Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 60
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
Biochem Biophys Res Commun ; 528(1): 62-70, 2020 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-32456796

RESUMO

The acquisition of chemoresistance is a major clinical challenge for pancreatic cancer (PC) treatment. Chemoresistance is largely attributed to aberrant DNA damage repair. However, the underlying mechanisms of chemoresistance in pancreatic cancer remain unclear. Here, we showed that CD147 was strongly correlated to DNA damage response (DDR) indices and poor prognosis in pancreatic ductal adenocarcinoma (PDAC) patients. CD147 knockdown or monoclonal antibodies improved the killing effects of gemcitabine in gemcitabine resistant cells, exhibiting reduced activation of ATM/p53. Moreover, we found the interaction of CD147 with ATM, ATR and p53, which was augmented in gemcitabine resistant cells. High CD147/p-ATM/p-ATR/p-p53 cytoplasmic expression associated with poor survival of PC patients. Our studies thus identify CD147 as a critical player in DDR programing that affects gemcitabine therapeutic outcomes of pancreatic cancer patients.


Assuntos
Proteínas Mutadas de Ataxia Telangiectasia/metabolismo , Basigina/metabolismo , Dano ao DNA , Desoxicitidina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Neoplasias Pancreáticas/diagnóstico , Neoplasias Pancreáticas/patologia , Proteína Supressora de Tumor p53/metabolismo , Animais , Carcinoma Ductal Pancreático/tratamento farmacológico , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Citoplasma/metabolismo , Desoxicitidina/farmacologia , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Instabilidade Genômica/genética , Humanos , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Prognóstico , Transdução de Sinais/efeitos dos fármacos , Análise de Sobrevida , Gencitabina
2.
Apoptosis ; 24(7-8): 673-685, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31177396

RESUMO

Chemotherapeutic resistance always results in poor clinical outcomes of cancer patients and its intricate mechanisms are large obstacles in overcoming drug resistance. CCR4-NOT transcription complex subunit 3 (CNOT3), a post-translational regulator, is suggested to be involved in cancer development and progression. However, its role in chemotherapeutic resistance is not well understood. In this study, after screening the CNOT3 mRNA in a cancer microarray database called Oncomine and examining the expression levels of CNOT3 mRNA in normal tissues and lung cancer tissues, we found that CNOT3 was up-regulated in lung cancer tissues. Besides, its high-expression was associated with poor prognosis of lung cancer patients. We also found higher expression level of CNOT3 and lower expression level of receptor-interacting protein kinase 3 (RIPK3) in cisplatin-resistant A549 (A549/DDP) cells, and knocking down CNOT3 expression could sensitize A549/DDP cells to cisplatin-induced apoptosis. We demonstrated that CNOT3 depletion up-regulated the expression level of RIPK3 and the enhanced apoptosis was mediated by the elevated RIPK3 to further trigger Caspase 8 activation. Taken together, our results reveal a role of CNOT3 in cisplatin resistance of lung cancer and provide a potential target for lung cancer therapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Cisplatino/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Pulmonares/patologia , Proteína Serina-Treonina Quinases de Interação com Receptores/genética , Fatores de Transcrição/metabolismo , Células A549 , Caspase 8/metabolismo , Proliferação de Células , Proteína de Domínio de Morte Associada a Fas/metabolismo , Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Prognóstico , Fatores de Transcrição/genética
3.
J Pathol ; 245(1): 41-52, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29431199

RESUMO

While the importance of protein N-glycosylation in cancer cell migration is well appreciated, the precise mechanisms by which N-acetylglucosaminyltransferase V (GnT-V) regulates cancer processes remain largely unknown. In the current study, we report that GnT-V-mediated N-glycosylation of CD147/basigin, a tumor-associated glycoprotein that carries ß1,6-N-acetylglucosamine (ß1,6-GlcNAc) glycans, is upregulated during TGF-ß1-induced epithelial-to-mesenchymal transition (EMT), which correlates with tumor metastasis in patients with hepatocellular carcinoma (HCC). Interruption of ß1,6-GlcNAc glycan modification of CD147/basigin decreased matrix metalloproteinase (MMP) expression in HCC cell lines and affected the interaction of CD147/basigin with integrin ß1. These results reveal that ß1,6-branched glycans modulate the biological function of CD147/basigin in HCC metastasis. Moreover, we showed that the PI3K/Akt pathway regulates GnT-V expression and that inhibition of GnT-V-mediated N-glycosylation suppressed PI3K signaling. In summary, ß1,6-branched N-glycosylation affects the biological function of CD147/basigin and these findings provide a novel approach for the development of therapeutic strategies targeting metastasis. © 2018 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Assuntos
Basigina/efeitos dos fármacos , Carcinoma Hepatocelular/metabolismo , Glicosilação/efeitos dos fármacos , N-Acetilglucosaminiltransferases/farmacologia , Fator de Crescimento Transformador beta1/efeitos dos fármacos , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/patologia , Metástase Neoplásica/patologia
4.
Exp Cell Res ; 357(1): 9-16, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28433697

RESUMO

Rab22a is a member of the Ras-related small GTPase family, which plays a key role in regulating the recycling of cargo proteins entering cells through clathrin-independent endocytosis (CIE). Rab22a is overexpressed in different cancer types, including liver cancer, malignant melanoma, ovarian cancer and osteosarcoma. However, its oncogenic role remains unknown. In this study, we found that silencing of Rab22a suppressed the migration and invasion of lung cancer cells. Furthermore, Rab22a interacts with CD147, and knockdown of Rab22a blocks CD147 recycling and promotes CD147 degradation. Taken together, our findings indicate that Rab22a enhances recycling of CD147, which is required for lung cancer cell migration and invasion,and targeting CD147 recycling may be a rational strategy for lung cancer therapy.


Assuntos
Basigina/metabolismo , Movimento Celular/fisiologia , Neoplasias Pulmonares/metabolismo , Proteínas rab de Ligação ao GTP/metabolismo , Linhagem Celular Tumoral , Endocitose , Endossomos/metabolismo , Humanos , Neoplasias Pulmonares/patologia , Transporte Proteico/fisiologia , Transferrina/metabolismo
5.
Int J Mol Sci ; 18(1)2017 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-28117675

RESUMO

Drug resistance remains a major clinical obstacle to successful treatment of cancer. As posttranslational modification is becoming widely recognized to affect the function of oncoproteins, targeting specific posttranslational protein modification provides an attractive strategy for anticancer drug development. CD147 is a transmembrane glycoprotein contributing to chemo-resistance of cancer cells in a variety of human malignancies. Ubiquitination is an important posttranslational modification mediating protein degradation. Degradation of oncoproteins, CD147 included, emerges as an attractive alternative for tumor inhibition. However, the ubiquitination of CD147 remains elusive. Here in this study, we found that deletion of the CD147 intracellular domain (CD147-ICD) prolonged the half-life of CD147 in HEK293T cells, and we identified that CD147-ICD interacts with FBXO22 using mass spectrometry and Western blot. Then, we demonstrated that FBXO22 mediates the polyubiquitination and degradation of CD147 by recognizing CD147-ICD. While knocking down of FBXO22 prolonged the half-life of CD147 in HEK293T cells, we found that FBXO22 regulates CD147 protein turnover in SMMC-7721, Huh-7 and A549 cells. Moreover, we found that the low level of FBXO22 contributes to the accumulation of CD147 and thereafter the cisplatin resistance of A549/DDP cells. To conclude, our study demonstrated that FBXO22 mediated the polyubiquitination and degradation of CD147 by interacting with CD147-ICD, and CD147 polyubiquitination by FBXO22 reversed cisplatin resistance of tumor cells.


Assuntos
Basigina/metabolismo , Cisplatino/farmacologia , Proteínas F-Box/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Ubiquitinação , Células A549 , Antineoplásicos/farmacologia , Basigina/genética , Sítios de Ligação/genética , Western Blotting , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Proteínas F-Box/genética , Deleção de Genes , Células HEK293 , Humanos , Espectrometria de Massas , Microscopia Confocal , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Poliubiquitina/metabolismo , Ligação Proteica , Proteólise , Interferência de RNA , Receptores Citoplasmáticos e Nucleares/genética
6.
Cancer Cell Int ; 16(1): 69, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27601938

RESUMO

BACKGROUND: The acquisition of inappropriate migratory feature is crucial for tumor metastasis. Rho-family GTPases including RhoA are molecular switches that play critical roles in regulating cell movement. We investigated the molecular mechanism underlying CD147 induced RhoA deactivation in hepatocellular carcinoma (HCC) cells. METHODS: Wound-healing assay was performed to study the cell motility. Analysis of RhoA activation in living cells was conducted using RhoA biosensor. Changes in the expression of certain genes were determined by quantitative real-time PCR. The expression of proteins was evaluated by Western blot. Cytoskeleton reorganization and focal adhesion formation were observed by immunofluorescence staining. Further investigation on the correlation between CD147 and p190-B RhoGAP (p190-B) in HCC tissues was performed by immunological histological chemistry analysis. RESULTS: CD147 promoted cell movement and suppressed RhoA activation. p190-B, a negative regulator of RhoA activity, was upregulated by CD147 at both mRNA and protein levels. This regulatory relationship was further confirmed by analyzing the expression pattern of CD147 and p190-B in human HCC tissues. Silencing of p190-B caused the increased formation of stress fiber and focal adhesion and blunted the impact of CD147 overexpression on cell movement, indicating that the regulatory effect of CD147 on cell movement is mediated, at least partially, by p190-B. CONCLUSIONS: These findings indicated that p190-B, a negative regulator of RhoA, is positively regulated by CD147 and contributes to the regulation of cell movement in HCC. CD147 plays critical roles in the motility of cancer cells and may be therefore a valuable drug target for anti-cancer therapy.

7.
Int J Mol Sci ; 17(11)2016 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-27834933

RESUMO

Hepatocellular carcinoma (HCC) is currently the third most common cause of cancer-related death in the Asia-Pacific region. Our previous work showed that knockdown of CD98 significantly inhibits malignant HCC cell phenotypes in vitro and in vivo. The level of CD98 in the membrane is tightly regulated to mediate complex processes associated with cell-cell communication and intracellular signaling. In addition, the intracellular domain of CD98 (CD98-ICD) seems to be of vital importance for recycling CD98 to the membrane after it is endocytosed. The intracellular and transmembrane domains of CD98 associate with ß-integrins (primarily ß1 but also ß3), and this association is essential for CD98 mediation of integrin-like signaling and complements dominant suppression of ß1-integrin. We speculated that isolated CD98-ICD would similarly suppress ß1-integrin activation and inhibit the malignant behaviors of cancer cells. In particular, the exact role of CD98-ICD has not been studied independently in HCC. In this study, we found that ectopic expression of CD98-ICD inhibited the malignant phenotypes of HCC cells, and the mechanism possibly involves ß1-integrin suppression. Moreover, the expression levels of CD98, ß1-integrin-A (the activated form of ß1-integrin) and Ki-67 were significantly increased in HCC tissues relative to those of normal liver tissues. Therefore, our preliminary study indicates that ectopic CD98-ICD has an inhibitory role in the malignant development of HCC, and shows that CD98-ICD acts as a dominant negative mutant of CD98 that attenuates ß1-integrin activation. CD98-ICD may emerge as a promising candidate for antitumor treatment.


Assuntos
Carcinoma Hepatocelular/metabolismo , Proteína-1 Reguladora de Fusão/metabolismo , Integrina beta1/metabolismo , Neoplasias Hepáticas/metabolismo , Animais , Sítios de Ligação/genética , Western Blotting , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Progressão da Doença , Citometria de Fluxo , Proteína-1 Reguladora de Fusão/genética , Humanos , Imuno-Histoquímica , Antígeno Ki-67/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , Microscopia Confocal , Transfecção , Transplante Heterólogo , Carga Tumoral
8.
Breast Cancer Res Treat ; 148(2): 423-36, 2014 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25348432

RESUMO

GINS2, a subunit of the GINS complex, is overexpressed in lung adenocarcinoma and metastatic breast tumor; however, its prognostic power and possible molecular mechanisms in breast cancer (BC) remain unclear. In this study, we aimed to explore the function of GINS2 in BC. The association between GINS2 transcript level and the clinical outcome of BC patients were estimated using Kaplan-Meier plots, multivariate cox regression analysis, forest plots, and receiver operating characteristics curves. Gene set enrichment analysis (GSEA) was performed to explore the mechanisms underlying the effects of the GINS2 transcript. High GINS2 transcript level was correlated with poor relapse free survival (log-rank P ≤ 0.001 in six cohorts; forest plot: total n = 1,420, total RR = 1.72, 95% CI 1.45-2.03; multivariate cox regression analysis: n = 906, HR 2.36, 95% CI 1.88-2.97), and distant metastasis free survival (log-rank P < 0.01 in 3 cohorts; forest plot: total n = 691, total RR 1.91, 95% CI 1.36-2.67; multivariate cox regression analysis: n = 442, HR 2.43, 95% CI 1.70-3.47). BC patients with higher GINS2 transcript levels showed poorer tamoxifen efficacy in a dose-dependent manner. GINS2 expression was significantly downregulated under mutated p53-depleted condition in MDA-468 and MDA-MB-231 cells, upregulated in mammary cancer stem cells (MaCSCs) (P = 0.003), and correlated with upregulated genes in mammary stem cells (GSEA: P < 0.01). Our study, for the first time, demonstrates that GINS2 is an independent prognostic marker and is associated with lung metastasis, histological grade, and endocrine therapy resistance in BC patients, which may attribute to mutant p53 and MaCSCs.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Proteínas Cromossômicas não Histona/genética , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias Pulmonares/genética , Células-Tronco Neoplásicas/metabolismo , Tamoxifeno/uso terapêutico , Antineoplásicos Hormonais/uso terapêutico , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/mortalidade , Neoplasias da Mama/patologia , Estudos de Coortes , Feminino , Seguimentos , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/mortalidade , Neoplasias Pulmonares/secundário , Metástase Linfática , Pessoa de Meia-Idade , Mutação/genética , Gradação de Tumores , Estadiamento de Neoplasias , Células-Tronco Neoplásicas/patologia , Análise de Sequência com Séries de Oligonucleotídeos , Prognóstico , RNA Mensageiro/genética , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Taxa de Sobrevida , Proteína Supressora de Tumor p53/genética
9.
Biochem J ; 449(2): 437-48, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23005037

RESUMO

Degradation of the basement membrane by MMPs (matrix metalloproteinases) is one of the most critical steps in tumour progression. CD147 is a tumour-associated antigen that plays a key regulatory role for MMP activities. In the present study, mass spectrum analysis demonstrated that the purified native CD147 from human lung cancer tissue was N-glycosylated and contained a series of high-mannose and complex-type N-linked glycan structures. Moreover, native glycosylated CD147 existed exclusively as oligomers in solution and directly stimulated MMP production more efficiently than non-glycosylated prokaryotic CD147. The glycosylation site mutation results indicated that, among three N-glycan attachment sites, the N152Q mutants were retained in the endoplasmic reticulum and unfolded protein response signalling was activated. This improper intracellular accumulation impaired its MMP-inducing activity. Increased ß1,6-branching of N-glycans as a result of overexpression of GnT-V (N-acetylglucosaminyltransferase V) plays an important role in tumour metastasis. In the present study, we identified CD147 as a target protein of GnT-V and found that overexpression of GnT-V resulted in an elevated level of CD147 at the plasma membrane and in cell-conditioned medium, thereby increasing the induction of MMPs. The present study reveals the important role of N-glycosylation of CD147 in its biological function and implied that targeting aberrant ß1,6-branching of N-glycans on CD147 would be valuable for the development of novel therapeutic modalities against carcinoma.


Assuntos
Basigina/metabolismo , Membrana Celular/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , N-Acetilglucosaminiltransferases/metabolismo , Basigina/química , Basigina/genética , Sítios de Ligação/genética , Western Blotting , Linhagem Celular Tumoral , Meios de Cultivo Condicionados/metabolismo , Retículo Endoplasmático/metabolismo , Técnicas de Silenciamento de Genes , Glicosilação , Células Hep G2 , Humanos , Neoplasias Pulmonares/metabolismo , Manose/química , Manose/metabolismo , Espectrometria de Massas , Metaloproteinases da Matriz/metabolismo , Microscopia Confocal , Mutação , N-Acetilglucosaminiltransferases/genética , Polissacarídeos/química , Polissacarídeos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Resposta a Proteínas não Dobradas
10.
Int J Mol Sci ; 15(4): 6356-77, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24739808

RESUMO

Glycosylation of glycoproteins is one of many molecular changes that accompany malignant transformation. Post-translational modifications of proteins are closely associated with the adhesion, invasion, and metastasis of tumor cells. CD147, a tumor-associated antigen that is highly expressed on the cell surface of various tumors, is a potential target for cancer diagnosis and therapy. A significant biochemical property of CD147 is its high level of glycosylation. Studies on the structure and function of CD147 glycosylation provide valuable clues to the development of targeted therapies for cancer. Here, we review current understanding of the glycosylation characteristics of CD147 and the glycosyltransferases involved in the biosynthesis of CD147 N-glycans. Finally, we discuss proteins regulating CD147 glycosylation and the biological functions of CD147 glycosylation.


Assuntos
Basigina/metabolismo , Basigina/química , Basigina/genética , Glicosilação , Glicosiltransferases/metabolismo , Humanos , Metaloproteinases da Matriz/metabolismo , Mutagênese Sítio-Dirigida , Neoplasias/metabolismo , Neoplasias/patologia
11.
MedComm (2020) ; 5(3): e512, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38469549

RESUMO

Therapeutic antibodies (Abs) improve the clinical outcome of cancer patients. However, on-target off-tumor toxicity limits Ab-based therapeutics. Cluster of differentiation 147 (CD147) is a tumor-associated membrane antigen overexpressed in cancer cells. Ab-based drugs targeting CD147 have achieved inadequate clinical benefits for liver cancer due to side effects. Here, by using glycoengineering and hypoxia-activation strategies, we developed a conditional Ab-dependent cellular cytotoxicity (ADCC)-enhanced humanized anti-CD147 Ab, HcHAb18-azo-PEG5000 (HAP18). Afucosylated ADCC-enhanced HcHAb18 Ab was produced by a fed-batch cell culture system. Azobenzene (Azo)-linked PEG5000 conjugation endowed HAP18 Ab with features of hypoxia-responsive delivery and selective targeting. HAP18 Ab potently inhibits the migration, invasion, and matrix metalloproteinase secretion, triggers the cytotoxicity and apoptosis of cancer cells, and induces ADCC, complement-dependent cytotoxicity, and Ab-dependent cellular phagocytosis under hypoxia. In xenograft mouse models, HAP18 Ab selectively targets hypoxic liver cancer tissues but not normal organs or tissues, and has potent tumor-inhibiting effects. HAP18 Ab caused negligible side effects and exhibited superior pharmacokinetics compared to those of parent HcHAb18 Ab. The hypoxia-activated ADCC-enhanced humanized HAP18 Ab safely confers therapeutic efficacy against liver cancer with improved selectivity. This study highlights that hypoxia activation is a promising strategy for improving the tumor targeting potential of anti-CD147 Ab drugs.

12.
J Biol Chem ; 287(7): 4759-72, 2012 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-22130661

RESUMO

Several lines of evidence suggest that HAb18G/CD147 interacts with the integrin variants α3ß1 and α6ß1. However, the mechanism of the interaction remains largely unknown. In this study, mammalian protein-protein interaction trap (MAPPIT), a mammalian two-hybrid method, was used to study the CD147-integrin ß1 subunit interaction. CD147 in human hepatocellular carcinoma (HCC) cells was interfered with by small hairpin RNA. Nude mouse xenograft model and metastatic model of HCC were used to detect the role of CD147 in carcinogenesis and metastasis. We found that the extracellular membrane-proximal domain of HAb18G/CD147 (I-type domain) binds at the metal ion-dependent adhesion site in the ßA domain of the integrin ß1 subunit, and Asp(179) in the I-type domain of HAb18G/CD147 plays an important role in the interaction. The levels of the proteins that act downstream of integrin, including focal adhesion kinase (FAK) and phospho-FAK, were decreased, and the cytoskeletal structures of HCC cells were rearranged bearing the HAb18G/CD147 deletion. Simultaneously, the migration and invasion capacities, secretion of matrix metalloproteinases, colony formation rate in vitro, and tumor growth and metastatic potential in vivo were decreased. These results indicate that the interaction of HAb18G/CD147 extracellular I-type domain with the integrin ß1 metal ion-dependent adhesion site motif activates the downstream FAK signaling pathway, subsequently enhancing the malignant properties of HCC cells.


Assuntos
Basigina/metabolismo , Carcinoma Hepatocelular/metabolismo , Movimento Celular , Integrina beta1/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Neoplasias/metabolismo , Transdução de Sinais , Motivos de Aminoácidos , Animais , Basigina/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Adesão Celular , Colagenases , Modelos Animais de Doenças , Quinase 1 de Adesão Focal/genética , Quinase 1 de Adesão Focal/metabolismo , Humanos , Integrina beta1/genética , Íons/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Metais/metabolismo , Camundongos , Camundongos Nus , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Estrutura Terciária de Proteína , Transplante Heterólogo
13.
Cell Biol Int ; 37(7): 643-52, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23733271

RESUMO

Calpains are a conserved family of calcium-dependent cysteine proteinases involved in various cellular functions. Two ubiquitous isoforms, µ- and m-calpain, are key members of the calpain family that play essential roles in regulating cell migration and invasion. However, it remains unclear whether they are involved in the progression of hepatocellular carcinoma (HCC). Here, we investigated the functions of µ- and m-calpain in the invasive and metastatic processes of human hepatoma cells. Our results indicated that the expression levels of calpains were elevated in HCC cells compared with those in normal hepatic cells. Our results indicated that small interfering RNA (siRNA)-mediated silencing of µ- and m-calpain expressions significantly suppressed the adhesive, migrative and invasive potentials of human hepatoma cells. The matrix metalloproteinases (MMPs) are key regulators of malignant tumour invasion and metastasis. siRNA-mediated down-regulation of µ- and m-calpain expressions also significantly attenuated MMP-2 and MMP-9 secretion. Thus µ- and m-calpain may play important roles in the invasion and metastasis of human hepatoma cells, and calpains may be drug targets for preventing HCC metastasis.


Assuntos
Calpaína/metabolismo , Calpaína/antagonistas & inibidores , Calpaína/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Adesão Celular , Linhagem Celular , Movimento Celular , Regulação para Baixo , Células Hep G2 , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo
14.
Cell Death Discov ; 9(1): 406, 2023 Nov 02.
Artigo em Inglês | MEDLINE | ID: mdl-37919290

RESUMO

Lung cancer is the leading cause of cancer-related mortality worldwide. CNOT3, a subunit of the CCR4-NOT complex, has recently been suggested to be overexpressed in lung cancer and involved in tumor malignancy. However, its precise role and the underlying mechanisms still need to be fully revealed. In the present study, we found in lung cancer cells the expression of CNOT3 could be regulated by EGFR signaling pathway and c-Jun, a transcription factor downstream of EGFR, transcriptionally regulated its expression. Interestingly, CNOT3 could inversely regulate the expression of c-Jun via modulating its translation. Thus, a feedback loop existed between c-Jun and CNOT3. CNOT3 reduction post EGFR blockade facilitated the drug-induced cell death, and simultaneously inhibited cell proliferation via impacting TSC1/mTOR axis. Whereas, further up-regulation of the CNOT3 expression was observed in gefitinib-resistant cells, which dampened gefitinib sensitivity. Mechanically, the elevation of CNOT3 was induced by the bypass activation of HER2/c-Jun signaling. Depleting CNOT3 in vitro and in vivo sensitized the drug-resistant cells to gefitinib treatment and inhibited metastatic progression. These results give novel insights into the role of CNOT3 in lung cancer malignancy and provide a theoretical basis for the development of therapeutic strategies to solve acquired resistance to EGFR-TKIs.

15.
J Pharm Anal ; 13(10): 1135-1152, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-38024852

RESUMO

Morphine is a frequently used analgesic that activates the mu-opioid receptor (MOR), which has prominent side effects of tolerance. Although the inefficiency of morphine in inducing the endocytosis of MOR underlies the development of morphine tolerance, currently, there is no effective therapy to treat morphine tolerance. In the current study, we aimed to develop a monoclonal antibody (mAb) precisely targeting MOR and to determine its therapeutic efficacy on morphine tolerance and the underlying molecular mechanisms. We successfully prepared a mAb targeting MOR, named 3A5C7, by hybridoma technique using a strategy of deoxyribonucleic acid immunization combined with cell immunization, and identified it as an immunoglobulin G mAb with high specificity and affinity for MOR and binding ability to antigens with spatial conformation. Treatment of two cell lines, HEK293T and SH-SY5Y, with 3A5C7 enhanced morphine-induced MOR endocytosis via a G protein-coupled receptor kinase 2 (GRK2)/ß-arrestin2-dependent mechanism, as demonstrated by immunofluorescence staining, flow cytometry, Western blotting, coimmunoprecipitation, and small interfering ribonucleic acid (siRNA)-based knockdown. This mAb also allowed MOR recycling from cytoplasm to plasma membrane and attenuated morphine-induced phosphorylation of MOR. We established an in vitro morphine tolerance model using differentiated SH-SY5Y cells induced by retinoic acid. Western blot, enzyme-linked immunosorbent assays, and siRNA-based knockdown revealed that 3A5C7 mAb diminished hyperactivation of adenylate cyclase, the in vitro biomarker of morphine tolerance, via the GRK2/ß-arrestin2 pathway. Furthermore, in vivo hotplate test demonstrated that chronic intrathecal administration of 3A5C7 significantly alleviated morphine tolerance in mice, and withdrawal jumping test revealed that both chronic and acute 3A5C7 intrathecal administration attenuated morphine dependence. Finally, intrathecal electroporation of silencing short hairpin RNA illustrated that the in vivo anti-tolerance and anti-dependence efficacy of 3A5C7 was mediated by enhanced morphine-induced MOR endocytosis via GRK2/ß-arrestin2 pathway. Collectively, our study provided a therapeutic mAb, 3A5C7, targeting MOR to treat morphine tolerance, mediated by enhancing morphine-induced MOR endocytosis. The mAb 3A5C7 demonstrates promising translational value to treat clinical morphine tolerance.

16.
J Biol Chem ; 286(10): 8197-8203, 2011 Mar 11.
Artigo em Inglês | MEDLINE | ID: mdl-21245143

RESUMO

Cyclophilin A (CyPA) is a ubiquitously distributed peptidylprolyl cis-trans isomerase (PPIase) that possesses diverse biological functions. Extracellular CyPA is a potent chemokine, which can directly induce leukocyte chemotaxis and contribute to the pathogenesis of inflammation-mediated diseases. Although it has been identified that the chemotaxis activity of CyPA is mediated through its cell surface signaling receptor CD147, the role of CyPA PPIase activity in this process is disputable, and the underlying molecular mechanism is still poorly understood. In this study, we present the first evidence that CyPA induces leukocyte chemotaxis through a direct binding with the ectodomain of CD147 (CD147(ECT)), independent of its PPIase activity. Although NMR study indicates that the CD147(ECT) binding site on CyPA overlaps with the PPIase active site, the PPIase inactive mutant CyPA(R55A) exhibits similar CD147(ECT) binding ability and chemotaxis activity to those of CyPA(WT). Furthermore, we have identified three key residues of CyPA involved in CD147(ECT) binding and found that mutations H70A, T107A, and R69A result in similar levels of reduction in CD147(ECT) binding ability and chemotaxis activity for CyPA, without affecting the PPIase activity. Our findings indicate that there exists a novel mechanism for CyPA to regulate cellular signaling processes, shedding new light on its applications in drug development and providing a new targeting site for drug design.


Assuntos
Basigina/metabolismo , Quimiotaxia/fisiologia , Ciclofilina A/metabolismo , Leucócitos/metabolismo , Substituição de Aminoácidos , Basigina/química , Basigina/genética , Sítios de Ligação , Ciclofilina A/química , Ciclofilina A/genética , Células HL-60 , Humanos , Mutação de Sentido Incorreto , Ressonância Magnética Nuclear Biomolecular , Ligação Proteica
17.
Biochem Biophys Res Commun ; 419(3): 517-22, 2012 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-22366034

RESUMO

HAb18G/CD147 is a transmembrane glycoprotein of the immunoglobulin superfamily (IgSF) and is reported to be correlated with invasion and metastasis of many cancers. The crystal structure of HAb18G/CD147 ectodomain has shown that it can form homodimers in crystal. However, the functional significance of HAb18G/CD147 dimerization remains unclear. In the present study, guided by the crystal structure, we performed extensive mutational and functional studies to identify residues critical for dimerization and molecular function of HAb18G/CD147. Fourteen mutants were purified and evaluated for their ability to form dimers in solution and in living cells. Subsequent functional validation revealed that K63E and S193A mutants, which disrupted CD147 dimerization both in solution and in living cells, showed clearly dominant-negative effects on MAPK activation, MMP2 induction and invasiveness in tumor cells. Taken together, the present study provides mutational and functional evidences demonstrating for the first time the functional importance of CD147 dimerization and its direct correlation with invasion and metastasis of tumor cells.


Assuntos
Basigina/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Invasividade Neoplásica , Basigina/genética , Linhagem Celular Tumoral , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Mutação , Multimerização Proteica
18.
Hepatology ; 54(6): 2012-24, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21809360

RESUMO

UNLABELLED: Tumor cells can move as individual cells in two interconvertible modes: mesenchymal mode and amoeboid mode. Cytoskeleton rearrangement plays an important role in the interconversion. Previously, we reported that HAb18G/CD147 and annexin II are interacting proteins involved in cytoskeleton rearrangement, yet the role of their interaction is unclear. In this study we found that the depletion of HAb18G/CD147 produced a rounded morphology, which is associated with amoeboid movement, whereas the depletion of annexin II resulted in an elongated morphology, which is associated with mesenchymal movement. The extracellular portion of HAb18G/CD147 can interact with a phosphorylation-inactive mutant of annexin II and inhibit its phosphorylation. HAb18G/CD147 inhibits Rho signaling pathways and amoeboid movement by inhibiting annexin II phosphorylation, promotes membrane localization of WAVE2 and Rac1 activation by way of the integrin-FAK-PI3K/PIP3 signaling pathway, and promotes the formation of lamellipodia and mesenchymal movement. CONCLUSION: These results suggest that the interaction of HAb18G/CD147 with annexin II is involved in the interconversion between mesenchymal and amoeboid movement of hepatocellular carcinoma cells.


Assuntos
Anexina A2/metabolismo , Basigina/fisiologia , Carcinoma Hepatocelular/fisiopatologia , Movimento Celular/imunologia , Neoplasias Hepáticas/fisiopatologia , Transdução de Sinais/fisiologia , Proteínas rac1 de Ligação ao GTP/fisiologia , Proteína rhoA de Ligação ao GTP/fisiologia , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Fosforilação , Transdução de Sinais/efeitos dos fármacos , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
19.
Cancer Commun (Lond) ; 42(8): 750-767, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35716012

RESUMO

BACKGROUND: The mechanism underlying colorectal cancer (CRC) initiation and progression remains elusive, and overall survival is far from satisfactory. Previous studies have shown that PDGFA-associated protein 1 (PDAP1) is upregulated in several cancers including CRC. Here, we aimed to identify the cause and consequence of PDAP1 dysregulation in CRC and evaluate its role as a potential therapeutic target. METHODS: Multi-omics data analysis was performed to identify potential key players in CRC initiation and progression. Immunohistochemistry (IHC) staining was applied to determine the expression pattern of PDAP1 in CRC tissues. Pdap1 conditional knockout mice were used to establish colitis and CRC mouse models. RNA sequencing, a phosphoprotein antibody array, western blotting, histological analysis, 5-bromo-2'-deoxyuridine (BrdU) incorporation assay, and interactome analysis were applied to identify the underlying mechanisms of PDAP1. A human patient-derived xenograft (PDX) model was used to assess the potential of PDAP1 as a therapeutic target. RESULTS: PDAP1 was identified as a potential key player in CRC development using multi-omics data analysis. PDAP1 was overexpressed in CRC cells and correlated with reduced overall survival. Further investigation showed that PDAP1 was critical for the regulation of cell proliferation, migration, invasion, and metastasis. Significantly, depletion of Pdap1 in intestinal epithelial cells impaired mucosal restitution in dextran sulfate sodium salt-induced colitis and inhibited tumor initiation and growth in colitis-associated cancers. Mechanistic studies showed that c-Myc directly transactivated PDAP1, which contributed to the high PDAP1 expression in CRC cells. PDAP1 interacted with the juxtamembrane domain of epidermal growth factor receptor (EGFR) and facilitated EGFR-mitogen-activated protein kinase (MAPK) signaling activation, which resulted in FOS-related antigen 1 (FRA-1) expression, thereby facilitating CRC progression. Notably, silencing of PDAP1 could hinder the growth of patient-derived xenografts that sustain high PDAP1 levels. CONCLUSIONS: PDAP1 facilitates mucosal restitution and carcinogenesis in colitis-associated cancer. c-Myc-driven upregulation of PDAP1 promotes proliferation, migration, invasion, and metastasis of CRC cells via the EGFR-MAPK-FRA-1 signaling axis. These findings indicated that PDAP1 inhibition is warranted for CRC patients with PDAP1 overexpression.


Assuntos
Colite , Neoplasias Colorretais , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Animais , Proliferação de Células , Colite/induzido quimicamente , Colite/complicações , Colite/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Camundongos
20.
Cell Biol Int ; 35(8): 811-7, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21395558

RESUMO

ßig-h3 is a TGF-ß (transforming growth factor ß)-induced ECM (extracellular matrix) protein that induces the secretion of MMPs (matrix metalloproteinases). However, the mechanism of induction is yet to be established. In this study, siRNAs (small interfering RNAs) targeted against ßig-h3 were transfected into SMMC-7721 cells [a HCC (human hepatocellular carcinoma) cell line] to knockdown the expression of ßig-h3. We found that NiCl2, a potent blocker of extracellular Ca2+ entry, reduced ßig-h3-induced secretion of MMP-2 and -9. Further investigation suggested that reduction in the levels of ßig-h3 decreased the secretion of MMP-2 and -9 that was enhanced by an increase in the concentration of extracellular Ca2+. SNAP (S-nitroso-N-acetylpenicillamine), a NO (nitric oxide) donor, and 8-Br-cGMP (8-bromo-cGMP) inhibited thapsigargin-induced Ca2+ entry and MMP secretion in the invasive potential of human SMMC-7721 cells. Further, the inhibitory effects of 8-Br-cGMP and SNAP could be significantly enhanced by down-regulating ßig-h3. ßig-h3 attenuates the negative regulation of NO/cGMP-sensitive store-operated Ca2+ entry. Our findings suggest that the expression of ßig-h3 might play an important role in the regulation of store-operated Ca2+ entry to increase the invasive potential of HCC cells.


Assuntos
Cálcio/metabolismo , Carcinoma Hepatocelular/metabolismo , Proteínas da Matriz Extracelular/genética , Proteínas da Matriz Extracelular/metabolismo , Neoplasias Hepáticas/metabolismo , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , GMP Cíclico/análogos & derivados , GMP Cíclico/farmacologia , Imunofluorescência , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Invasividade Neoplásica , Níquel/farmacologia , Interferência de RNA , RNA Interferente Pequeno , Reação em Cadeia da Polimerase Via Transcriptase Reversa , S-Nitroso-N-Acetilpenicilamina/metabolismo , Tapsigargina/farmacologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA