Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
1.
Sci Rep ; 13(1): 20420, 2023 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-37990044

RESUMO

Previously, we have identified a novel human metastasis-inducing lncRNA (named SKAI1BC), that suppresses the KAI1/CD82 metastasis-suppressing gene and is upregulated in triple negative breast cancer and melanoma derived cell lines. Modeling of the SKAI1BC lncRNA secondary structure and its potential interaction with Inforna compounds, led us to identify several compounds that might bind the SKAI1BC lncRNA. We found that these compounds inhibit metastasis invasion and cell migration in culture, in all eight types of solid human cancers tested: several of which are the most lethal and/or frequent human malignancies. Moreover, in most cases, the mechanism of action of several of our compounds involves enhancement of KAI1/CD82 RNA level depending on the specific compound and the human tumor type. With the epigenetic inactivation of KAI1/CD82 in at least ten additional solid human cancers, this implies a very good chance to broaden the spectrum of human cancers affected by our compounds. This is the first time that modeling of a large lncRNA (> 700 bp) secondary structure followed by its potential interaction with Inforna like compounds database has led to the identification of potential biologically active small molecule drugs.


Assuntos
Melanoma , RNA Longo não Codificante , Humanos , RNA Longo não Codificante/genética , Proteína Kangai-1/genética , Proteína Kangai-1/metabolismo , Genes Supressores de Tumor , Linhagem Celular , Melanoma/tratamento farmacológico , Melanoma/genética , Metástase Neoplásica
2.
Comb Chem High Throughput Screen ; 26(5): 1001-1014, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-35713138

RESUMO

OBJECTIVE: Malignant melanoma with gastric cancer is one of the most malignant tumors. However, there have been no reports on the effects of KAI1 and miRNA-633 on the survival and prognosis of patients with malignant melanoma with gastric cancer. METHODS: Fifty patients with malignant melanoma and gastric cancer were collected from October 2017 to December 2019. The clinical parameters included clinical information, such as sex, age, tumor size, and tumor staging. RT-qPCR was used to detect the expression of KAI1 and miRNA- 633. The role of KAI1 and miRNA-633 on the overall survival of melanoma was explored by the Pearson chi-square test, Spearman-rho correlation test, Univariate and multivariate cox regression analyses, and Kaplan-Meier method. Furthermore, the bioinformatic analysis was used to verify the role of KAI1 and miRNA-633 on malignant melanoma with gastric cancer. RESULTS: The expression of KAI1 and miRNA-633 was significantly related with the tumor size and staging of tumor (p<0.05) based on the Pearson chi-square test. Spearman's correlation coefficient displayed that KAI1 was significantly correlated with the miRNA-633 (ρ=-0.439, p=0.001). The result of multivariate cox proportional regression analysis showed that KAI1 (HR =0.109, 95% CI: 0.031-0.375, p< 0.001), and miRNA-633 (HR = 13.315, 95% CI: 3.844-46.119, p<0.001) were significantly associated with overall survival. CONCLUSION: The low expression level of KAI1 and high expression of miRNA-633 are significantly correlated with the poor overall survival prognosis of malignant melanoma with gastric cancer, to provide a basis for KAI1 and miRNA-633 to become novel molecular targets for malignant melanoma with gastric cancer.


Assuntos
Melanoma , MicroRNAs , Neoplasias Gástricas , Humanos , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/genética , MicroRNAs/genética , Proteína Kangai-1/genética , Proteína Kangai-1/análise , Proteína Kangai-1/metabolismo , Melanoma/diagnóstico , Melanoma/genética , Biomarcadores Tumorais/metabolismo , Estadiamento de Neoplasias , Melanoma Maligno Cutâneo
3.
Biochem Biophys Res Commun ; 629: 121-127, 2022 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-36116374

RESUMO

CD82, a tetraspanin superfamily member, has been identified to be glycosylated at three specific residues (Asn129, Asn157, and Asn198). However, CD82 post-translational modification and its effect on colorectal cancer (CRC) metastasis remain unclear. Here, we constructed various deficient mutants of CD82 N-glycosylation in SW620 cells and demonstrated that the Asn157 site is necessary for CD82 glycosylation in CRC cells migration and LN-dependent adhesion in vitro. Furthermore, we found that CD82 N-glycosylation at the Asn157 site leads to lower expression levels of vimentin and claudin-1 but higher expression levels of E-cadherin, which are the EMT markers; also, there are lower expression levels of phospho-GSK3ß and less ß-catenin transportation to the nucleus. These findings suggest that CD82 N-glycosylation at the Asn157 site inhibits EMT by down-regulating the Wnt/ß-catenin pathway. Moreover, we reported that CD82 with N-glycosylation at a single site of the Asn157 reduces lung metastases in vivo. The results indicate that N-glycosylation of CD82 at the Asn157 site regulates CRC metastasis and adhesion. These observations suggest that the N-glycosylation of CD82 might be a potential therapeutic target for CRC.


Assuntos
Neoplasias do Colo , Neoplasias Colorretais , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Claudina-1/metabolismo , Neoplasias do Colo/genética , Neoplasias Colorretais/patologia , Transição Epitelial-Mesenquimal/fisiologia , Glicogênio Sintase Quinase 3 beta/metabolismo , Glicosilação , Humanos , Proteína Kangai-1/genética , Proteína Kangai-1/metabolismo , Vimentina/metabolismo , Via de Sinalização Wnt , beta Catenina/metabolismo
4.
J Int Med Res ; 50(8): 3000605221116758, 2022 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-35942551

RESUMO

OBJECTIVE: This study aimed to investigate the correlation between KAI1 (CD82) and miR-633 expression and prognosis and survival time of patients with melanoma combined with colorectal cancer (CRC). METHODS: Clinical and follow-up data of melanoma and CRC patients were recorded, and the expression levels of KAI1 and miR-633 were detected. Pearson chi-square tests and Spearman correlation coefficient were used to analyze the relationship between prognosis and related parameters in these patients. Cox proportional risk regression and receiver operating characteristic curve analyses were used. RESULTS: Overall, 195 patients were included. KAI1 and miR-633 expression levels were significantly correlated with the prognosis of patients with melanoma combined with CRC. Spearman correlation analysis showed that the expression levels of KAI1 and miR-633 were significantly correlated with the prognosis of patients. Multivariate Cox regression analysis suggested that low expression levels of KAI1 and high expression levels of miR-633 indicated shorter survival time for patients. CONCLUSIONS: KAI1 expression was significantly correlated with melanoma and CRC patient prognosis. When KAI1 expression levels were low, the patient survival time was poor.


Assuntos
Neoplasias Colorretais , Proteína Kangai-1/metabolismo , Melanoma , MicroRNAs , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Humanos , Proteína Kangai-1/análise , Proteína Kangai-1/genética , Melanoma/genética , Melanoma/patologia , MicroRNAs/genética , Estadiamento de Neoplasias , Prognóstico
5.
Acta Biochim Biophys Sin (Shanghai) ; 54(3): 400-408, 2022 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-35538033

RESUMO

Tetraspanin CD82 often participates in regulating the function of epidermal growth factor receptor (EGFR) and hepatocyte growth factor receptor (c-Met). Palmitoylation is a post-translational modification that contributes to tetraspanin web formation and affects tetraspanin-dependent cell signaling. However, the molecular mechanisms by which CD82 palmitoylation affects the localization and stability of EGFR and c-Met have not yet been elucidated. This study focuses on the expression and distribution of EGFR and c-Met in breast cancer as well as the related metabolic pathways and molecular mechanisms associated with different CD82 palmitoylation site mutations. The results show that CD82 with a palmitoylation mutation at Cys5+Cys74 can promote the internalization of EGFR. EGFR is internalized and strengthened by direct binding to CD82 with the tubulin assistance and located at the recycling endosome. After studying the recycling pathway marker proteins Rab11a and FIP2, we found that formation of the EGFR/CD82/Rab11a/FIP2 complex promotes the internalization and metabolism of EGFR through the recycling pathway and results in the re-expression of EGFR and CD82 on the cell membrane.


Assuntos
Proteína Kangai-1 , Lipoilação , Receptores ErbB/genética , Receptores ErbB/metabolismo , Proteína Kangai-1/genética , Proteína Kangai-1/metabolismo , Mutação , Fosforilação
6.
Technol Cancer Res Treat ; 20: 15330338211045204, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34605330

RESUMO

Background: Pancreatic cancer (PC) has a poor prognosis and is prone to liver metastasis. The KAI1/CD82 gene inhibits PC metastasis. This study aimed to explore differential metabolites and enrich the pathways in serum samples between PC and liver metastasis nude mouse models stably expressing KAI1/CD82. Methods: KAI1/CD82-PLV-EF1α-MCS-IRES-Puro vector and PANC1 cell line stably expressing KAI1/CD82 were constructed for the first time. This cell line was used to construct 3 PC nude mouse models and 3 liver metastasis nude mouse models. The different metabolites and Kyoto encyclopedia of genes and genomes (KEGG) and human metabolome database (HMDB) enrichment pathways were analyzed using the serum samples of the 2 groups of nude mouse models on the basis of untargeted ultra-performance liquid chromatography-tandem mass spectrometry platform. Results: KAI1/CD82-PLV-EF1α-MCS-IRES-Puro vector and PANC1 cell line stably expressing KAI1/CD82 were constructed successfully, and all nude mouse models survived and developed cancers. Among the 1233 metabolites detected, 18 metabolites (9 upregulated and 9 downregulated) showed differences. In agreement with the literature data, the most significant differences between both groups were found in the levels of bile acids (taurocholic acid, chenodeoxycholic acid), glycine, prostaglandin E2, vitamin D, guanosine monophosphate, and inosine. Bile recreation, primary bile acid biosynthesis, and purine metabolism KEGG pathways and a series of HMDB pathways (P < .05) contained differential metabolites that may be associated with liver metastasis from PC. However, the importance of these metabolites on PC liver metastases remains to be elucidated. Conclusions: Our findings suggested that the metabolomic approach may be a useful method to detect potential biomarkers in PC.


Assuntos
Biomarcadores Tumorais/sangue , Proteína Kangai-1/metabolismo , Neoplasias Hepáticas/sangue , Neoplasias Pancreáticas/sangue , Animais , Linhagem Celular Tumoral , Ácido Quenodesoxicólico/sangue , Bases de Dados Genéticas , Dinoprostona/sangue , Modelos Animais de Doenças , Feminino , Glicina/sangue , Guanosina Monofosfato/sangue , Humanos , Inosina/sangue , Proteína Kangai-1/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/secundário , Redes e Vias Metabólicas , Metabolômica , Camundongos , Camundongos Nus , Transplante de Neoplasias , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patologia , Ácido Taurocólico/sangue , Vitamina D/sangue
7.
J Hematol Oncol ; 14(1): 148, 2021 09 16.
Artigo em Inglês | MEDLINE | ID: mdl-34530889

RESUMO

BACKGROUND: Little is known about endogenous inhibitors of angiogenic growth factors. In this study, we identified a novel endogenous anti-angiogenic factor expressed in pericytes and clarified its underlying mechanism and clinical significance. METHODS: Herein, we found Kai1 knockout mice showed significantly enhanced angiogenesis. Then, we investigated the anti-angiogenic roll of Kai1 in vitro and in vivo. RESULTS: KAI1 was mainly expressed in pericytes rather than in endothelial cells. It localized at the membrane surface after palmitoylation by zDHHC4 enzyme and induced LIF through the Src/p53 pathway. LIF released from pericytes in turn suppressed angiogenic factors in endothelial cells as well as in pericytes themselves, leading to inhibition of angiogenesis. Interestingly, KAI1 had another mechanism to inhibit angiogenesis: It directly bound to VEGF and PDGF and inhibited activation of their receptors. In the two different in vivo cancer models, KAI1 supplementation significantly inhibited tumor angiogenesis and growth. A peptide derived from the large extracellular loop of KAI1 has been shown to have anti-angiogenic effects to block the progression of breast cancer and retinal neovascularization in vivo. CONCLUSIONS: KAI1 from PC is a novel molecular regulator that counterbalances the effect of angiogenic factors.


Assuntos
Proteína Kangai-1/metabolismo , Neovascularização Patológica/metabolismo , Neovascularização Fisiológica , Animais , Feminino , Proteína Kangai-1/genética , Masculino , Microdomínios da Membrana/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neovascularização Patológica/genética , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Reproduction ; 162(3): 227-236, 2021 08 11.
Artigo em Inglês | MEDLINE | ID: mdl-34280124

RESUMO

A successful pregnancy requires sufficient decidualization of endometrial stromal cells (ESCs). CD82, a metastasis suppressor, is a critical regulator for trophoblast invasion but the effect in decidualization was largely unknown. Here we reported that there was a high level of CD82 in DSC by the immunohistochemistry staining and flow cytometer analysis. Stimulation with prostaglandin E2 (PGE2) elevated the expression of CD82 in ESCs. In contrast, celecoxib, a selective COX-2 inhibitor, significantly downregulated the expression of CD82 in decidual stromal cells (DSCs). Bioinformatics analysis and further research showed that recombinant human interleukin (IL)-1ß protein (rhIL-1ß) upregulated CD82 in ESCs. Of note, blocking IL-1ß signaling with anti-human IL-1ß neutralizing antibody could reverse the stimulatory effect of PGE2 on CD82 in ESCs. Silencing CD82 resulted in the decease of the decidualization markers PRL and IGFBP1 mRNA levels in DSCs. More importantly, we observed rhIL-1ß also upregulated the expression of COX-2, and the upregulation of PRL and IGFBP1 induced by rhIL-1ß could be abolished by celecoxib in ESCs or CD82 deficiency in DSCs. This study suggests that CD82 should be a novel promotor for decidualization under a positive regulation of the COX-2/PGE2/IL-1ß positive feedback loop.


Assuntos
Decídua , Proteína Kangai-1 , Células Estromais , Células Cultivadas , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Decídua/metabolismo , Feminino , Humanos , Interleucina-1beta/metabolismo , Proteína Kangai-1/genética , Proteína Kangai-1/metabolismo , Gravidez , Células Estromais/metabolismo , Trofoblastos/metabolismo
9.
Mol Med Rep ; 23(2)2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33655331

RESUMO

The specific role and mechanism of ferroptosis in the development of pancreatic cancer (PC) remain to be elucidated. The present study aimed to investigate the effects of the overexpression of the KAI1 gene on the ferroptosis of the human PC cell line MIA PaCa­2. MIA PaCa­2 cells infected with pCMV­KAI1 and selected by G418 and KAI1 protein were analyzed by western blotting. The MIA PaCa­2 cells with a stable expression of the KAI1 gene were termed MIA PaCa­2­KAI1. The proliferative capacities of MIA PaCa­2 and MIA PaCa­2­KAI1 cells were detected using Cell Counting Kit­8. The reactive oxygen species (ROS) in the cells were compared by flow cytometry. The expressions of ferroportin (FPN) and glutathione peroxidase 4 (GPX4) protein were analyzed by western blotting. The KAI1 stable expression cell line was confirmed and relabeled as MIA PaCa­2­KAI1. No significant differences in the proliferation of MIA PaCa­2 and MIA PaCa­2­KAI1 were identified. Following treatment with a ferroptosis blocker, the increase in the proliferation of MIA PaCa­2­KAI1 (from 2.06±0.02 to 2.75±0.02) was more evident compared with MIA PaCa­2 (from 2.94±0.02 to 2.95±0.02; P<0.05). The ROS in MIA PaCa­2­KAI1 was significantly higher compared with MIA PaCa­2 (P<0.05). FPN and GPX4 protein demonstrated higher expression levels in MIA PaCa­2­KAI1 compared with MIA PaCa­2. Moreover, KAI1 exerted an obvious promotion effect on FPN expression. This study identified that the high expression of the KAI1 gene promoted the occurrence of ferroptosis in PC cells through its extensive effect on FPN and GPX4. KAI1­induced ferroptosis did not significantly inhibit the proliferation of PC cells.


Assuntos
Proteínas de Transporte de Cátions/genética , Ferroptose , Proteína Kangai-1/metabolismo , Neoplasias Pancreáticas/fisiopatologia , Fosfolipídeo Hidroperóxido Glutationa Peroxidase/genética , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Humanos , Proteína Kangai-1/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Espécies Reativas de Oxigênio/metabolismo
10.
J Diabetes Res ; 2021: 4012893, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34988229

RESUMO

The human leucocyte antigen (HLA) association with type 1 diabetes (T1D) is well known but there are limited studies investigating the association between ß-cell autoantibodies and HLA genes. We evaluated the prevalence of GAD65 and IA-2 autoantibodies (GADA and IA2A) in 252 T1D patients from North India and investigated the genetic association of GADA and IA2A with HLA class I and class II genes/haplotypes. GADA and IA2A were detected in 50.79% and 15.87% of T1D patients, respectively, while only 8.73% had both GADA and IA2A. HLA-DRB1∗03 was observed to be significantly higher in GADA+ T1D patients as compared to GADA- (91.41% vs. 66.13%, Bonferroni-corrected P (P c) = 1.11 × 10-5; OR = 5.45; 95% CI: 2.67-11.08). Similarly, HLA-DQB1∗02 was found to be significantly increased in GADA+ patients (94.53%, P c = 2.19 × 10-5; OR = 6.27; 95% CI: 2.7-14.49) as compared to GADA- (73.39%). The frequencies of HLA-DRB1∗04 and DQB1∗03 were increased in IA2A+ patients (45.0% and 52.5%, respectively) as compared to that in IA2A- (25.94% and 33.96%, respectively). Further, the frequency of DRB1∗03-DQB1∗02 haplotype was found to be significantly increased in GADA+ T1D patients as compared to GADA- (60.55% vs. 41.94%, P = 3.94 × 10-5; OR = 2.13; 95%CI = 1.49-3.03). Similarly, HLA-DRB1∗04-DQB1∗03 haplotype was found to be significantly increased in IA2A+ T1D patients compared to IA2A- patients (22.5% vs. 12.97%; P = 0.041; OR = 1.95; 95%CI = 1.08-3.52). None of the HLA class I genes (HLA-A, B, and Cw) was found to be associated with GADA or IA2A in people with T1D. Our findings suggest that HLA-DRB1∗03/DQB1∗02 and HLA-DRB1∗04/DQB1∗03 might play an important role in the development of GADA and IA2A, respectively.


Assuntos
Autoantígenos/genética , Diabetes Mellitus Tipo 1/genética , Glutamato Descarboxilase/genética , Proteína Kangai-1/genética , Fragmentos de Peptídeos/genética , Adolescente , Adulto , Autoanticorpos/análise , Autoanticorpos/sangue , Autoanticorpos/genética , Autoantígenos/análise , Criança , Pré-Escolar , Diabetes Mellitus Tipo 1/epidemiologia , Feminino , Glutamato Descarboxilase/análise , Antígenos HLA , Humanos , Índia/epidemiologia , Proteína Kangai-1/análise , Masculino , Pessoa de Meia-Idade , Fragmentos de Peptídeos/análise
11.
BMC Cancer ; 20(1): 1211, 2020 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-33298014

RESUMO

BACKGROUND: Tetraspanin CD82 is a tumor metastasis suppressor that is known to down regulate in various metastatic cancers. However, the exact mechanism by which CD82 prevents cancer metastasis is unclear. This study aims to identify genes that are regulated by CD82 in human prostate cell lines. METHODS: We used whole human genome microarray to obtain gene expression profiles in a normal prostate epithelial cell line that expressed CD82 (PrEC-31) and a metastatic prostate cell line that does not express CD82 (PC3). Then, siRNA silencing was used to knock down CD82 expression in PrEC-31 while CD82 was re-expressed in PC3 to acquire differentially-expressed genes in the respective cell line. RESULTS: Differentially-expressed genes with a P < 0.05 were identified in 3 data sets: PrEC-31 (+CD82) vs PrEC-31(-CD82), PC3-57 (+CD82) vs. PC3-5 V (-CD82), and PC3-29 (+CD82) vs. PC3-5 V (-CD82). Top 25 gene lists did not show overlap within the data sets, except (CALB1) the calcium binding protein calbindin 1 which was significantly up-regulated (2.8 log fold change) in PrEC-31 and PC3-29 cells that expressed CD82. Other most significantly up-regulated genes included serine peptidase inhibitor kazal type 1 (SPINK1) and polypeptide N-acetyl galactosaminyl transferase 14 (GALNT14) and most down-regulated genes included C-X-C motif chemokine ligand 14 (CXCL14), urotensin 2 (UTS2D), and fibroblast growth factor 13 (FGF13). Pathways related with cell proliferation and angiogenesis, migration and invasion, cell death, cell cycle, signal transduction, and metabolism were highly enriched in cells that lack CD82 expression. Expression of two mutually inclusive genes in top 100 gene lists of all data sets, runt-related transcription factor (RUNX3) and trefoil factor 3 (TFF3), could be validated with qRT-PCR. CONCLUSION: Identification of genes and pathways regulated by CD82 in this study may provide additional insights into the role that CD82 plays in prostate tumor progression and metastasis, as well as identify potential targets for therapeutic intervention.


Assuntos
Adenocarcinoma/genética , Regulação Neoplásica da Expressão Gênica/genética , Proteína Kangai-1/fisiologia , Proteínas de Neoplasias/fisiologia , Neoplasias da Próstata/genética , Adenocarcinoma/secundário , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Ontologia Genética , Humanos , Proteína Kangai-1/antagonistas & inibidores , Proteína Kangai-1/genética , Masculino , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Interferência de RNA , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/farmacologia , Análise Serial de Tecidos
12.
Skelet Muscle ; 10(1): 34, 2020 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-33243288

RESUMO

BACKGROUND: Tetraspanins are a family of proteins known to assemble protein complexes at the cell membrane. They are thought to play diverse cellular functions in tissues by modifying protein-binding partners, thus bringing complexity and diversity in their regulatory networks. Previously, we identified the tetraspanin KAI/CD82 as a prospective marker for human muscle stem cells. CD82 expression appeared decreased in human Duchenne muscular dystrophy (DMD) muscle, suggesting a functional link to muscular dystrophy, yet whether this decrease is a consequence of dystrophic pathology or a compensatory mechanism in an attempt to rescue muscle from degeneration is currently unknown. METHODS: We studied the consequences of loss of CD82 expression in normal and dystrophic skeletal muscle and examined the dysregulation of downstream functions in mice aged up to 1 year. RESULTS: Expression of CD82 is important to sustain satellite cell activation, as in its absence there is decreased cell proliferation and less efficient repair of injured muscle. Loss of CD82 in dystrophic muscle leads to a worsened phenotype compared to control dystrophic mice, with decreased pulmonary function, myofiber size, and muscle strength. Mechanistically, decreased myofiber size in CD82-/- dystrophic mice is not due to altered PTEN/AKT signaling, although increased phosphorylation of mTOR at Ser2448 was observed. CONCLUSION: Basal CD82 expression is important to dystrophic muscle, as its loss leads to significantly weakened myofibers and impaired muscle function, accompanied by decreased satellite cell activity that is unable to protect and repair myofiber damage.


Assuntos
Proteína Kangai-1/metabolismo , Distrofia Muscular de Duchenne/metabolismo , Células Satélites de Músculo Esquelético/metabolismo , Animais , Proliferação de Células , Células Cultivadas , Feminino , Proteína Kangai-1/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Força Muscular , Distrofia Muscular de Duchenne/genética , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Células Satélites de Músculo Esquelético/fisiologia , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo
13.
Mol Med Rep ; 22(5): 3994-4002, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-33000220

RESUMO

Previous studies have shown that (GM3), a ganglioside, suppresses hepatoma cell motility and migration by inhibiting phosphorylation of EGFR and the activity of the PI3K/AKT signaling pathway. Therefore, the aim of the present study was to investigate whether the combined treatment of CD82 with gangliosides can exert a synergistic inhibitory effect on cell motility and migration. Epidermal growth factor receptor (EGFR) signaling was studied for its role in the mechanism through which CD82 and gangliosides synergistically inhibit the motility and migration of SW620 human colon adenocarcinoma cells. GM3 and/or GM2 treatment, and/or overexpression of CD82 was performed in SW620 cells. High-performance thin layer chromatography, reverse transcription-quantitative PCR, western blotting and flow cytometry assays were used to confirm the content changes of GM2, GM3 and CD82. In addition, the phosphorylation of EGFR, MAPK and Akt were evaluated by western blot analysis. SW620 cell motility was investigated using wound healing analysis and chemotaxis migration assay. The combination of GM3 and GM2 with CD82 was found to markedly suppress EGF-stimulated SW620 cell motility compared with the individual factors or combination of GM2 or GM3 with CD82 by inhibiting the phosphorylation of EGFR. The results suggested that CD82 in combination with either GM2 or GM3 can exert a synergistic inhibitory effect on cell motility and migration; however, the synergistic mechanisms elicited by GM2 or GM3 with CD82 differ.


Assuntos
Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Neoplasias do Colo/metabolismo , Gangliosídeo G(M2)/farmacologia , Gangliosídeo G(M3)/farmacologia , Proteína Kangai-1/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Tirosina/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Receptores ErbB/metabolismo , Humanos , Proteína Kangai-1/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Transfecção
14.
Asian Pac J Cancer Prev ; 21(9): 2549-2553, 2020 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-32986351

RESUMO

OBJECTIVE: KAI1 (CD82) is a metastasis suppressor gene known to be down-regulated in carcinomas of breast, prostate and many other organs. The mechanism of KAI1 down-regulation is complex and not well understood. Here, we investigate the role of 8 SNPs (not previously studied) in KAI1 gene that could influence its expression in tumor tissue samples of breast cancer patients from the Eastern province of Saudi Arabia. METHODS: Single nucleotide polymorphisms (SNPs) in KAI1 gene were selected from the NCBI website (dbSNP) and were then filtered for those SNPs causing stop codon mutations (rs139889503 and rs150533529) or nonsynonymous mutation in the 5'-UTR (rs11541048, rs77359459, rs115500759, rs182579675, rs200238062, and rs372733853). SNPs genotyping was performed using TaqMan SNP Genotyping Assay and the results were correlated with KAI1 protein expression profile by immunohistochemistry (IHC) on formalin-fixed paraffin-embedded (FFPE) samples of breast cancer and control none-neoplastic tissues. RESULTS: KAI1 expression by IHC was observed in all none-neoplastic breast tissue samples and only in 35% out of the 59 breast cancer tissue samples. None of the samples was homozygous for the stop codon allele A in the SNP rs139889503 or allele T in the SNP rs150533529. The SNPs in the 5-UTR, rs11541048, rs115500759, and rs182579675, were only present in the homozygous state for the G and C alleles respectively in both cancer and control samples. The other SNPs in the 5'-UTR (rs77359459, rs200238062, and rs372733853) had no significant difference in the allele distribution between KAI1 expressing or none-expressing tissue samples. CONCLUSION: Our findings showed no significant effect of the studied SNPs on down-regulation of KAI1 expression.
.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/patologia , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Proteína Kangai-1/genética , Polimorfismo de Nucleotídeo Único , Neoplasias da Mama/epidemiologia , Neoplasias da Mama/genética , Estudos de Casos e Controles , Feminino , Seguimentos , Humanos , Prognóstico , Arábia Saudita/epidemiologia
15.
Biochem Biophys Res Commun ; 533(3): 338-345, 2020 12 10.
Artigo em Inglês | MEDLINE | ID: mdl-32958256

RESUMO

We have previously demonstrated that the peptide mimicking small extracellular ring domain of CD82 (CD82EC1-mP) could inhibit tumor cell motility and metastasis. However, its acting mechanism is not understood. Here, we reported that the cell motility-inhibitory function of CD82EC1-mP was involved in the downregulation of epithelial-mesenchymal transition (EMT). Both vimentin and E-cadherin are EMT makers. We found that CD82EC1-mP could inhibit the expression of vimentin, but promot the expression of E-cadherin, suggesting that CD82EC1-mP suppressed EMT. Hippo/YAP and Wnt/ß-catenin are both key signal pathways that regulate the EMT process. The futher studies showed that CD82EC1-mP couled activate GSK3ß, promote the phosphorylation of ß-catenin, and inhibit the ß-catenin nuclear location. Moreover, CD82EC1-mP couled activate Hipoo kinase cascade, promote the phosphorylation of YAP, and inhibit the YAP nuclear location. These results suggested that CD82EC1-mP inhibited invation and matestasis via inhibiting EMT through downregulating Wnt pathway and upregulating Hippo pathway.


Assuntos
Antineoplásicos/farmacologia , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica , Proteína Kangai-1/genética , Peptídeos/farmacologia , Proteínas Serina-Treonina Quinases/genética , Via de Sinalização Wnt/efeitos dos fármacos , Células A549 , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Antígenos CD/genética , Antígenos CD/metabolismo , Antineoplásicos/síntese química , Caderinas/agonistas , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Glicogênio Sintase Quinase 3 beta/genética , Glicogênio Sintase Quinase 3 beta/metabolismo , Células Hep G2 , Via de Sinalização Hippo , Humanos , Proteína Kangai-1/antagonistas & inibidores , Proteína Kangai-1/química , Proteína Kangai-1/metabolismo , Mimetismo Molecular , Células PC-3 , Peptídeos/síntese química , Fosforilação , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Vimentina/antagonistas & inibidores , Vimentina/genética , Vimentina/metabolismo , Proteínas de Sinalização YAP , beta Catenina/genética , beta Catenina/metabolismo
16.
Indian J Pathol Microbiol ; 63(3): 405-411, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32769329

RESUMO

OBJECTIVE: Renal oncocytoma (RO) and chromophobe renal cell carcinoma (ChRCC) originate from the same cell origin, that is, the intercalated cells of the collecting duct.[1] In most cases, there are clear morphologic differences between RO and ChRCC; however, in some instances, overlapping features may be encountered and the differentiation between the two entities becomes difficult.[2] Several immunohistochemical markers with different expression patterns in ChRCC and RO have been described to rule out this dilemma. MATERIALS AND METHODS: About 47 primary renal neoplasms that had been diagnosed as RO or ChRCC were submitted for immunohistochemical staining of amylase α-1A (AMY1A), MOC 31, and CD 82. The sensitivity, specificity, positive predictive value (PPV), negative predictive value (NPV), and diagnostic accuracy have been analyzed. RESULTS: AMY1A positivity was observed in all RO cases in our work with 91.7% sensitivity and 100% specificity in the diagnosis of RO. The PPV of its expression was (100%) and NPV (97.2%) with a diagnostic accuracy of 97.9%. A significant high expression of MOC 31 was observed in ChRCC compared to its expression in RO with a statistical significance (P < 0.001). In addition, we obtained 82.9% sensitivity and 91.7% specificity of MOC 31 expression in the diagnosis of ChRCC. The positive predictive value (PPV) was (96.7%), negative predictive value (NPV) (64.7%) with diagnostic accuracy (85.1%). In our studied cases, we detected positive immunoexpression of CD 82 in 10 cases (83.3%) of ChRCC. However, it was lost in all RO cases (100%). CD 82 sensitivity and specificity in differentiating ChRCC from RO were 100% and 83.3%, respectively. CONCLUSION: We propose MOC 31 and CD 82 as negative immunostains for RO, as these markers are commonly expressed in ChRCC. In conjunction with AMY1A strong immunopositivity in RO cases, we provide a triple panel of biomarkers (AMY1A, MOC 31, and CD 82) for the distinction between RO and ChRCC.


Assuntos
Adenoma Oxífilo/diagnóstico , Carcinoma de Células Renais/diagnóstico , Proteína Kangai-1/genética , Neoplasias Renais/diagnóstico , Glicoproteínas de Membrana/genética , alfa-Amilases Salivares/genética , Adenoma Oxífilo/genética , Adulto , Idoso , Biomarcadores Tumorais/genética , Carcinoma de Células Renais/genética , Diagnóstico Diferencial , Feminino , Humanos , Imuno-Histoquímica , Neoplasias Renais/genética , Masculino , Pessoa de Meia-Idade , Valor Preditivo dos Testes , Reprodutibilidade dos Testes , Sensibilidade e Especificidade
17.
Cell Death Differ ; 27(12): 3289-3306, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32555380

RESUMO

Kaposi's sarcoma (KS), a highly angiogenic and invasive vascular tumor, is the most common AIDS-associated cancer caused by KS-associated herpesvirus (KSHV) infection. We have recently shown that KSHV-encoded viral interferon regulatory factor 1 (vIRF1) contributes to KSHV-induced cell motility (PLoS Pathog. 15:e1007578, 2019). However, the role of vIRF1 in KSHV-induced angiogenesis remains unknown. Here, using two in vivo angiogenesis models including the chick chorioallantoic membrane assay (CAM) and the matrigel plug angiogenesis assay in mice, we show that vIRF1 promotes angiogenesis by upregulating CUB domain (for complement C1r/C1s, Uegf, Bmp1) containing protein 1 (CDCP1). Mechanistically, vIRF1 enhances the expression of transcription factor lymphoid enhancer-binding factor 1 (Lef1) and binds to Lef1 to promote CDCP1 transcription. Meanwhile, vIRF1 degrades metastasis suppressor CD82 through an ubiquitin-proteasome pathway by recruiting E3 ubiquitin ligase AMFR to CD82, which protects CDCP1 from CD82-mediated, palmitoylation-dependent degradation. CDCP1 activates AKT signaling, which is required for vIRF1-induced cell motility but not angiogenesis. Our results illustrate that, by hijacking Lef1 and CD82, vIRF1 upregulates CDCP1 to promote angiogenesis and cell invasion. These novel findings demonstrate the vIRF1 targets multiple cellular proteins and pathways to promote the pathogenesis of KS, which could be attractive therapeutic targets for KSHV-induced malignancies.


Assuntos
Herpesvirus Humano 8/fisiologia , Fatores Reguladores de Interferon/metabolismo , Proteína Kangai-1/metabolismo , Fator 1 de Ligação ao Facilitador Linfoide/metabolismo , Neovascularização Patológica/patologia , Sarcoma de Kaposi/patologia , Proteínas Virais/metabolismo , Animais , Carcinogênese , Movimento Celular , Regulação da Expressão Gênica , Células Endoteliais da Veia Umbilical Humana , Humanos , Fatores Reguladores de Interferon/genética , Proteína Kangai-1/genética , Fator 1 de Ligação ao Facilitador Linfoide/genética , Masculino , Camundongos , Camundongos Nus , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Neovascularização Patológica/virologia , Sarcoma de Kaposi/genética , Sarcoma de Kaposi/metabolismo , Sarcoma de Kaposi/virologia , Transdução de Sinais , Ubiquitina-Proteína Ligases , Proteínas Virais/genética
18.
Oncogene ; 39(19): 3910-3925, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32203165

RESUMO

A principal challenge in treating acute myeloid leukemia (AML) is chemotherapy refractory disease. As such, there remains a critical need to identify key regulators of chemotherapy resistance in AML. In this study, we demonstrate that the membrane scaffold, CD82, contributes to the chemoresistant phenotype of AML. Using an RNA-seq approach, we identified the increased expression of the tetraspanin family member, CD82, in response to the chemotherapeutic, daunorubicin. Analysis of the TARGET and BEAT AML databases identifies a correlation between CD82 expression and overall survival of AML patients. Moreover, using a combination of cell lines and patient samples, we find that CD82 overexpression results in significantly reduced cell death in response to chemotherapy. Investigation of the mechanism by which CD82 promotes AML survival in response to chemotherapy identified a crucial role for enhanced protein kinase c alpha (PKCα) signaling and downstream activation of the ß1 integrin. In addition, analysis of ß1 integrin clustering by super-resolution imaging demonstrates that CD82 expression promotes the formation of dense ß1 integrin membrane clusters. Lastly, evaluation of survival signaling following daunorubicin treatment identified robust activation of p38 mitogen-activated protein kinase (MAPK) downstream of PKCα and ß1 integrin signaling when CD82 is overexpressed. Together, these data propose a mechanism where CD82 promotes chemoresistance by increasing PKCα activation and downstream activation/clustering of ß1 integrin, leading to AML cell survival via activation of p38 MAPK. These observations suggest that the CD82-PKCα signaling axis may be a potential therapeutic target for attenuating chemoresistance signaling in AML.


Assuntos
Integrina beta1/genética , Proteína Kangai-1/genética , Leucemia Mieloide Aguda/tratamento farmacológico , Proteína Quinase C-alfa/genética , Adulto , Idoso , Daunorrubicina/efeitos adversos , Daunorrubicina/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HL-60 , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Masculino , Pessoa de Meia-Idade , RNA-Seq , Proteínas Quinases p38 Ativadas por Mitógeno/genética
19.
Nat Commun ; 11(1): 333, 2020 01 16.
Artigo em Inglês | MEDLINE | ID: mdl-31949145

RESUMO

Cutaneous malignant melanoma is an aggressive cancer of melanocytes with a strong propensity to metastasize. We posit that melanoma cells acquire metastatic capability by adopting an embryonic-like phenotype, and that a lineage approach would uncover metastatic melanoma biology. Using a genetically engineered mouse model to generate a rich melanoblast transcriptome dataset, we identify melanoblast-specific genes whose expression contribute to metastatic competence and derive a 43-gene signature that predicts patient survival. We identify a melanoblast gene, KDELR3, whose loss impairs experimental metastasis. In contrast, KDELR1 deficiency enhances metastasis, providing the first example of different disease etiologies within the KDELR-family of retrograde transporters. We show that KDELR3 regulates the metastasis suppressor, KAI1, and report an interaction with the E3 ubiquitin-protein ligase gp78, a regulator of KAI1 degradation. Our work demonstrates that the melanoblast transcriptome can be mined to uncover targetable pathways for melanoma therapy.


Assuntos
Perfilação da Expressão Gênica , Melanoma/genética , Melanoma/metabolismo , Neoplasias Cutâneas/genética , Neoplasias Cutâneas/metabolismo , Transcriptoma , Animais , Linhagem Celular Tumoral , Retículo Endoplasmático , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Proteína Kangai-1/genética , Proteína Kangai-1/metabolismo , Pulmão/patologia , Melanócitos/metabolismo , Melanoma/patologia , Camundongos , Camundongos Endogâmicos C57BL , Metástase Neoplásica/genética , Segunda Neoplasia Primária/patologia , Fenótipo , Receptores de Peptídeos/genética , Receptores de Peptídeos/metabolismo , Neoplasias Cutâneas/patologia , Ubiquitina-Proteína Ligases/metabolismo , Melanoma Maligno Cutâneo
20.
Asian Pac J Cancer Prev ; 20(11): 3321-3329, 2019 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-31759355

RESUMO

BACKGROUND: Metastasis represents a deadly aspect of any cancer including breast cancer, given its high prevalence; treatment of metastatic breast cancer remains a clinically unmet need, which necessitates the exploration of metastasis suppressor genes (MSGs). KAI-1/CD82 is an important member of MSGs; the role of KAI1 has been well explored in prostate cancer, however its role in breast cancer is not fully explored and in fact the results of breast cancer studies are contentious. Thus, the present study aimed to investigate expression of KAI1 at both transcriptional and translational levels in the tissue of breast cancer patients and benign breast disease. Further, we analysed the relationship between expression levels of KAI1 and clinicopathological parameters in breast cancer patients. MATERIALS AND METHODS: mRNA expression was studied by Real time PCR and protein expression was analyzed by both Western blot and Immunohistochemistry. RESULTS: The results of the study indicate that KAI1 expression was remarkably decreased in breast cancer both at the gene and the protein levels (P < 0.05) compared to benign breast disease. In addition, KAI1 expression levels were strongly associated with axillary lymph node status and advanced T stage (p < 0.05), however no association was found with tumor grade, age, menopausal status and receptor status like ER, PR and Her2. CONCLUSION: Low expression of KAI1 might be helpful for predicting the lymph node metastasis and T staging, thus predicts malignant prognosis of breast cancer.
.


Assuntos
Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/patologia , Proteína Kangai-1/metabolismo , Linfonodos/patologia , Adulto , Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Estudos de Casos e Controles , Feminino , Seguimentos , Humanos , Proteína Kangai-1/genética , Linfonodos/metabolismo , Metástase Linfática , Estadiamento de Neoplasias
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...