Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
1.
EMBO J ; 35(8): 866-80, 2016 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-26988033

RESUMO

Autophagy, mediated by a number of autophagy-related (ATG) proteins, plays an important role in the bulk degradation of cellular constituents. Beclin-1 (also known as Atg6 in yeast) is a core protein essential for autophagic initiation and other biological processes. The activity of Beclin-1 is tightly regulated by multiple post-translational modifications, including ubiquitination, yet the molecular mechanism underpinning its reversible deubiquitination remains poorly defined. Here, we identified ubiquitin-specific protease 19 (USP19) as a positive regulator of autophagy, but a negative regulator of type I interferon (IFN) signaling.USP19 stabilizes Beclin-1 by removing the K11-linked ubiquitin chains of Beclin-1 at lysine 437. Moreover, we foundthat USP19 negatively regulates type IIFNsignaling pathway, by blockingRIG-I-MAVSinteraction in a Beclin-1-dependent manner. Depletion of eitherUSP19 or Beclin-1 inhibits autophagic flux and promotes type IIFNsignaling as well as cellular antiviral immunity. Our findings reveal novel dual functions of theUSP19-Beclin-1 axis by balancing autophagy and the production of type IIFNs.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Autofagia/fisiologia , Endopeptidases/metabolismo , Proteínas de Membrana/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Proteínas Reguladoras de Apoptose/genética , Proteínas Reguladoras de Apoptose/imunologia , Proteína Beclina-1 , Linhagem Celular/virologia , Proteína DEAD-box 58 , RNA Helicases DEAD-box/metabolismo , Endopeptidases/genética , Endopeptidases/imunologia , Células HeLa/metabolismo , Interações Hospedeiro-Patógeno/imunologia , Humanos , Vírus da Influenza A/patogenicidade , Interferon Tipo I/metabolismo , Lisina/metabolismo , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Estabilidade Proteica , Receptores Imunológicos , Transdução de Sinais/fisiologia , Ubiquitinação
2.
PLoS Pathog ; 13(7): e1006503, 2017 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-28732079

RESUMO

Myeloid-derived suppressor cells (MDSCs) are expanded in tumor microenvironments, including that of Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC). The link between MDSC expansion and EBV infection in NPC is unclear. Here, we show that EBV latent membrane protein 1 (LMP1) promotes MDSC expansion in the tumor microenvironment by promoting extra-mitochondrial glycolysis in malignant cells, which is a scenario for immune escape initially suggested by the frequent, concomitant detection of abundant LMP1, glucose transporter 1 (GLUT1) and CD33+ MDSCs in tumor sections. The full process has been reconstituted in vitro. LMP1 promotes the expression of multiple glycolytic genes, including GLUT1. This metabolic reprogramming results in increased expression of the Nod-like receptor family protein 3 (NLRP3) inflammasome, COX-2 and P-p65 and, consequently, increased production of IL-1ß, IL-6 and GM-CSF. Finally, these changes in the environment of malignant cells result in enhanced NPC-derived MDSC induction. One key step is the physical interaction of LMP1 with GLUT1 to stabilize the GLUT1 protein by blocking its K48-ubiquitination and p62-dependent autolysosomal degradation. This work indicates that LMP1-mediated glycolysis regulates IL-1ß, IL-6 and GM-CSF production through the NLRP3 inflammasome, COX-2 and P-p65 signaling pathways to enhance tumor-associated MDSC expansion, which leads to tumor immunosuppression in NPC.


Assuntos
Carcinoma/fisiopatologia , Infecções por Vírus Epstein-Barr/fisiopatologia , Herpesvirus Humano 4/metabolismo , Células Supressoras Mieloides/citologia , Neoplasias Nasofaríngeas/fisiopatologia , Proteínas da Matriz Viral/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/virologia , Linhagem Celular Tumoral , Proliferação de Células , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/metabolismo , Infecções por Vírus Epstein-Barr/virologia , Regulação Neoplásica da Expressão Gênica , Glicólise , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Herpesvirus Humano 4/genética , Interações Hospedeiro-Patógeno , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Células Supressoras Mieloides/metabolismo , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/metabolismo , Neoplasias Nasofaríngeas/virologia , Transdução de Sinais , Proteínas da Matriz Viral/genética
3.
J Transl Med ; 13: 166, 2015 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-25990580

RESUMO

BACKGROUND: The immune non-recognition is often the underlying cause of failure in tumor immunotherapeutic. This is because most tumor-related antigens are poorly immunogenic, and fail to arouse an efficient immune response against cancers. Here we synthesized a novel TLR7 agonist, and developed a safe and effective immunotherapeutic vaccine by conjugating this TLR7 agonist with the pluripotency antigen OCT4. METHODS: Purified recombinant OCT4 protein was covalently linked with a novel TLR7 agonist to form a TLR7-OCT4 conjugate (T7-OCT4). After conjugation, the in vitro release of IL-12 and IFN-γ was observed in spleen lymphocytes. Mice were immunized with TLR7-OCT4, and the release of IFN-γ, the percentages of CD3+/CD8+ T cells and the OCT4-specific cytotoxicity rates were measured. The immunized mice were challenged with mouse embryonic carcinoma (EC), and the tumor volume and tumor weight were determined. Blood routine examination was performed to evaluate the biosafety of TLR7 agonist and TLR7-OCT4 conjugate in mice. RESULTS: T7-OCT4 conjugate significantly increased the in vitro release of IL-12 and IFN-γ by mouse spleen lymphocytes. In addition, the release of IFN-γ, the percentages of CD3+/CD8+ T cells and the tumor-specific cytotoxicity rates in immunized mice were significantly higher. Importantly, in EC xenografted mice, immunization with T7-OCT4 conjugate decreased the growth of the tumor dramatically up to 90 %, as compared to mice immunized with OCT4 protein or TLR7 agonist alone. Furthermore, blood routine examination demonstrated that no abnormalities of the blood cells and components in the blood fluids were detected by T7-OCT4 and TLR7 agonist injections. CONCLUSIONS: Our results showed that conjugating OCT4 protein to the novel TLR7 agonist produced a vaccine which is effective and safe in preventing tumor growth in mice. Our results suggest that this type of vaccine formulation has great potentiality in preventive vaccines against OCT4 expressing tumors.


Assuntos
Carcinoma Embrionário/metabolismo , Glicoproteínas de Membrana/química , Fator 3 de Transcrição de Octâmero/química , Neoplasias Testiculares/metabolismo , Receptor 7 Toll-Like/química , Animais , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/citologia , Carcinoma Embrionário/prevenção & controle , Interferon gama/metabolismo , Interleucina-12/metabolismo , Linfócitos/citologia , Masculino , Glicoproteínas de Membrana/agonistas , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Transplante de Neoplasias , Proteínas Recombinantes/metabolismo , Baço/metabolismo , Linfócitos T Citotóxicos/citologia , Neoplasias Testiculares/prevenção & controle , Fatores de Tempo , Receptor 7 Toll-Like/agonistas
4.
Nat Commun ; 14(1): 6069, 2023 09 28.
Artigo em Inglês | MEDLINE | ID: mdl-37770465

RESUMO

The PI3K/AKT pathway plays an essential role in tumour development. NOD-like receptors (NLRs) regulate innate immunity and are implicated in cancer, but whether they are involved in PI3K/AKT pathway regulation is poorly understood. Here, we report that NLRP6 potentiates the PI3K/AKT pathway by binding and destabilizing p85α, the regulatory subunit of PI3K. Mechanistically, NLRP6 recruits the E3 ligase RBX1 to p85α and ubiquitinates lysine 256 on p85α, which is recognized by the autophagy cargo receptor OPTN, causing selective autophagic degradation of p85α and subsequent activation of the PI3K/AKT pathway by reducing PTEN stability. We further show that loss of NLRP6 suppresses cell proliferation, colony formation, cell migration, and tumour growth in glioblastoma cells in vitro and in vivo. Disruption of the NLRP6/p85α interaction using the Pep9 peptide inhibits the PI3K/AKT pathway and generates potent antitumour effects. Collectively, our results suggest that NLRP6 promotes p85α degradation via selective autophagy to drive tumorigenesis, and the interaction between NLRP6 and p85α can be a promising therapeutic target for tumour treatment.


Assuntos
Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Humanos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Transdução de Sinais , Carcinogênese , Autofagia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo
6.
Ann Clin Lab Sci ; 52(6): 895-903, 2022 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-36564068

RESUMO

OBJECTIVE: Primary ovarian insufficiency (POI), which refers to the occurrence of ovarian insufficiency before the age of 40, is indicated by menstrual cycle changes as a precursor and is accompanied by menstrual disorders, elevated gonadotropin levels, and decreased estrogen levels. The incidence of POI is reportedly increasing worldwide and this disease markedly reduces the quality of life and affects the physical and mental health of patients. Treatment options for POI include hormone replacement therapy; however, its efficacy remains unsatisfactory. Therefore, exploring hormonal drugs with superior curative effects and clarifying the molecular mechanism underlying POI pathogenesis could afford new directions for POI therapy. METHODS: 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide and lactate dehydrogenase assays were used to detect the effects of melatonin (MT) on cell survival and mortality. Flow cytometry was performed to examine the effect of MT on apoptosis. The impact of MT on autophagosome formation was examined using electron microscopy, whereas the expression of autophagy-related proteins and phosphatidylinositol-3-kinase (PI3K)/Akt/mammalian target of rapamycin (mTOR) pathway-related proteins following MT intervention was detected by western blotting. RESULTS: (1) MT exerted a protective effect on ovarian granulosa cells subjected to serum starvation. (2) MT inhibited serum starvation-induced apoptosis of ovarian granulosa cells. (3) MT inhibited serum starvation-induced autophagosome formation in ovarian granulosa cells. (4) MT inhibited the expression of autophagy-related proteins LC3II/I and Agt5. (5) MT suppressed autophagy in ovarian granulosa cells by activating the PI3K/Akt/mTOR signaling pathway. CONCLUSION: Collectively, our results demonstrate that MT can inhibit excessive autophagy in ovarian granulosa cells by activating the PI3K/Akt/mTOR pathway, thereby exerting its protective effect against POI.


Assuntos
Melatonina , Insuficiência Ovariana Primária , Feminino , Humanos , Fosfatidilinositol 3-Quinase/metabolismo , Fosfatidilinositol 3-Quinase/farmacologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Melatonina/farmacologia , Insuficiência Ovariana Primária/prevenção & controle , Qualidade de Vida , Serina-Treonina Quinases TOR/metabolismo , Autofagia , Proteínas Relacionadas à Autofagia/farmacologia , Apoptose
7.
Sci Bull (Beijing) ; 66(8): 794-802, 2021 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-36654136

RESUMO

Ubiquitination plays a crucial role in retinoic acid-inducible gene I (RIG-I)-induced antiviral responses. However, the precise regulatory mechanisms of RIG-I activity mediated by conjugated and unanchored ubiquitin chains remain to be determined. In this study, we discovered that T55 of RIG-I was required for its binding ability for the unanchored ubiquitin chains. Experimental and mathematical analysis showed that unanchored ubiquitin chains associated with RIG-I were essential for sustained activation of type I interferon (IFN) signaling. Transcriptomics study revealed that the binding of RIG-I with unanchored ubiquitin chains additionally regulated the expression of a subset of metabolic and cell fate decision genes. Moreover, we found that ubiquitin-specific peptidase 21 (USP21) and USP3 deubiquitinate conjugated and unanchored ubiquitin chains on RIG-I respectively. Taken together, characterization of the regulation mode and functions of conjugated ubiquitination and the unconjugated ubiquitin chain-binding of RIG-I may provide means to fine-tune RIG-I-mediated type I IFN signaling.

8.
Clin Transl Med ; 11(9): e553, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34586728

RESUMO

BACKGROUND: Despite the development of new treatment protocols for glioblastoma (GBM), temozolomide (TMZ) resistance remains a primary hindrance. Previous studies, including our study, have shown that aberrant N6-methyladenosine (m6 A) modification is implicated in GBM pathobiology. However, the roles and precise mechanisms of m6 A modification in the regulation of TMZ resistance in GBM remain unclear. METHODS: m6 A individual-nucleotide-resolution cross-linking and immunoprecipitation sequencing (miCLIP-seq) was performed to identify m6 A modification of transcripts in TMZ-resistant and -sensitive tumors. To explore the role of METTL3 in TMZ resistance, TMZ-resistant GBM cells were transfected with METTL3 shRNA or overexpression lentivirus and then assessed by cell viability, tumor sphere formation, and apoptosis assays. An intracranial GBM xenograft model was developed to verify the effect of METTL3 depletion during TMZ treatment in vivo. ATAC-seq, ChIP-qPCR, and dual-luciferase reporter assays were carried out to verify the role of SOX4/EZH2 in the modulation of METTL3 expression upon TMZ treatment. RESULTS: We demonstrated that TMZ treatment upregulated the expression of the m6 A methyltransferase METTL3, thereby increasing m6 A modification of histone modification-related gene transcripts. METTL3 is required to maintain the features of GBM stem cells. When combined with TMZ, METTL3 silencing suppressed orthotopic TMZ-resistant xenograft growth in a cooperative manner. Mechanistically, TMZ induced a SOX4-mediated increase in chromatin accessibility at the METTL3 locus by promoting H3K27ac levels and recruiting RNA polymerase II. Moreover, METTL3 depletion affected the deposition of m6 A on histone modification-related gene transcripts, such as EZH2, leading to nonsense-mediated mRNA decay. We revealed an important role of EZH2 in the regulation of METTL3 expression, which was via an H3K27me3 modification-independent manner. CONCLUSIONS: Our findings uncover the fundamental mechanisms underlying the interplay of m6 A RNA modification and histone modification in TMZ resistance and emphasize the therapeutic potential of targeting the SOX4/EZH2/METTL3 axis in the treatment of TMZ-resistant GBM.


Assuntos
Adenosina/análogos & derivados , Antineoplásicos Alquilantes/farmacologia , Resistencia a Medicamentos Antineoplásicos , Glioblastoma/genética , Código das Histonas/genética , Temozolomida/farmacologia , Adenosina/genética , Adenosina/metabolismo , Animais , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C
9.
Adv Sci (Weinh) ; 8(20): e2101895, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34432948

RESUMO

Liquid-liquid phase-separated (LLPS) transcriptional factor assemblies at super-enhancers (SEs) provide a conceptual framework for underlying transcriptional control in mammal cells. However, the mechanistic understanding of LLPS in aberrant transcription driven by dysregulation of SEs in human malignancies is still elusive. By integrating SE profiling and core regulatory circuitry (CRC) calling algorithm, the CRC of metastatic and chemo-resistant osteosarcoma is delineated. CRC components, HOXB8 and FOSL1, produce dense and dynamic phase-separated droplets in vitro and liquid-like puncta in cell nuclei. Disruption of CRC phase separation decreases the chromatin accessibility in SE regions and inhibits the release of RNA polymerase II from the promoter of SE-driven genes. Importantly, absence of CRC key component causes a reduction in osteosarcoma tumor growth and metastasis. Moreover, it is shown that CRC condensates can be specifically attenuated by the H3K27 demethylase inhibitor, GSK-J4. Pharmacological inhibition of the CRC phase separation results in metastasis suppression and re-sensitivity to chemotherapy drugs in patient-derived xenograft model. Taken together, this study reveals a previously unknown mechanism that CRC factors formed LLPS condensates, and provides a phase separation-based pharmacological strategy to target undruggable CRC components for the treatment of metastatic and chemo-resistant osteosarcoma.


Assuntos
Proteínas de Homeodomínio/genética , Histona Desmetilases com o Domínio Jumonji/genética , Osteossarcoma/tratamento farmacológico , Proteínas Proto-Oncogênicas c-fos/genética , Animais , Benzazepinas/farmacologia , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Núcleo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Cromatina/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Elementos Facilitadores Genéticos/genética , Feminino , Humanos , Histona Desmetilases com o Domínio Jumonji/antagonistas & inibidores , Masculino , Camundongos , Osteossarcoma/genética , Osteossarcoma/patologia , Pirimidinas/farmacologia , RNA Polimerase II/genética , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Front Immunol ; 11: 1906, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32973789

RESUMO

T cell exhaustion is an obstacle to immunotherapy for solid tumors. An understanding of the mechanism by which T cells develop this phenotype in solid tumors is needed. Here, hypoxia, a feature of the tumor microenvironment, causes T cell exhaustion (TExh) by inducing a mitochondrial defect. Upon exposure to hypoxia, activated T cells with a TExh phenotype are characterized by mitochondrial fragmentation, decreased ATP production, and decreased mitochondrial oxidative phosphorylation activity. The TExh phenotype is correlated with the downregulation of the mitochondrial fusion protein mitofusin 1 (MFN1) and upregulation of miR-24. Overexpression of miR-24 alters the transcription of many metabolism-related genes including its target genes MYC and fibroblast growth factor 11 (FGF11). Downregulation of MYC and FGF11 induces TExh differentiation, reduced ATP production and a loss of the mitochondrial mass in T cell receptor (TCR)-stimulated T cells. In addition, we determined that MYC regulates the transcription of FGF11 and MFN1. In nasopharyngeal carcinoma (NPC) tissues, the T cells exhibit an increased frequency of exhaustion and loss of mitochondrial mass. In addition, inhibition of miR-24 signaling decreases NPC xenograft growth in nude mice. Our findings reveal a mechanism for T cell exhaustion in the tumor environment and provide potential strategies that target mitochondrial metabolism for cancer immunotherapy.


Assuntos
Linfócitos do Interstício Tumoral/metabolismo , Mitocôndrias/metabolismo , Carcinoma Nasofaríngeo/metabolismo , Neoplasias Nasofaríngeas/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Linfócitos T/metabolismo , Microambiente Tumoral , Animais , Estudos de Casos e Controles , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Feminino , Fatores de Crescimento de Fibroblastos/genética , Fatores de Crescimento de Fibroblastos/metabolismo , GTP Fosfo-Hidrolases/genética , GTP Fosfo-Hidrolases/metabolismo , Regulação Neoplásica da Expressão Gênica , Humanos , Linfócitos do Interstício Tumoral/imunologia , Linfócitos do Interstício Tumoral/patologia , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Mitocôndrias/genética , Mitocôndrias/imunologia , Mitocôndrias/patologia , Dinâmica Mitocondrial , Proteínas de Transporte da Membrana Mitocondrial/genética , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/imunologia , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/imunologia , Neoplasias Nasofaríngeas/patologia , Fenótipo , Proteínas Proto-Oncogênicas c-myc/genética , Transdução de Sinais , Linfócitos T/imunologia , Linfócitos T/patologia , Hipóxia Tumoral
11.
Oncogenesis ; 9(7): 65, 2020 Jul 06.
Artigo em Inglês | MEDLINE | ID: mdl-32632113

RESUMO

Galectin-9 (Gal-9) is known to enhance the expansion of myeloid-derived suppressor cells (MDSCs) in murine models. Its contribution to the expansion of MDSCs in human malignancies remain to be investigated. We here report that Gal-9 expression in nasopharyngeal carcinoma (NPC) cells enhances the generation of MDSCs (CD33+CD11b+HLA-DR-) from CD33+ bystander cells. The underlying mechanisms involve both the intracellular and secreted Gal-9. Inside carcinoma cells, Gal-9 up-regulates the expression of a variety of pro-inflammatory cytokines which are critical for MDSC differentiation, including IL-1ß and IL-6. This effect is mediated by accelerated STING protein degradation resulting from direct interaction of the Gal-9 carbohydrate recognition domain 1 with the STING C-terminus and subsequent enhancement of the E3 ubiquitin ligase TRIM29-mediated K48-linked ubiquitination of STING. Moreover, we showed that extracellular Gal-9 secreted by carcinoma cells can enter the myeloid cells and trigger the same signaling cascade. Consistently, high concentrations of tumor and plasma Gal-9 are associated with shortened survival of NPC patients. Our findings unearth that Gal-9 induces myeloid lineage-mediated immunosuppression in tumor microenvironments by suppressing STING signaling.

12.
Cell Death Differ ; 26(11): 2314-2328, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30816302

RESUMO

Stimulator of interferon genes (STING), a major adaptor protein in antiviral innate immune signaling, is considered as one of the most important regulators of antiviral and antitumor immunity. Although STING agonists are now intensively studied in clinical trials as a new class of adjuvants to boost cancer immunotherapy, the tumor-intrinsic role of the STING pathway in shaping the tumor microenvironment remains controversial. Here, we discovered that STING plays a vital role in regulation of myeloid-derived suppressor cell (MDSC) differentiation and antitumor immunity in Epstein-Barr virus (EBV)-associated nasopharyngeal carcinoma (NPC). Mechanistic analyses reveal that STING represses NPC-derived MDSC induction by enhancing SOCS1 expression in both tumor cells and MDSCs. SOCS1 physically interacts with STAT3 through its SH2 domain to prevent STAT3 phosphorylation and dimerization, resulting in reduced MDSC induction via inhibition of GM-CSF and IL-6 production. Notably, reduced tumoral STING expression was found to be significantly associated with a poor prognosis for NPC patients. Our findings reveal a novel mechanism linking STING to tumor microenvironmental cytokine production and MDSC induction.


Assuntos
Proteínas de Membrana/metabolismo , Células Supressoras Mieloides/citologia , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/patologia , Microambiente Tumoral/fisiologia , Sistemas CRISPR-Cas , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Dimerização , Infecções por Vírus Epstein-Barr/patologia , Técnicas de Inativação de Genes , Fator Estimulador de Colônias de Granulócitos e Macrófagos/antagonistas & inibidores , Células HEK293 , Herpesvirus Humano 4/imunologia , Humanos , Interleucina-6/antagonistas & inibidores , Proteínas de Membrana/genética , Células Supressoras Mieloides/imunologia , Carcinoma Nasofaríngeo/imunologia , Carcinoma Nasofaríngeo/virologia , Neoplasias Nasofaríngeas/imunologia , Neoplasias Nasofaríngeas/virologia , Fosforilação/fisiologia , Prognóstico , Fator de Transcrição STAT3/metabolismo , Proteína 1 Supressora da Sinalização de Citocina/metabolismo , Microambiente Tumoral/efeitos dos fármacos
13.
Cell Rep ; 23(3): 838-851, 2018 Apr 17.
Artigo em Inglês | MEDLINE | ID: mdl-29669288

RESUMO

p38 signaling is broadly involved in controlling inflammation and stress-induced cell death; however, the mechanisms controlling its activity have seldom been studied. Here, we report that TRIM9 short isoform (TRIM9s) potentiates p38 signaling by stabilizing MKK6. Mechanistic studies revealed that TRIM9s promotes the K63-linked ubiquitination of MKK6 at Lys82, thus inhibiting the degradative K48-linked ubiquitination of MKK6 at the same lysine. MKK6 could also stabilize TRIM9s by promoting the phosphorylation of TRIM9s at Ser76/80 via p38, thereby blocking the ubiquitin-proteasome pathway. Further functional analyses showed that p38 signaling plays a critical role in suppressing glioblastoma progression. Co-reduction of MKK6 and TRIM9s is significantly associated with overall poor survival of glioblastoma patients. We identify a positive feedback loop in p38 signaling generated by MKK6-TRIM9s, which suppresses glioblastoma progression, and we provide insights into the mechanisms by which TRIM9s and MKK6 potentiate p38 signaling through mutual stabilization.


Assuntos
Glioblastoma/patologia , MAP Quinase Quinase 6/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Transdução de Sinais , Proteínas com Motivo Tripartido/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Animais , Linhagem Celular Tumoral , Glioblastoma/tratamento farmacológico , Glioblastoma/mortalidade , Humanos , MAP Quinase Quinase 6/deficiência , MAP Quinase Quinase 6/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas do Tecido Nervoso/antagonistas & inibidores , Proteínas do Tecido Nervoso/genética , Fosforilação , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Interferência de RNA , RNA Interferente Pequeno/metabolismo , RNA Interferente Pequeno/uso terapêutico , Taxa de Sobrevida , Proteínas com Motivo Tripartido/antagonistas & inibidores , Proteínas com Motivo Tripartido/genética , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética , Ubiquitinação , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Cell Death Dis ; 8(5): e2831, 2017 05 25.
Artigo em Inglês | MEDLINE | ID: mdl-28542145

RESUMO

Tripartite motif-containing protein 45 (TRIM45) belongs to a large family of RING-finger-containing E3 ligases, which are highly expressed in the brain. However, little is known regarding the role of TRIM45 in cancer biology, especially in human glioma. Here, we report that TRIM45 expression is significantly reduced in glioma tissue samples. Overexpression of TRIM45 suppresses proliferation and tumorigenicity in glioblastoma cells in vitro and in vivo. In addition, CRISPR/Cas9-mediated knockout of TRIM45 promotes proliferation and inhibits apoptosis in glioblastoma cells. Further mechanistic analyses show that TRIM45 interacts with and stabilizes p53. TRIM45 conjugates K63-linked polyubiquitin chain to the C-terminal six lysine residues of p53, thereby inhibiting the availability of these residues to the K48-linked polyubiquitination that targets p53 for degradation. These findings suggest that TRIM45 is a novel tumor suppressor that stabilizes and activates p53 in glioma.


Assuntos
Encéfalo/metabolismo , Lisina/metabolismo , Proteínas Repressoras/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Ubiquitina-Proteína Ligases/metabolismo , Ubiquitinação , Animais , Apoptose , Biocatálise , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Regulação para Baixo , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Glioma/patologia , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Modelos Biológicos , Estabilidade Proteica , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Repressoras/genética , Transdução de Sinais
15.
Oncol Lett ; 14(5): 6001-6011, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29113238

RESUMO

Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) is a rapid, reliable and widely used method of studying gene expression profiles that requires appropriate normalization for accurate and reliable results. Reference genes are usually used to normalize mRNA levels; however, the expression levels of these reference genes may vary between cell types, developmental stages, species and experimental conditions. Therefore, a normalization strategy is an important precondition for reliable conclusions, with endogenous controls requiring determination for every experimental system. In the present study, 18 reference genes used in various prior studies were analyzed to determine their applicability in bladder cancer. A total of 35 matched malignant and non-malignant bladder cancer (specifically transitional cell carcinoma) tissue specimens were examined. RNA and cDNA quality was stringently controlled. Candidate reference genes were assessed using SYBR-Green RT-qPCR. mRNA abundance was compared and reference genes with distinct ranges of expression to possible target genes were excluded. Genes that were differentially expressed in matched non-cancerous and cancerous samples were also excluded, using quantification cycle analysis. Subsequently, the stability of the selected reference genes was analyzed using three different methods: geNorm, NormFinder and BestKeeper. The rarely used ribosomal protein S23 (RPS23) was the most stable single reference gene, with RPS23, tumor protein, translationally controlled 1 and RPS13 comprising the optimal reference gene set for all the bladder samples. These stable reference genes should be employed in normalization and quantification of transcript levels in future expression studies of bladder cancer-associated genes.

17.
Oncotarget ; 7(3): 2754-64, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26700620

RESUMO

To develop a routine and effectual procedure of detecting bladder cancer (BlCa), an optimized combination of epigenetic biomarkers that work synergistically with high sensitivity and specificity is necessary. In this study, methylation levels of seven biomarkers (EOMES, GDF15, NID2, PCDH17, POU4F2, TCF21, and ZNF154) in 148 individuals-which including 58 urothelial cell carcinoma (UCC) patients, 20 infected urinary calculi (IUC) patients, 20 kidney cancer (KC) patients,20 prostate cancer (PC) patients, and 30 healthy volunteers (HV)-were quantified by qMSP using the urine sediment DNA. Receiver operating characteristic (ROC) curves were generated for each biomarker. The combining predictors of possible combinations were calculated through logistic regression model. Subsequently, ROC curves of the three best performing combinations were constructed. Then, we validated the three best performing combinations and POU4F2 in another 72 UCC, 21 IUC, 26 KC and 22 PC, and 23 HV urine samples. The combination of POU4F2/PCDH17 has yielded the highest sensitivity and specificity of 90.00% and 93.96% in all the 312 individuals, showing the capability of detecting BlCa effectively among pathologically varied sample groups.


Assuntos
Biomarcadores Tumorais/urina , Caderinas/genética , Metilação de DNA/genética , Fator de Transcrição Brn-3B/genética , Neoplasias da Bexiga Urinária/diagnóstico , Neoplasias da Bexiga Urinária/urina , Biomarcadores Tumorais/genética , China , DNA/genética , DNA/urina , Humanos , Neoplasias Renais/urina , Masculino , Neoplasias da Próstata/urina , Sensibilidade e Especificidade , Neoplasias da Bexiga Urinária/genética , Cálculos Urinários/urina , Urotélio/patologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa