Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Elife ; 122024 May 13.
Artigo em Inglês | MEDLINE | ID: mdl-38738857

RESUMO

Enhanced protein synthesis is a crucial molecular mechanism that allows cancer cells to survive, proliferate, metastasize, and develop resistance to anti-cancer treatments, and often arises as a consequence of increased signaling flux channeled to mRNA-bearing eukaryotic initiation factor 4F (eIF4F). However, the post-translational regulation of eIF4A1, an ATP-dependent RNA helicase and subunit of the eIF4F complex, is still poorly understood. Here, we demonstrate that IBTK, a substrate-binding adaptor of the Cullin 3-RING ubiquitin ligase (CRL3) complex, interacts with eIF4A1. The non-degradative ubiquitination of eIF4A1 catalyzed by the CRL3IBTK complex promotes cap-dependent translational initiation, nascent protein synthesis, oncogene expression, and cervical tumor cell growth both in vivo and in vitro. Moreover, we show that mTORC1 and S6K1, two key regulators of protein synthesis, directly phosphorylate IBTK to augment eIF4A1 ubiquitination and sustained oncogenic translation. This link between the CRL3IBTK complex and the mTORC1/S6K1 signaling pathway, which is frequently dysregulated in cancer, represents a promising target for anti-cancer therapies.


Assuntos
Fator de Iniciação 4A em Eucariotos , Alvo Mecanístico do Complexo 1 de Rapamicina , Biossíntese de Proteínas , Proteínas Quinases S6 Ribossômicas 70-kDa , Transdução de Sinais , Ubiquitinação , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , Fator de Iniciação 4A em Eucariotos/metabolismo , Fator de Iniciação 4A em Eucariotos/genética , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/genética
2.
Cell Death Dis ; 15(1): 65, 2024 01 17.
Artigo em Inglês | MEDLINE | ID: mdl-38233384

RESUMO

Mitochondria are highly dynamic organelles capable of altering their sizes and shapes to maintain metabolic balance through coordinated fission and fusion processes. In various cancer types, mitochondrial hyperfragmentation has been frequently observed, contributing to the progression of cancer toward metastasis. Inverted formin 2 (INF2), which resides in the endoplasmic reticulum (ER), has been found to accelerate actin polymerization and drive mitochondrial fission. In this study, we demonstrate that INF2 expression is significantly upregulated in endometrial cancer (EC) and is associated with a poor prognosis in EC patients. INF2 promotes anchorage-dependent and independent EC cell growth in part by facilitating mitochondrial fission. Furthermore, in conditions of energy stress, AMP-activated protein kinase (AMPK) phosphorylates INF2 at Ser1077, leading to increased localization of INF2 to the ER and enhanced recruitment of the dynamin-related protein 1 (DRP1) to mitochondria. This AMPK-mediated phosphorylation of INF2 at Ser1077 facilitates mitochondrial division and promotes EC cell growth. Pathological examination using immunohistochemical analyses revealed a positive correlation between AMPK activity and phosphorylated INF2 (Ser1077) in EC specimens. Collectively, our findings uncover novel molecular mechanisms involving the AMPK-INF2 axis, which regulates mitochondrial dynamics and malignant cell growth in EC.


Assuntos
Proteínas Quinases Ativadas por AMP , Neoplasias do Endométrio , Feminino , Humanos , Proteínas Quinases Ativadas por AMP/metabolismo , Dinaminas/metabolismo , Neoplasias do Endométrio/genética , Neoplasias do Endométrio/metabolismo , Forminas/metabolismo , Mitocôndrias/metabolismo , Dinâmica Mitocondrial , Proteínas Mitocondriais/genética , Proteínas Mitocondriais/metabolismo , Fosforilação
4.
Cell Death Differ ; 30(2): 475-487, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36481790

RESUMO

Blockade of programmed cell death 1 (PD-1)/programmed cell death 1 ligand (PD-L1) has evolved into one of the most promising immunotherapy strategies for cancer patients. Tumor cells frequently overexpress PD-L1 to evade T cell-mediated immune surveillance. However, the specific genetic alterations that drive aberrant overexpression of PD-L1 in cancer cells remain poorly understood. The gene encoding the E3 ubiquitin ligase substrate-binding adaptor SPOP is frequently mutated in endometrial cancer (EC). Here, we report that SPOP negatively regulates PD-L1 expression at the transcriptional level. Wild-type SPOP binds to IRF1, a primary transcription factor responsible for the inducible expression of PD-L1, and subsequently triggers its ubiquitin- proteasomal degradation to suppress IRF1-mediated transcriptional upregulation of PD-L1. In contrast, EC-associated SPOP mutants lose their capacity to degrade IRF1 but stabilize IRF1, and upregulate PD-L1 expression. EC-associated SPOP mutations accelerate xenograft tumor growth partially by increasing IRF1 and PD-L1 expression. Together, we identify SPOP as a negative regulator of the IRF1-PD-L1 axis and characterize the critical roles of IRF1 and PD-L1 in SPOP mutation-driven tumor immune evasion in EC.


Assuntos
Neoplasias do Endométrio , Proteínas Nucleares , Evasão Tumoral , Feminino , Humanos , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Linhagem Celular Tumoral , Neoplasias do Endométrio/genética , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Mutação/genética , Proteínas Nucleares/metabolismo
5.
Cell Commun Signal ; 20(1): 177, 2022 11 14.
Artigo em Inglês | MEDLINE | ID: mdl-36376931

RESUMO

BACKGROUND: Loss-of-function (LOF) mutations of JAK1, a member of the JAK kinase family, were frequently observed in EC, indicating that JAK1 may act as a tumor suppressor, at least in EC. However, the mechanism of JAK1 mediated regulation of tumorigenesis remains poorly understood. METHODS: The genetic alterations of JAK1 in EC using latest sequencing dataset of EC deposited in TCGA database. The RNA-Seq dataset of EC and normal endometrial tissues from TCGA cohort was analyzed. The expression of JAK1 in EC and normal endometrial tissues were investigated using immunohistochemistry. The expression levels of genes in endometrial cancer cells were detected by quantitative reverse transcription-PCR (RT-qPCR) and western blotting. JAK1 protein was efficiently depleted by the two shRNAs. HIF1/2-α protein was efficiently depleted by siRNAs. JAK1 overexpressed EC cells were generated by an expressing plasmid. The proliferation and migration ability of cancer cells were evaluated by CCK8, colony formation assays and transwell assays. The global transcriptomic changes in JAK1-depleted KLE cells were investigated using RNA-Seq. Gene Ontology (GO) Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis were used to identify the most significant pathways that were altered in JAK1-depleted KLE cells. The physical association between HIF-1/2α and JAK1 using co-immunoprecipitation (co-IP) assays. RESULTS: In the present study, we found that JAK1 was frequently mutated and downregulated in EC. JAK1 knockdown promotes EC cell proliferation and migration. JAK1 overexpression reduces EC cell proliferation and migration. We examined the transcriptional profiling changes in JAK1-depleted EC cells and unexpectedly found that the hypoxia inducible factor (HIF) pathway was activated. Mechanistically, JAK1 interacts with HIF-1/2α, and reduces HIF1/2-α protein expression under hypoxia. HIF-1/2α knockdown reverses the JAK1 knockdown-induced growth and migration of EC cells under hypoxia. JAK1 knockdown or pharmacological inhibition of JAK1 kinase activity by Ruxolitinib upregulates transcription of HIF target genes under hypoxia. JAK1 overexpression downregulates transcription of HIF target genes under hypoxia. CONCLUSIONS: These findings provide novel insights into the functional link between JAK1 LOF mutations and abnormal HIF pathway activation in EC and suggest that pharmacological inhibition of HIF1/2 represents a promising therapeutic strategy targeting JAK1-mutated ECs. Video Abstract.


Assuntos
Neoplasias do Endométrio , Humanos , Feminino , Janus Quinase 1 , Neoplasias do Endométrio/genética , Transdução de Sinais , Proliferação de Células , RNA Interferente Pequeno/metabolismo , Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Hipóxia Celular , Regulação Neoplásica da Expressão Gênica , Linhagem Celular Tumoral
6.
Cell Oncol (Dordr) ; 45(5): 861-872, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35951287

RESUMO

BACKGROUND: Metastasis is still the major cause of endometrial cancer (EC)-related death. Because of their biological function and regenerative properties, exosomes have been applied to therapeutic regimens. SERPINA5 expression is downregulated in several tumors and linked to tumor cell migration and invasion. However, the expression and biological functions of SERPINA5 in EC remain unclear. METHODS: The levels of SERPINA5 in plasma exosomes were determined with ELISAs. SERPINA5 expression in EC and its relationship with survival outcomes were analyzed using the TCGA database and clinical EC tissue samples. The effect of SERPINA5 overexpression or exosomal SERPINA5 on EC metastasis was examined by cell migration and invasion assays in vitro. Mechanistically, overexpression of SERPINA5 or high exosomal SERPINA5 levels mediated the regulation of the integrin ß1/FAK signaling pathway in EC cell lines. The therapeutic effect of exosomal SERPINA5 was determined with xenograft models. RESULTS: This study revealed that the level of exosomal SERPINA5 was increased in the circulating plasma of EC patients. In addition, the expression of SERPINA5 was decreased in EC patients with distant metastasis, and low expression of SERPINA5 indicated worse survival. In addition, SERPINA5 was elevated in normal tissues adjacent to EC tumors. Moreover, overexpression of SERPINA5 inhibited metastatic potential of EC cell lines in vitro. Furthermore, SERPINA5 loaded on secreted exosomes reduced the metastatic ability of EC cells. Notably, overexpression of SERPINA5 or high exosomal SERPINA5 levels suppressed EC metastatic potential by suppressing integrin ß1/FAK signaling pathway activation. Finally, exosomal SERPINA5 impeded tumor growth and metastasis in xenograft models. CONCLUSIONS: Our findings revealed that a low level of SERPINA5 expression indicated poor survival outcomes in EC and that exogenous SERPINA5 loading of exosomes may be a novel therapeutic strategy for metastatic EC.


Assuntos
Neoplasias do Endométrio , Exossomos , MicroRNAs , Feminino , Humanos , Exossomos/metabolismo , Integrina beta1 , Linhagem Celular Tumoral , Movimento Celular , Transdução de Sinais , Neoplasias do Endométrio/metabolismo , MicroRNAs/metabolismo , Proliferação de Células , Inibidor da Proteína C/metabolismo
7.
Oncogene ; 41(21): 3000-3010, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35459779

RESUMO

Members of the Inhibitor of Apoptosis Protein (IAP) family are essential for cell survival and appear to neutralize the cell death machinery by binding pro-apoptotic caspases. dcaf12 was recently identified as an apoptosis regulator in Drosophila. However, the underlying molecular mechanisms are unknown. Here we revealed that human DCAF12 homolog binds multiple IAPs, including XIAP, cIAP1, cIAP2, and BRUCE, through recognition of BIR domains in IAPs. The pro-apoptotic function of DCAF12 is dependent on its capacity to bind IAPs. In response to apoptotic stimuli, DCAF12 translocates from the nucleus to the cytoplasm, where it blocks the interaction between XIAP and pro-apoptotic caspases to facilitate caspase activation and apoptosis execution. Similarly, DCAF12 suppresses NF-κB activation in an IAP binding-dependent manner. Moreover, DCAF12 acts as a tumor suppressor to restrict the malignant phenotypes of cancer cells. Together, our results suggest that DCAF12 is an evolutionarily conserved IAP antagonist.


Assuntos
Proteínas Inibidoras de Apoptose , NF-kappa B , Apoptose , Caspases/metabolismo , Sobrevivência Celular , Humanos , Proteínas Inibidoras de Apoptose/química , Proteínas Inibidoras de Apoptose/genética , NF-kappa B/metabolismo , Domínios Proteicos , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo
8.
Cell Death Differ ; 29(6): 1228-1239, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-34987184

RESUMO

p62/SQSTM1 is a selective autophagy receptor that drives ubiquitinated cargos towards autophagic degradation. This receptor is also a stress-induced scaffold protein that helps cells to cope with oxidative stress through activation of the Nrf2 pathway. Functional disorders of p62 are closely associated with multiple neurodegenerative diseases and cancers. The gene encoding the E3 ubiquitin ligase substrate-binding adapter SPOP is frequently mutated in prostate cancer (PCa), but the molecular mechanisms underlying how SPOP mutations contribute to PCa tumorigenesis remain poorly understood. Here, we report that cytoplasmic SPOP binds and induces the non-degradative ubiquitination of p62 at residue K420 within the UBA domain. This protein modification decreases p62 puncta formation, liquid phase condensation, dimerization, and ubiquitin-binding capacity, thereby suppressing p62-dependent autophagy. Moreover, we show that SPOP relieves p62-mediated Keap1 sequestration, which ultimately decreases Nrf2-mediated transcriptional activation of antioxidant genes. We further show that PCa-associated SPOP mutants lose the capacity to ubiquitinate p62 and instead promote autophagy and the redox response in a dominant-negative manner. Thus, our findings indicate oncogenic roles of autophagy and Nrf2 activation in the tumorigenesis of SPOP-mutated PCa.


Assuntos
Fator 2 Relacionado a NF-E2 , Proteínas Nucleares , Neoplasias da Próstata , Proteínas Repressoras , Proteína Sequestossoma-1 , Humanos , Masculino , Autofagia/fisiologia , Carcinogênese , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Mutação , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Neoplasias da Próstata/genética , Neoplasias da Próstata/metabolismo , Proteínas Repressoras/genética , Proteínas Repressoras/metabolismo , Proteína Sequestossoma-1/genética , Proteína Sequestossoma-1/metabolismo , Ubiquitina/genética , Ubiquitina/metabolismo
9.
Int J Biol Sci ; 18(2): 800-808, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35002526

RESUMO

The RHO GTPase family has been suggested to play critical roles in cell growth, migration, and polarization. Regulators and effectors of RHO GTPases have been extensively explored in recent years. However, little attention has been given to RHO family interacting cell polarization regulators (RIPORs), a recently discovered protein family of RHO regulators. RIPOR proteins, namely, RIPOR1-3, bind directly to RHO proteins (A, B and C) via a RHO-binding motif and exert suppressive effects on RHO activity, thereby negatively influencing RHO-regulated cellular functions. In addition, RIPORs are phosphorylated by upstream protein kinases under chemokine stimulation, and this phosphorylation affects not only their subcellular localization but also their interaction with RHO proteins, altering the activation of RHO downstream targets and ultimately impacting cell polarity and migration. In this review, we provide an overview of recent studies on the function of RIPOR proteins in regulating RHO-dependent directional movement in immune responses and other pathophysiological functions.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias/patologia , Proteínas rho de Ligação ao GTP/metabolismo , Autofagia , Diferenciação Celular , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Neoplasias/etiologia , Linfócitos T/imunologia , Proteínas rho de Ligação ao GTP/genética
10.
Cell Death Differ ; 29(4): 758-771, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-34743205

RESUMO

The cystine/glutamate antiporter SLC7A11 (commonly known as xCT) functions to import cystine for glutathione biosynthesis, thereby protecting cells from oxidative stress and ferroptosis, a regulated form of non-apoptotic cell death driven by the accumulation of lipid-based reactive oxygen species (ROS). p14ARF, a well-established tumor suppressor, promotes ferroptosis by inhibiting NRF2-mediated SLC7A11 transcription. Here, we demonstrate the crucial role of Cullin 2 RING E3 ligase (CRL2)-KLHDC3 E3 ubiquitin ligase complex in regulating p14ARF protein stability. KLHDC3 acts as a CRL2 adaptor that specifically recognizes a C-terminal degron in p14ARF and triggers p14ARF for ubiquitin-proteasomal degradation. This regulation mode is absent in the murine p14ARF homolog, p19arf which lacks the C-terminal degron. We also show that KLHDC3 suppresses ferroptosis in vitro and supports tumor growth in vivo by relieving p14ARF-mediated suppression of SLC7A11 transcription. Overall, these findings reveal that the protein stability and pro-ferroptotic function of p14ARF are controlled by a CRL2 E3 ubiquitin ligase complex, and suggest that suppression of the p14ARF-NRF2-SLC7A11 regulatory pathway by KLHDC3 overexpression likely contributes to cancer progression.


Assuntos
Proteínas de Ciclo Celular , Ferroptose , Proteína Supressora de Tumor p14ARF , Ubiquitina-Proteína Ligases , Animais , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Cistina , Camundongos , Proteína Supressora de Tumor p14ARF/metabolismo , Ubiquitina-Proteína Ligases/genética
12.
Connect Tissue Res ; 60(2): 189-199, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-29852784

RESUMO

AIM: Isoquercitrin is widely present in fruits, vegetables and medicinal herbs. As a natural phytoestrogen, isoquercitrin has been considered a possible osteoporosis prevention option to avoid the risk of hormone therapy. MATERIALS AND METHODS: The cell proliferation of osteoblasts and bone mesenchymal stem cells (BMSCs) was examined by cell counting kit-8 (CCK-8). The osteogenic differentiation was evaluated by real-time qPCR, ALP staining and Alizarin Red S staining. Small interfering RNA (siRNA) was used to knockdown the expression of runt-related transcription factor 2 (RUNX2). RESULTS: The cell proliferation of osteoblasts and BMSCs was promoted by isoquercitrin at low concentrations. High concentrations of isoquercitrin promoted the osteogenic differentiation via RUNX2 expression in osteoblasts and via the bone morphogenetic protein (BMP) pathway in BMSCs. Inhibition of RUNX2 expression in osteoblasts by siRNA or addition of noggin to the culture medium of BMSCs reduced the effects of osteogenic differentiation induced by isoquercitrin. CONCLUSIONS: These data suggest that isoquercitrin is a natural potential osteoinductive compound and might be valuable for the prevention/treatment of bone disorders.


Assuntos
Proteínas Morfogenéticas Ósseas/metabolismo , Diferenciação Celular/efeitos dos fármacos , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Células-Tronco Mesenquimais/citologia , Osteoblastos/citologia , Osteoblastos/metabolismo , Osteogênese/efeitos dos fármacos , Quercetina/análogos & derivados , Animais , Calcificação Fisiológica/efeitos dos fármacos , Morte Celular/efeitos dos fármacos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Estrogênios/farmacologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Camundongos , Osteoblastos/efeitos dos fármacos , Quercetina/química , Quercetina/farmacologia , Transdução de Sinais/efeitos dos fármacos
13.
Am J Cancer Res ; 9(12): 2797-2812, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31911863

RESUMO

Dysregulation of the ubiquitin-proteasome pathway is closely associated with cancer initiation and progression. SPOP is an adapter protein of the CUL3-based E3 ubiquitin ligase complexes. Several whole genome/exome sequencing studies on endometrial cancers (ECs) revealed that the SPOP gene is frequently mutated. However, how SPOP mutations contribute to EC remains poorly understood. In this study, transcription factor ZBTB3 was identified as a proteolytic substrate for the SPOP-CUL3-RBX1 E3 ubiquitin ligase complex. SPOP specifically recognizes two Ser/Thr (S/T)-rich degrons located in ZBTB3 and triggers the degradation of ZBTB3 via the ubiquitin-proteasome pathway. By contrast, EC-associated SPOP mutants are defective in regulating ZBTB3 stability. SPOP inactivation promotes endometrial cell proliferation, migration, and invasion partly through ZBTB3 accumulation. Sonic hedgehog (SHH) was found to be a transcriptional target of ZBTB3. SPOP inactivation leads to ZBTB3-dependent SHH upregulation in EC cells. RUSKI-43, a small molecule inhibitor of SHH, suppresses cell proliferation, migration, and invasion in SPOP-depleted or EC-associated SPOP mutant-overexpressed EC cells. Our data indicate that pharmacological inhibition of SHH represents a possible treatment strategy for SPOP-mutated ECs.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA