondrial outer membrane, where it is activated by largely unknown mechanisms. We found that Bim is regulated by formation of large protein complexes containing dynein light chain 1 (DLC1). Bim rapidly inserted into cardiolipin-containing membranes in vitro and recruited DLC1 to the membrane. Bim binding to DLC1 induced the formation of large Bim complexes on lipid vesicles, on isolated mitochondria, and in intact cells. Native gel electrophoresis and gel filtration showed Bim-containing mitochondrial complexes of several hundred kilodaltons in all cells tested. Bim unable to form complexes was consistently more active than complexed Bim, which correlated with its substantially reduced binding to anti-apoptotic Bcl-2 proteins. At endogenous levels, Bim surprisingly bound only anti-apoptotic Mcl-1 but not Bcl-2 or Bcl-XL, recruiting only Mcl-1 into large complexes. Targeting of DLC1 by RNAi in human cell lines induced disassembly of Bim-Mcl-1 complexes and the proteasomal degradation of Mcl-1 and sensitized the cells to the Bcl-2/Bcl-XL inhibitor ABT-737. Regulation of apoptosis at mitochondria thus extends beyond the interaction of monomers of proapoptotic and anti-apoptotic Bcl-2 family members but involves more complex structures of proteins at the mitochondrial outer membrane, and targeting complexes may be a novel therapeutic strategy.


Assuntos
Apoptose/genética , Proteína 11 Semelhante a Bcl-2/metabolismo , Dineínas/metabolismo , Mitocôndrias/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Animais , Proteína 11 Semelhante a Bcl-2/genética , Células CACO-2 , Linhagem Celular Tumoral , Regulação da Expressão Gênica , Células HeLa , Humanos , Células MCF-7 , Camundongos , Ligação Proteica , Multimerização Proteica/genética , Estabilidade Proteica , Interferência de RNA , Proteína X Associada a bcl-2/genética
3.
J Biol Chem ; 297(4): 101161, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34480897

RESUMO

Cell migration is an essential physiological process, and aberrant migration of epithelial cells underlies many pathological conditions. However, the molecular mechanisms governing cell migration are not fully understood. We report here that growth factor-induced epithelial cell migration is critically dependent on the crosstalk of two molecular switches, namely phosphorylation switch (P-switch) and transcriptional switch (T-switch). P-switch refers to dynamic interactions of deleted in liver cancer 1 (DLC1) and PI3K with tensin-3 (TNS3), phosphatase and tensin homolog (PTEN), C-terminal tension, and vav guanine nucleotide exchange factor 2 (VAV2) that are dictated by mitogen-activated protein kinase kinase 1/2-extracellular signal-regulated protein kinase 1/2-dependent phosphorylation of TNS3, PTEN, and VAV2. Phosphorylation of TNS3 and PTEN on specific Thr residues led to the switch of DLC1-TNS3 and PI3K-PTEN complexes to DLC1-PTEN and PI3K-TNS3 complexes, whereas Ser phosphorylation of VAV2 promotes the transition of the PI3K-TNS3/PTEN complexes to PI3K-VAV2 complex. T-switch denotes an increase in C-terminal tension transcription/expression regulated by both extracellular signal-regulated protein kinase 1/2 and signal transducer and activator of transcription 3 (STAT3) via interleukin-6-Janus kinase-STAT3 signaling pathway. We have found that, the P-switch is indispensable for both the initiation and continuation of cell migration induced by growth factors, whereas the T-switch is only required to sustain cell migration. The interplay of the two switches facilitated by the interleukin-6-Janus kinase-STAT3 pathway governs a sequence of dynamic protein-protein interactions for sustained cell migration. That a similar mechanism is employed by both normal and tumorigenic epithelial cells to drive their respective migration suggests that the P-switch and T-switch are general regulators of epithelial cell migration and potential therapeutic targets.


Assuntos
Movimento Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 1/metabolismo , MAP Quinase Quinase 2/genética , MAP Quinase Quinase 2/metabolismo , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo
4.
J Biol Chem ; 295(2): 645-656, 2020 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-31806702

RESUMO

Deleted-in-liver cancer 1 (DLC1) exerts its tumor suppressive function mainly through the Rho-GTPase-activating protein (RhoGAP) domain. When activated, the domain promotes the hydrolysis of RhoA-GTP, leading to reduced cell migration. DLC1 is kept in an inactive state by an intramolecular interaction between its RhoGAP domain and the DLC1 sterile α motif (SAM) domain. We have shown previously that this autoinhibited state of DLC1 may be alleviated by tensin-3 (TNS3) or PTEN. We show here that the TNS3/PTEN-DLC1 interactions are mediated by the C2 domains of the former and the SAM domain of the latter. Intriguingly, the DLC1 SAM domain was capable of binding to specific peptide motifs within the C2 domains. Indeed, peptides containing the binding motifs were highly effective in blocking the C2-SAM domain-domain interaction. Importantly, when fused to the tat protein-transduction sequence and subsequently introduced into cells, the C2 peptides potently promoted the RhoGAP function in DLC1, leading to decreased RhoA activation and reduced tumor cell growth in soft agar and migration in response to growth factor stimulation. To facilitate the development of the C2 peptides as potential therapeutic agents, we created a cyclic version of the TNS3 C2 domain-derived peptide and showed that this peptide readily entered the MDA-MB-231 breast cancer cells and effectively inhibited their migration. Our work shows, for the first time, that the SAM domain is a peptide-binding module and establishes the framework on which to explore DLC1 SAM domain-binding peptides as potential therapeutic agents for cancer treatment.


Assuntos
Neoplasias da Mama/metabolismo , Proliferação de Células , Proteínas Ativadoras de GTPase/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Movimento Celular , Feminino , Proteínas Ativadoras de GTPase/química , Células HEK293 , Humanos , Modelos Moleculares , PTEN Fosfo-Hidrolase/química , PTEN Fosfo-Hidrolase/metabolismo , Fragmentos de Peptídeos/química , Fragmentos de Peptídeos/metabolismo , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Mapas de Interação de Proteínas , Motivo Estéril alfa , Tensinas/química , Tensinas/metabolismo , Proteínas Supressoras de Tumor/química , Proteína rhoA de Ligação ao GTP/química
5.
Mol Cancer ; 20(1): 141, 2021 11 02.
Artigo em Inglês | MEDLINE | ID: mdl-34727930

RESUMO

BACKGROUND: DLC1, a tumor suppressor gene that is downregulated in many cancer types by genetic and nongenetic mechanisms, encodes a protein whose RhoGAP and scaffolding activities contribute to its tumor suppressor functions. The role of the DLC1 START (StAR-related lipid transfer; DLC1-START) domain, other than its binding to Caveolin-1, is poorly understood. In other START domains, a key function is that they bind lipids, but the putative lipid ligand for DLC1-START is unknown. METHODS: Lipid overlay assays and Phosphatidylserine (PS)-pull down assays confirmed the binding of DLC1-START to PS. Co-immunoprecipitation studies demonstrated the interaction between DLC1-START and Phospholipase C delta 1 (PLCD1) or Caveolin-1, and the contribution of PS to those interactions. Rho-GTP, cell proliferation, cell migration, and/or anchorage-independent growth assays were used to investigate the contribution of PS and PLCD1, or the implications of TCGA cancer-associated DLC1-START mutants, to DLC1 functions. Co-immunoprecipitations and PS-pull down assays were used to investigate the molecular mechanisms underlying the impaired functions of DLC1-START mutants. A structural model of DLC1-START was also built to better understand the structural implications of the cancer-associated mutations in DLC1-START. RESULTS: We identified PS as the lipid ligand for DLC1-START and determined that DLC1-START also binds PLCD1 protein in addition to Caveolin-1. PS binding contributes to the interaction of DLC1 with Caveolin-1 and with PLCD1. The importance of these activities for tumorigenesis is supported by our analysis of 7 cancer-associated DLC1-START mutants, each of which has reduced tumor suppressor function but retains wildtype RhoGAP activity. Our structural model of DLC1-START indicates the mutants perturb different elements within the structure, which is correlated with our experimental findings that the mutants are heterogenous with regard to the deficiency of their binding properties. Some have reduced PS binding, others reduced PLCD1 and Caveolin-1 binding, and others are deficient for all of these properties. CONCLUSION: These observations highlight the importance of DLC1-START for the tumor suppressor function of DLC1 that is RhoGAP-independent. They also expand the versatility of START domains, as DLC1-START is the first found to bind PS, which promotes the binding to other proteins.


Assuntos
Caveolina 1/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Fosfatidilserinas/metabolismo , Fosfolipase C delta/metabolismo , Domínios e Motivos de Interação entre Proteínas , Proteínas Supressoras de Tumor/metabolismo , Sítios de Ligação , Proteínas de Transporte , Caveolina 1/química , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Proteínas Ativadoras de GTPase/genética , Humanos , Modelos Moleculares , Mutação , Fosfolipase C delta/química , Ligação Proteica , Conformação Proteica , Relação Estrutura-Atividade , Proteínas Supressoras de Tumor/genética
6.
Exp Lung Res ; 47(4): 173-182, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33678109

RESUMO

Lung adenocarcinoma (LUAD), one of the most common cancers, is a major threat to people's health due to its high mortality, and the survival of most patients suffering LUAD remains poor. This study aimed to explore the mechanism of Deleted in Liver Cancer 1 (DLC1) as a tumor suppressor underlying the occurrence and progression of LUAD. As revealed by bioinformatics analysis and qRT-PCR, DLC1 was significantly down-regulated in LUAD tumor tissue and cells. A series of cellular experiments including CCK-8, wound healing and Transwell assays were performed to detect the effect of DLC1 on the biological function of LUAD cells. It was found that overexpressing DLC1 significantly inhibited LUAD cell proliferative, migratory and invasive abilities, while knockdown of DLC1 promoted these abilities. Gene Set Enrichment Analysis (GSEA) and dual-luciferase assay were used to explore the downstream signaling pathway of DLC1, finding that DLC1 could remarkably inhibit the activity of mitogen-activated protein kinase (MAPK) signaling pathway. Western blot implemented for MAPK signaling pathway-related proteins further identified that DLC1 restrained the activation of MAPK/ERK signaling pathway. Furthermore, rescue experiments suggested that DLC1 inhibited LUAD cell proliferation and invasion by suppressing the MAPK/ERK signaling pathway. Overall, our study discussed the DLC1-dependent mechanism involved in LUAD. We found that the up-regulation of DLC1 may inhibit the malignant progression of LUAD by suppressing MAPK signaling pathway, which supports the view that DLC1 may serve as a molecular target for the targeted therapy of LUAD patients.


Assuntos
Adenocarcinoma de Pulmão , Proteínas Ativadoras de GTPase , Neoplasias Pulmonares , Proteínas Supressoras de Tumor , Adenocarcinoma de Pulmão/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Proteínas Ativadoras de GTPase/genética , Humanos , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica , Proteínas Supressoras de Tumor/genética
7.
Exp Cell Res ; 386(2): 111732, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31770531

RESUMO

Acquisition of features of mesenchymal cells represents a key step of metastatic progression of cancer cells and searching for mechanisms underlying the acquisition will help design novel clinical strategies for suppressing the metastatic progression. The Deleted in Liver Cancer-1 (DLC-1) gene is a p122/RhoGAP tumor/metastatic suppressor gene. However, the mechanism underlying DLC-1's inhibition of metastasis still remains largely unknown. In this study, we revealed that the DLC-1-deficient, but not the DLC-1-competent, human non-small cell lung carcinoma cells (NSCLCs) could acquire the TGF-ß1-induced expression of CD105, a common surface marker of mesenchymal stem cells, with consequent increase in CD105-associated cell motility. Interestingly, the induced CD105 expression and cell motility were subjected to the inhibition by the DLC-1-RhoA-Rock1 signaling through inhibiting the serine phosphorylation at a linker region, but not at the C-terminus, of the Smad3 protein and Smad3 protein nuclear translocation down the canonical TGF-ß1 signaling. In addition, the evidence suggested that DLC-1 very likely exerted its inhibitory effects on the TGF-ß1 signaling and the associated CD105 acquisition in both the cytoplasm and the nucleus. Consistent to the in vitro findings, a reverse correlation between CD105 and DLC-1 in protein expression was identified in primary NSCLC tissues and their surrounding non-tumor tissues. In summary, this study revealed a novel anti-metastasis mechanism governed by the DLC-1 tumor/metastasis suppressor, thus helping design new diagnostic and therapeutic approaches for NSCLCs.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/genética , Endoglina/genética , Proteínas Ativadoras de GTPase/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Fator de Crescimento Transformador beta1/genética , Proteínas Supressoras de Tumor/genética , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral , Movimento Celular , Núcleo Celular/metabolismo , Proliferação de Células , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Citosol/metabolismo , Endoglina/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/patologia , Metástase Linfática , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/patologia , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Fosforilação , Transporte Proteico , Transdução de Sinais , Proteína Smad3/genética , Proteína Smad3/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Quinases Associadas a rho/genética , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
8.
Dig Dis Sci ; 66(12): 4374-4383, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33439397

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) is the third leading cause of cancer-associated mortality worldwide. CircZKSCAN1 (hsa_circ_0001727) was reported to be related to HCC development. The present study aims to elucidate the potential role and molecular mechanism of circZKSCAN1 in the regulation of HCC progression. METHODS: CircZKSCAN1, miR-873-5p, and downregulation of deleted in liver cancer 1 (DLC1) in HCC tissues and cells were detected by RT-qPCR. Correlation between circZKSCAN1 expression and overall survival rate was measured by Kaplan-Meier survival analysis. The effects of circZKSCAN1, miR-873-5p, and DLC1 on proliferation, migration, and invasion were analyzed by CCK-8 and transwell assays, respectively. CyclinD1, Matrix metalloproteinase (MMP)-9, MMP-2, and DLC1 in HCC cells were detected by Western blot assay. The binding relationship between miR-873-5p and circZKSCAN1 or DLC1 was predicted by the Circinteractome or Starbase, and then confirmed by dual-luciferase reporter assays, respectively. Tumor volume and tumor weight were measured in vivo. RESULTS: CircZKSCAN1 was downregulated in HCC tissues and cells. Kaplan-Meier survival analysis suggested that there was a positive correlation between circZKSCAN1 expression and overall survival rate. Functionally, circZKSCAN1 blocked proliferation, migration, and invasion of HCC cells. MiR-873-5p was a target miRNA of circZKSCAN1, and miR-873-5p directly bound with DLC1. Rescue experiments confirmed that miR-873-5p overexpression or DLC1 knockdown attenuated the suppressive effects of circZKSCAN1 on HCC tumor growth in vitro. Besides, circZKSCAN1 inhibited HCC cell growth in vivo. CONCLUSIONS: This study firstly revealed that circZKSCAN1 curbed HCC progression via modulating miR-873-5p/DLC1 axis, providing a potential therapeutic target for HCC treatment.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Fatores de Transcrição Kruppel-Like/genética , Neoplasias Hepáticas/metabolismo , MicroRNAs/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/genética
9.
J Cell Mol Med ; 24(18): 10648-10662, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32725802

RESUMO

Enhancer of zeste homolog 2 (EZH2), an oncogene, is a commonly up-regulated epigenetic factor in human cancer. Hepatocellular carcinoma deletion gene 1 (DLC1) is an antioncogene that is either expressed at low levels or not expressed in many malignant tumours. Curcumin is a promising anticancer drug that has antitumour effects in many tumours, but its mechanism of action is unclear. Our research demonstrated that EZH2 was up-regulated in breast cancer (BC) tissues and cells, whereas DLC1 was down-regulated, and the expression of EZH2 and DLC1 was negatively correlated in BC. By analysing the characteristics of clinical cases, we found that positive expression of EZH2 and negative expression of DLC1 may be predictors of poor prognosis in patients with triple-negative breast cancer (TNBC). Moreover, knockdown of EZH2 expression restored the expression of DLC1 and inhibited the migration, invasion and proliferation, promoted the apoptosis, and blocked the cell cycle of MDA-MB-231 cells. Furthermore, we found that curcumin restored the expression of DLC1 by inhibiting EZH2; it also inhibited the migration, invasion and proliferation of MDA-MB-231 cells, promoted their apoptosis and blocked the cell cycle. Finally, xenograft tumour models were used to demonstrate that curcumin restored DLC1 expression by inhibiting EZH2 and also inhibited the growth and promoted the apoptosis of TNBC cells. In conclusion, our results suggest that curcumin can inhibit the migration, invasion and proliferation, promote the apoptosis, block the cycle of TNBC cells and restore the expression of DLC1 by inhibiting the expression of EZH2.


Assuntos
Adenocarcinoma/patologia , Antineoplásicos Fitogênicos/farmacologia , Curcumina/farmacologia , Proteína Potenciadora do Homólogo 2 de Zeste/antagonistas & inibidores , Proteínas Ativadoras de GTPase/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias de Mama Triplo Negativas/patologia , Proteínas Supressoras de Tumor/biossíntese , Adulto , Idoso , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/genética , Divisão Celular/efeitos dos fármacos , Divisão Celular/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proteína Potenciadora do Homólogo 2 de Zeste/biossíntese , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Feminino , Proteínas Ativadoras de GTPase/genética , Técnicas de Silenciamento de Genes , Código das Histonas , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Pessoa de Meia-Idade , Invasividade Neoplásica , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Prognóstico , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Proteínas Supressoras de Tumor/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Plant J ; 99(3): 556-570, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31004552

RESUMO

Meiosis is critical for sexual reproduction and the generation of new allelic variations in most eukaryotes. In this study, we report the isolation of a meiotic gene, DLC1, using a map-based cloning strategy. The dlc1 mutant is sterile in both male and female gametophytes due to an earlier defect in the leptotene chromosome and subsequent abnormalities at later stages. DLC1 is strongly expressed in the pollen mother cells (PMCs) and tapetum and encodes a nucleus-located rice type-B response regulator (RR) with transcriptional activity. Further investigations showed that DLC1 interacts with all five putative rice histidine phosphotransfer proteins (HPs) in yeast and planta cells, suggesting a possible participation of the two-component signalling systems (TCS) in rice meiosis. Our results demonstrated that DLC1 is required for rice meiosis and fertility, providing useful information for the role of TCS in rice meiosis.


Assuntos
Meiose/genética , Prófase Meiótica I/genética , Oryza/genética , Proteínas de Plantas/genética , Pólen/metabolismo , Clonagem Molecular , Regulação da Expressão Gênica no Desenvolvimento , Regulação da Expressão Gênica de Plantas , Mutação , Oryza/crescimento & desenvolvimento , Infertilidade das Plantas/genética , Proteínas de Plantas/metabolismo , Plantas Geneticamente Modificadas , Pólen/citologia , Pólen/crescimento & desenvolvimento
11.
J Cell Physiol ; 235(6): 4999-5007, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31773748

RESUMO

Deleted in liver cancer-1 (DLC1), a potential tumor suppressor, acts as a GTPase-activating protein for Rho family members. In many human cancers, the DLC1 expression is frequently downregulated or inactivated, which allows cancer cells to proliferate and disseminate. In this review, we describe the characteristics and other members of the DLC1 family and delineate the signal pathways DLC1 involved in regulating cancer cell growth, colony formation, apoptosis, senescence, autophagy, migration and invasion. In addition, we explore the clinical data of DLC1 and the mechanisms that natural products upregulate the DLC1 expression to inhibit cancer. Despite these insights, many important unanswered questions remain about the exact mechanisms of DLC1-mediated cancer suppression.


Assuntos
Biomarcadores Tumorais/genética , Proliferação de Células/genética , Proteínas Ativadoras de GTPase/genética , Neoplasias/genética , Proteínas Supressoras de Tumor/genética , Apoptose/genética , Autofagia/genética , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Neoplasias/patologia , Transdução de Sinais/genética
12.
BMC Cancer ; 20(1): 1064, 2020 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-33148199

RESUMO

BACKGROUND: Immunomodulatory activities of human mesenchymal stromal /stem cells (hMSCs) has been widely recognized as the most critical function of hMSCs for exerting its therapeutic effects. However, the detailed mechanisms responsible for regulating the immunomodulation of hMSCs still remain largely unknown. Previous studies revealed that the Notch1 protein exerted a pro-immunomodulatory function probably through interacting with the protein(s) subjective to proteasome-mediated protein degradation. The DLC-1 protein represents a well characterized tumor suppressor subjective to proteasome-mediated degradation. However, the detailed signaling pathway of Notch1 and the involvement of DLC-1 in regulating the immunomodulation of hMSCs have not been studied before. METHODS: The transfection with cDNA or siRNA into hMSCs assisted by co-culture of hMSCs with peripheral blood mononuclear cells and small molecule inhibitors of signaling proteins, followed by immunoprecipitation, Western blotting, RT-PCR, and flowcytometry, were employed to characterize the Notch1 signaling, to identify DLC-1 as a candidate proteasome-targeted protein, and to characterize DLC-1 signaling pathway and its interaction with the Notch1 signaling, in the regulation of immunomodulation of hMSCs, specifically, the inhibition of pro-inflammatory CD4+-Th1 lymphocytes, and the release of immunomodulatory molecule IDO1. STATISTICAL ANALYSIS: One-way ANOVA was utilized as a statistical tool to analyze the data presented as means ± SEM of at least three separate experiments. RESULTS: The present study revealed that the Notch1-Hey1 axis, but not the Notch1-Hes1 axis, was likely responsible for mediating the pro-immunomodulatory function of the Notch1 signaling. The DLC-1 protein was found subjective to proteasome-mediated protein degradation mediated by the DDB1 and FBXW5 E3 ligases and served as an inhibitor of the immunomodulation of hMSCs through inhibiting Rock1, but not Rock2, downstream the DLC-1 signaling. The Notch1 signaling in the Notch1-Hey1 pathway and the DLC-1 signaling in the DLC-1-Rock1-FBXW5 pathway exhibited a mutual exclusion interaction in the regulation of immunomodulation of hMSCs. CONCLUSIONS: The present study uncovers a novel function of DLC-1 tumor suppressor in regulating the immunomodulation of hMSCs. It also proposes a novel mutual exclusion mechanism between the DLC-1 signaling and the Notch1 signaling that is possibly responsible for fine-tuning the immunomodulation of hMSCs with different clinical implications in hMSCs therapy.


Assuntos
Proteínas F-Box/metabolismo , Proteínas Ativadoras de GTPase/metabolismo , Imunomodulação , Leucócitos Mononucleares/imunologia , Células-Tronco Mesenquimais/imunologia , Receptor Notch1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Quinases Associadas a rho/metabolismo , Células Cultivadas , Técnicas de Cocultura , Proteínas F-Box/genética , Proteínas Ativadoras de GTPase/genética , Genes Supressores de Tumor , Humanos , Leucócitos Mononucleares/citologia , Leucócitos Mononucleares/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Receptor Notch1/genética , Transdução de Sinais , Proteínas Supressoras de Tumor/genética , Quinases Associadas a rho/genética
13.
Clin Sci (Lond) ; 134(4): 419-434, 2020 02 28.
Artigo em Inglês | MEDLINE | ID: mdl-32065214

RESUMO

Cancer-derived exosomal miRNAs play an important role in the development of metastasis, but the effects and underlying mechanisms remain unclear. In the present study, we investigated the miRNA expression profiles of 5 paired serum exosomal samples from metastatic colorectal cancer (mCRC) and non-mCRC patients via RNA sequencing. After we evaluated the differentially expressed miRNAs in 80 CRC patients, miR-106b-3p was selected as a metastasis-associated miRNA of CRC. We showed that the expression level of serum exosomal miR-106b-3p was significantly higher in CRC patients with metastasis than those without metastasis. Additionally, high serum exosomal miR-106b-3p expression in patients was correlated with a poor prognosis. Coculture of low-metastatic CRC cells with high-metastatic CRC cell-derived exosomes promoted cell migration, invasion, and epithelial-to-mesenchymal transition (EMT), which was caused by the transport and transduction of miR-106b-3p in vitro. Moreover, exosomal miR-106b-3p promoted lung metastasis of CRC cells in vivo. In addition, we demonstrated that miR-106b-3p regulated metastasis by targeting deleted in liver cancer-1 (DLC-1). A negative correlation was also identified between miR-106b-3p and DLC-1 expression in human CRC tumour tissues and in mouse lung metastatic lesions. Collectively, our study indicated that metastasis-associated miR-106b-3p from serum exosomes could be used as a potential prognostic biomarker and therapeutic target for CRC patients.


Assuntos
Neoplasias Colorretais/genética , Exossomos/genética , Proteínas Ativadoras de GTPase/genética , MicroRNAs/metabolismo , Proteínas Supressoras de Tumor/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Neoplasias Colorretais/sangue , Progressão da Doença , Regulação para Baixo , Transição Epitelial-Mesenquimal/genética , Proteínas Ativadoras de GTPase/metabolismo , Regulação Neoplásica da Expressão Gênica , Células HEK293 , Humanos , Neoplasias Pulmonares/secundário , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/sangue , MicroRNAs/genética , Invasividade Neoplásica , Metástase Neoplásica , Prognóstico , Reprodutibilidade dos Testes , Proteínas Supressoras de Tumor/metabolismo
14.
Biochim Biophys Acta Mol Cell Res ; 1865(1): 1-11, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28982593

RESUMO

The hepatitis B virus core protein (HBc), also named core antigen, is well-known for its key role in viral capsid formation and virus replication. Recently, studies showed that HBc has the potential to control cell biology activity by regulating host gene expression. Here, we utilized miRNA microarray to identify 24 upregulated miRNAs and 21 downregulated miRNAs in HBc-expressed HCC cells, which were involved in multiple biological processes, including cell motility. Consistently, the in vitro transwell assay and the in vivo tail-vein injection model showed HBc promotion on HCC metastasis. Further, the miRNA-target gene network analysis displayed that the deleted in liver cancer (DLC-1) gene, an important negative regulator for cell motility, was potentially targeted by several differentially expressed miRNAs in HBc-introduced cells. Introduction of miRNAs mimics or inhibitors and 3'UTR luciferase activity assay proved that miR-382-5p efficiently suppressed DLC-1 expression and its 3'-UTR luciferase reporter activity. Importantly, cotransfection of miR-382-5p mimics/inhibitors and the DLC-1 expression vector almost abrogated HBc promotion on cell motility, indicating that the miR-382-5p/DLC-1 axis is important for mediating HBc-enhanced HCC motility. Clinical HCC samples also showed a negative correlation between miR-382-5p and DLC-1 expression level. Furthermore, HBc-positive HCC tissues showed high miR-382-5p level and reduced DLC-1 expression. In conclusion, our findings revealed that HBc promoted HCC motility by regulating the miR-382-5p/DLC-1 axis, which might provide a novel target for clinical diagnosis and treatment.


Assuntos
Carcinoma Hepatocelular/patologia , Movimento Celular , Proteínas Ativadoras de GTPase/genética , Antígenos do Núcleo do Vírus da Hepatite B/fisiologia , Neoplasias Hepáticas/patologia , MicroRNAs/genética , Proteínas Supressoras de Tumor/genética , Carcinoma Hepatocelular/complicações , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Hepatite B/complicações , Hepatite B/genética , Hepatite B/patologia , Humanos , Neoplasias Hepáticas/complicações , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/virologia , Metástase Neoplásica , Transdução de Sinais/genética
15.
J Biol Chem ; 293(37): 14407-14416, 2018 09 14.
Artigo em Inglês | MEDLINE | ID: mdl-30045871

RESUMO

Many newly synthesized cellular proteins pass through the Golgi complex from where secretory transport carriers sort them to the plasma membrane and the extracellular environment. The formation of these secretory carriers at the trans-Golgi network is promoted by the protein kinase D (PKD) family of serine/threonine kinases. Here, using mathematical modeling and experimental validation of the PKD activation and substrate phosphorylation kinetics, we reveal that the expression level of the PKD substrate deleted in liver cancer 1 (DLC1), a Rho GTPase-activating protein that is inhibited by PKD-mediated phosphorylation, determines PKD activity at the Golgi membranes. RNAi-mediated depletion of DLC1 reduced PKD activity in a Rho-Rho-associated protein kinase (ROCK)-dependent manner, impaired the exocytosis of the cargo protein horseradish peroxidase, and was associated with the accumulation of the small GTPase RAB6 on Golgi membranes, indicating a protein-trafficking defect. In summary, our findings reveal that DLC1 maintains basal activation of PKD at the Golgi and Golgi secretory activity, in part by down-regulating Rho-ROCK signaling. We propose that PKD senses cytoskeletal changes downstream of DLC1 to coordinate Rho signaling with Golgi secretory function.


Assuntos
Proteínas Ativadoras de GTPase/metabolismo , Proteína Quinase C/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Rede trans-Golgi/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática , Exocitose , Proteínas Ativadoras de GTPase/genética , Células HEK293 , Humanos , Membranas Intracelulares/metabolismo , Modelos Biológicos , Fosforilação , Interferência de RNA , Transdução de Sinais , Especificidade por Substrato , Proteínas Supressoras de Tumor/genética , Proteínas rab de Ligação ao GTP/metabolismo , Quinases Associadas a rho/metabolismo
16.
J Cell Biochem ; 120(4): 6078-6089, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30362160

RESUMO

Colorectal cancer (CRC) is the third most common type of cancer. MicroRNAs have been reported to participate in the progression of various cancers. In previous studies, miR-301a-3p expression was shown to be upregulated in CRC tissues. However, the underlying mechanism of miR-301a-3p in CRC has not yet been elucidated. Herein, the level of miR-301a-3p was found to be significantly upregulated in CRC clinical tissues and cell lines (HT29 and SW620). In addition, overexpression of miR-301a-3p obviously promoted cell proliferation, migration and invasion, and inhibited cell apoptosis in CRC cells. Meanwhile, upregulated miR-301a-3p expression also enhanced the expressions of Bax, caspase-3, caspase-9, matrix metalloproteinase (MMP)-2, and MMP-9, while the expression of Bax-2 was decreased. Furthermore, deleted in liver cancer-1 (DLC-1) and runt-related transcription factor 3 (RUNX3) were verified to be direct target genes of miR-301a-3p. Furthermore, overexpression of DLC-1 and RUNX3 revealed antitumor effects in CRC cell lines with the inhibition of cell proliferation, migration and invasion, and the induction of cell apoptosis. In addition, the expressions of Bax, caspase-3, caspase-3, MMP-2, and MMP-9 could be decreased after upregulating the expressions of DLC-1 and RUNX3, along with the upregulation of Bax-2. Moreover, overexpression of miR-301a-3p could promote the growth of xenograft tumors and liver metastasis in vivo, along with reducing the expressions of DLC-1 and RUNX3. Overall, miR-301a-3p might act as a tumor inducer in CRC cells through negatively regulating DLC-1 and RUNX3.


Assuntos
Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , MicroRNAs/metabolismo , Animais , Apoptose/genética , Apoptose/fisiologia , Western Blotting , Caspase 3/genética , Caspase 3/metabolismo , Proliferação de Células/genética , Proliferação de Células/fisiologia , Neoplasias Colorretais/genética , Subunidade alfa 3 de Fator de Ligação ao Core/genética , Feminino , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Marcação In Situ das Extremidades Cortadas , Masculino , Metaloproteinase 2 da Matriz/genética , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/genética , Camundongos , Camundongos Nus , MicroRNAs/genética , Pessoa de Meia-Idade , Invasividade Neoplásica/genética , Invasividade Neoplásica/patologia , Proteínas Supressoras de Tumor/genética , Proteínas Supressoras de Tumor/metabolismo , Cicatrização/genética , Cicatrização/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto
17.
Exp Cell Res ; 370(2): 292-302, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-29964052

RESUMO

Induction of cellular senescence represents a novel strategy to inhibit aberrant proliferation of cancer cells. Resveratrol is gaining attention for its cancer preventive and suppressive properties. Tumor suppressor gene DLC1 is shown to induce apoptosis, suppress migration and invasion in various cancer cells. However, the function of DLC1 in cancer cellular senescence is unclear. This study was designed to investigate the biological role of DLC1 in resveratrol induced cancer cellular senescence. Our results showed that resveratrol inhibited proliferation of cancer cell lines (MCF-7, MDA-MB-231 and H1299) and induced senescence along with increase of SA-ß-gal activity and regulation of senescence-associated molecular markers p38MAPK, p-p38MAPK, p27, p21, Rb and p-Rb protein. The underlying mechanism was that resveratrol induced mitochondrial dysfunction with reduction of mitochondrial membrane potential, down-regulation of MT-ND1, MT-ND6 and ATPase8 in transcript level and down-regulation of PGC-1α in protein level to result in ROS production. With ROS elevation, resveratrol decreased DNMT1 and increased DLC1 expression significantly. However, after ROS scavenger NAC was added to the cancer cells treated by resveratrol, DNMT1, DLC1 and senescence-associated molecular markers were reversed. This reveals that resveratrol induced cancer cellular senescence through DLC1 in a ROS-dependent manner. Silencing DLC1 markedly attenuated SA-ß-gal activity and p38MAPK, p27 and p21 protein levels, and increased Rb expression, indicating that resveratrol promoted senescence via targeting DLC1. Moreover, DLC1 promoted senescence through FoxO3a/NF-κB signaling mediated by SIRT1 after resveratrol treatment. Finally, resveratrol increased ROS production to induce DNA damage with p-CHK1 up-regulation and result in cancer cellular senescence. This is the first time to investigate resveratrol induced cancer cellular senescence by primarily targeting DLC1. Induction of cellular senescence by resveratrol may represent a novel anticancer mechanism.


Assuntos
Senescência Celular/efeitos dos fármacos , Proteínas Ativadoras de GTPase/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Resveratrol/farmacologia , Proteínas Supressoras de Tumor/metabolismo , Dano ao DNA/efeitos dos fármacos , Genes Mitocondriais/efeitos dos fármacos , Humanos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Supressoras de Tumor/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
J Biol Chem ; 291(39): 20353-71, 2016 09 23.
Artigo em Inglês | MEDLINE | ID: mdl-27481945

RESUMO

RHO GTPase-activating proteins (RHOGAPs) are one of the major classes of regulators of the RHO-related protein family that are crucial in many cellular processes, motility, contractility, growth, differentiation, and development. Using database searches, we extracted 66 distinct human RHOGAPs, from which 57 have a common catalytic domain capable of terminating RHO protein signaling by stimulating the slow intrinsic GTP hydrolysis (GTPase) reaction. The specificity of the majority of the members of RHOGAP family is largely uncharacterized. Here, we comprehensively investigated the sequence-structure-function relationship between RHOGAPs and RHO proteins by combining our in vitro data with in silico data. The activity of 14 representatives of the RHOGAP family toward 12 RHO family proteins was determined in real time. We identified and structurally verified hot spots in the interface between RHOGAPs and RHO proteins as critical determinants for binding and catalysis. We have found that the RHOGAP domain itself is nonselective and in some cases rather inefficient under cell-free conditions. Thus, we propose that other domains of RHOGAPs confer substrate specificity and fine-tune their catalytic efficiency in cells.


Assuntos
Proteínas Ativadoras de GTPase/química , Proteínas rho de Ligação ao GTP/química , Proteínas Ativadoras de GTPase/genética , Proteínas Ativadoras de GTPase/metabolismo , Humanos , Domínios Proteicos , Relação Estrutura-Atividade , Proteínas rho de Ligação ao GTP/genética , Proteínas rho de Ligação ao GTP/metabolismo
19.
Mol Cell Biochem ; 425(1-2): 47-58, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27830358

RESUMO

The heterogeneity of breast cancer makes it a challenging solid tumor to diagnose and treat. A tumor suppressor Deleted in Liver Cancer 1 (DLC1) has been reported to be down-regulated or even silenced in several kinds of cancer including breast cancer. Curcumin has been reported to modulate the growth of tumor cells through regulation of multiple cell signaling pathways and modulate epigenetic changes by CpG demethylation of many tumor suppressor genes. This study was designed to investigate the effect of curcumin on the expression of Deleted in Liver Cancer 1 (DLC1) in human breast cancer cell line MDA-MB-361 and the underlying mechanism in vitro and in vivo. Curcumin induced DLC1 expression in a dose-dependent manner. In curcumin-treated cells, methylation of DLC1 promoter was reduced and active forms of RhoA and Cdc42 were also decreased. DLC1 expression was closely related to tumor cell growth, demonstrated by Ki67 staining. Curcumin inhibited DNA methyltransferase 1 expression through down-regulation of transcription factor Sp1. Consistent with the in vitro data, in vivo administration of curcumin inhibited the growth of implanted MDA-MB-361 cells and induced DLC1 expression in tumor tissue. In MDA-MB-361 cells, curcumin down-regulates the expression of Sp1 to inhibit the expression of DNA methyltransferase 1, thus subsequently reducing hypermethylation of DLC1 promoter to induce DLC1 expression.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Curcumina/farmacologia , Metilação de DNA/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Proteínas Ativadoras de GTPase/biossíntese , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Regiões Promotoras Genéticas , Proteínas Supressoras de Tumor/biossíntese , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , DNA de Neoplasias/genética , Feminino , Proteínas Ativadoras de GTPase/genética , Humanos , Proteínas Supressoras de Tumor/genética
20.
Zhonghua Yi Xue Za Zhi ; 97(6): 412-417, 2017 Feb 14.
Artigo em Chinês | MEDLINE | ID: mdl-28219126

RESUMO

Objective: To detect the methylation status of DLC-1 gene in the patients with myelodysplastic syndrome(MDS), the effect of abnormal methylation of DLC-1 gene on the expression of DLC-1 gene, the clinical significance of methylation of DLC-1 gene in MDS patients, and the effect of decitabine on DLC-1 gene expression. Methods: A total of 43 MDS patients were treated in Fujian Medical University Union Hospital from 2013 to 2015. Methylation status of DLC-1 gene in MDS patients were detected by the methylation specific PCR(MSP). The expression of DLC-1 gene mRNA was determined with real-time fluorescence quantitative PCR(RTFQ-PCR). MDS patients were divided into 5 groups (very low-risk, low-risk, intermediate-risk, high-risk and very high-risk, n=0, 8, 7, 18, 10) according to WPSS classification. And the clinical significance of methylation of DLC-1 gene in patients with MDS were investigated. In order to investigate the change in gene methylation and expression of DLC-1 gene after treatment with decitabine, methylation statuses of DLC-1 gene in MDS patients before and after be treated with decitabine were detected by the bisulfite sequencing PCR(BSP). The expressions of DLC-1 gene mRNA of these patients were determined with RTFQ-PCR. Results: Hypermethylation of CpG island of DLC-1 gene was observed in 55.16%(22/43)MDS patients. The expressions of DLC-1 gene mRNA in methylation positive patients were significantly lower than that in methylation negative patients (0.32±0.06 vs 0.91±0.11)(P=0.008). For MDS patients, the DLC-1 methylation rate of intermediate-and high-risk patient was 21/35, which was significantly higher than that of low-risk patient(1/8, P=0.006). The methylation status of DLC-1 gene were monitored in 8 patients before and after treatment with the decitabine (decitabine 20 mg/m(2,) d1-d5/d28, more than 4 courses) , the methylation rate of DLC-1 gene dropped from 57.50%±5.11% to 14.13%±2.07% after treatment(P=0.010). The expression of DLC-1 gene increased after treatment with decitabine(0.67±0.08 vs 0.28±0.06, P=0.015). Conclusions: Methylation of DLC-1 gene is common in MDS patients and may be associated with poor prognosis. Decitabine may activate the expression of DLC-1 gene by demethylation, which may be one of the mechanisms for the treatment of patients with MDS.


Assuntos
Metilação de DNA , Síndromes Mielodisplásicas , Azacitidina/análogos & derivados , Ilhas de CpG , Decitabina , Proteínas Ativadoras de GTPase , Expressão Gênica , Humanos , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , RNA Mensageiro , Proteínas Supressoras de Tumor
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA