Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Oncogene ; 42(12): 861-868, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36788350

RESUMO

Experimental studies on immunoglobulin superfamily (IgSF) member EWI2 reveal that it suppresses a variety of solid malignant tumors including brain, lung, skin, and prostate cancers in animal models and inhibits tumor cell movement and growth in vitro. While EWI2 appears to support myeloid leukemia in mouse models and maintain leukemia stem cells. Bioinformatics analyses suggest that EWI2 gene expression is downregulated in glioblastoma but upregulated in melanoma, pancreatic cancer, and liver cancer. The mechanism of action for EWI2 is linked to its inhibition of growth factor receptors and cell adhesion proteins through its associated tetraspanin-enriched membrane domains (TEMDs), by altering the cell surface clustering and endolysosome trafficking/turnover of these transmembrane proteins. Recent studies also show that EWI2 modulates the nuclear translocation of ERK and TFEB to change the activities of these gene expression regulators. For EWI2 relatives including FPRP, IgSF3, and CD101, although their roles in malignant diseases are not fully clear and remain to be determined experimentally, FPRP and IgSF3 likely promote the progression of solid malignant tumors while CD101 seems to modulate immune cells of tumor microenvironment. Distinctive from other tumor regulators, the impacts of EWI subfamily members on solid malignant tumors are likely to be context dependent. In other words, the effect of a given EWI subfamily member on a tumor probably depends on the molecular network and composition of TEMDs in that tumor. Collectively, EWI2 and its relatives are emerged as important regulators of malignant diseases with promising potentials to become anti-cancer therapeutics and cancer therapy targets.


Assuntos
Antígenos CD , Proteínas de Membrana , Neoplasias , Animais , Humanos , Masculino , Camundongos , Imunoglobulinas/genética , Melanoma , Proteínas de Membrana/metabolismo , Neoplasias/metabolismo , Neoplasias da Próstata , Tetraspaninas/genética , Microambiente Tumoral , Antígenos CD/metabolismo
2.
Cell Mol Life Sci ; 79(7): 389, 2022 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-35773608

RESUMO

EWI2 is a transmembrane immunoglobulin superfamily (IgSF) protein that physically associates with tetraspanins and integrins. It inhibits cancer cells by influencing the interactions among membrane molecules including the tetraspanins and integrins. The present study revealed that, upon EWI2 silencing or ablation, the elevated movement and proliferation of cancer cells in vitro and increased cancer metastatic potential and malignancy in vivo are associated with (i) increases in clustering, endocytosis, and then activation of EGFR and (ii) enhancement of Erk MAP kinase signaling. These changes in signaling make cancer cells (i) undergo partial epithelial-to-mesenchymal (EMT) for more tumor progression and (ii) proliferate faster for better tumor formation. Inhibition of EGFR or Erk kinase can abrogate the cancer cell phenotypes resulting from EWI2 removal. Thus, to inhibit cancer cells, EWI2 prevents EGFR from clustering and endocytosis to restrain its activation and signaling.


Assuntos
Antígenos CD , Endocitose , Receptores ErbB , Proteínas de Membrana , Neoplasias , Antígenos CD/metabolismo , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Transição Epitelial-Mesenquimal , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Integrinas/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia
3.
Cancer Lett ; 536: 215641, 2022 06 28.
Artigo em Inglês | MEDLINE | ID: mdl-35339615

RESUMO

As a partner of tetraspanins, EWI2 suppresses glioblastoma, melanoma, and prostate cancer; but its role in lung cancer has not been investigated. Bioinformatics analysis reveals that EWI2 gene expression is up regulated in lung adenocarcinoma and higher expression of EWI2 mRNA may predict poorer overall survival. However, experimental analysis shows that EWI2 protein is actually downregulated constantly in the tissues of lung adenocarcinoma and lung squamous cell carcinoma. Forced expression of EWI2 in human lung adenocarcinoma cells reduces total cellular and cell surface levels of various integrins and growth factor receptors, which initiates the outside-in motogenic and mitogenic signaling. These reductions result in the decreases in 1) cell-matrix adhesion, cell movement, and cell transformation in vitro and 2) tumor growth, burden, and metastasis in vivo, and result from the increases in lysosomal trafficking and proteolytic degradation of theses membrane receptors. EWI2 elevates lysosome formation by promoting nuclear retention of TFEB, the master transcription factor driving lysosomogenesis. In conclusion, EWI2 as a lung cancer suppressor attenuates lung cancer cells in a comprehensive fashion by inhibiting both tumor growth and tumor metastasis; EWI2 as an endolysosome regulator promotes lysosome activity to enhance lysosomal degradation of growth factor receptors and integrins and then reduce their levels and functions; and EWI2 can become a promising therapeutic candidate given its accessibility at the cell surface, dual inhibition on growth factor receptors and integrins, and broad-spectrum anti-cancer activity. More importantly, our observations also provide a novel therapeutic strategy to bypass the resistance to EGFR inhibitors.


Assuntos
Adenocarcinoma de Pulmão , Antígenos CD/metabolismo , Neoplasias Pulmonares , Proteínas de Membrana/metabolismo , Adenocarcinoma de Pulmão/genética , Adenocarcinoma de Pulmão/metabolismo , Linhagem Celular Tumoral , Humanos , Integrinas/genética , Integrinas/metabolismo , Neoplasias Pulmonares/metabolismo , Lisossomos/metabolismo , Masculino , Receptores de Fatores de Crescimento/metabolismo
4.
J Extracell Vesicles ; 9(1): 1692417, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31807237

RESUMO

Tumour metastasis suppressor KAI1/CD82 inhibits tumour cell movement. As a transmembrane protein, tetraspanin CD82 bridges the interactions between membrane microdomains of lipid rafts and tetraspanin-enriched microdomains (TEMs). In this study, we found that CD82 and other tetraspanins contain cholesterol recognition/interaction amino-acid consensus (CRAC) sequences in their transmembrane domains and revealed that cholesterol binding of CD82 determines its interaction with lipid rafts but not with TEMs. Functionally, CD82 needs cholesterol binding to inhibit solitary migration, collective migration, invasion and infiltrative outgrowth of tumour cells. Importantly, CD82-cholesterol/-lipid raft interaction not only promotes extracellular release of lipid raft components such as cholesterol and gangliosides but also facilitates extracellular vesicle (EV)-mediated release of ezrin-radixin-moesin (ERM) protein Ezrin. Since ERM proteins link actin cytoskeleton to the plasma membrane, we show for the first time that cell movement can be regulated by EV-mediated releases, which disengage the plasma membrane from cytoskeleton and then impair cell movement. Our findings also conceptualize that interactions between membrane domains, in this case converge of lipid rafts and TEMs by CD82, can change cell movement. Moreover, CD82 coalescences with both lipid rafts and TEMs are essential for its inhibition of tumour cell movement and for its enhancement of EV release. Finally, our study underpins that tetraspanins as a superfamily of functionally versatile molecules are cholesterol-binding proteins. Abbreviations: Ab: antibody; CBM: cholesterol-binding motif; CCM: cholesterol consensus motif; CRAC/CARC: cholesterol recognition or interaction amino-acid consensus; CTxB: cholera toxin B subunit; ECM: extracellular matrix; ERM: ezrin, radixin and moesin; EV: extracellular vesicles; FBS: foetal bovine serum; mAb: monoclonal antibody; MST: microscale thermophoresis; pAb: polyclonal antibody; and TEM: tetraspanin-enriched microdomain.

5.
Geroscience ; 41(2): 209-227, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-31037472

RESUMO

There is significant overlap between the cellular and molecular mechanisms of aging and pathways contributing to carcinogenesis, including the role of genome maintenance pathways. In the field of geroscience analysis of novel genetic mouse models with either a shortened, or an extended, lifespan provides a unique opportunity to evaluate the synergistic roles of longevity assurance pathways in cancer resistance and regulation of lifespan and to develop novel targets for interventions that both delay aging and prevent carcinogenesis. There is a growing need for robust assays to assess the susceptibility of cancer in these models. The present review focuses on a well-characterized method frequently used in cancer research, which can be adapted to study resilience to genotoxic stress and susceptibility to genotoxic stress-induced carcinogenesis in geroscience research namely, chemical carcinogenesis induced by treatment with 7,12-dimethylbenz(a)anthracene (DMBA). Recent progress in understanding how longer-living mice may achieve resistance to chemical carcinogenesis and how these pathways are modulated by anti-aging interventions is reviewed. Strain-specific differences in sensitivity to DMBA-induced carcinogenesis are also explored and contrasted with mouse lifespan. The clinical relevance of inhibition of DMBA-induced carcinogenesis for the pathogenesis of mammary adenocarcinomas in older human subjects is discussed. Finally, the potential role of insulin-like growth factor-1 (IGF-1) in the regulation of pathways responsible for cellular resilience to DMBA-induced mutagenesis is discussed.


Assuntos
9,10-Dimetil-1,2-benzantraceno/farmacologia , Envelhecimento/genética , Dano ao DNA , Neoplasias Mamárias Experimentais/induzido quimicamente , Animais , Carcinogênese/genética , Modelos Animais de Doenças , Feminino , Geriatria , Humanos , Longevidade/genética , Masculino , Neoplasias Mamárias Experimentais/genética , Camundongos , Ratos Sprague-Dawley , Pesquisa , Roedores
6.
Cell Mol Life Sci ; 75(18): 3423-3439, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29589089

RESUMO

Tetraspanins co-emerged with multi-cellular organisms during evolution and are typically localized at the cell­cell interface, [corrected] and form tetraspanin-enriched microdomains (TEMs) by associating with each other and other membrane molecules. Tetraspanins affect various biological functions, but how tetraspanins engage in multi-faceted functions at the cellular level is largely unknown. When cells interact, the membrane microextrusions at the cell-cell interfaces form dynamic, digit-like structures between cells, which we term digitation junctions (DJs). We found that (1) tetraspanins CD9, CD81, and CD82 and (2) TEM-associated molecules integrin α3ß1, CD44, EWI2/PGRL, and PI-4P are present in DJs of epithelial, endothelial, and cancer cells. Tetraspanins and their associated molecules also regulate the formation and development of DJs. Moreover, (1) actin cytoskeleton, RhoA, and actomyosin activities and (2) growth factor receptor-Src-MAP kinase signaling, but not PI-3 kinase, regulate DJs. Finally, we showed that DJs consist of various forms in different cells. Thus, DJs are common, interactive structures between cells, and likely affect cell adhesion, migration, and communication. TEMs probably modulate various cell functions through DJs. Our findings highlight that DJ morphogenesis reflects the transition between cell-matrix adhesion and cell-cell adhesion and involves both cell-cell and cell-matrix adhesion molecules.


Assuntos
Junções Intercelulares/metabolismo , Tetraspaninas/metabolismo , Citoesqueleto de Actina/efeitos dos fármacos , Actinas/metabolismo , Animais , Antígenos CD/genética , Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Citocalasina D/farmacologia , Cães , Humanos , Células Madin Darby de Rim Canino , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Microscopia Confocal , Tetraspaninas/química
7.
Oncotarget ; 9(18): 14382-14396, 2018 Mar 06.
Artigo em Inglês | MEDLINE | ID: mdl-29581851

RESUMO

Heart failure (HF) is the final common pathway of various cardiovascular diseases. Although it is well documented that reduction of cardiac angiogenesis contributes to the progression from adaptive cardiac hypertrophy to HF, the molecular mechanisms remain unknown. In the present study, we found that cardiac expression of miR-124 was increased in patients and mice with HF. Recombinant adeno-associated virus (rAAV)-mediated miR-124 over-expression aggravated angiotensin II (Ang II) infusion-induced cardiac dysfunction and abnormal cardiac angiogenesis in mice. In vitro, transfection of miR-124 mimics significantly promoted apoptosis and reduced viability, migration, tube formation, and nitric oxide release in endothelial cells. In addition, CD151 was identified as a direct target of miR-124. Endothelial cell injury caused by CD151 silencing was mimicked by miR-124 over-expression. Re-expression of CD151 attenuated miR-124-mediated suppression of cardiac angiogenesis and cardiac dysfunction in Ang II-treated mice. Our observations suggest that miR-124 is an important negative regulator of cardiac angiogenesis and cardiac function, likely by suppressing the expression of CD151 in heart cells. Modulation of miR-124 levels may provide new strategies and targets for HF therapy.

8.
Oncotarget ; 8(4): 6496-6512, 2017 Jan 24.
Artigo em Inglês | MEDLINE | ID: mdl-28030805

RESUMO

Tissue inhibitor of metalloproteinases-1 (TIMP-1) is a widely secreted protein that regulates cell motility, proliferation, and apoptosis. Although it is recognized that TIMP-1-tetraspanin CD63 regulates epithelial cell apoptosis and proliferation, how TIMP-1 controls cell motility is not well understood. In this study, we identify tetraspanin CD82 (also called KAI1) as a component of the promiscuous TIMP-1 interacting protein complex on cell surface of human pancreatic adenocarcinoma cells. CD82 directly binds to TIMP-1 N-terminal region through its large extracellular loop and co-localizes with TIMP-1 in both cancer cell lines and clinical samples. Moreover, CD82 facilitates membrane-bound TIMP-1 endocytosis, which significantly contributes to the anti-migration effect of TIMP-1. CD82 silencing partially eliminates these functions. TIMP-1 and CD82 expression status in patients with pancreatic ductal adenocarcinoma (PDAC) might demonstrate future usefulness as a differentiation marker and give us new insight into tumorigenic metastatic potential.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Proteína Kangai-1/metabolismo , Neoplasias Pancreáticas/metabolismo , Inibidor Tecidual de Metaloproteinase-1/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Biomarcadores Tumorais/química , Biomarcadores Tumorais/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/secundário , Membrana Celular/metabolismo , Movimento Celular , Endocitose , Feminino , Humanos , Proteína Kangai-1/química , Proteína Kangai-1/genética , Células MCF-7 , Masculino , Pessoa de Meia-Idade , Simulação de Acoplamento Molecular , Invasividade Neoplásica , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Ligação Proteica , Domínios e Motivos de Interação entre Proteínas , Transporte Proteico , Interferência de RNA , Fatores de Tempo , Inibidor Tecidual de Metaloproteinase-1/química , Inibidor Tecidual de Metaloproteinase-1/genética , Transfecção
9.
Oncotarget ; 7(15): 20966-80, 2016 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-26980709

RESUMO

Drug-induced aberrant DNA methylation is the first identified epigenetic marker involved in chemotherapy resistance. Understanding how the aberrant DNA methylation is acquired would impact cancer treatment in theory and practice. In this study we systematically investigated whether and how ERα propelled aberrant global DNA hypermethylation in the context of breast cancer drug resistance. Our data demonstrated that anticancer drug paclitaxel (PTX) augmented ERα binding to the DNMT1 and DNMT3b promoters to activate DNMT1 and DNMT3b genes, enhancing the PTX resistance of breast cancer cells. In support of these observations, estrogen enhanced multi-drug resistance of breast cancer cells by up-regulation of DNMT1 and DNMT3b genes. Nevertheless, the aberrant global DNA hypermethylation was dominantly induced by ERα-activated-DNMT1, since DNMT1 over-expression significantly increased global DNA methylation and DNMT1 knockdown reversed the ERα-induced global DNA methylation. Altering DNMT3b expression had no detectable effect on global DNA methylation. Consistently, the expression level of DNMT1 was positively correlated with ERα in 78 breast cancer tissue samples shown by our immunohistochemistry (IHC) analysis and negatively correlated with relapse-free survival (RFS) and distance metastasis-free survival (DMFS) of ERα-positive breast cancer patients. This study provides a new perspective for understanding the mechanism underlying drug-resistance-facilitating aberrant DNA methylation in breast cancer and other estrogen dependent tumors.


Assuntos
Neoplasias da Mama/patologia , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Metilação de DNA/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Receptor alfa de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Recidiva Local de Neoplasia/patologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Biomarcadores Tumorais/genética , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/genética , Proliferação de Células/efeitos dos fármacos , DNA (Citosina-5-)-Metiltransferase 1/genética , Epigenômica , Receptor alfa de Estrogênio/genética , Feminino , Humanos , Metástase Linfática , Invasividade Neoplásica , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/genética , Paclitaxel/farmacologia , Prognóstico , Regiões Promotoras Genéticas/genética , Taxa de Sobrevida , Células Tumorais Cultivadas
10.
Cancer Metastasis Rev ; 34(4): 619-33, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26335499

RESUMO

Tetraspanin CD82 suppresses the progression and metastasis of a wide range of solid malignant tumors. However, its roles in tumorigenesis and hematopoietic malignancy remain unclear. Ubiquitously expressed CD82 restrains cell migration and cell invasion by modulating both cell-matrix and cell-cell adhesiveness and confining outside-in pro-motility signaling. This restraint at least contributes to, if not determines, the metastasis-suppressive activity and, also likely, the physiological functions of CD82. As a modulator of cell membrane heterogeneity, CD82 alters microdomains, trafficking, and topography of the membrane by changing the membrane molecular landscape. The functional activities of membrane molecules and the cytoskeletal interaction of the cell membrane are subsequently altered, followed by changes in cellular functions. Given its pathological and physiological importance, CD82 is a promising candidate for clinically predicting and blocking tumor progression and metastasis and also an emerging model protein for mechanistically understanding cell membrane organization and heterogeneity.


Assuntos
Adesão Celular/genética , Genes Supressores de Tumor , Proteína Kangai-1/genética , Microdomínios da Membrana/metabolismo , Invasividade Neoplásica/genética , Neoplasias/patologia , Movimento Celular/genética , Junções Célula-Matriz/genética , Citoesqueleto , Humanos , Proteína Kangai-1/biossíntese , Transdução de Sinais/genética
11.
Biochim Biophys Acta ; 1850(6): 1261-6, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25727396

RESUMO

BACKGROUND: Physical cues of cellular environment affect cell fate and differentiation. For example, an environment with high stiffness drives mesenchymal stem cells (MSCs) to undergo osteogenic differentiation, while low stiffness leads to lipogenic differentiation. Such effects could be independent of chemical/biochemical inducers. SCOPE OF REVIEW: Stiffness and/or topography of cellular environment can control MSC differentiation and fate determination. In addition, physical factors such as tension, which resulted from profound cytoskeleton reorganization during MSC differentiation, affect the gene expression essential for the differentiation. Although physical cues control MSC lineage specification probably by reorganizing and tuning cytoskeleton, the full mechanism is largely unclear. It also remains elusive how physical signals are sensed by cells and transformed into biochemical and biological signals. More importantly, it becomes pivotal to define explicitly the physical cue(s) essential for cell differentiation and fate decision. With a focus on MSC, we present herein current understanding of the interplay between i) physical cue and factors and ii) MSC differentiation and fate determination. MAJOR CONCLUSIONS: Biophysical cues can initiate or strengthen the biochemical signaling for MSC fate determination and differentiation. Physical properties of cellular environment direct the structural adaptation and functional coupling of the cells to their environment. GENERAL SIGNIFICANCE: These observations not only open a simple avenue to engineer cell fate in vitro, but also start to reveal the physical elements that regulate and determine cell fate.


Assuntos
Linhagem da Célula , Mecanotransdução Celular , Células-Tronco Mesenquimais/fisiologia , Nicho de Células-Tronco , Animais , Forma Celular , Citoesqueleto/fisiologia , Matriz Extracelular/metabolismo , Humanos , Células-Tronco Mesenquimais/metabolismo , Estimulação Física , Pressão , Estresse Mecânico
12.
Curr Cancer Drug Targets ; 15(2): 99-115, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25553514

RESUMO

Deregulated c-MYC (hereafter MYC) is widely expressed in many human tumors. Myctransgenic mouse models produce diverse tumors. MYC is a strong driver of tumorigenesis and is required for tumor maintenance. MYC is therefore an attractive target for cancer treatment. However, genetic and pharmacological approaches for the targeted inactivation of MYC for the treatment of MYC-overexpressing tumors have been shown to be unsatisfactory. MYC expression is regulated by different mechanisms at transcriptional, post-transcriptional, and post-translational levels. Turnover of MYC protein is an important step that influences the expression and function of MYC. MYC turnover is predominantly controlled by the GSK3/FBW7 axis that is regulated by multiple elements. Small molecule inhibitors (SMIs) can influence the stability and activity of MYC protein by targeting the axis and its regulator elements. Wang et al. (2004) first introduced the concept of MYC-mediated synthetic lethality (MYC-SL) induced by TRAIL and DR5-agonists. Researchers have turned to synthetic lethality as a treatment strategy for MYC-overexpressing tumors. MYC function is closely associated with MYC levels. Two strategies have been developed to treat MYC-overexpressing tumors by upregulating or downregulating MYC. An SMI can induce MYC-SL by increasing MYC expression through the inactivation of the GSK3ß/FBW7 axis and CK1. Elevated MYC levels lead to DNA damage, senescence, and apoptosis. An SMI can also induce MYC-SL by decreasing MYC expression through the activation of the GSK3ß/FBW7 axis, the inactivation of PP2A inhibitors, and the inhibition of ARK5, AUK-A, Brd4, CDK1, CDK2, CHK1, and SAE1/2. Reduced MYC levels cause tumor regression. Some SMIs have entered phase I and II clinical trials. SMIs may be used in the near future to treat cancers co-overexpressing MYC and the corresponding MYC-SL genes.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias/tratamento farmacológico , Proteínas Proto-Oncogênicas c-myc/antagonistas & inibidores , Animais , Antineoplásicos/metabolismo , Sistemas de Liberação de Medicamentos/métodos , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Resultado do Tratamento
13.
Curr Cancer Drug Targets ; 15(1): 53-70, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25564254

RESUMO

Deregulated c-MYC expression is found in many human malignancies. MYC activation induces multiple lineages of hematological malignancies in single Myc transgenic mice. MYC inactivation causes tumor regression. MYC is therefore an attractive target for cancer treatment. However, little progress has been made in the development and application of targeted MYC inactivation in clinical practice. In double Myc transgenic mouse models, Myc-driven leukemogenesis and lymphomagenesis can be accelerated by transduction of non-MYC oncogenes, leading to dual addiction to MYC and the non-MYC oncogenes. Wang et al. (2004) first established the concept of MYC-mediated synthetic lethality (MYC-SL). MYC overexpression sensitized cells to TRAILand DR5-agonist-induced apoptosis. This suggests that MYC-dependent tumor cells may be killed by targeting partner oncogenes of MYC. Many small molecule inhibitors (SMIs) have been proven to induce MYC-SL by targeting AUK-B, Brd4, CDK1, CHK1, MCL-1, the mTOR/4E-BP1/eIF4E pathway, and PIM1/2. Compared with conventional treatment approaches, SMI-induced MYC-SL displays highly selective anticancer activity and much lower cytotoxicity to normal cells. SMI-induced MYC-SL can reverse eIF4F- and PIM2-induced multiple chemoresistance. The combination of an SMI with chemotherapeutic agents can elevate chemotherapy efficacy by enhancing chemosensitivity. This combination will be a promising novel approach to treating MYC-dependent tumors by inducing MYC-SL.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Hematológicas/tratamento farmacológico , Proteínas Proto-Oncogênicas c-myc/metabolismo , Animais , Neoplasias Hematológicas/metabolismo , Humanos , Camundongos , Proteínas Proto-Oncogênicas c-myc/efeitos dos fármacos , Transdução de Sinais
14.
Circulation ; 130(17): 1493-504, 2014 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-25149363

RESUMO

BACKGROUND: Angiogenesis is crucial for many pathological processes and becomes a therapeutic strategy against diseases ranging from inflammation to cancer. The regulatory mechanism of angiogenesis remains unclear. Although tetraspanin CD82 is widely expressed in various endothelial cells (ECs), its vascular function is unknown. METHODS AND RESULTS: Angiogenesis was examined in Cd82-null mice with in vivo and ex vivo morphogenesis assays. Cellular functions, molecular interactions, and signaling were analyzed in Cd82-null ECs. Angiogenic responses to various stimuli became markedly increased upon Cd82 ablation. Major changes in Cd82-null ECs were enhanced migration and invasion, likely resulting from the upregulated expression of cell adhesion molecules such as CD44 and integrins at the cell surface and subsequently elevated outside-in signaling. Gangliosides, lipid raft clustering, and CD44-membrane microdomain interactions were increased in the plasma membrane of Cd82-null ECs, leading to less clathrin-independent endocytosis and then more surface presence of CD44. CONCLUSIONS: Our study reveals that CD82 restrains pathological angiogenesis by inhibiting EC movement, that lipid raft clustering and cell adhesion molecule trafficking modulate angiogenic potential, that transmembrane protein modulates lipid rafts, and that the perturbation of CD82-ganglioside-CD44 signaling attenuates pathological angiogenesis.


Assuntos
Células Endoteliais/metabolismo , Receptores de Hialuronatos/metabolismo , Proteína Kangai-1/metabolismo , Microdomínios da Membrana/metabolismo , Neovascularização Patológica/metabolismo , Animais , Moléculas de Adesão Celular/metabolismo , Linhagem Celular , Movimento Celular/fisiologia , Citoesqueleto/metabolismo , Endocitose/fisiologia , Células Endoteliais/patologia , Gangliosídeos/metabolismo , Proteína Kangai-1/genética , Microdomínios da Membrana/patologia , Camundongos Knockout , Neovascularização Patológica/genética , Neovascularização Patológica/patologia , Transporte Proteico/fisiologia , Transdução de Sinais/fisiologia
15.
Mol Med Rep ; 7(3): 836-42, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23292489

RESUMO

Previous studies have shown that the tetraspanin CD151 is essential for pathological or physiological angiogenesis. However, the cellular signaling mechanism and the role of the CD151 YRSL sorting motif in in vitro vasculogenesis remains unknown. In this study, the results showed that both CD151 and CD151-ARSA gene delivery were capable of increasing the expression of CD151 at the protein level in human umbilical vein endothelial cells (HUVECs). Moreover, there was no significant difference in CD151 protein expression between the CD151 group and the CD151-ARSA group. Overexpression of CD151 promoted HUVEC cell proliferation, migration and capillary network formation in vitro. However, in the CD151-ARSA group, the abilities of cell proliferation, migration and capillary network formation were all decreased, compared with the CD151 group. Furthermore, the activation of PI3K, Akt and ERK signaling pathways was attenuated in the CD151-ARSA mutant group compared with the CD151 group. This study suggests that the YRSL motif of CD151 plays a key role in CD151-induced angiogenesis. Our observations provide insights into a new mechanism of CD151 regulating angiogenesis via vesicle trafficking.


Assuntos
Neovascularização Fisiológica/fisiologia , Tetraspanina 24/metabolismo , Motivos de Aminoácidos , Movimento Celular , Proliferação de Células , Dependovirus/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Vetores Genéticos/genética , Vetores Genéticos/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Mutação , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Tetraspanina 24/genética , Transfecção
16.
J Virol ; 87(6): 3435-46, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23302890

RESUMO

Human papillomavirus type 16 (HPV16) is the primary etiologic agent for cervical cancer. The infectious entry of HPV16 into cells occurs via a so-far poorly characterized clathrin- and caveolin-independent endocytic pathway, which involves tetraspanin proteins and actin. In this study, we investigated the specific role of the tetraspanin CD151 in the early steps of HPV16 infection. We show that surface-bound HPV16 moves together with CD151 within the plane of the membrane before they cointernalize into endosomes. Depletion of endogenous CD151 did not affect binding of viral particles to cells but resulted in reduction of HPV16 endocytosis. HPV16 uptake is dependent on the C-terminal cytoplasmic region of CD151 but does not require its tyrosine-based sorting motif. Reexpression of the wild-type CD151 but not mutants affecting integrin functions restored virus internalization in CD151-depleted cells. Accordingly, short interfering RNA (siRNA) gene knockdown experiments confirmed that CD151-associated integrins (i.e., α3ß1 and α6ß1/4) are involved in HPV16 infection. Furthermore, palmitoylation-deficient CD151 did not support HPV16 cell entry. These data show that complex formation of CD151 with laminin-binding integrins and integration of the complex into tetraspanin-enriched microdomains are critical for HPV16 endocytosis.


Assuntos
Endocitose , Papillomavirus Humano 16/fisiologia , Tetraspanina 24/metabolismo , Internalização do Vírus , Linhagem Celular , Análise Mutacional de DNA , Técnicas de Silenciamento de Genes , Humanos , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Tetraspanina 24/genética
17.
PLoS One ; 7(12): e51797, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23251627

RESUMO

To determine how tetraspanin KAI1/CD82, a tumor metastasis suppressor, inhibits cell migration, we assessed which cellular events critical for motility are altered by KAI1/CD82 and how KAI1/CD82 regulates these events. We found that KAI1/CD82-expressing cells typically exhibited elongated cellular tails and diminished lamellipodia. Live imaging demonstrated that the polarized protrusion and retraction of the plasma membrane became deficient upon KAI1/CD82 expression. The deficiency in developing these motility-related cellular events was caused by poor formations of actin cortical network and stress fiber and by aberrant dynamics in actin organization. Rac1 activity was reduced by KAI1/CD82, consistent with the diminution of lamellipodia and actin cortical network; while the growth factor-stimulated RhoA activity was blocked by KAI1/CD82, consistent with the loss of stress fiber and attenuation in cellular retraction. Upon KAI1/CD82 expression, Rac effector cofilin was not enriched at the cell periphery to facilitate lamellipodia formation while Rho kinase exhibited a significantly lower activity leading to less retraction. Phosphatidylinositol 4, 5-biphosphate, which initiates actin polymerization from the plasma membrane, became less detectable at the cell periphery in KAI1/CD82-expressing cells. Moreover, KAI1/CD82-induced phenotypes likely resulted from the suppression of multiple signaling pathways such as integrin and growth factor signaling. In summary, at the cellular level KAI1/CD82 inhibited polarized protrusion and retraction events by disrupting actin reorganization; at the molecular level, KAI1/CD82 deregulated Rac1, RhoA, and their effectors cofilin and Rho kinase by perturbing the plasma membrane lipids.


Assuntos
Actinas/metabolismo , Movimento Celular/fisiologia , Extensões da Superfície Celular/fisiologia , Proteína Kangai-1/metabolismo , Adesão Celular/efeitos dos fármacos , Adesão Celular/fisiologia , Linhagem Celular Tumoral , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Membrana Celular/fisiologia , Movimento Celular/efeitos dos fármacos , Extensões da Superfície Celular/metabolismo , Genes Supressores de Tumor , Células HT29 , Humanos , Integrinas/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Masculino , Lipídeos de Membrana/metabolismo , Fosfatidilinositol 4,5-Difosfato/farmacologia , Pseudópodes/efeitos dos fármacos , Pseudópodes/metabolismo , Pseudópodes/fisiologia , Transdução de Sinais/efeitos dos fármacos , Fibras de Estresse/metabolismo , Proteínas rac1 de Ligação ao GTP/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo
18.
PLoS One ; 7(6): e38464, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22679508

RESUMO

Alterations in tetraspanin CO-029 expression are associated with the progression and metastasis of cancers in the digestive system. However, how CO-029 promotes cancer metastasis is still poorly understood. To determine the mechanism, we silenced CO-029 expression in HT29 colon cancer cells and found that the CO-029 knockdown significantly reduced cell migratory ability. The diminished cell migration was accompanied by the upregulation of both integrin-dependent cell-matrix adhesion on laminin and calcium-dependent cell-cell adhesion. The cell surface levels of laminin-binding integrin α3ß1 and fibronectin-integrin α5ß1 were increased while the level of CD44 was decreased upon CO-029 silencing. These changes contribute to the altered cell-matrix adhesion. The deregulated cell-cell adhesion results, at least partially, from increased activity of cadherins and reduced level of MelCAM. In conclusion, CO-029 functions as a regulator of both cell-matrix and cell-cell adhesion. During colon cancer progression, CO-029 promotes cancer cell movement by deregulating cell adhesions.


Assuntos
Movimento Celular/fisiologia , Tetraspaninas/metabolismo , Western Blotting , Adesão Celular/genética , Adesão Celular/fisiologia , Movimento Celular/genética , Neoplasias Colorretais/metabolismo , Citometria de Fluxo , Células HT29 , Humanos , Receptores de Hialuronatos/genética , Receptores de Hialuronatos/metabolismo , Imunoprecipitação , Integrina alfa3beta1/genética , Integrina alfa3beta1/metabolismo , Integrina alfa5beta1/genética , Integrina alfa5beta1/metabolismo , Laminina/genética , Laminina/metabolismo , Interferência de RNA , Tetraspaninas/genética
19.
Biochem J ; 437(3): 399-411, 2011 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-21609323

RESUMO

The tumour suppressor EWI2 associates with tetraspanins and regulates tumour cell movement and proliferation. The short cytoplasmic domain of EWI2 is positively charged; five out of the ten residues of this domain are basic. In the present study we demonstrated that the EWI2 cytoplasmic tail interacts specifically with negatively charged PIPs (phosphatidylinositol phosphates), but not with other membrane lipids. The PIPs that interact with EWI2 cytoplasmic tail include PtdIns5P, PtdIns4P, PtdIns3P, PtdIns(3,5)P(2) and PtdIns(3,4)P2. The binding affinity of PIPs to the EWI2 tail, however, is not solely based on charge because PtdIns5P, PtdIns4P and PtdIns3P have a higher affinity to EWI2 than PtdIns(3,5)P(2) and PtdIns(3,4)P(2) do. Mutation of either of two basic residue clusters in the EWI2 cytoplasmic tail abolishes PIP binding, and PIP binding is also determined by the position of basic residues in the EWI2 cytoplasmic tail. In addition, EWI2 is constitutively palmitoylated at the cytoplasmic cysteine residues located at the N-terminal of those basic residues. The PIP interaction is not required for, but appears to regulate, the palmitoylation, whereas palmitoylation is neither required for nor regulates the PIP interaction. Functionally, the PIP interaction regulates the stability of EWI2 proteins, whereas palmitoylation is needed for tetraspanin-EWI2 association and EWI2-dependent inhibition of cell migration and lamellipodia formation. For cell-cell adhesion and cell proliferation, the PIP interaction functions in opposition to the palmitoylation. In conclusion, the EWI2 cytoplasmic tail actively engages with the cell membrane via PIP binding and palmitoylation, which play differential roles in EWI2 functions.


Assuntos
Proteínas de Transporte/metabolismo , Regulação da Expressão Gênica/fisiologia , Lipoilação , Proteínas de Membrana/metabolismo , Fosfolipídeos/metabolismo , Animais , Proteínas de Transporte/genética , Adesão Celular , Proliferação de Células , Proteínas de Membrana/genética , Camundongos , Células NIH 3T3 , Fosfatos de Fosfatidilinositol/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA