Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Int J Mol Sci ; 23(19)2022 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-36232466

RESUMO

The zinc fingers and homeobox (ZHX) family includes ZHX1, ZHX2, and ZHX3, and their proteins have similar unique structures, containing two C2H2-type zinc finger motifs and four or five HOX-like homeodomains. The members of the ZHX family can form homodimers or heterodimers with each other or with a subunit of nuclear factor Y. Previous studies have suggested that ZHXs can function as positive or negative transcriptional regulators. Recent studies have further revealed their biological functions and underlying mechanisms in cancers. This review summarized the advances of ZHX-mediated functions, including tumor-suppressive and oncogenic functions in cancer formation and progression, the molecular mechanisms, and regulatory functions, such as cancer cell proliferation, migration, invasion, and metastasis. Moreover, the differential expression levels and their association with good or poor outcomes in patients with various malignancies and differential responses to chemotherapy exert opposite functions of oncogene or tumor suppressors. Therefore, the ZHXs act as a double-edged sword in cancers.


Assuntos
Genes Homeobox , Neoplasias , Proteínas de Homeodomínio/metabolismo , Humanos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Fatores de Transcrição/metabolismo , Dedos de Zinco
2.
Br J Cancer ; 123(9): 1445-1455, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32814835

RESUMO

BACKGROUND: E2F transcription factors are considered to be important drivers of tumour growth. E2F7 is an atypical E2F factor, and its role in glioblastoma remains undefined. METHODS: E2F7 expression was examined in patients by IHC and qRT-PCR. The overall survival probability was determined by statistical analyses. MTT assay, colony formation, cell-cycle assay, cell metastasis and the in vivo model were employed to determine the functional role of E2F7 in glioblastoma. Chromatin immunoprecipitation, luciferase assay and western blot were used to explore the underlying mechanisms. RESULTS: E2F7 was found to be up-regulated in glioblastoma patients, and high E2F7 expression was associated with poor overall survival in glioblastoma patients. Functional studies showed that E2F7 promoted cell proliferation, cell-cycle progression, cell metastasis and tumorigenicity abilities in vitro and in vivo. E2F7 promoted the transcription of EZH2 by binding to its promoter and increased H3K27me3 level. EZH2 recruited H3K27me3 to the promoter of PTEN and inhibited PTEN expression, and then activated the AKT/mTOR signalling pathway. In addition, restored expression of EZH2 recovered the abilities of cell proliferation and metastasis in E2F7-silencing cells. CONCLUSION: Collectively, our findings indicate that E2F7 promotes cell proliferation, cell metastasis and tumorigenesis via EZH2-mediated PTEN/AKT/mTOR pathway in glioblastoma.


Assuntos
Neoplasias Encefálicas/patologia , Fator de Transcrição E2F7/fisiologia , Proteína Potenciadora do Homólogo 2 de Zeste/fisiologia , Glioblastoma/patologia , PTEN Fosfo-Hidrolase/genética , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Proliferação de Células/genética , Células Cultivadas , Progressão da Doença , Regulação Neoplásica da Expressão Gênica , Glioblastoma/genética , Glioblastoma/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Nus , Metástase Neoplásica , PTEN Fosfo-Hidrolase/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/fisiologia , Serina-Treonina Quinases TOR/metabolismo
3.
Br J Cancer ; 123(5): 730-741, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32528119

RESUMO

BACKGROUND: The anticancer potential of ibuprofen has created a broad interest to explore the clinical benefits of ibuprofen in cancer therapy. However, the current understanding of the molecular mechanisms involved in the anticancer potential of ibuprofen remains limited. METHODS: Cancer stemness assays to validate ibuprofen function in vitro and in vivo. Histone modification assays to check the effect of ibuprofen on histone acetylation/methylation, as well as the activity of HDAC and KDM6A/B. Inhibitors' in vivo assays to evaluate therapeutic effects of various inhibitors' combination manners. RESULTS: In our in vitro studies, we report that ibuprofen diminishes cancer cell stemness properties that include reducing the ALDH + subpopulation, side population and sphere formation in three cancer types. In our in vivo studies, we report that ibuprofen decreases tumour growth, metastasis and prolongs survival. In addition, our results showed that ibuprofen inhibits inflammation-related stemness gene expression (especially ICAM3) identified by a high-throughput siRNA platform. In regard to the underlying molecular mechanism of action, we report that ibuprofen reduces HDACs and histone demethylase (KDM6A/B) expression that mediates histone acetylation and methylation, and suppresses gene expression via a COX2-dependent way. In regard to therapeutic strategies, we report that ibuprofen combined HDAC/HDM inhibitors prevents cancer progression in vivo. CONCLUSIONS: The aforementioned findings suggest a molecular model that explains how ibuprofen diminishes cancer cell stemness properties. These may provide novel targets for therapeutic strategies involving ibuprofen in the prevention of cancer progression.


Assuntos
Ciclo-Oxigenase 2/metabolismo , Histonas/metabolismo , Ibuprofeno/farmacologia , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Células A549 , Acetilação/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Feminino , Células Hep G2 , Histona Desacetilases/metabolismo , Humanos , Molécula 3 de Adesão Intercelular/metabolismo , Metilação/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Camundongos Nus , Camundongos SCID , Metástase Neoplásica , Neoplasias/metabolismo , Células-Tronco Neoplásicas/metabolismo , Distribuição Aleatória
4.
Int J Mol Sci ; 20(21)2019 Oct 30.
Artigo em Inglês | MEDLINE | ID: mdl-31671668

RESUMO

6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3 (PFKFB3), a glycolytic enzyme highly expressed in cancer cells, has been reported to participate in regulating metabolism, angiogenesis, and autophagy. Although anti-cancer drug oxaliplatin (Oxa) effectively inhibits cell proliferation and induces apoptosis, the growing resistance and side-effects make it urgent to improve the therapeutic strategy of Oxa. Although Oxa induces the autophagy process, the role of PFKFB3 in this process remains unknown. In addition, whether PFKFB3 affects the cytotoxicity of Oxa has not been investigated. Here, we show that Oxa-inhibited cell proliferation and migration concomitant with the induction of apoptosis and autophagy in SW480 cells. Both inhibition of autophagy by small molecule inhibitors and siRNA modification decreased the cell viability loss and apoptosis induced by Oxa. Utilizing quantitative PCR and immunoblotting, we observed that Oxa increased PFKFB3 expression in a time- and dose-dependent manner. Meanwhile, suppression of PFKFB3 attenuated both the basal and Oxa-induced autophagy, by monitoring the autophagic flux and phosphorylated-Ulk1, which play essential roles in autophagy initiation. Moreover, PFKFB3 inhibition further inhibited the cell proliferation/migration, and cell viability decreased by Oxa. Collectively, the presented data demonstrated that PFKFB3 inhibition attenuated Oxa-induced autophagy and enhanced its cytotoxicity in colorectal cancer cells.


Assuntos
Neoplasias do Colo/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Oxaliplatina/farmacologia , Fosfofrutoquinase-2/antagonistas & inibidores , Autofagia/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Neoplasias do Colo/tratamento farmacológico , Relação Dose-Resposta a Droga , Humanos , Fosfofrutoquinase-2/genética , Fosfofrutoquinase-2/metabolismo , RNA Interferente Pequeno/farmacologia , Bibliotecas de Moléculas Pequenas/farmacologia , Fatores de Tempo , Regulação para Cima
5.
Biochim Biophys Acta Mol Basis Dis ; 1864(8): 2566-2578, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29729315

RESUMO

ICAM3 was reported to promote metastasis in tumors. However, the underlying mechanism remains elusive. Here, we disclosed that the expression of ICAM3 was closely correlated with the TNM stage of human breast and lung cancer, as well as the dominant overexpression in high aggressive tumor cell lines (231 and A549 cells). Moreover, the knockdown of ICAM3 inhibited tumor metastasis whereas the ectopic expression of ICAM3 promoted tumor metastasis both in vitro and in vivo. In addition, exploration of the underlying mechanism demonstrated that ICAM3 not only binds to LFA-1 with its extracellular domain and structure protein ERM but also to lamellipodia with its intracellular domain which causes a tension that pulls cells apart (metastasis). Furthermore, ICAM3 extracellular or intracellular mutants alternatively abolished ICAM3 mediated tumor metastasis in vitro and in vivo. As a therapy strategy, LFA-1 antibody or Lifitegrast restrained tumor metastasis via targeting ICAM3-LFA-1 interaction. In summary, the aforementioned findings suggest a model of ICAM3 in mediating tumor metastasis. This may provide a promising target or strategy for the prevention of tumor metastasis.


Assuntos
Antígenos CD/metabolismo , Neoplasias da Mama/metabolismo , Moléculas de Adesão Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Neoplasias Pulmonares/metabolismo , Antígeno-1 Associado à Função Linfocitária/metabolismo , Proteínas de Neoplasias/metabolismo , Fatores de Transcrição/metabolismo , Células A549 , Animais , Antígenos CD/genética , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Moléculas de Adesão Celular/genética , Proteínas de Ligação a DNA/genética , Feminino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Antígeno-1 Associado à Função Linfocitária/genética , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Metástase Neoplásica , Proteínas de Neoplasias/genética , Fatores de Transcrição/genética
6.
J Biol Inorg Chem ; 23(6): 939-947, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30014256

RESUMO

Arsenic trioxide (As2O3) induces cell apoptosis and reduces the invasive and metastatic activities in various cancer types. However, the role of As2O3 in ovarian cancer angiogenesis remains unclear. In this study, we investigated the role of As2O3 in ovarian cancer angiogenesis and found that a low concentration of As2O3 causes no effects on epithelial ovarian cancer cell viability or apoptosis. Moreover, we found that As2O3-treated epithelial ovarian cancer cells demonstrate a reduced tube formation of endothelial cells in Matrigel. In addition, As2O3-treated epithelial ovarian cancer cells show a decreased VEGFA, VEGFR2 and CD31 mRNA expression. As per the underlying mechanisms involved in As2O3 treatment, we found that As2O3 inhibits VEGFA and VEGFR2 expression that thereby inhibits the VEGFA-VEGFR2-PI3K/ERK signaling pathway. This leads to a suppression in both VEGFA synthesis and angiogenesis-related gene expression. A decreased VEGFA synthesis and secretion also inhibits the VEGFA-VEGFR2-PI3K/ERK signaling pathway in human umbilical vein endothelial cells (HUVECs). In summary, our results may provide strategies for the use of As2O3 in the prevention of tumor angiogenesis.


Assuntos
Apoptose , Trióxido de Arsênio/farmacologia , Carcinoma Epitelial do Ovário/irrigação sanguínea , Neovascularização Patológica/prevenção & controle , Neoplasias Ovarianas/irrigação sanguínea , Trióxido de Arsênio/administração & dosagem , Carcinoma Epitelial do Ovário/metabolismo , Carcinoma Epitelial do Ovário/patologia , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Feminino , Humanos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , RNA Mensageiro/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
7.
Int J Mol Sci ; 19(9)2018 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-30231491

RESUMO

Gut dysbiosis is associated with colitis-associated colorectal carcinogenesis, and the genetic deficiency of the Muc2 gene causes spontaneous development of colitis and colorectal cancer. Whether there are changes of gut microbiota and a linkage between the changes of microbiota and intestinal pathology in Muc2-/- mice are unclear. Muc2-/- and Muc2+/+ mice were generated by backcrossing from Muc2+/- mice, and the fecal samples were collected at different dates (48th, 98th, 118th, 138th, and 178th day). Gut microbiota were analyzed by high-throughput sequencing with the universal 16S rRNA primers (V3⁻V5 region). All mice were sacrificed at day 178 to collect colonic tissue and epithelial cells for the analysis of histopathology and inflammatory cytokines. On the 178th day, Muc2-/- mice developed colorectal chronic colitis, hyperplasia, adenomas and adenocarcinomas, and inflammatory cytokines (e.g., cyclooxygenase 2 (COX-2), interleukin 6 (IL-6), tumor necrosis factor-α (TNF-α), interleukin 1 ß (IL-1ß), i-kappa-B-kinase ß (IKKß)) were significantly increased in colonic epithelial cells of Muc2-/- mice. In general, structural segregation of gut microbiota was observed throughout the experimental time points between the Muc2-/- and Muc2+/+ mice. Impressively, in Muc2-/- mice, Alpha diversities reflected by Shannon and Chao indexes were higher, the phylum of Firmicutes was enriched and Bacteroidetes was decreased, and Desulfovibrio, Escherichia, Akkermansia, Turicibacter, and Erysipelotrichaceae were significantly increased, but Lactobacilli and Lachnospiraceae were significantly decreased. Moreover, the abundance of Ruminococcaceae and butyrate-producing bacteria was significantly higher in the Muc2-/- mice. There were significant differences of gut microbiota between Muc2-/- and Muc2+/+ mice. The dynamic changes of microbiota might contribute to the development of colitis and colitis-associated colorectal carcinogenesis. Therefore, this study revealed specific functional bacteria in the development of colitis and colitis-associated colorectal carcinogenesis, which will benefit the development of preventive and therapeutic strategies for chronic inflammation and its malignant transformation.


Assuntos
Colite/genética , Colite/microbiologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/microbiologia , Microbioma Gastrointestinal , Deleção de Genes , Mucina-2/genética , Animais , Carcinogênese/genética , Carcinogênese/patologia , Colite/complicações , Colite/patologia , Neoplasias Colorretais/etiologia , Neoplasias Colorretais/patologia , Disbiose/complicações , Disbiose/genética , Disbiose/microbiologia , Disbiose/patologia , Masculino , Camundongos
8.
Int J Mol Sci ; 18(4)2017 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-28441730

RESUMO

Colorectal cancer is one of the most common malignancies and is the second-leading cause of cancer-related death world-wide, which is linked to genetic mutations, epigenetic alterations, and oncogenic signaling activation. MicroRNAs, one of the categories of epigenetics, have been demonstrated significant roles in carcinogenesis and progression through regulating of oncogenic signaling pathways, stem cells, epithelial-mesenchymal transition, and metastasis. This review summarizes the roles of microRNAs in the regulating of Wnt, Ras, TGF-ß, and inflammatory signaling pathways, stemness, and epithelial-mesenchymal transition, for carcinogenesis and metastasis in colorectal cancer. Improving our understanding of the mechanisms of regulatory interactions of microRNAs with signaling pathways in colorectal cancer formation and progression will aid in determining the genes responsible for colorectal cancer initiation, progression, metastasis, and recurrence and, finally, in developing personalized approaches for cancer prevention and therapy.


Assuntos
Neoplasias Colorretais/patologia , MicroRNAs/metabolismo , Neoplasias Colorretais/genética , Transição Epitelial-Mesenquimal , Redes Reguladoras de Genes , Humanos , Metástase Neoplásica , Células-Tronco Neoplásicas/citologia , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais , Fator de Crescimento Transformador beta/metabolismo , Proteínas Wnt/metabolismo , Proteínas ras/metabolismo
9.
Int J Mol Sci ; 18(4)2017 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-28350360

RESUMO

Colorectal cancer (CRC) is one of the most common malignant diseases worldwide, but the involved signaling pathways and driven-genes are largely unclear. This study integrated four cohorts profile datasets to elucidate the potential key candidate genes and pathways in CRC. Expression profiles GSE28000, GSE21815, GSE44076 and GSE75970, including 319 CRC and 103 normal mucosa, were integrated and deeply analyzed. Differentially expressed genes (DEGs) were sorted and candidate genes and pathways enrichment were analyzed. DEGs-associated protein-protein interaction network (PPI) was performed. Firstly, 292 shared DEGs (165 up-regulated and 127 down-regulated) were identified from the four GSE datasets. Secondly, the DEGs were clustered based on functions and signaling pathways with significant enrichment analysis. Thirdly, 180 nodes/DEGs were identified from DEGs PPI network complex. Lastly, the most significant 2 modules were filtered from PPI, 31 central node genes were identified and most of the corresponding genes are involved in cell cycle process, chemokines and G protein-coupled receptor signaling pathways. Taken above, using integrated bioinformatical analysis, we have identified DEGs candidate genes and pathways in CRC, which could improve our understanding of the cause and underlying molecular events, and these candidate genes and pathways could be therapeutic targets for CRC.


Assuntos
Neoplasias Colorretais/genética , Biologia Computacional/métodos , Redes Reguladoras de Genes , Transcriptoma , Análise por Conglomerados , Bases de Dados Genéticas , Regulação Neoplásica da Expressão Gênica , Predisposição Genética para Doença , Humanos , Análise de Sequência com Séries de Oligonucleotídeos
10.
Int J Mol Sci ; 18(10)2017 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-28991193

RESUMO

Sphingosine kinase 1 (Sphk1) is a highly conserved lipid kinase that phosphorylates sphingosine to form sphingosine-1-phosphate (S1P). Growing studies have demonstrated that Sphk1 is overexpressed in various types of solid cancers and can be induced by growth factors, cytokines, and carcinogens, leading to the increase of S1P production. Subsequently, the increased Sphk1/S1P facilitates cancer cell proliferation, mobility, angiogenesis, invasion, and metastasis. Therefore, Sphk1/S1P signaling plays oncogenic roles. This review summarizes the features of Sphk1/S1P signaling and their functions in colorectal cancer cell growth, tumorigenesis, and metastasis, as well as the possible underlying mechanisms.


Assuntos
Neoplasias Colorretais/enzimologia , Neoplasias Colorretais/metabolismo , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Animais , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Humanos , Lisofosfolipídeos/metabolismo , Transdução de Sinais/fisiologia , Esfingosina/análogos & derivados , Esfingosina/metabolismo
11.
Am J Clin Exp Urol ; 11(6): 578-593, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38148933

RESUMO

Prostate cancer is a health-threaten disease in men worldwide, however, lacking is the reliable biomarkers for patient management. Aberrant metabolic events including glucose metabolism are involved in prostate cancer progression. To examine the involvement of glucose metabolic pathways in prostate cancer, we analyzed the expression profiles of glucose transporter family genes using multiple RNA-seq datasets. Our results showed that three SLC2A family genes (SLC2A4/5/9) were significantly downregulated in primary prostate cancers compared to their benign compartments. These down-regulated expressions were inversely correlated with their gene promoter methylation and genome abnormalities. Among these three SLC2A genes, only SLC2A4 showed a significantly reverse correlation with all clinicopathological parameters, including TNM stage, disease relapse, Gleason score, disease-specific survival, and progression-free interval. In addition, the expression levels of these three genes were strongly correlated with anti-cancer immune cell filtration in primary prostate cancers. In a group of patients with early-onset prostate cancers, SLC2A4 also showed a strong negative correlation with multiple clinicopathological parameters, such as tumor mutation burden, biochemical relapse, pre-surgical PSA levels, and Gleason score but a positive correlation with progression-free interval after surgery. In metastatic castration-resistant prostate cancers (CRPC), SLC2A9 gene expression but not SLC2A4 or SLC2A5 genes showed a significant correlation with androgen receptor (AR) activity score and neuroendocrinal (NE) activity score. Meanwhile, SLC2A2/9/13 expression was significantly elevated in CRPC tumors with neuroendocrinal features compared to those without NE features. On the other hand, SLC2A10 and SlC2A12 gene expression were significantly reduced in NEPC tumors compared to CRPC tumors. Consistently, SLC2A10/12 expression levels were significantly reduced in castrated animals carrying the LuCaP35 xenograft models. Survival outcome analysis revealed that SLC2A4 expression in primary tumors is a favorable prognostic factor and SLC2A6 is a worse prognostic factor for disease-specific survival and progression-free survival in prostate cancer patients. In conclusion, our results suggest that SLC2A4/6 expressions are strong prognostic factors for prostate cancer progression and survival. The significance of SLC2A2/9/13 over-expression during NEPC progression needs more investigation.

12.
Mol Biol Rep ; 39(3): 2515-22, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21667252

RESUMO

With the ultimate aim of producing an RNA interference-mediated transgenic pig that is resistant to porcine reproductive and respiratory syndrome virus (PRRSV), we have investigated the effect of RNA interference (RNAi) on silencing the expression of viral genes in the MARC-145 cell line. Twenty small interfering RNAs (siRNAs) were designed and screened for their ability to suppress the expression of the genes ORF1b, 5, 6, and 7 from the highly virulent isolate, PRRSV-JXwn06. Of these siRNAs, the four most effective were selected and four short hairpin RNA (shRNA) expression vectors (pGenesil-1-1b-135, pGenesil-1-1b-372, pGenesil-1-6-135, and pGenesil-1-6-169) targeting ORF1b and ORF6 were constructed and delivered into MARC-145 cells. These cells were then infected with JXwn06. All four vectors inhibited the PRRSV-specific cytopathic effect (CPE). The virus titers in cells transfected with pGenesil-1-1b-135, pGenesil-1-1b-372, pGenesil-1-6-135, and pGenesil-1-6-169 were lower than that of control cells by approximately 150-, 600-, 2.3- and 1.7-fold, respectively. In addition, the expression levels of ORF1 and ORF6 were reduced compared with controls. The unglycosylated membrane protein M, encoded by ORF6, was not detectable in cells transfected with shRNA expression vectors. These results verified that RNAi can effectively inhibit PRRSV-JXwn06 replication in cultured cells in vitro. The four shRNA expression vectors are an initial step in the production of transgenic pigs with PRRSV resistance.


Assuntos
Regulação Viral da Expressão Gênica/genética , Genes Virais/genética , Vírus da Síndrome Respiratória e Reprodutiva Suína/fisiologia , Interferência de RNA , Replicação Viral/genética , Animais , Western Blotting , Linhagem Celular , Vetores Genéticos/genética , Haplorrinos , Vírus da Síndrome Respiratória e Reprodutiva Suína/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transfecção
13.
Am J Cancer Res ; 12(4): 1824-1842, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35530294

RESUMO

Acyl-coenzyme A synthetase medium chain family member 1 (ACSM1) is a medium chain Acyl-CoA Synthetase family member and plays an important role in fatty acid metabolism. The oncogenic roles of ACSM1 are largely unknown. Using comprehensive approaches, we analyzed gene expression profiles and genomic datasets and identified that the expression of ACSM1 was specifically increased in prostate cancer in comparison to the adjacent non-tumor tissues. The increased expression of ACSM1 was associated with increased risks of poor prognosis and shorter survival time. Moreover, genomic copy number alterations of ACSM1, including deletion, amplification, and amino acid changes were frequently observed in prostate cancers, although these mutations did not correlate with gene expression levels. However, ACSM1 gene amplifications were significantly corrected with increased risks of prostate cancer metastasis, and ACSM1 genetic alterations were significantly associated with worse disease-free. And progress-free survival. Gene function stratification and gene set enrichment analysis revealed that the oncogenic roles of ACSM1 in prostate cancer were mainly through metabolic pathways and extracellular matrix (ECM)-receptor interaction signaling pathways, but not associated with microenvironmental immunological signaling pathways, and that ACSM1 expression was not associated with immune cell infiltration in the cancer microenvironment or prostate cancer immune subtypes. In conclusion, the present work has demonstrated that ACSM1 can be specifically and significantly elevated in prostate cancer. ACSM1 gene expression and genomic amplification exhibit important clinical significance through metabolic and ECM-receptor interaction signaling pathways. Thus, ACSM1 may be a novel oncogene and serve as a biomarker for prostate cancer screening and prognosis prediction, and/or a therapeutic target.

14.
Immunogenetics ; 63(10): 667-78, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21660591

RESUMO

In humans, classical MHC class II molecules include DQ, DR, and DP, which are similar in structure but consist of distinct α- and ß-chains. The genes encoding these molecules are all located in the MHC class II gene region. In non-mammalian vertebrates such as chickens, only a single class II α-chain gene corresponding to the human DRA has been identified. Here, we report a characterization of the duck MHC class II α-chain (Anpl-DRA) encoding gene, which contains four exons encoding a typical signal peptide, a peptide-binding α1 domain, an immunoglobulin-like α2 domain, and Tm/Cyt, respectively. This gene is present in the duck genome as a single copy and is highly expressed in the spleen. Sequencing of cDNA and genomic DNA of the Anpl-DRA of different duck individuals/strains revealed low levels of genetic polymorphism, especially in the same strain, although most duck individuals have two different alleles. Otherwise, we found that the duck gene is located next to class II ß genes, which is the same as in humans but different from the situation in chickens.


Assuntos
Patos/imunologia , Genes MHC da Classe II , Sequência de Aminoácidos , Animais , Sequência de Bases , Patos/genética , Éxons , Dosagem de Genes , Dados de Sequência Molecular , Filogenia , Polimorfismo Genético , Baço/metabolismo
15.
Am J Cancer Res ; 11(5): 2062-2080, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34094669

RESUMO

Cancer cells prone to utilize aerobic glycolysis other than oxidative phosphorylation to sustain its continuous cell activity in the stress microenvironment. Meanwhile, cancer cells generally suffer from genome instability, and both radiotherapy and chemotherapy may arouse DNA strand break, a common phenotype of genome instability. Glycolytic enzyme PFKFB3 (6-Phosphofructo-2-kinase/fructose-2,6-bisphosphatase isoform 3), plays essential roles in variety physiology and pathology processes, and generally maintain high level in cancer cells. Although this protein has been reported to involve in genome instability, its role remains unclear and controversial. Here, we showed that PFK-15, a PFKFB3 inhibitor, obviously induced apoptosis, cell viability loss, and inhibited cell proliferation/migration. Besides, PFK-15 was also found to induce necroptosis, as it not only up-regulated the phosphorylated RIP1, RIP3 and MLKL, but also enhanced the interaction between RIP3 and RIP1/MLKL, all of which are characterization of necroptosis induction. Both genetically and pharmacologically deprivation of necroptosis attenuated the cytotoxic effect of PFK-15. Besides, PFK-15 increased the γ-H2AX level and micronuclei formation, markers for genome instability, and inhibition of necroptosis attenuated these phenotypes. Collectively, the presented data demonstrated that PFK-15 induced genome instability and necroptosis, and deprivation of necroptosis attenuated cytotoxicity and genotoxicity of PFK-15 in colorectal cancer cells, thereby revealing a more intimate relationship among PFKFB3, necroptosis and genome instability.

16.
Fish Shellfish Immunol ; 28(1): 40-8, 2010 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19782140

RESUMO

In the present study, we have annotated both the immunoglobulin heavy (IgH) and light (IgL) chain genes in the stickleback (Gasterosteus aculeatus), based on the recently released genome data. The IgH gene locus is arranged in a configuration of (V(n)-D-J-C(zeta)-D(3)-J(4)-C(mu)-C(delta))(3)-V(6)-D-J-C(zeta), which is structurally different from any of the known teleost IgH loci. The mu genes consistently exhibit a 4-CH encoding structure and all the zeta genes encode only three CH domains (lacking the equivalent exon of the zebrafish zetaCH2). As in many other teleosts, the stickleback delta genes contain multiple CH exons, but exist as three copies. The members of four V(H) gene families, containing 47 segments, were interspersed in the germline. The stickleback IgL chain genes are also organized in multiple clusters and located in three chromosomes (10, 11, and 15). Sequence and phylogenetic analyses revealed that two isotypes, L1 (kappa, including two subgroups, 1A and 1B) and L2 (sigma) could be identified. The transcriptional orientations of the V(L) segments were found to be either the same (only in L2 isotype) or opposite to (in L1A, 1B and 2 isotypes) those of the J(L) and C(L) segments, indicating that these segments would undergo rearrangement by deletion or inversion when expressed.


Assuntos
Genes de Imunoglobulinas/genética , Imunoglobulinas/genética , Smegmamorpha/genética , Sequência de Aminoácidos , Animais , Mapeamento Cromossômico , Genes de Cadeia Pesada de Imunoglobulina/genética , Genes de Cadeia Leve de Imunoglobulina/genética , Genoma/genética , Cadeias Pesadas de Imunoglobulinas/genética , Cadeias Pesadas de Imunoglobulinas/imunologia , Cadeias Leves de Imunoglobulina/genética , Cadeias Leves de Imunoglobulina/imunologia , Família Multigênica/genética , Filogenia , Smegmamorpha/imunologia
17.
Oncogene ; 39(14): 2975-2986, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32034306

RESUMO

Cancer metabolism research has recently been revived and its focus expanded from glucose and the Warburg's effects on other nutrients, such as glutamine. The underlying mechanism of oncogenic alterations of glutaminolysis remains unclear. Genetic alterations of EGFR are observed in ~50% of glioblastoma (GBM) patients, and have been found to play important roles in the metabolic abnormalities of GBM. In this study, we found that glutamine metabolism was upregulated after EGFR activation in a GDH1 (glutamate dehydrogenase 1)-dependent manner. Knockdown of GDH1 significantly reduced the cell proliferation, colony formation and tumorigenesis abilities of glioblastoma cells. Furthermore, we showed that GDH1-mediated glutaminolysis was involved in EGF-promoted cell proliferation. EGFR triggered the phosphorylation of ELK1 at Ser 383 through activating MEK/ERK signaling. Phosphorylated ELK1 enriched in the promoter of GDH1 to activate the transcription of GDH1, which then promoted glutamine metabolism. In addition, EGFR activation did not accelerate glutaminolysis in ELK1 knockdown or ELK1 Ser383-mutated cells. Collectively, our findings indicate that EGFR phosphorylates ELK1 to activate GDH1 transcription and glutaminolysis through MEK/ERK pathway, providing new insight into oncogenic alterations of glutamine metabolism.


Assuntos
Glioblastoma/genética , Glutamato Desidrogenase/genética , Glutamina/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Transcrição Gênica/genética , Proteínas Elk-1 do Domínio ets/genética , Animais , Carcinogênese/genética , Linhagem Celular , Linhagem Celular Tumoral , Proliferação de Células/genética , Receptores ErbB/genética , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/metabolismo , Células HEK293 , Humanos , Masculino , Camundongos , Camundongos Nus , Oncogenes/genética , Regulação para Cima/genética
18.
Mol Biol Rep ; 36(2): 415-21, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18157703

RESUMO

Insulin-like growth factor-I (IGF-I) plays a fundamental role in postnatal mammalian growth, development, and metabolism. The mammalian IGF-I gene contains at least six exons from which several different mRNAs are transcribed. In this study, IGF-I mRNA in Songliao black pig liver was investigated using RLM-RACE. Using a 3'-RACE technique, we determined that all mRNA transcripts lacked exon 5 sequence and contained only exon 6 sequence. Using a 5'-RACE technique, we investigated the presence of class 1 and class 2 IGF-I mRNAs. In several other species, the class 1 and class 2 IGF-I polypeptides are generated from mRNAs containing exon 1 or exon 2, respectively. Both class 1 and class 2 IGF-I mRNAs were identified in Songliao black pig liver. Transcription is initiatated upstream of exons 1 and 2 at multiple dispersed start sites to yield two distinct IGF-I mRNA transcript classes which differ in the precursor peptides predicted from their individual leader sequences. Tissue distribution of Songliao black pig class 1 and class 2 IGF-I mRNA was investigated by real-time RT-PCR. Both classes of IGF-I mRNA were expressed in a variety of tissues, however, class 1 IGF-I mRNA was more abundant than class 2 in all tissues.


Assuntos
Fator de Crescimento Insulin-Like I/genética , RNA Mensageiro/genética , Animais , Clonagem Molecular/métodos , Éxons , Reação em Cadeia da Polimerase/métodos , Sus scrofa , Distribuição Tecidual
19.
Hematol Oncol Stem Cell Ther ; 12(3): 146-154, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-30796884

RESUMO

OBJECTIVE/BACKGROUND: Idiopathic myelofibrosis (IM) is a clonal hematological malignancy originating from pluripotent hematopoietic stem cells (HSC). HSC are very rare potent cells that reside in the bone marrow (BM) and at a lower level in peripheral blood (PB). Previous studies showed that IM PB CD34+ cells contain not only BM repopulating cells belonging to the malignant clone but also residual normal HSC. METHODS: In the current study, we separated the subpopulations of IM PB CD34+ cells using IL-3Rα/CD123 labeling and further characterized them by genetic and functional analyses. RESULTS: We differentiated IM PB CD34+ cells into three subpopulations (IL-3Rαhigh, IL-3Rαlow, and IL-3Rαnegative). IL-3Rαhigh CD34+ cell subgroup represents a small population in IM PB CD34+ cells which was not seen in normal G-CSF mobilized CD34+ cells. IM IL-3Rαhigh CD34+ cells contained significant higher percentage of cells bearing marker chromosome detected by fluorescence in situ hybridization (FISH) analysis. In the absence of growth factors, IM IL-3Rαhigh CD34+ cells exhibited abnormal colony forming ability and carried greater percentage of JAK2V617F mutant allele compared with IL-3Rαlow and IL-3Rαnegative CD34+ cells. CONCLUSION: These data indicate that IL-3Rαhigh CD34+ cells from IM enriched for the malignant progenitor cells and IL-3Rα/CD123 may be a potential biomarker and therapeutic target for IM. Our findings will be further validated in future studies with a larger sample size and serial transplant in murine models.


Assuntos
Células-Tronco Hematopoéticas/patologia , Mielofibrose Primária/patologia , Antígenos CD34/análise , Antígenos CD34/genética , Células Cultivadas , Aberrações Cromossômicas , Células-Tronco Hematopoéticas/metabolismo , Humanos , Hibridização in Situ Fluorescente , Subunidade alfa de Receptor de Interleucina-3/análise , Subunidade alfa de Receptor de Interleucina-3/genética , Janus Quinase 2/genética , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Mutação Puntual , Mielofibrose Primária/genética
20.
Oncogene ; 38(4): 497-517, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30115975

RESUMO

The serine protease PRSS8 has shown important physiological and pathological functions, but its roles in cancer initiation and progression are unclear. We developed and dynamically characterized a conditional knockout Prss8fl/fl, p-Villin-Cre+ mouse model. We found that genetic deficiency of the Prss8 gene caused spontaneous colitis and an inflamed rectum at an early age and caused intestinal tumors at a late age, which were linked to increased intestinal cell proliferation and migration but decreased cell differentiation. Increased PRSS8 expression inhibited cancer cell growth and metastasis in nude mice and inhibited cancer cell migration, invasion, colony formation and tumor sphere formation in vitro, but decreased PRSS8 expression facilitated malignancies in vivo and in vitro. Gene profiling on manipulated cancer cells and intestinal epithelial cells of Prss8 mouse models, gene set enrichment analysis and mechanistic studies revealed that PRSS8 targeted the Wnt/ß-catenin, epithelial-mesenchymal transition, and stem cell signaling pathways, which were further supported by the results from the TCGA data mining and validated by immunohistochemical staining on colorectal cancer tissue microarrays. In conclusion, PRSS8 is a novel tumor suppressor that plays critical roles in the suppression of colorectal carcinogenesis and metastasis.


Assuntos
Colite/genética , Neoplasias Colorretais/genética , Proteínas de Neoplasias/fisiologia , Serina Endopeptidases/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patologia , Adenoma/genética , Animais , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Cruzamentos Genéticos , Neoplasias Duodenais/genética , Transição Epitelial-Mesenquimal/genética , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Genes Supressores de Tumor , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Nus , Camundongos Transgênicos , Metástase Neoplásica , Proteínas de Neoplasias/deficiência , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/fisiologia , Interferência de RNA , RNA Interferente Pequeno/genética , Serina Endopeptidases/deficiência , Serina Endopeptidases/genética , Organismos Livres de Patógenos Específicos , Proteínas Supressoras de Tumor/antagonistas & inibidores , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/genética , Via de Sinalização Wnt
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA