Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
J Cell Mol Med ; 28(17): e70035, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39245790

RESUMO

Diabetes-related bone loss represents a significant complication that persistently jeopardizes the bone health of individuals with diabetes. Primary cilia proteins have been reported to play a vital role in regulating osteoblast differentiation in diabetes-related bone loss. However, the specific contribution of KIAA0753, a primary cilia protein, in bone loss induced by diabetes remains unclear. In this investigation, we elucidated the pivotal role of KIAA0753 as a promoter of osteoblast differentiation in diabetes. RNA sequencing demonstrated a marked downregulation of KIAA0753 expression in pro-bone MC3T3 cells exposed to a high glucose environment. Diabetes mouse models further validated the downregulation of KIAA0753 protein in the femur. Diabetes was observed to inhibit osteoblast differentiation in vitro, evidenced by downregulating the protein expression of OCN, OPN and ALP, decreasing primary cilia biosynthesis, and suppressing the Hedgehog signalling pathway. Knocking down KIAA0753 using shRNA methods was found to shorten primary cilia. Conversely, overexpression KIAA0753 rescued these changes. Additional insights indicated that KIAA0753 effectively restored osteoblast differentiation by directly interacting with SHH, OCN and Gli2, thereby activating the Hedgehog signalling pathway and mitigating the ubiquitination of Gli2 in diabetes. In summary, we report a negative regulatory relationship between KIAA0753 and diabetes-related bone loss. The clarification of KIAA0753's role offers valuable insights into the intricate mechanisms underlying diabetic bone complications.


Assuntos
Diferenciação Celular , Proteínas Associadas aos Microtúbulos , Osteoblastos , Transdução de Sinais , Animais , Humanos , Masculino , Camundongos , Linhagem Celular , Cílios/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Diabetes Mellitus Experimental/genética , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/genética , Camundongos Endogâmicos C57BL , Osteoblastos/metabolismo , Osteogênese/genética , Proteínas Associadas aos Microtúbulos/metabolismo
2.
Brief Bioinform ; 22(6)2021 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-34013324

RESUMO

Non-small cell lung cancer (NSCLC) is characterized by relatively rapid response to systemic treatments yet inevitable resistance and predisposed to distant metastasis. We thus aimed at performing sequencing analysis to determine genomic events and underlying mechanisms concerning drug resistance in NSCLC. We performed targeted sequencing of 40 medication-relevant genes on plasma samples from 98 NSCLC patients and analyzed impact of genetic alterations on clinical presentation as well as response to systemic treatments. Profiling of multi-omics data from 1024 NSCLC tissues in public datasets was carried out for comparison and validation of identified molecular events implicated in resistance. A genetic association of CYP2D6 deletion with drug resistance was identified through circulating tumor DNA (ctDNA) profiling and response assessment. FCGR3A amplification was potentially involved in resistance to EGFR inhibitors. We further verified our findings in tissue samples and focused on potential resistance mechanisms, which uncovered that depleted CYP2D6 affected a set of genes involved in EMT, oncogenic signaling as well as inflammatory pathways. Tumor microenvironment analysis revealed that NSCLC with CYP2D6 loss manifested increased levels of immunomodulatory gene expressions, PD-L1 expression, relatively high mutational burden and lymphocyte infiltration. DNA methylation alterations were also found to be correlated with mRNA expressions and copy numbers of CYP2D6. Finally, MEK inhibitors were identified by CMap as the prospective therapeutic drugs for CYP2D6 deletion. These analyses identified novel resistance mechanisms to systemic NSCLC treatments and had significant implications for the development of new treatment strategies.


Assuntos
Biomarcadores Tumorais , Carcinoma Pulmonar de Células não Pequenas/genética , Resistencia a Medicamentos Antineoplásicos/genética , Variação Genética , Neoplasias Pulmonares/genética , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/efeitos adversos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/diagnóstico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/mortalidade , Biologia Computacional/métodos , Variações do Número de Cópias de DNA , Metilação de DNA , Bases de Dados Genéticas , Epigênese Genética , Feminino , Genômica/métodos , Humanos , Neoplasias Pulmonares/diagnóstico , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/mortalidade , Masculino , Pessoa de Meia-Idade , Anotação de Sequência Molecular , Mutação , Prognóstico , Transcriptoma
3.
Exp Cell Res ; 402(1): 112551, 2021 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-33675808

RESUMO

Long noncoding RNAs (lncRNAs) are known to play a crucial role in the onset and progression of cervical cancer (CC). Here, the results of RNA microarray and RNA-sequencing dataset analysis showed that lncRNA DLEU2 was significantly upregulated in CC tissues. Clinicopathologic analysis indicated that lncRNA DLEU2 was closely related to tumor topography. Functional experiments and bioinformatics analysis revealed that lncRNA DLEU2 promoted CC cell proliferation and accelerated the cell cycle. Mechanistically, lncRNA DLEU2 promoted the progression of the cell cycle and inhibited the activity of the Notch signaling pathway by inhibiting p53 expression. Additionally, lncRNA DLEU2 probably interacted with ZFP36 Ring Finger Protein (ZFP36) to inhibit the expression of p53. In conclusion, this study revealed the function of lncRNA DLEU2 in CC tumorigenesis, suggesting new therapeutic targets in CC.


Assuntos
Carcinogênese/genética , RNA Longo não Codificante/genética , Tristetraprolina/genética , Proteína Supressora de Tumor p53/genética , Neoplasias do Colo do Útero/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Inativação Gênica , Humanos , Receptores Notch/genética , Transdução de Sinais/genética , Neoplasias do Colo do Útero/patologia
4.
J Gastroenterol Hepatol ; 36(1): 186-195, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32424838

RESUMO

BACKGROUND AND AIM: Long noncoding RNA TRPM2-AS has emerged as a novel regulator in cancer initiation and progression of various cancers. However, the function and underlying mechanism of TRPM2-AS in the progression of gastric cancer (GC) remain poorly understood. METHODS: GEO and TCGA databases were used for isolation of differential lncRNA expression. TRPM2-AS expression levels in GC tissues and cells were measured by quantitative polymerase chain reaction method. TRPM2-AS subcellular location was detected by fluorescence in situ hybridization analysis. The functional roles of TRPM2-AS in cells were analyzed by loss and gain function assays. RESULTS: By using bioinformatics and quantitative polymerase chain reaction methods, TRPM2-AS expression levels were proved to be upregulated in GSE70880 dataset, TCGA database, and 26 GC tissues, which was partly induced by SP1. The results of clinical assays showed that TRPM2-AS could be an indicator for early-stage GC diagnosis. Fluorescence in situ hybridization analysis showed that TRPM2-AS was located in both nucleus and cytoplasm. Functional experiments displayed that knockdown of TRPM2-AS inhibited proliferation, migration, and invasion in GC cells. Furthermore, depression of TRPM2-AS suppressed cell growth though promotion of cell apoptosis. The expression levels of cleaved PARP, caspase 9, caspase 3, and Bax were significantly increased in BGC823 with TRPM2-AS knockdown. In addition, knockdown of TRPM2-AS reduced and phosphorylate signal transducer and activator of transcription 3 and increased and phosphorylate p38 mitogen-activated protein kinase. CONCLUSIONS: This study demonstrated that SP1-regulated TRPM2-AS is involved in GC cell apoptosis probably via p38 mitogen-activated protein kinase and signal transducer and activator of transcription 3 pathways, indicating that TRPM2-AS might be a potential therapeutic target in GC.


Assuntos
Apoptose/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , RNA Longo não Codificante/metabolismo , Fator de Transcrição STAT3/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Canais de Cátion TRPM/metabolismo , Linhagem Celular Tumoral , Progressão da Doença , Humanos , Terapia de Alvo Molecular , Transdução de Sinais/genética , Transdução de Sinais/fisiologia , Neoplasias Gástricas/terapia
5.
J Cell Biochem ; 120(10): 17898-17911, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31135068

RESUMO

Gastric cancer (GC) is a prevalent malignant cancer of digestive system, identification of novel diagnostic and prognostic biomarkers for GC is urgently demanded. The aim of this study was to determine potential long noncoding RNAs (lncRNAs) associated with the pathogenesis and prognosis of GC. Raw noncoding RNA microarray data (GSE53137, GSE70880, and GSE99417) was downloaded from Gene Expression Omnibus (GEO) database. Differentially expressed genes between GC and adjacent normal gastric tissue samples were screened by an integrated analysis of multiple gene expression profile after gene reannotation and batch normalization. Differentially expressed genes were further confirmed by The Cancer Genome Atlas (TCGA) database. Competing endogenous RNA (ceRNA) network, Gene Ontology term and Kyoto Encyclopedia of Genes and Genomes pathway, survival analysis were extensively applied to identify hub lncRNAs and discover potential biomarkers related to diagnosis and prognosis of GC. In total of 246 integrated differential genes including 15 lncRNAs and 241 messenger RNAs (mRNAs) were obtained after intersections of differential genes between GEO and TCGA database. ceRNA network comprised of three lncRNAs (UCA1, HOTTIP, and HMGA1P4), 26 microRNAs (miRNAs) and 72 mRNAs. Functional analysis revealed that three lncRNAs were mainly dominated in cell cycle and cellular senescence. Survival analysis showed that HMGA1P4 was statistically related to the overall survival rate. For the first time, we identified that HMGA1P4, a target of miR-301b/miR-508, is involved in cell cycle and senescence process by regulating CCNA2 in GC. Finally, the expression levels of three lncRNAs were validated to be upregulated in GC tissues. Thus, three lncRNAs including UCA1, HOTTIP, and HMGA1P4 may contribute to GC development and their potential functions might be associated with the prognosis of GC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Genoma Humano , RNA Longo não Codificante/genética , Neoplasias Gástricas/genética , Bases de Dados Genéticas , Feminino , Ontologia Genética , Redes Reguladoras de Genes , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Prognóstico , Mapas de Interação de Proteínas/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Transcriptoma
6.
BMC Med Inform Decis Mak ; 19(1): 198, 2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31640686

RESUMO

BACKGROUND: Breast cancer causes hundreds of thousands of deaths each year worldwide. The early stage diagnosis and treatment can significantly reduce the mortality rate. However, the traditional manual diagnosis needs intense workload, and diagnostic errors are prone to happen with the prolonged work of pathologists. Automatic histopathology image recognition plays a key role in speeding up diagnosis and improving the quality of diagnosis. METHODS: In this work, we propose a breast cancer histopathology image classification by assembling multiple compact Convolutional Neural Networks (CNNs). First, a hybrid CNN architecture is designed, which contains a global model branch and a local model branch. By local voting and two-branch information merging, our hybrid model obtains stronger representation ability. Second, by embedding the proposed Squeeze-Excitation-Pruning (SEP) block into our hybrid model, the channel importance can be learned and the redundant channels are thus removed. The proposed channel pruning scheme can decrease the risk of overfitting and produce higher accuracy with the same model size. At last, with different data partition and composition, we build multiple models and assemble them together to further enhance the model generalization ability. RESULTS: Experimental results show that in public BreaKHis dataset, our proposed hybrid model achieves comparable performance with the state-of-the-art. By adopting the multi-model assembling scheme, our method outperforms the state-of-the-art in both patient level and image level accuracy for BACH dataset. CONCLUSIONS: We propose a novel compact breast cancer histopathology image classification scheme by assembling multiple compact hybrid CNNs. The proposed scheme achieves promising results for the breast cancer image classification task. Our method can be used in breast cancer auxiliary diagnostic scenario, and it can reduce the workload of pathologists as well as improve the quality of diagnosis.


Assuntos
Neoplasias da Mama/patologia , Processamento de Imagem Assistida por Computador , Redes Neurais de Computação , Algoritmos , Mama/patologia , Neoplasias da Mama/classificação , Carcinoma in Situ , Simulação por Computador , Compressão de Dados , Conjuntos de Dados como Assunto , Feminino , Humanos , Invasividade Neoplásica , Sensibilidade e Especificidade
7.
Biochem Biophys Res Commun ; 499(2): 338-344, 2018 05 05.
Artigo em Inglês | MEDLINE | ID: mdl-29574154

RESUMO

In order to better understand the mechanisms underlying the development of papillary thyroid carcinoma (PTC), and to identify new potential biomarkers, high-resolution label-free mass spectrometry was performed on PTC tissues and adjacent normal thyroid tissues from six patients. In this process, 2788 proteins were identified, out of which 49 proteins presented significant differences between PTC tissues and adjacent normal thyroid tissues. Gene ontology revealed that the majority of these proteins are involved in the catalytic activity and binding. We selected three proteins with differential expressions: PDZ and LIM domain 5 (PDLIM5), PDLIM1 and ALDH1A1; Protein expressions were further verified by RT-PCR and western blot. Among these, expression of PDLIM5 and PDLIM1 was up-regulated, while that of ALDH1A1 was down-regulated in PTC tissues. Next, we confirmed their expression through quantitative dot blot (QDB) technique. We found that knockdown of PDLIM5 expression in the B-CPAP cell line could inhibit the migration, invasion and proliferation of PTC cells. In addition, PDLIM5 knockdown reduced Ras and Phospho-ERK1/2 expression. Thus, we suggested that PDLIM5 promotes PTC via activation of the Ras-ERK pathway. Our research provides new molecular insight into the function of PDLIM5, which may assist in studying the mechanism of PTC. In addition, PDLIM5 could be further explored as a potential candidate for PTC treatment.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Biomarcadores Tumorais/metabolismo , Carcinoma Papilar/metabolismo , Proteínas com Domínio LIM/metabolismo , Proteômica , Neoplasias da Glândula Tireoide/metabolismo , Western Blotting , Carcinoma Papilar/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases , Invasividade Neoplásica , Proteínas de Neoplasias/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Reprodutibilidade dos Testes , Coloração e Rotulagem , Câncer Papilífero da Tireoide , Neoplasias da Glândula Tireoide/patologia , Proteínas ras/metabolismo
8.
Biochem Biophys Res Commun ; 488(1): 196-203, 2017 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-28487109

RESUMO

C-C chemokine receptor type 4 has been reported to correlate with lung cancer. However, the role of CCR4 in human non-small cell lung cancer patients is not well defined. Here, we demonstrated that increased expression of CCR4 was associated with clinical stage and CCR4 was an independent risk factor for overall survival in NSCLC patients. Moreover, tumor-infiltrating Treg cells were higher expression than matched adjacent tissues in CCR4+ NSCLC. Higher expression of chemokine CCL17 and CCL22 could recruit Treg cells to tumor sites in NSCLC. Treg in TIL exhibit a higher level of suppressive activity on effector T cells than matched adjacent tissues in NSCLC patients. Significant NK cell reduction was observed in tumor regions compared to non-tumor regions. NK cells demonstrated that reduced the killing capacity against target cells and the expression of CD69 + in vitro. The addition of Treg cells from NSCLC patients efficiently inhibited the anti-tumor ability of autologous NK cells. Treatment with anti-TGF-ß antibody restored the impaired cytotoxic activity of T cells and NK cells from tumor tissues. Our results indicate that TGF-ß plays an important role in impaired Teff cells and NK cells. It will therefore be valuable to develop therapeutic strategies against CCR4 and TGF-ß pathway for therapy of NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Quimiotaxia , Células Matadoras Naturais/metabolismo , Neoplasias Pulmonares/metabolismo , Receptores CCR4/metabolismo , Linfócitos T/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade
9.
Int J Mol Sci ; 18(6)2017 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-28555007

RESUMO

B-Myb is a transcription factor that is overexpressed and plays an oncogenic role in several types of human cancers. However, its potential implication in lung cancer remains elusive. In the present study, we have for the first time investigated the expression profile of B-Myb and its functional impact in lung cancer. Expression analysis by quantificational real-time polymerase chain reaction (qRT-PCR) and immunohistochemistry demonstrated that B-Myb expression is aberrantly overexpressed in non-small cell lung cancer (NSCLC), and positively correlated with pathologic grade and clinical stage of NSCLC. A gain-of-function study revealed that overexpression of B-Myb significantly increases lung cancer cell growth, colony formation, migration, and invasion. Conversely, a loss-of-function study showed that knockdown of B-Myb decreases cell growth, migration, and invasion. B-Myb overexpression also promoted tumor growth in vivo in a NSCLC xenograft nude mouse model. A molecular mechanistic study by RNA-sequencing (RNA-seq) analysis showed that B-Myb overexpression causes up-regulation of various downstream genes (e.g., COL11A1, COL6A1, FN1, MMP2, NID1, FLT4, INSR, and CCNA1) and activation of multiple critical pathways (e.g., extracellular signal-regulated kinases (ERK) and phosphorylated-protein kinase B (Akt) signaling pathways) involved in cell proliferation, tumorigenesis, and metastasis. Collectively, our results indicate a tumor-promoting role for B-Myb in NSCLC and thus imply its potential as a target for the diagnosis and/or treatment of NSCLC.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Proteínas de Ciclo Celular/metabolismo , Transativadores/metabolismo , Carcinoma Pulmonar de Células não Pequenas/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/fisiologia , Proliferação de Células/genética , Proliferação de Células/fisiologia , MAP Quinases Reguladas por Sinal Extracelular/genética , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Análise Serial de Tecidos , Transativadores/genética
10.
Int J Mol Sci ; 18(3)2017 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-28257042

RESUMO

We previously identified proline-rich protein 11 (PRR11) as a novel cancer-related gene that is implicated in the regulation of cell cycle and tumorigenesis. Our recent study demonstrated that PRR11 and its adjacent gene, kinetochore associated 2 (SKA2), constitute a classic head-to-head gene pair that is coordinately regulated by nuclear factor Y (NF-Y). In the present study, we further show that the PRR11-SKA2 bidirectional transcription unit is an indirect target of the tumor suppressor p53. A luciferase reporter assay revealed that overexpression of wild type p53, but not mutant p53, significantly represses the basal activity and NF-Y mediated transactivation of the PRR11-SKA2 bidirectional promoter. Deletion and mutation analysis of the PRR11-SKA2 promoter revealed that p53-mediated PRR11-SKA2 repression is dependent on the presence of functional NF-Y binding sites. Furthermore, a co-immunoprecipitation assay revealed that p53 associates with NF-Y in lung cancer cells, and a chromatin immunoprecipitation assay showed that p53 represses PRR11-SKA2 transcription by reducing the binding amount of NF-Y in the PRR11-SKA2 promoter region. Consistently, the ability of p53 to downregulate PRR11-SKA2 transcription was significantly attenuated upon siRNA-mediated depletion of nuclear factor Y subunit beta (NF-YB). Notably, lung cancer patients with lower expression of either PRR11 or SKA2 along with wild type p53 exhibited the best overall survival compared with others with p53 mutation and/or higher expression of either PRR11 or SKA2. Taken together, our results demonstrate that p53 negatively regulates the expression of the PRR11-SKA2 bidirectional transcription unit through NF-Y, suggesting that the inability to repress the PRR11-SKA2 bidirectional transcription unit after loss of p53 might contribute to tumorigenesis.


Assuntos
Fator de Ligação a CCAAT/metabolismo , Proteínas Cromossômicas não Histona/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Proteínas/genética , Transcrição Gênica , Proteína Supressora de Tumor p53/metabolismo , Sítios de Ligação , Linhagem Celular Tumoral , Humanos , Neoplasias Pulmonares/mortalidade , Prognóstico , Regiões Promotoras Genéticas , Ligação Proteica
11.
Biochim Biophys Acta ; 1849(9): 1133-44, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26162986

RESUMO

Head-to-head gene pairs represent a unique feature of gene organization in eukaryotes, accounting for >10% of genes in the human genome. Identification and functional analysis of such gene pairs is only in its infancy. Recently, we identified PRR11 as a novel cancer-related gene that is implicated in cell cycle and lung cancer. Here we demonstrate that PRR11 is oriented in a head-to-head configuration with its neighboring gene, SKA2. 5'-RACE assay revealed that the intergenic spacer region between the two genes is <500 bp. Serial luciferase reporter assays demonstrated that a minimal 80-bp intergenic region functions as a core bidirectional promoter to drive basal transcription in both the PRR11 and SKA2 orientations. EMSA and ChIP assays demonstrated that NF-Y binds to and directly transactivates the PRR11-SKA2 bidirectional promoter. SiRNA-mediated NF-Y depletion significantly downregulated PRR11 and SKA2 expression. Expression of both PRR11 and SKA2 was significantly upregulated in lung cancer. Expression of the two genes was highly correlated with each other and with NF-Y expression. Remarkably, high expression of both PRR11 and SKA2 was associated with poorer prognosis in lung cancer patients compared with high expression of one gene or low expression of both genes. Knockdown of PRR11 and/or SKA2 remarkably reduced cell proliferation, migration, and invasion in lung cancer cells. Thus, the PRR11-SKA2 bidirectional transcription unit, which is a novel direct target of NF-Y, is essential for the accelerated proliferation and motility of lung cancer cells and may represent a potential target in the diagnosis and/or treatment of human lung cancer.


Assuntos
Fator de Ligação a CCAAT/fisiologia , Proteínas Cromossômicas não Histona/genética , Regulação da Expressão Gênica , Neoplasias Pulmonares/genética , Regiões Promotoras Genéticas , Proteínas/genética , Animais , Sequência de Bases , Linhagem Celular Tumoral , Cromatina/genética , DNA/genética , Humanos , Dados de Sequência Molecular , Homologia de Sequência do Ácido Nucleico , Transcrição Gênica
12.
BMC Cancer ; 16: 545, 2016 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-27464624

RESUMO

BACKGROUND: Directly targeting therapeutic suicide gene to a solid tumor is a hopeful approach for cancer gene therapy. Treatment of a solid tumor by an effective vector for a suicide gene remains a challenge. Given the lack of effective treatments, we constructed a bifidobacterial recombinant thymidine kinase (BF-rTK) -ganciclovir (GCV) targeting system (BKV) to meet this requirement and to explore antitumor mechanisms. METHODS: Bifidobacterium (BF) or BF-rTK was injected intratumorally with or without ganciclovir in a human colo320 intestinal xenograft tumor model. The tumor tissues were analyzed using apoptosis antibody arrays, real time PCR and western blot. The colo320 cell was analyzed by the gene silencing method. Autophagy and necroptosis were also detected in colo320 cell. Meanwhile, three human digestive system xenograft tumor models (colorectal cancer colo320, gastric cancer MKN-45 and liver cancer SSMC-7721) and a breast cancer (MDA-MB-231) model were employed to validate the universality of BF-rTK + GCV in solid tumor gene therapy. The survival rate was evaluated in three human cancer models after the BF-rTK + GCV intratumor treatment. The analysis of inflammatory markers (TNF-α) in tumor indicated that BF-rTK + GCV significantly inhibited TNF-α expression. RESULTS: The results suggested that BF-rTK + GCV induced tumor apoptosis without autophagy and necroptosis occurrence. The apoptosis was transduced by multiple signaling pathways mediated by FasL and TNFR2 and mainly activated the mitochondrial control of apoptosis via Bid and Bim, which was rescued by silencing Bid or/and Bim. However, BF + GCV only induced apoptosis via Fas/FasL signal pathway accompanied with increased P53 expression. We further found that BF-rTK + GCV inhibited the expression of the inflammatory maker of TNF-α. However, BF-rTK + GCV did not result in necroptosis and autophagy. CONCLUSIONS: BF-rTK + GCV induced tumor apoptosis mediated by FasL and TNFR2 through the mitochondrial control of apoptosis via Bid and Bim without inducing necroptosis and autophagy. Furthermore, BF-rTK + GCV showed to repress the inflammation of tumor through downregulating TNF-α expression. Survival analysis results of multiple cancer models confirmed that BF-rTK + GCV system has a wide field of application in solid tumor gene therapy.


Assuntos
Bifidobacterium/fisiologia , Proteína Ligante Fas/genética , Ganciclovir/administração & dosagem , Neoplasias/terapia , Receptores Tipo II do Fator de Necrose Tumoral/genética , Timidina Quinase/genética , Animais , Apoptose , Linhagem Celular Tumoral , Terapia Combinada , Proteína Ligante Fas/metabolismo , Ganciclovir/uso terapêutico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Genes Transgênicos Suicidas , Terapia Genética , Vetores Genéticos/administração & dosagem , Humanos , Camundongos , Neoplasias/genética , Receptores Tipo II do Fator de Necrose Tumoral/metabolismo , Proteínas Recombinantes/genética , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Int J Mol Sci ; 17(6)2016 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-27275821

RESUMO

The herpes simplex virus thymidine kinase/ganciclovir (HSV TK/GCV) system is one of the best studied cancer suicide gene therapy systems. Our previous study showed that caspase 3 expression was upregulated and bladder tumor growth was significantly reduced in rats treated with a combination of Bifidobacterium (BF) and HSV TK/GCV (BF-rTK/GCV). However, it was raised whether the BF-mediated recombinant thymidine kinase combined with ganciclovir (BF-rTK/GCV) was safe to administer via venous for cancer gene therapy. To answer this question, the antitumor effects of BF-rTK/GCV were mainly evaluated in a xenograft nude mouse model bearing MKN-45 gastric tumor cells. The immune response, including analysis of cytokine profiles, was analyzed to evaluate the safety of intramuscular and intravenous injection of BF-rTK in BALB/c mice. The results suggested that gastric tumor growth was significantly inhibited in vivo by BF-rTK/GCV. However, the BF-rTK/GCV had no effect on mouse body weight, indicating that the treatment was safe for the host. The results of cytokine profile analysis indicated that intravenous injection of a low dose of BF-rTK resulted in a weaker cytokine response than that obtained with intramuscular injection. Furthermore, immunohistochemical analysis showed that intravenous administration did not affect the expression of immune-associated TLR2 and TLR4. Finally, the BF-rTK/GCV inhibited vascular endothelial growth factor (VEGF) expression in mouse model, which is helpful for inhibiting of tumor angiogenesis. That meant intravenous administration of BF-rTK/GCV was an effective and safe way for cancer gene therapy.


Assuntos
Bifidobacterium/fisiologia , Ganciclovir/farmacologia , Terapia Genética , Vetores Genéticos/genética , Herpesvirus Humano 1/genética , Neoplasias/genética , Neoplasias/patologia , Timidina Quinase/genética , Administração Intravenosa , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Biomarcadores , Caspase 8/metabolismo , Linhagem Celular Tumoral , Citocinas/genética , Citocinas/metabolismo , Modelos Animais de Doenças , Ganciclovir/administração & dosagem , Expressão Gênica , Terapia Genética/métodos , Vetores Genéticos/administração & dosagem , Vetores Genéticos/efeitos adversos , Humanos , Masculino , Camundongos , Neoplasias/metabolismo , Neoplasias/terapia , Transdução de Sinais/efeitos dos fármacos , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo , Carga Tumoral/efeitos dos fármacos , Carga Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
14.
Tumour Biol ; 36(4): 2875-83, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25492484

RESUMO

Metastasis is the leading cause of death in patients with breast cancer and aberrantly expressed microRNAs (miRNAs) are highly associated with this process. A previous study has shown that miR-335 is downregulated in breast cancer and can suppress tumor invasion and metastasis. Emerging evidences indicate that c-Met is implicated in cell scattering, migration, and invasion. However, little is known about the relationship between miR-335 expression and c-Met alteration in breast cancer. In the present study, we found that miR-335 expression was downregulated and c-Met protein expression was upregulated in two human breast cell lines. MiR-335 was found to negatively regulate c-Met protein level by directly targeting its 3' untranslated region (UTR). Forced expression of miR-335 decreased c-Met expression at protein levels and consequently diminished hepatocyte growth factor (HGF)-induced phosphorylation of c-Met and subsequently inhibited HGF promotion of breast cancer cell migration in a c-Met-dependent manner. MiR-335 expression was increased after 5-aza-2'-deoxycytidine (5-AZA-CdR) treatment, and 5-AZA-CdR treatment resulted in the same phenotype as the effect of miR-335 overexpression. Taken together, these results demonstrate that miR-335 suppresses breast cancer cell migration by negatively regulating the HGF/c-Met pathway.


Assuntos
Neoplasias da Mama/genética , Fator de Crescimento de Hepatócito/metabolismo , MicroRNAs/biossíntese , Proteínas Proto-Oncogênicas c-met/biossíntese , Azacitidina/administração & dosagem , Azacitidina/análogos & derivados , Neoplasias da Mama/patologia , Movimento Celular/efeitos dos fármacos , Proliferação de Células/genética , Decitabina , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , MicroRNAs/genética , Invasividade Neoplásica/genética , Proteínas Proto-Oncogênicas c-met/genética
15.
Can J Microbiol ; 61(2): 124-30, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25587774

RESUMO

A resident plasmid, pBIF10, was isolated from Bifidobacterium longum B200304, and the full-length sequence of pBIF10 was analyzed. In this sequence, we identified at least 17 major open reading frames longer than 200 bp. A tetracycline resistance gene, tetQ, was identified and verified to confer antibiotic resistance to tetracycline. The plasmid replicon with replication protein B gene (repB) and a typical iteron was identified in pBIF10. An artificial clone vector was constructed with the replicon of pBIF10; the results showed that repB controlled plasmid replication in other bifidobacteria host cells at low transformation frequency. Taken together, the analysis and characterization of pBIF10 provided necessary information for the understanding of antibiotic resistance mediated by a plasmid in a Bifidobacterium strain. GC% and repB sequence analyses indicated that pBIF10 was a molecular hybrid of at least 2 other bacterial genera plasmids.


Assuntos
Proteínas de Bactérias/genética , Bifidobacterium/genética , Plasmídeos/genética , Resistência a Tetraciclina/genética , Antibacterianos/química , Sequência de Bases , Fezes , Transferência Genética Horizontal , Vetores Genéticos , Humanos , Dados de Sequência Molecular , Fases de Leitura Aberta , Análise de Sequência de DNA
16.
J Biol Chem ; 288(41): 29680-91, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-23995842

RESUMO

Cullin-RING ubiquitin ligases (CRLs) are the largest family of E3 ligases and require cullin neddylation for their activation. The NEDD8-activating enzyme inhibitor MLN4924 reportedly blocked cullin neddylation and inactivated CRLs, which resulted in apoptosis induction and tumor suppression. However, CRL roles in ovarian cancer cell survival and the ovarian tumor repressing effects of MLN4924 are unknown. We show here that CRL4 components are highly expressed in human epithelial ovarian cancer tissues. MLN4924-induced DNA damage, cell cycle arrest, and apoptosis in ovarian cancer cells in a time- and dose-dependent manner. In addition, MLN4924 sensitized ovarian cancer cells to other chemotherapeutic drug treatments. Depletion of CRL4 components Roc1/2, Cul4a, and DDB1 had inhibitory effects on ovarian cancer cells similar to MLN4924 treatment, which suggested that CRL4 inhibition contributed to the chemotherapeutic effect of MLN4924 in ovarian cancers. We also investigated for key CRL4 substrate adaptors required for ovarian cancer cells. Depleting Vprbp/Dcaf1 did not significantly affect ovarian cancer cell growth, even though it was expressed by ovarian cancer tissues. However, depleting Cdt2/Dcaf2 mimicked the pharmacological effects of MLN4924 and caused the accumulation of its substrate, CDT1, both in vitro and in vivo. MLN4924-induced DNA damage and apoptosis were partially rescued by Cdt1 depletion, suggesting that CRL4(CDT2) repression and CDT1 accumulation were key biochemical events contributing to the genotoxic effects of MLN4924 in ovarian cancer cells. Taken together, these results indicate that CRL4(CDT2) is a potential drug target in ovarian cancers and that MLN4924 may be an effective anticancer agent for targeted ovarian cancer therapy.


Assuntos
Ciclopentanos/farmacologia , Neoplasias Epiteliais e Glandulares/tratamento farmacológico , Proteínas Nucleares/genética , Neoplasias Ovarianas/tratamento farmacológico , Pirimidinas/farmacologia , Ubiquitina-Proteína Ligases/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Western Blotting , Carcinoma Epitelial do Ovário , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Dano ao DNA , Relação Dose-Resposta a Droga , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Imuno-Histoquímica , Camundongos , Camundongos Nus , Neoplasias Epiteliais e Glandulares/genética , Neoplasias Epiteliais e Glandulares/patologia , Proteínas Nucleares/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/patologia , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Ubiquitina-Proteína Ligases/metabolismo
17.
Mol Cell Biochem ; 392(1-2): 95-107, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24633962

RESUMO

MCPH1, initially identified as an hTERT repressor, has recently been implicated in mediating DNA damage response and maintaining chromosome integrity. This study is to investigate its potential role in the onset of cervical cancer. In the study, decreased expression of MCPH1 was observed in 19 of 31 cases (61.3%) at mRNA level and 44 of 63 cases (69.8%) at protein level of cervical tumor tissues compared with the paired nontumor tissues. Reduced MCPH1 protein expression was significantly associated with high-tumor grade (1 vs. 3 P = 0.013; 2 vs. 3 P = 0.047). In addition to inhibit SiHa cell migration and invasion, the overexpression of MCPH1 inhibited cervical cancer cells growth through inducing S phase arrest and mitochondrial apoptosis. Further analysis demonstrated cyclinA2/CDK2, CDC25C-cyclinB/CDC2, and p53/p21 pathways were involved in the MCPH1 overexpression-induced S phase arrest. Moreover, the overexpression of MCPH1 activated mitochondrial apoptosis through regulating several apoptosis-related proteins such as p53, Bcl-2, Bax, cytochrome c, caspase-3, and PARP-1. Our findings indicate that downregulated MCPH1 correlates with tumor progression in cervical cancer, and MCPH1 has an important role in regulating cell growth through regulating the cell cycle and apoptosis. Thus, it may be a crucial tumor suppressor gene and a novel candidate therapeutic target for cervical cancer.


Assuntos
Caspase 3/metabolismo , Proteínas de Ciclo Celular/fisiologia , Divisão Celular/fisiologia , Citocromos c/metabolismo , Proteínas do Tecido Nervoso/fisiologia , Neoplasias do Colo do Útero/patologia , Sequência de Bases , Linhagem Celular , Proteínas do Citoesqueleto , Primers do DNA , Feminino , Humanos , Proteínas do Tecido Nervoso/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais , Neoplasias do Colo do Útero/enzimologia , Neoplasias do Colo do Útero/metabolismo
18.
Med Oncol ; 41(7): 174, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38869721

RESUMO

Cervical cancer (CC), one of the most aggressive tumors in women, has high risk rates of recurrence and metastasis. It is essential to study the key genes and proteins involved in CC development. IRTKS, a member of the IRSp53 family, has been reported as a tumor promoter in gastric and breast cancers. However, the biological role of IRTKS in CC is still unclear. The purpose of this study was to explore the biological function of IRTKS in CC cells in vitro and the effect of IRTKS on tumorigenesis in vivo. Siha and Hela cells were treated with si-RNA and plasmids. Cell proliferation and growth were detected by CCK8, colony formation assay and nude mouse tumorigenicity assay, respectively. Transwell assay was used to analyze cell migration and invasion. The expression of epithelial-mesenchymal transition (EMT)-related proteins was determined by western blot. IRTKS was highly expressed in CC. IRTKS contributed to the proliferation of CC cells in vitro and in vivo. Furthermore, IRTKS facilitated the migration and invasion of CC cells and modulated EMT. IRTKS plays a crucial role in CC tumorigenesis, suggesting it may be a potential key gene for new therapeutic strategies in CC.


Assuntos
Movimento Celular , Proliferação de Células , Transição Epitelial-Mesenquimal , Camundongos Nus , Neoplasias do Colo do Útero , Animais , Feminino , Humanos , Camundongos , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Progressão da Doença , Transição Epitelial-Mesenquimal/genética , Células HeLa , Camundongos Endogâmicos BALB C , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/genética , Neoplasias do Colo do Útero/metabolismo
19.
Front Immunol ; 15: 1390650, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39221250

RESUMO

Different host proteins target different HIV proteins and antagonize their functions, depending on the stage of the HIV life cycle and the stage of infection. Concurrently, HIV proteins also target and antagonize various different host proteins to facilitate HIV replication within host cells. The preceding quite specific area of knowledge in HIV pathogenesis, however, remains insufficiently understood. We therefore propose, in this review article, to examine and discuss the HIV proteins that counteract those host restriction proteins which results directly in increased infectivity of HIV. We elaborate on HIV proteins that antagonize host cellular proteins to promote HIV replication, and thus HIV infection. We examine the functions and mechanisms via which Nef, Vif, Vpu, Env, Vpr, and Vpx counteract host proteins such as Ser5, PSGL-1, IFITMS, A3G, tetherin, GBP5, SAMHD1, STING, HUSH, REAF, and TET2 to increase HIV infectivity. Nef antagonizes three host proteins, viz., Ser5, PSGL1, and IFITIMs, while Vpx also antagonizes three host restriction factors, viz., SAMHD1, STING, and HUSH complex; therefore, these proteins may be potential candidates for therapeutic intervention in HIV infection. Tetherin is targeted by Vpu and Env, PSGL1 is targeted by Nef and Vpu, while Ser5 is targeted by Nef and Env proteins. Finally, conclusive remarks and future perspectives are also presented.


Assuntos
Infecções por HIV , HIV-1 , Interações Hospedeiro-Patógeno , Proteínas do Vírus da Imunodeficiência Humana , Humanos , Infecções por HIV/metabolismo , Infecções por HIV/virologia , Infecções por HIV/imunologia , Proteínas do Vírus da Imunodeficiência Humana/metabolismo , HIV-1/fisiologia , Replicação Viral , Animais , Fatores de Restrição Antivirais
20.
Mol Immunol ; 173: 10-19, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39004021

RESUMO

Adjuvant is a major supplementary component of vaccines to boost adaptive immune responses. To select an efficient adjuvant from the heat-labile toxin B subunit (LTB) of E. coli, four LTB mutants (numbered LTB26, LTB34, LTB57, and LTB85) were generated by multi-amino acid random replacement. Mice have been intranasally vaccinated with human rotavirus VP8 admixed. Among the four mutants, enzyme-linked immunosorbent assay (ELISA) revealed that LTB26 had enhanced mucosal immune adjuvanticity compared to LTB, showing significantly enhanced immune responses in both serum IgG and mucosal sIgA levels. The 3D modeling analysis suggested that the enhanced immune adjuvanticity of LTB26 might be due to the change of the first LTB α-helix to a ß-sheet. The molecular mechanism was studied using transcriptomic and flow cytometric (FCM) analysis. The transcriptomic data demonstrated that LTB26 enhanced immune response by enhancing B cell receptor (BCR) and major histocompatibility complex (MHC) II+-related pathways. Furthermore, LTB26 promoted Th1 and Th2-type immune responses which were confirmed by detecting IFN-γ and IL-4 expression levels. Immunohistochemical analysis demonstrated that LTB26 enhanced both Th1 and Th2 type immunity. Therefore, LTB26 was a potent mucosal immune adjuvant meeting the requirement for use in human clinics in the future.


Assuntos
Adjuvantes Imunológicos , Enterotoxinas , Proteínas de Escherichia coli , Animais , Feminino , Humanos , Camundongos , Antígenos Virais/imunologia , Antígenos Virais/genética , Toxinas Bacterianas/imunologia , Toxinas Bacterianas/genética , Proteínas do Capsídeo/imunologia , Proteínas do Capsídeo/genética , Enterotoxinas/imunologia , Enterotoxinas/genética , Escherichia coli/imunologia , Proteínas de Escherichia coli/imunologia , Proteínas de Escherichia coli/genética , Imunidade nas Mucosas/imunologia , Imunoglobulina G/imunologia , Camundongos Endogâmicos BALB C , Mutação , Rotavirus/imunologia , Infecções por Rotavirus/imunologia , Vacinas contra Rotavirus/imunologia , Vacinas contra Rotavirus/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA