Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
J Biol Chem ; 299(5): 104675, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-37028761

RESUMEN

MafA and c-Maf are close members of the Maf transcription factor family and indicators of poor prognosis of multiple myeloma (MM). Our previous study finds that the ubiquitin ligase HERC4 induces c-Maf degradation but stabilizes MafA, and the mechanism is elusive. In the present study, we find that HERC4 interacts with MafA and mediates its K63-linked polyubiquitination at K33. Moreover, HERC4 inhibits MafA phosphorylation and its transcriptional activity triggered by glycogen synthase kinase 3ß (GSK3ß). The K33R MafA variant prevents HERC4 from inhibiting MafA phosphorylation and increases MafA transcriptional activity. Further analyses reveal that MafA can also activate the STAT3 signaling, but it is suppressed by HERC4. Lastly, we demonstrate that lithium chloride, a GSK3ß inhibitor, can upregulate HERC4 and synergizes dexamethasone, a typical anti-MM drug, in inhibiting MM cell proliferation and xenograft growth in nude mice. These findings thus highlight a novel regulation of MafA oncogenic activity in MM and provide the rationale by targeting HERC4/GSK3ß/MafA for the treatment of MM.


Asunto(s)
Glucógeno Sintasa Quinasa 3 beta , Factores de Transcripción Maf de Gran Tamaño , Mieloma Múltiple , Poliubiquitina , Ubiquitina-Proteína Ligasas , Ubiquitinación , Animales , Humanos , Ratones , Proliferación Celular , Dexametasona/farmacología , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Cloruro de Litio/farmacología , Factores de Transcripción Maf de Gran Tamaño/antagonistas & inhibidores , Factores de Transcripción Maf de Gran Tamaño/metabolismo , Ratones Desnudos , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Fosforilación , Poliubiquitina/metabolismo , Factor de Transcripción STAT3/metabolismo , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Biochem Biophys Res Commun ; 513(4): 1013-1018, 2019 06 11.
Artículo en Inglés | MEDLINE | ID: mdl-31010679

RESUMEN

The GPCR-like transmembrane protein Smoothened (Smo) is an indispensable transducer in Hedgehog (Hh) pathway, its hyperactivation leads to several human cancers, including non-small cell lung cancer (NSCLC). The mechanism governing Smo stability still remains elusive. Here, we perform a modifier screening in Drosophila, and find that the E3 ligase dHerc4 degrades dSmo. Depletion of dherc4 increases dSmo protein and activates Hh pathway. In addition, we reveal that HERC4 is downregulated in NSCLC samples, negative correlating with Smo. HERC4 interacts with Smo reciprocally in NSCLC cells. Finally, we show that knockdown of herc4 activates Hh pathway and promotes NSCLC cell proliferation. Taken together, our studies have demonstrated that HERC4 acts as a tumor suppressor via destabilizing the oncoprotein Smo, and provided HERC4 as a promising therapeutic target for NSCLC treatment.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias Pulmonares/patología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Carcinoma de Pulmón de Células no Pequeñas/metabolismo , Regulación hacia Abajo , Drosophila , Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Receptor Smoothened/metabolismo , Células Tumorales Cultivadas , Ubiquitina-Proteína Ligasas/metabolismo
3.
J Virol ; 92(13)2018 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-29669830

RESUMEN

In humans, homologous to the E6-AP carboxyl terminus (HECT) and regulator of chromosome condensation 1 (RCC1)-like domain-containing protein 5 (HERC5) is an interferon-induced protein that inhibits replication of evolutionarily diverse viruses, including human immunodeficiency virus type 1 (HIV-1). To better understand the origin, evolution, and function of HERC5, we performed phylogenetic, structural, and functional analyses of the entire human small-HERC family, which includes HERC3, HERC4, HERC5, and HERC6. We demonstrated that the HERC family emerged >595 million years ago and has undergone gene duplication and gene loss events throughout its evolution. The structural topology of the RCC1-like domain and HECT domains from all HERC paralogs is highly conserved among evolutionarily diverse vertebrates despite low sequence homology. Functional analyses showed that the human small HERCs exhibit different degrees of antiviral activity toward HIV-1 and that HERC5 provides the strongest inhibition. Notably, coelacanth HERC5 inhibited simian immunodeficiency virus (SIV), but not HIV-1, particle production, suggesting that the antiviral activity of HERC5 emerged over 413 million years ago and exhibits species- and virus-specific restriction. In addition, we showed that both HERC5 and HERC6 are evolving under strong positive selection, particularly blade 1 of the RCC1-like domain, which we showed is a key determinant of antiviral activity. These studies provide insight into the origin, evolution, and biological importance of the human restriction factor HERC5 and the other HERC family members.IMPORTANCE Intrinsic immunity plays an important role as the first line of defense against viruses. Studying the origins, evolution, and functions of proteins responsible for effecting this defense will provide key information about virus-host relationships that can be exploited for future drug development. We showed that HERC5 is one such antiviral protein that belongs to an evolutionarily conserved family of HERCs with an ancient marine origin. Not all vertebrates possess all HERC members, suggesting that different HERCs emerged at different times during evolution to provide the host with a survival advantage. Consistent with this, two of the more recently emerged HERC members, HERC5 and HERC6, displayed strong signatures of having been involved in an ancient evolutionary battle with viruses. Our findings provide new insights into the evolutionary origin and function of the HERC family in vertebrate evolution, identifying HERC5 and possibly HERC6 as important effectors of intrinsic immunity in vertebrates.


Asunto(s)
Antivirales/metabolismo , Organismos Acuáticos , Evolución Molecular , Infecciones por VIH/virología , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Virales/metabolismo , Infecciones por VIH/genética , VIH-1/fisiología , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Filogenia , Conformación Proteica , Selección Genética , Proteínas Virales/genética
4.
Redox Biol ; 73: 103220, 2024 07.
Artículo en Inglés | MEDLINE | ID: mdl-38838551

RESUMEN

Temozolomide (TMZ) is a widely utilized chemotherapy treatment for patients with glioblastoma (GBM), although drug resistance constitutes a major therapeutic hurdle. Emerging evidence suggests that ferroptosis-mediated therapy could offer an appropriate alternative treatment option against cancer cells that are resistant to certain drugs. However, recurrent gliomas display robust ferroptosis resistance, although the precise mechanism of resistance remains elusive. In the present work, we report that proline rich protein 11 (PRR11) depletion significantly sensitizes GBM cells to TMZ by inducing ferroptosis. Mechanistically, PRR11 directly binds to and stabilizes dihydroorotate dehydrogenase (DHODH), which leads to glioma ferroptosis-resistant in a DHODH-dependent manner in vivo and in vitro. Furthermore, PRR11 inhibits HERC4 and DHODH binding, by suppressing the recruitment of E3 ubiquitin ligase HERC4 and polyubiquitination degradation of DHODH at the K306 site, which maintains DHODH protein stability. Importantly, downregulated PRR11 increases lipid peroxidation and alters DHODH-mediated mitochondrial morphology, thereby promoting ferroptosis and increasing TMZ chemotherapy sensitivity. In conclusion, our results reveal a mechanism via which PRR11 drives ferroptosis resistance and identifies ferroptosis induction and TMZ as an attractive combined therapeutic strategy for GBM.


Asunto(s)
Dihidroorotato Deshidrogenasa , Resistencia a Antineoplásicos , Ferroptosis , Glioblastoma , Temozolomida , Humanos , Ferroptosis/efectos de los fármacos , Ferroptosis/genética , Glioblastoma/metabolismo , Glioblastoma/tratamiento farmacológico , Glioblastoma/genética , Glioblastoma/patología , Temozolomida/farmacología , Resistencia a Antineoplásicos/genética , Línea Celular Tumoral , Ratones , Dihidroorotato Deshidrogenasa/metabolismo , Animales , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/genética
5.
Biochim Biophys Acta Gen Subj ; 1868(4): 130557, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38181892

RESUMEN

BACKGROUND: HERC4 has been reported to have functions in several types of tumors, but its roles in ovarian cancer have not been studied yet. METHODS: Primary tissues from ovarian cancer patients and cell lines were collected for real-time PCR. Kaplan-Meier Plotter was used to predict the prognosis of ovarian cancer patients. HERC4 was overexpressed in cells by lentivirus, and CCK-8 assay was performed to evaluate cell viability. Real-time PCR and Western blot were carried out to analyze the mRNA and protein expression, respectively. Xenograft tumor models were established to analyze HERC4 function in vivo. RESULTS: Firstly, we found that HERC4 was significantly downregulated in ovarian cancer. We then found that ovarian cancer patients with high HERC4 expression had significantly higher overall survival and progression-free survival rates compared with patients with low expression. Then, HERC4 was overexpressed in ovarian cancer cells, and we found that overexpression of HERC4 significantly inhibited ovarian cancer cell growth, as well as the expression of the target protein SMO, and the key proteins in the downstream hedgehog signaling pathway. Finally, the xenograft tumor models revealed that overexpression of HERC4 significantly inhibited tumor growth in vivo. CONCLUSIONS: Overall, these results indicate that overexpression of HERC4 inhibits cell proliferation of ovarian cancer in vitro and in vivo, suggesting that HERC4 may serve as an effective target for the treatment of ovarian cancer.


Asunto(s)
Proteínas Hedgehog , Neoplasias Ováricas , Humanos , Femenino , Proteínas Hedgehog/metabolismo , Proteínas Hedgehog/farmacología , Línea Celular Tumoral , Transducción de Señal , Neoplasias Ováricas/genética , Proliferación Celular , Receptor Smoothened/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
6.
Elife ; 112022 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-35244538

RESUMEN

The establishment of pregnancy in human necessitates appropriate decidualization of stromal cells, which involves steroids regulated periodic transformation of endometrial stromal cells during the menstrual cycle. However, the potential molecular regulatory mechanism underlying the initiation and maintenance of decidualization in humans is yet to be fully elucidated. In this investigation, we document that SOX4 is a key regulator of human endometrial stromal cells decidualization by directly regulating FOXO1 expression as revealed by whole genomic binding of SOX4 assay and RNA sequencing. Besides, our immunoprecipitation and mass spectrometry results unravel that SOX4 modulates progesterone receptor (PGR) stability through repressing E3 ubiquitin ligase HERC4-mediated degradation. More importantly, we provide evidence that dysregulated SOX4-HERC4-PGR axis is a potential cause of defective decidualization and recurrent implantation failure in in-vitro fertilization (IVF) patients. In summary, this study evidences that SOX4 is a new and critical regulator for human endometrial decidualization, and provides insightful information for the pathology of decidualization-related infertility and will pave the way for pregnancy improvement.


Asunto(s)
Decidua , Receptores de Progesterona , Decidua/metabolismo , Endometrio , Femenino , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Humanos , Embarazo , Estabilidad Proteica , Receptores de Progesterona/metabolismo , Factores de Transcripción SOXC/genética , Factores de Transcripción SOXC/metabolismo , Células del Estroma/metabolismo
7.
Transl Cancer Res ; 10(1): 349-360, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35116265

RESUMEN

BACKGROUND: Hepatic carcinoma is one of the most malignant cancers worldwide. Salvador 1 (SAV1) plays a key role in a variety of human carcinogenesis. This study investigated the role of SAV1 and HERC4 in hepatocellular carcinoma (HCC). METHODS: SAV1 and HERC4 expressions in HCC tissues were examined using RT-qPCR assay. The regulatory effect of HERC4 on SAV1 was verified by co-immunoprecipitation (Co-IP), RT-qPCR, Western blot, and immunofluorescent assays in HEP3B and Huh 7 cell lines. In addition, functional experimental verification was performed through Edu staining, colony formation, and Transwell assay. Finally, Xenograft tumor model was finally used in nude mice. RESULTS: Clinical features showed significant difference with SAV1 and HERC4 expression. HERC4 was found to be upregulated, while SAV1 was downregulated in HCC. Patients with high HERC4 or low SAV1 had a worse prognosis. Results showed that HERC4 could notably decreased the expression level of SAV1 in HCC cells. Our results showed that overexpression HERC4 could reverse the inhibitory effects of SAV1 on HCC cell proliferation, migration, and invasion. SAV1 overexpression repressed tumor growth and enhance caspase 3 expression. CONCLUSION: SAV1 can be directly downregulated by HERC4, indicating that the HERC4/SAV1 axis might have great promise for targeted therapies of HCC.

8.
J Mol Cell Biol ; 11(9): 791-803, 2019 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-30925584

RESUMEN

Hedgehog (Hh) signalling plays conserved roles in controlling embryonic development; its dysregulation causes many diseases including cancers. The G protein-coupled receptor Smoothened (Smo) is the key signal transducer of the Hh pathway, whose posttranslational regulation has been shown to be critical for its accumulation and activation. Ubiquitination has been reported an essential posttranslational regulation of Smo. Here, we identify a novel E3 ligase of Smo, Herc4, which binds to Smo, and regulates Hh signalling by controlling Smo ubiquitination and degradation. Interestingly, our data suggest that Herc4-mediated Smo degradation is regulated by Hh in PKA-primed phosphorylation-dependent and independent manners.


Asunto(s)
Proteínas de Drosophila/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal , Receptor Smoothened/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Animales , Drosophila , Proteínas de Drosophila/genética , Técnicas de Silenciamiento del Gen , Lisosomas/metabolismo , Fenotipo , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Estabilidad Proteica , Proteolisis , Ubiquitina-Proteína Ligasas/genética , Ubiquitinación
9.
DNA Cell Biol ; 36(6): 490-500, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28430527

RESUMEN

Recently, more and more evidences unveiled that ubiquitin-proteasome system (UPS) makes an important contribution to the occurrence and development of cancer. HERC4 is one identified Ubiqutin ligase E3, a member of UPS. Although some studies showed that HERC4 abnormally expresses in many cancer cells, till now, nothing has been reported about the function of HERC4 in the development of hepatoma carcinoma. To this end, in this study, we studied the function of HERC4 for the first time in hepatoma carcinoma cells. We detected the expression of HERC4 in tumor and normal tissues, and in hepatoma carcinoma cell lines by using qRT-PCR, Western blot, immunohistochemistry, and immunofluorescence. The data showed that tumor tissues expressed higher HERC4 than normal ones. HERC4 was expressed, although to a different extent, in hepatoma carcinoma cell lines. Colony formation assay, CCK-8 assay, EdU assay, wound healing assay, and FACS indicated that HERC4 plays a role in cell proliferative and migration ability. HERC4 overexpression increases the proliferative and migration ability and reduces apoptosis of hepatoma carcinoma cells; in contrast, knockdown of HERC4 decreases the proliferative and migration ability and increases the apoptosis rate of hepatoma carcinoma cells. Taken together, our findings showed that HERC4 has an effect on the occurrence and development of hepatoma carcinoma by promoting hepatoma carcinoma cell proliferation and migration, and by reducing cell apoptosis, further providing another therapeutic target for the intervention of related diseases.


Asunto(s)
Carcinoma Hepatocelular/patología , Movimiento Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas/patología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo , Apoptosis , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular , Técnicas de Silenciamiento del Gen , Humanos , Ubiquitina-Proteína Ligasas/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA