Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
1.
Cell ; 154(2): 311-324, 2013 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-23830207

RESUMEN

Tumor cells metastasize to distant organs through genetic and epigenetic alterations, including changes in microRNA (miR) expression. Here we find miR-22 triggers epithelial-mesenchymal transition (EMT), enhances invasiveness and promotes metastasis in mouse xenografts. In a conditional mammary gland-specific transgenic (TG) mouse model, we show that miR-22 enhances mammary gland side-branching, expands the stem cell compartment, and promotes tumor development. Critically, miR-22 promotes aggressive metastatic disease in MMTV-miR-22 TG mice, as well as compound MMTV-neu or -PyVT-miR-22 TG mice. We demonstrate that miR-22 exerts its metastatic potential by silencing antimetastatic miR-200 through direct targeting of the TET (Ten eleven translocation) family of methylcytosine dioxygenases, thereby inhibiting demethylation of the mir-200 promoter. Finally, we show that miR-22 overexpression correlates with poor clinical outcomes and silencing of the TET-miR-200 axis in patients. Taken together, our findings implicate miR-22 as a crucial epigenetic modifier and promoter of EMT and breast cancer stemness toward metastasis.


Asunto(s)
Neoplasias de la Mama/patología , Ensamble y Desensamble de Cromatina , Transición Epitelial-Mesenquimal , Regulación Neoplásica de la Expresión Génica , MicroARNs/metabolismo , Metástasis de la Neoplasia , Células Madre Neoplásicas/metabolismo , 5-Metilcitosina/análogos & derivados , Animales , Neoplasias de la Mama/metabolismo , Citosina/análogos & derivados , Citosina/metabolismo , Humanos , Ratones , Ratones Transgénicos , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas/metabolismo , Interferencia de ARN , Trasplante Heterólogo
2.
Cell ; 144(2): 187-99, 2011 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-21241890

RESUMEN

PTEN is a frequently mutated tumor suppressor gene that opposes the PI3K/AKT pathway through dephosphorylation of phosphoinositide-3,4,5-triphosphate. Recently, nuclear compartmentalization of PTEN was found as a key component of its tumor-suppressive activity; however its nuclear function remains poorly defined. Here we show that nuclear PTEN interacts with APC/C, promotes APC/C association with CDH1, and thereby enhances the tumor-suppressive activity of the APC-CDH1 complex. We find that nuclear exclusion but not phosphatase inactivation of PTEN impairs APC-CDH1. This nuclear function of PTEN provides a straightforward mechanistic explanation for the fail-safe cellular senescence response elicited by acute PTEN loss and the tumor-suppressive activity of catalytically inactive PTEN. Importantly, we demonstrate that PTEN mutant and PTEN null states are not synonymous as they are differentially sensitive to pharmacological inhibition of APC-CDH1 targets such as PLK1 and Aurora kinases. This finding identifies a strategy for cancer patient stratification and, thus, optimization of targeted therapies. PAPERCLIP:


Asunto(s)
Cadherinas/metabolismo , Senescencia Celular , Fosfohidrolasa PTEN/metabolismo , Complejos de Ubiquitina-Proteína Ligasa/metabolismo , Ciclosoma-Complejo Promotor de la Anafase , Animales , Antígenos CD , Aurora Quinasas , Línea Celular Tumoral , Núcleo Celular/metabolismo , Humanos , Masculino , Ratones , Trasplante de Neoplasias , Fosfohidrolasa PTEN/genética , Monoéster Fosfórico Hidrolasas/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Trasplante Heterólogo
3.
Int J Mol Sci ; 21(5)2020 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-32138313

RESUMEN

Altered gene expression is the primary molecular mechanism responsible for the pathological processes of human diseases, including cancer. MicroRNAs (miRNAs) are virtually involved at the post-transcriptional level and bind to 3' UTR of their target messenger RNA (mRNA) to suppress expression. Dysfunction of miRNAs disturbs expression of oncogenic or tumor-suppressive target genes, which is implicated in cancer pathogenesis. As such, a large number of miRNAs have been found to be downregulated or upregulated in human cancers and to function as oncomiRs or oncosuppressor miRs. Notably, the molecular mechanism underlying the dysregulation of miRNA expression in cancer has been recently uncovered. The genetic deletion or amplification and epigenetic methylation of miRNA genomic loci and the transcription factor-mediated regulation of primary miRNA often alter the landscape of miRNA expression in cancer. Dysregulation of the multiple processing steps in mature miRNA biogenesis can also cause alterations in miRNA expression in cancer. Detailed knowledge of the regulatory mechanism of miRNAs in cancer is essential for understanding its physiological role and the implications of cancer-associated dysfunction and dysregulation. In this review, we elucidate how miRNA expression is deregulated in cancer, paying particular attention to the cancer-associated transcriptional and post-transcriptional factors that execute miRNA programs.


Asunto(s)
MicroARNs/metabolismo , Neoplasias/genética , Animales , Epigénesis Genética/genética , Epigénesis Genética/fisiología , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Humanos , MicroARNs/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo
4.
Biochem Biophys Res Commun ; 496(3): 826-833, 2018 02 12.
Artículo en Inglés | MEDLINE | ID: mdl-29378184

RESUMEN

Breast cancer is the most frequently diagnosed life-threatening cancer in women. Triple-negative breast cancer (TNBC) has an aggressive clinical behavior, but the treatment of TNBC remains challenging. MicroRNAs (miRNAs) have emerged as a potential target for the diagnosis, therapy and prognosis of breast cancer. However, the precise role of miRNAs and their targets in breast cancer remain to be elucidated. Here we show that miR-218 is downregulated and miR-129 is upregulated in TNBC samples and their expressions confer prognosis to patients. Gain-of-function and loss-of-function analysis reveals that miR-218 has a tumor suppressive activity, while miR-129 acts as an oncomir in breast cancer. Notably, miR-218 and miR-129 directly target Lamin B1 and Lamin A, respectively, which are also found to be deregulated in human breast tumors. Finally, we demonstrate Lamins as the major factors in reliable miR-218 and miR-129 functions for breast cancer progression. Our findings uncover a new miRNA-mediated regulatory network for different Lamins and provide a potential therapeutic target for breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Regulación Neoplásica de la Expresión Génica , Laminas/metabolismo , MicroARNs/metabolismo , Proliferación Celular , Humanos , Células MCF-7 , Invasividad Neoplásica/patología
5.
Curr Opin Hematol ; 21(4): 276-82, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24870972

RESUMEN

PURPOSE OF REVIEW: This review highlights recent insights into the roles of microRNAs (miRNAs) in pathogenesis of myeloid malignancies and tantalising prospects of miRNA therapy. RECENT FINDINGS: New roles for miRNAs in biological and disease processes are constantly being discovered. Although great effort has been put into identifying and cataloguing aberrantly expressed miRNAs in leukaemia, very little is known about the functional consequences of their deregulation in myeloid malignancies. This review will discuss the significance of powerful oncogenic miRNAs such as miR-22 in self-renewal and transformation of haematopoietic stem cells, as well as their ability to induce epigenetic alterations in the pathogenesis of the stem cell disorder myelodysplastic syndromes and myeloid leukaemia. SUMMARY: Improved understanding of biological roles of miRNAs in the pathogenesis of haematological malignancies will allow rational stratification of patients and provide new therapeutic entries for the treatment of myelodysplastic syndromes and leukaemia.


Asunto(s)
Transformación Celular Neoplásica/genética , Leucemia Mieloide/genética , MicroARNs/genética , Síndromes Mielodisplásicos/genética , Animales , Humanos , Leucemia Mieloide/terapia , Terapia Molecular Dirigida , Síndromes Mielodisplásicos/terapia
6.
J Cancer Res Clin Oncol ; 149(9): 6351-6360, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-36752909

RESUMEN

PURPOSE: Ubiquitin-conjugating enzyme E2 C (UBE2C) is known to show a causal relationship with cancer development and advancement. The role of UBE2C is to control the mitotic spindle checkpoint. Excess UBE2C has been identified in patients with advanced prostate cancer. The objective of the present study was to examine positive connections between the expression of UBE2C and prognostic factors for prostate cancer. METHODS: Prostate cancer patients' clinical data were analysed. Tissue microarrays (TMAs) were also performed for human prostate cancer tissues (n = 335) and adjacent non-neoplastic tissues (n = 22). TMA slides were incubated with antibodies against UBE2C. Cores were scored by a pathologist who was blind to cancer results. RESULTS: Of 335 prostate cancer patients, 200 could be assessed for biochemical recurrence, clinical recurrence, and overall survival. Human prostate cancer tissues showed higher expression of UBE2C than adjacent non-neoplastic tissues. High expression level of UBE2C showed a strong positive relationship with a high prostate-specific antigen (PSA), Gleason's score, and pathological stage of prostate cancer. Patients with a higher UBE2C grade demonstrated greater lymphatic engagement of prostate cancer than those with a lower UBE2C grade. CONCLUSION: The expression of UBE2C has positive correlations with several prognostic factors for prostate cancer. Thus, investigating the expression level of UBE2C staining is a promising tool for predicting prostate cancer prognosis.


Asunto(s)
Neoplasias de la Próstata , Enzimas Ubiquitina-Conjugadoras , Masculino , Humanos , Pronóstico
7.
Pharmaceuticals (Basel) ; 16(12)2023 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-38139829

RESUMEN

BACKGROUND: Ubiquitin-specific protease 11 (USP11), one of the principal phosphatase and tensin homolog (PTEN) deubiquitinases, can reserve PTEN polyubiquitination to maintain PTEN protein integrity and inhibit PI3K/AKT pathway activation. The aim of the current study was to investigate the associations between immunohistochemical USP11 staining intensities and prognostic indicators in individuals with prostate cancer. METHODS: Tissue microarrays (TMAs) were performed for human prostate cancer and normal tissue (control) samples. Data on patient's age, Gleason score, plasma prostate-specific antigen (PSA) titer, disease stage, and presence of seminal vesicles, lymph nodes, and surgical margin involvement were collected. A pathologist who was blinded to the clinical outcome data scored the TMA for USP11 staining intensity as either positive or negative. RESULTS: Cancerous tissues exhibited lower USP11 staining intensity, whereas the neighboring benign peri-tumoral tissues showed higher USP11 staining intensity. The degree of USP11 staining intensity was lower in patients with a higher PSA titer, higher Gleason score, or more advanced disease stage. Patients who showed positive USP11 staining were more likely to have more optimal clinical and biochemical recurrence-free survival statistics. CONCLUSIONS: USP11 staining intensity in patients with prostate cancer is negatively associated with several prognostic factors such as an elevated PSA titer and a high Gleason score. It also reflects both biochemical and clinical recurrence-free survival in such patients. Thus, USP11 staining is a valuable prognostic factor in patients with prostate cancer.

8.
EMBO J ; 27(13): 1863-74, 2008 Jul 09.
Artículo en Inglés | MEDLINE | ID: mdl-18566590

RESUMEN

The tumour suppressor p53, which accumulates in response to DNA damage and induces cell-cycle arrest and apoptosis, has a key function in the maintenance of genome integrity. Under normal conditions, the antiproliferative effects of p53 are inhibited by MDM2, a ubiquitin ligase that promotes p53 ubiquitination and degradation. MDM2 is also self-ubiquitinated and degraded. Here, we show that the tumour suppressor RASSF1A regulates G(1)-S cell-cycle progression in a p53-dependent manner by promoting MDM2 self-ubiquitination and preventing p53 degradation. Importantly, RASSF1A associates with MDM2 and death-domain-associated protein (DAXX) in the nucleus, thereby disrupting the interactions between MDM2, DAXX, and the deubiquitinase, HAUSP, and enhancing the self-ubiquitin ligase activity of MDM2. Moreover, RASSF1A partially contributes to p53-dependent checkpoint activation at early time points in response to DNA damage. These findings reveal a new and important function for RASSF1A in regulating the p53-MDM2 pathway.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas c-mdm2/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Línea Celular Tumoral , Proteínas Co-Represoras , Fibroblastos/metabolismo , Humanos , Ratones , Chaperonas Moleculares , Peptidasa Específica de Ubiquitina 7 , Ubiquitinación
9.
Exp Mol Med ; 54(11): 1814-1821, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36385557

RESUMEN

PTEN is among the most commonly lost or mutated tumor suppressor genes in human cancer. PTEN, a bona fide lipid phosphatase that antagonizes the highly oncogenic PI3K-AKT-mTOR pathway, is considered a major dose-dependent tumor suppressor. Although PTEN function can be compromised by genetic mutations in inherited syndromes and cancers, posttranslational modifications of PTEN may also play key roles in the dynamic regulation of its function. Notably, deregulated ubiquitination and deubiquitination lead to detrimental impacts on PTEN levels and subcellular partitioning, promoting tumorigenesis. While PTEN can be targeted by HECT-type E3 ubiquitin ligases for nuclear import and proteasomal degradation, studies have shown that several deubiquitinating enzymes, including HAUSP/USP7, USP10, USP11, USP13, OTUD3 and Ataxin-3, can remove ubiquitin from ubiquitinated PTEN in cancer-specific contexts and thus reverse ubiquitination-mediated PTEN regulation. Researchers continue to reveal the precise molecular mechanisms by which cancer-specific deubiquitinases of PTEN regulate its roles in the pathobiology of cancer, and new methods of pharmacologically for modulating PTEN deubiquitinases are critical areas of investigation for cancer treatment and prevention. Here, we assess the mechanisms and functions of deubiquitination as a recently appreciated mode of PTEN regulation and review the link between deubiquitinases and PTEN reactivation and its implications for therapeutic strategies.


Asunto(s)
Neoplasias , Fosfatidilinositol 3-Quinasas , Humanos , Fosfatidilinositol 3-Quinasas/metabolismo , Ubiquitinación , Ubiquitina/metabolismo , Carcinogénesis/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Enzimas Desubicuitinizantes , Tioléster Hidrolasas/metabolismo , Ubiquitina Tiolesterasa/genética , Ubiquitina Tiolesterasa/metabolismo , Peptidasa Específica de Ubiquitina 7/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo
10.
Pharmaceuticals (Basel) ; 14(8)2021 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-34451875

RESUMEN

BACKGROUND: The mTOR signaling pathway is inactivated by AMPK's tumor-suppressing function. It is recognized that ubiquitin conjugating enzyme 2O (UBE2O), which directly targets AMPK for ubiquitination and degradation, is intensified in human cancers. METHODS: This study investigated the clinical data about prostate cancer. Examination was also carried out into tissue microarrays (TMA) of human prostate cancer (n = 382) and adjacent non-neoplastic tissues around prostate cancer (n = 61). The TMA slides were incubated with antibodies against UBE2O, and the cores were scored by the pathologist blind to cancer results. RESULTS: Very strong positive correlations were identified between the expression of UBE2O staining and high PSA and pathological stage of prostate cancer. Cox's proportional hazard analysis established correlations between the following: (1) positive surgical margin and biochemical recurrence free survival, (2) PSA grade and clinical recurrence free survival, (3) regional lymph node positive and clinical recurrence free survival, (4) adjuvant treatment and overall survival, and (5) pathological T stage and overall survival. CONCLUSION: There is a positive correlation between the expression of UBE2O staining and prognosis for prostate cancer. Thus, a prostate cancer prognosis can be assessed with the expression of UBE2O staining.

11.
Cell Metab ; 33(12): 2380-2397.e9, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34879239

RESUMEN

Accelerated glycolysis is the main metabolic change observed in cancer, but the underlying molecular mechanisms and their role in cancer progression remain poorly understood. Here, we show that the deletion of the long noncoding RNA (lncRNA) Neat1 in MMTV-PyVT mice profoundly impairs tumor initiation, growth, and metastasis, specifically switching off the penultimate step of glycolysis. Mechanistically, NEAT1 directly binds and forms a scaffold bridge for the assembly of PGK1/PGAM1/ENO1 complexes and thereby promotes substrate channeling for high and efficient glycolysis. Notably, NEAT1 is upregulated in cancer patients and correlates with high levels of these complexes, and genetic and pharmacological blockade of penultimate glycolysis ablates NEAT1-dependent tumorigenesis. Finally, we demonstrate that Pinin mediates glucose-stimulated nuclear export of NEAT1, through which it exerts isoform-specific and paraspeckle-independent functions. These findings establish a direct role for NEAT1 in regulating tumor metabolism, provide new insights into the Warburg effect, and identify potential targets for therapy.


Asunto(s)
Neoplasias de la Mama , MicroARNs , ARN Largo no Codificante , Animales , Neoplasias de la Mama/genética , Línea Celular Tumoral , Proliferación Celular/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Glucólisis , Humanos , Ratones , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo
12.
Nat Commun ; 10(1): 3716, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31420536

RESUMEN

The Anaphase Promoting Complex (APC) coactivator Cdh1 drives proper cell cycle progression and is implicated in the suppression of tumorigenesis. However, it remains elusive how Cdh1 restrains cancer progression and how tumor cells escape the inhibition of Cdh1. Here we report that Cdh1 suppresses the kinase activity of c-Src in an APC-independent manner. Depleting Cdh1 accelerates breast cancer cell proliferation and cooperates with PTEN loss to promote breast tumor progression in mice. Hyperactive c-Src, on the other hand, reciprocally inhibits the ubiquitin E3 ligase activity of APCCdh1 through direct phosphorylation of Cdh1 at its N-terminus, which disrupts the interaction between Cdh1 and the APC core complex. Furthermore, pharmacological inhibition of c-Src restores APCCdh1 tumor suppressor function to repress a panel of APCCdh1 oncogenic substrates. Our findings reveal a reciprocal feedback circuit of Cdh1 and c-Src in the crosstalk between the cell cycle machinery and the c-Src signaling pathway.


Asunto(s)
Ciclosoma-Complejo Promotor de la Anafase/metabolismo , Proteínas Cdh1/metabolismo , Proteínas Proto-Oncogénicas pp60(c-src)/metabolismo , Animales , Neoplasias de la Mama , Carcinogénesis , Proteínas Cdh1/genética , Línea Celular Tumoral , Proliferación Celular , Femenino , Humanos , Células MCF-7 , Ratones , Ratones Noqueados , Trasplante de Neoplasias , Fosfohidrolasa PTEN/genética , Ubiquitina-Proteína Ligasas/metabolismo
13.
JCI Insight ; 4(13)2019 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-31292296

RESUMEN

Ubiquitin-conjugating enzyme E2O (UBE2O) is expressed preferentially in metabolic tissues, but its role in regulating energy homeostasis has yet to be defined. Here we find that UBE2O is markedly upregulated in obese subjects with type 2 diabetes and show that whole-body disruption of Ube2o in mouse models in vivo results in improved metabolic profiles and resistance to high-fat diet-induced (HFD-induced) obesity and metabolic syndrome. With no difference in nutrient intake, Ube2o-/- mice were leaner and expended more energy than WT mice. In addition, hyperinsulinemic-euglycemic clamp studies revealed that Ube2o-/- mice were profoundly insulin sensitive. Through phenotype analysis of HFD mice with muscle-, fat-, or liver-specific knockout of Ube2o, we further identified UBE2O as an essential regulator of glucose and lipid metabolism programs in skeletal muscle, but not in adipose or liver tissue. Mechanistically, UBE2O acted as a ubiquitin ligase and targeted AMPKα2 for ubiquitin-dependent degradation in skeletal muscle; further, muscle-specific heterozygous knockout of Prkaa2 ablated UBE2O-controlled metabolic processes. These results identify the UBE2O/AMPKα2 axis as both a potent regulator of metabolic homeostasis in skeletal muscle and a therapeutic target in the treatment of diabetes and metabolic disorders.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Resistencia a la Insulina , Síndrome Metabólico/metabolismo , Obesidad/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Animales , Línea Celular , Diabetes Mellitus Tipo 2/complicaciones , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Femenino , Glucosa/metabolismo , Humanos , Insulina/metabolismo , Metabolismo de los Lípidos , Masculino , Síndrome Metabólico/etiología , Ratones , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Mioblastos Esqueléticos , Obesidad/etiología , Cultivo Primario de Células , Proteolisis , Enzimas Ubiquitina-Conjugadoras/análisis , Enzimas Ubiquitina-Conjugadoras/genética , Ubiquitinación , Regulación hacia Arriba
14.
Nat Commun ; 10(1): 636, 2019 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-30733438

RESUMEN

PTEN is a lipid phosphatase that antagonizes the PI3K/AKT pathway and is recognized as a major dose-dependent tumor suppressor. The cellular mechanisms that control PTEN levels therefore offer potential routes to therapy, but these are as yet poorly defined. Here we demonstrate that PTEN plays an unexpected role in regulating its own stability through the transcriptional upregulation of the deubiquitinase USP11 by the PI3K/FOXO pathway, and further show that this feedforward mechanism is implicated in its tumor-suppressive role, as mice lacking Usp11 display increased susceptibility to PTEN-dependent tumor initiation, growth and metastasis. Notably, USP11 is downregulated in cancer patients, and correlates with PTEN expression and FOXO nuclear localization. Our findings therefore demonstrate that PTEN-PI3K-FOXO-USP11 constitute the regulatory feedforward loop that improves the stability and tumor suppressive activity of PTEN.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Tioléster Hidrolasas/metabolismo , Animales , Células Cultivadas , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Imagen por Resonancia Magnética , Masculino , Ratones , Ratones Noqueados , Ratones Desnudos , Fosfohidrolasa PTEN/genética , Fosfatidilinositol 3-Quinasas/genética , Transducción de Señal/genética , Transducción de Señal/fisiología , Tioléster Hidrolasas/genética
15.
Cancer Res ; 66(5): 2562-9, 2006 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-16510573

RESUMEN

Mammalian sterile 20-like kinase 1 (Mst1) is activated by both caspase-mediated cleavage and phosphorylation in response to apoptotic stimuli, including Fas ligation. Here, we examined the possible role of the tumor suppressor RASSF1A in Mst1 activation and Mst1-mediated apoptosis induced by death receptor signaling. Immunoprecipitation and immunofluorescence analyses revealed that Mst1 was associated with RASSF1A in cultured mammalian cells, with both proteins colocalizing to microtubules throughout the cell cycle. Whereas purified recombinant RASSF1A inhibited the kinase activity of purified recombinant Mst1 in vitro, overexpression of RASSF1A increased the kinase activity of Mst1 in intact cells, suggesting that regulation of Mst1 by RASSF1A in vivo involves more than the simple association of the two proteins. Both the activation of Mst1 and the incidence of apoptosis induced by Fas ligation were markedly reduced in cells depleted of RASSF1A by RNA interference and were increased by restoration of RASSF1A expression in RASSF1A-deficient cells. Moreover, the stimulatory effect of RASSF1A overexpression on Fas-induced apoptosis was inhibited by depletion of Mst1. These findings indicate that RASSF1A facilitates Mst1 activation and thereby promotes apoptosis induced by death receptor signaling.


Asunto(s)
Apoptosis/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Línea Celular Tumoral , Activación Enzimática , Células HeLa , Humanos , Inmunoprecipitación , Péptidos y Proteínas de Señalización Intracelular , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , ARN Interferente Pequeño/genética , Serina-Treonina Quinasa 3 , Transfección , Proteínas Supresoras de Tumor/biosíntesis , Proteínas Supresoras de Tumor/genética , Receptor fas/metabolismo
16.
Mol Cells ; 41(3): 168-178, 2018 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-29562734

RESUMEN

Intracellular communication via ubiquitin (Ub) signaling impacts all aspects of cell biology and regulates pathways critical to human development and viability; therefore aberrations or defects in Ub signaling can contribute to the pathogenesis of human diseases. Ubiquitination consists of the addition of Ub to a substrate protein via coordinated action of E1-activating, E2-conjugating and E3-ligating enzymes. Approximately 40 E2s have been identified in humans, and most are thought to be involved in Ub transfer; although little information is available regarding the majority of them, emerging evidence has highlighted their importance to human health and disease. In this review, we focus on recent insights into the pathogenetic roles of E2s (particularly the ubiquitin-conjugating enzyme E2O [UBE2O]) in debilitating diseases and cancer, and discuss the tantalizing prospect that E2s may someday serve as potential therapeutic targets for human diseases.


Asunto(s)
Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Ubiquitina/metabolismo , Ubiquitinación/genética , Humanos
17.
Mol Cell Oncol ; 4(3): e1304846, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28616582

RESUMEN

Ubiquitin-conjugating enzyme E2O (UBE2O) is upregulated in human cancers. We have demonstrated that genetic deletion or pharmacological blockade of UBE2O reduces tumorigenesis through inhibiting the mammalian target of rapamycin complex 1-hypoxia-inducible factor 1-α pathway. Critically, UBE2O targets adenosine monophosphate (AMP)-activated protein kinase-α 2 (AMPKα2) for ubiquitination and degradation. We thus suggest the UBE2O-AMPKα2 axis as a potential therapeutic target for cancer.

18.
Front Oncol ; 7: 331, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29441324

RESUMEN

Myeloid malignancies, including myelodysplastic syndromes and acute myeloid leukemia, are clonal diseases arising in hematopoietic stem or progenitor cells. In recent years, microRNA (miRNA) expression profiling studies have revealed close associations of miRNAs with cytogenetic and molecular subtypes of myeloid malignancies, as well as outcome and prognosis of patients. However, the roles of miRNA deregulation in the pathogenesis of myeloid malignancies and how they cooperate with protein-coding gene variants in pathological mechanisms leading to the diseases have not yet been fully understood. In this review, we focus on recent insights into the role of miRNAs in the development and progression of myeloid malignant diseases and discuss the prospect that miRNAs may serve as a potential therapeutic target for leukemia.

19.
Cancer Cell ; 31(2): 208-224, 2017 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-28162974

RESUMEN

UBE2O is localized in the 17q25 locus, which is known to be amplified in human cancers, but its role in tumorigenesis remains undefined. Here we show that Ube2o deletion in MMTV-PyVT or TRAMP mice profoundly impairs tumor initiation, growth, and metastasis, while switching off the metabolic reprogramming of tumor cells. Mechanistically, UBE2O specifically targets AMPKα2 for ubiquitination and degradation, and thereby promotes activation of the mTOR-HIF1α pathway. Notably, inactivation of AMPKα2, but not AMPKα1, abrogates the tumor attenuation caused by UBE2O loss, while treatment with rapamycin or inhibition of HIF1α ablates UBE2O-dependent tumor biology. Finally, pharmacological blockade of UBE2O inhibits tumorigenesis through the restoration of AMPKα2, suggesting the UBE2O-AMPKα2 axis as a potential cancer therapeutic target.


Asunto(s)
Proteínas Quinasas Activadas por AMP/fisiología , Neoplasias/etiología , Enzimas Ubiquitina-Conjugadoras/fisiología , Proteínas Quinasas Activadas por AMP/antagonistas & inhibidores , Animales , Antígenos de Neoplasias/metabolismo , Progresión de la Enfermedad , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/fisiología , Ratones , Ratones Endogámicos C57BL , Proteínas de Neoplasias/metabolismo , Neoplasias/tratamiento farmacológico , Serina-Treonina Quinasas TOR/fisiología , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Ubiquitinación
20.
Cancer Discov ; 7(4): 424-441, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28174173

RESUMEN

BRAF drives tumorigenesis by coordinating the activation of the RAS/RAF/MEK/ERK oncogenic signaling cascade. However, upstream pathways governing BRAF kinase activity and protein stability remain undefined. Here, we report that in primary cells with active APCFZR1, APCFZR1 earmarks BRAF for ubiquitination-mediated proteolysis, whereas in cancer cells with APC-free FZR1, FZR1 suppresses BRAF through disrupting BRAF dimerization. Moreover, we identified FZR1 as a direct target of ERK and CYCLIN D1/CDK4 kinases. Phosphorylation of FZR1 inhibits APCFZR1, leading to elevation of a cohort of oncogenic APCFZR1 substrates to facilitate melanomagenesis. Importantly, CDK4 and/or BRAF/MEK inhibitors restore APCFZR1 E3 ligase activity, which might be critical for their clinical effects. Furthermore, FZR1 depletion cooperates with AKT hyperactivation to transform primary melanocytes, whereas genetic ablation of Fzr1 synergizes with Pten loss, leading to aberrant coactivation of BRAF/ERK and AKT signaling in mice. Our findings therefore reveal a reciprocal suppression mechanism between FZR1 and BRAF in controlling tumorigenesis.Significance: FZR1 inhibits BRAF oncogenic functions via both APC-dependent proteolysis and APC-independent disruption of BRAF dimers, whereas hyperactivated ERK and CDK4 reciprocally suppress APCFZR1 E3 ligase activity. Aberrancies in this newly defined signaling network might account for BRAF hyperactivation in human cancers, suggesting that targeting CYCLIN D1/CDK4, alone or in combination with BRAF/MEK inhibition, can be an effective anti-melanoma therapy. Cancer Discov; 7(4); 424-41. ©2017 AACR.See related commentary by Zhang and Bollag, p. 356This article is highlighted in the In This Issue feature, p. 339.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/genética , Proteínas Cdh1/genética , Melanoma/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Animales , Carcinogénesis/genética , Proteínas Cdh1/metabolismo , Línea Celular Tumoral , Ciclina D1/genética , Dimerización , Células HeLa , Humanos , Melanocitos/metabolismo , Melanocitos/patología , Melanoma/tratamiento farmacológico , Melanoma/patología , Ratones , Complejos Multiproteicos/genética , Fosforilación/genética , Inhibidores de Proteínas Quinasas/administración & dosificación , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal/efectos de los fármacos , Ubiquitina-Proteína Ligasas/genética , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA