Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Environ Sci Technol ; 50(14): 7671-8, 2016 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-27338715

RESUMO

Microcystin-LR (MC-LR) is a potent hepatotoxin that is often associated with blooms of cyanobacteria. Experiments were conducted to evaluate the efficiency of the chlorine/UV process for MC-LR decomposition and detoxification. Chlorinated MC-LR was observed to be more photoactive than MC-LR. LC/MS analyses confirmed that the arginine moiety represented an important reaction site within the MC-LR molecule for conditions of chlorination below the chlorine demand of the molecule. Prechlorination activated MC-LR toward UV254 exposure by increasing the product of the molar absorption coefficient and the quantum yield of chloro-MC-LR, relative to the unchlorinated molecule. This mechanism of decay is fundamentally different than the conventional view of chlorine/UV as an advanced oxidation process. A toxicity assay based on human liver cells indicated MC-LR degradation byproducts in the chlorine/UV process possessed less cytotoxicity than those that resulted from chlorination or UV254 irradiation applied separately. MC-LR decomposition and detoxification in this combined process were more effective at pH 8.5 than at pH 7.5 or 6.5. These results suggest that the chlorine/UV process could represent an effective strategy for control of microcystins and their associated toxicity in drinking water supplies.


Assuntos
Cloro , Cianobactérias/química , Espectrometria de Massas , Oxirredução , Abastecimento de Água
2.
Cell Metab ; 36(1): 193-208.e8, 2024 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-38171333

RESUMO

Metabolic reprogramming is key for cancer development, yet the mechanism that sustains triple-negative breast cancer (TNBC) cell growth despite deficient pyruvate kinase M2 (PKM2) and tumor glycolysis remains to be determined. Here, we find that deficiency in tumor glycolysis activates a metabolic switch from glycolysis to fatty acid ß-oxidation (FAO) to fuel TNBC growth. We show that, in TNBC cells, PKM2 directly interacts with histone methyltransferase EZH2 to coordinately mediate epigenetic silencing of a carnitine transporter, SLC16A9. Inhibition of PKM2 leads to impaired EZH2 recruitment to SLC16A9, and in turn de-represses SLC16A9 expression to increase intracellular carnitine influx, programming TNBC cells to an FAO-dependent and luminal-like cell state. Together, these findings reveal a new metabolic switch that drives TNBC from a metabolically heterogeneous-lineage plastic cell state to an FAO-dependent-lineage committed cell state, where dual targeting of EZH2 and FAO induces potent synthetic lethality in TNBC.


Assuntos
Neoplasias de Mama Triplo Negativas , Humanos , Neoplasias de Mama Triplo Negativas/metabolismo , Linhagem Celular Tumoral , Mutações Sintéticas Letais , Glicólise , Carnitina
3.
Cancers (Basel) ; 15(21)2023 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-37958341

RESUMO

Heat shock factor 1 (HSF1) is a transcription factor crucial for regulating heat shock response (HSR), one of the significant cellular protective mechanisms. When cells are exposed to proteotoxic stress, HSF1 induces the expression of heat shock proteins (HSPs) to act as chaperones, correcting the protein-folding process and maintaining proteostasis. In addition to its role in HSR, HSF1 is overexpressed in multiple cancer cells, where its activation promotes malignancy and leads to poor prognosis. The mechanisms of HSF1-induced tumorigenesis are complex and involve diverse signaling pathways, dependent on cancer type. With its important roles in tumorigenesis and tumor progression, targeting HSF1 offers a novel cancer treatment strategy. In this article, we examine the basic function of HSF1 and its regulatory mechanisms, focus on the mechanisms involved in HSF1's roles in different cancer types, and examine current HSF1 inhibitors as novel therapeutics to treat cancers.

4.
Biochem Biophys Rep ; 32: 101364, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36237442

RESUMO

Aberrant expression of protein arginine methyltransferases (PRMTs) has been implicated in a number of brain tumors, but the role of PRMT1 in medulloblastoma, the most common malignant pediatric brain tumor, remains unexplored. By examining the publicly available databases of pediatric brain tumor collection, we found that PRMT1 was predominantly expressed in medulloblastomas across all the pediatric brain tumors and that the high-level expression of PRMT1 correlated with poor survival of medulloblastoma patients. To determine the role of PRMT1 in medulloblastoma cells, we established an inducible knockdown system and demonstrated that PRMT1 depletion decreased medulloblastoma cell proliferation and induced cell apoptosis. Furthermore, the diamidine compounds, previously shown to exhibit selective PRMT1 inhibition, suppressed medulloblastoma cell viability in a dose-dependent manner. Finally, we observed induction of medulloblastoma cell apoptosis by the potent diamidine compounds at low micromolar concentrations. Together, our results suggest that PRMT1 could be an actionable therapeutic target in medulloblastoma.

5.
Cells ; 10(11)2021 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-34831357

RESUMO

Targeting the hedgehog (HH) pathway to treat aggressive cancers of the brain, breast, pancreas, and prostate has been ongoing for decades. Gli gene amplifications have been long discovered within malignant glioma patients, and since then, inhibitors against HH pathway-associated molecules have successfully reached the clinical stage where several of them have been approved by the FDA. Albeit this success rate implies suitable progress, clinically used HH pathway inhibitors fail to treat patients with metastatic or recurrent disease. This is mainly due to heterogeneous tumor cells that have acquired resistance to the inhibitors along with the obstacle of effectively targeting the tumor microenvironment (TME). Severe side effects such as hyponatremia, diarrhea, fatigue, amenorrhea, nausea, hair loss, abnormal taste, and weight loss have also been reported. Furthermore, HH signaling is known to be involved in the regulation of immune cell maturation, angiogenesis, inflammation, and polarization of macrophages and myeloid-derived suppressor cells. It is critical to determine key mechanisms that can be targeted at different levels of tumor development and progression to address various clinical issues. Hence current research focus encompasses understanding how HH controls TME to develop TME altering and combinatorial targeting strategies. In this review, we aim to discuss the pros and cons of targeting HH signaling molecules, understand the mechanism involved in treatment resistance, reveal the role of the HH pathway in anti-tumor immune response, and explore the development of potential combination treatment of immune checkpoint inhibitors with HH pathway inhibitors to target HH-driven cancers.


Assuntos
Proteínas Hedgehog/antagonistas & inibidores , Transdução de Sinais , Microambiente Tumoral , Animais , Antineoplásicos/farmacologia , Proteínas Hedgehog/metabolismo , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Imunidade/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
6.
FASEB Bioadv ; 3(6): 459-469, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-34124601

RESUMO

Vismodegib, a Smoothened antagonist, is clinically approved for treatment of human basal cell carcinoma (BCC), in the clinical trials of medulloblastoma (MB) and other cancers. However, a significant proportion of these tumors fail to respond to Vismodegib after a period of treatment. Here, we find that AMPK agonists, A769662, and Metformin, can inhibit GLI1 activity and synergize with Vismodegib to suppress MB cell growth in vitro and in vivo. Furthermore, combination of AMPK agonists with Vismodegib is effective in overcoming Vismodegib-resistant MB. This is the first report demonstrating that combining AMPK agonist (Metformin) and SHH pathway inhibitor (Vismodegib) confers synergy for MB treatment and provides an effective chemotherapeutic regimen that can be used to overcome resistance to Vismodegib in SHH-driven cancers.

7.
Biochem Biophys Res Commun ; 403(1): 103-7, 2010 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-21040707

RESUMO

While p21 is well known to inhibit cyclin-CDK activity in the nucleus and it has also been demonstrated to have oncogenic properties in different types of human cancers. In vitro studies showed that the oncogenic function of p21is closely related to its cytoplasmic localization. However, it is unclear whether cytoplasmic p21 contributes to tumorigenesis in vivo. To address this question, we generated transgenic mice expressing the Akt-phosphorylated form of p21 (p21T145D) in the mammary epithelium. The results showed that Akt-activated p21 was expressed in the cytoplasm of mammary epithelium. Overexpression of Akt-activated p21 accelerated tumor onset and promoted lung metastasis in MMTV/neu mice, providing evidence that p21, especially cytoplasmic phosphorylated p21, has an oncogenic role in promoting mammary tumorigenesis and metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Transformação Celular Neoplásica/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Epitélio/metabolismo , Neoplasias Pulmonares/secundário , Neoplasias Mamárias Experimentais/metabolismo , Animais , Neoplasias da Mama/patologia , Transformação Celular Neoplásica/patologia , Epitélio/patologia , Feminino , Humanos , Neoplasias Pulmonares/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Transgênicos , Proteínas Proto-Oncogênicas c-akt/metabolismo
8.
Clin Cancer Res ; 15(3): 752-7, 2009 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-19188143

RESUMO

Forkhead O transcription factors (FOXO) play a pivotal role in the regulation of a myriad of cellular functions including cell cycle arrest, cell death, and protection from stress stimuli. Activation of cell survival pathways such as phosphoinositide-3-kinase/AKT/IKK or RAS/mitogen-activated protein kinase are known to phosphorylate FOXOs at different sites which cause FOXOs nuclear exclusion and degradation, resulting in the suppression of FOXO's transcriptional activity. Perturbation of FOXO's function leads to deregulated cell proliferation and accumulation of DNA damage, resulting in diseases such as cancer. Emerging evidence shows that active FOXO proteins are crucial for keeping cells in check; and inactivation of FOXO proteins is associated with tumorigenesis, including breast cancer, prostate cancer, glioblastoma, rhabdomyosarcoma, and leukemia. Moreover, clinically used drugs like paclitaxel, imatinib, and doxorubicin have been shown to achieve their therapeutic effects through activation of FOXO3a and FOXO3a targets. In this review, we will focus the novel functions of FOXOs revealed in recent studies and further highlight FOXOs as new therapeutic targets in a broad spectrum of cancers.


Assuntos
Fatores de Transcrição Forkhead/fisiologia , Regulação Neoplásica da Expressão Gênica , Neoplasias/terapia , Núcleo Celular/metabolismo , Sistemas de Liberação de Medicamentos , Proteína Forkhead Box O3 , Fatores de Transcrição Forkhead/genética , Humanos , Modelos Biológicos , Neoplasias/tratamento farmacológico , Neoplasias/genética , Proteínas Quinases/fisiologia , Transdução de Sinais/efeitos dos fármacos
9.
Nat Commun ; 11(1): 4642, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32934200

RESUMO

Epigenetic regulation plays an important role in governing stem cell fate and tumorigenesis. Lost expression of a key DNA demethylation enzyme TET2 is associated with human cancers and has been linked to stem cell traits in vitro; however, whether and how TET2 regulates mammary stem cell fate and mammary tumorigenesis in vivo remains to be determined. Here, using our recently established mammary specific Tet2 deletion mouse model, the data reveals that TET2 plays a pivotal role in mammary gland development and luminal lineage commitment. We show that TET2 and FOXP1 form a chromatin complex that mediates demethylation of ESR1, GATA3, and FOXA1, three key genes that are known to coordinately orchestrate mammary luminal lineage specification and endocrine response, and also are often silenced by DNA methylation in aggressive breast cancers. Furthermore, Tet2 deletion-PyMT breast cancer mouse model exhibits enhanced mammary tumor development with deficient ERα expression that confers tamoxifen resistance in vivo. As a result, this study elucidates a role for TET2 in governing luminal cell differentiation and endocrine response that underlies breast cancer resistance to anti-estrogen treatments.


Assuntos
Diferenciação Celular , Proteínas de Ligação a DNA/metabolismo , Estradiol/metabolismo , Estrogênios/metabolismo , Glândulas Mamárias Animais/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/fisiopatologia , Linhagem da Célula , Metilação de DNA , Proteínas de Ligação a DNA/genética , Dioxigenases , Sistema Endócrino/metabolismo , Epigênese Genética , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Feminino , Humanos , Glândulas Mamárias Animais/fisiopatologia , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas/genética
10.
Mol Cell Biol ; 26(19): 7269-82, 2006 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16980628

RESUMO

Gene amplification and protein overexpression of MDM2, which is often found in certain types of cancers, indicate that MDM2 plays an important role in tumorigenesis. Interestingly, several clinical reports have demonstrated that amplification of the MDM2 gene correlates with the metastatic stage. Using an antibody array assay, we identified E-cadherin as an MDM2-binding protein and confirmed that E-cadherin is a substrate for the MDM2 E3 ubiquitin ligase. We demonstrate that MDM2 interacts in vivo with E-cadherin, resulting in its ubiquitination and degradation. This regulation appears to be clinically relevant, as we found a significant correlation between high MDM2 and low E-cadherin protein levels in resected tumor specimens recovered from breast cancer patients with lymph node metastases. Ectopic expression of MDM2 in breast cancer cells was found to disrupt cell-cell contacts and enhance cell motility and invasive potential. We found that E-cadherin and MDM2 colocalized on the plasma membrane and in the early endosome, where ubiquitin moieties were attached to E-cadherin. Blocking endocytosis with dominant-negative mutants of dynamin abolished the association of MDM2 with E-cadherin, prevented E-cadherin degradation, and attenuated cell motility as observed by fluorescence microscopy. Thus, we provide evidence to support a novel role for MDM2 in regulating cell adhesions by a mechanism that involves degrading and down-regulating the expression of E-cadherin via an endosome pathway. This novel MDM2-regulated pathway is likely to play a biologically relevant role in cancer metastasis.


Assuntos
Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Caderinas/metabolismo , Movimento Celular , Processamento de Proteína Pós-Traducional , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Neoplasias da Mama/ultraestrutura , Endocitose/fisiologia , Endossomos/ultraestrutura , Feminino , Expressão Gênica , Perfilação da Expressão Gênica , Células HeLa , Humanos , Linfonodos/patologia , Invasividade Neoplásica , Metástase Neoplásica , Ligação Proteica , Transporte Proteico , Células Tumorais Cultivadas , Ubiquitina/metabolismo
11.
Nucleic Acids Res ; 35(20): 6984-94, 2007.
Artigo em Inglês | MEDLINE | ID: mdl-17940099

RESUMO

FOXO3a is a transcription factor of the FOXO family. The FOXO proteins participate in multiple signaling pathways, and their transcriptional activity is regulated by several post-translational mechanisms, including phosphorylation, acetylation and ubiquitination. Because these post-translational modification sites are located within the C-terminal basic region of the FOXO DNA-binding domain (FOXO-DBD), it is possible that these post-translational modifications could alter the DNA-binding characteristics. To understand how FOXO mediate transcriptional activity, we report here the 2.7 A crystal structure of the DNA-binding domain of FOXO3a (FOXO3a-DBD) bound to a 13-bp DNA duplex containing a FOXO consensus binding sequence (GTAAACA). Based on a unique structural feature in the C-terminal region and results from biochemical and mutational studies, our studies may explain how FOXO-DBD C-terminal phosphorylation by protein kinase B (PKB) or acetylation by cAMP-response element binding protein (CBP) can attenuate the DNA-binding activity and thereby reduce transcriptional activity of FOXO proteins. In addition, we demonstrate that the methyl groups of specific thymine bases within the consensus sequence are important for FOXO3a-DBD recognition of the consensus binding site.


Assuntos
DNA/química , Fatores de Transcrição Forkhead/química , Fatores de Transcrição Forkhead/metabolismo , Processamento de Proteína Pós-Traducional , Sequência de Aminoácidos , Cristalografia por Raios X , DNA/metabolismo , Proteína Forkhead Box O3 , Humanos , Dados de Sequência Molecular , Estrutura Terciária de Proteína , Alinhamento de Sequência
12.
Cancer Res ; 67(10): 4564-71, 2007 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-17495324

RESUMO

Myeloid cell leukemia-1 (Mcl-1), an antiapoptotic Bcl-2 family member, is overexpressed in many types of human cancer and associates with cell immortalization, malignant transformation, and chemoresistance. Glycogen synthase kinase-3beta (GSK-3beta), a key component of the Wnt signaling pathway, is involved in multiple physiologic processes such as protein synthesis, tumorigenesis, and apoptosis. Here, we report that expression of Mcl-1 was correlated with phosphorylated GSK-3beta (p-GSK-3beta) at Ser(9) (an inactivated form of GSK-3beta) in multiple cancer cell lines and primary human cancer samples. In addition, Mcl-1 was strikingly linked with poor prognosis of human breast cancer, in which the high level of Mcl-1 was related to high tumor grade and poor survival of breast cancer patients. Furthermore, we found that activation of GSK-3beta could down-regulate Mcl-1 and was required for proteasome-mediated Mcl-1 degradation. Under some physiologic conditions, such as UV irradiation, anticancer drug treatment, and inhibition of growth factor pathways, Mcl-1 was down-regulated through activation of GSK-3beta. Our results indicate that Mcl-1 stabilization by GSK-3beta inactivation could be involved in tumorigenesis and serve as a useful prognostic marker for human breast cancer.


Assuntos
Neoplasias da Mama/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Neoplasias da Mama/enzimologia , Linhagem Celular Tumoral , Regulação para Baixo , Ativação Enzimática , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Glicogênio Sintase Quinase 3 beta , Humanos , Fator de Crescimento Insulin-Like I/farmacologia , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/biossíntese , Fosforilação , Prognóstico , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Estaurosporina/farmacologia , Raios Ultravioleta
13.
Cell Metab ; 29(4): 993-1002.e6, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30527740

RESUMO

Mitochondria are dynamic organelles that have been linked to stem cell homeostasis. However, the mechanisms involved in mitochondrial regulation of stem cell fate determination remain elusive. Here we discover that epithelial-mesenchymal transition (EMT), a key process in cancer progression, induces mitochondrial fusion through regulation of the miR200c-PGC1α-MFN1 pathway. EMT-activated MFN1 forms a complex with PKCζ and is required for PKCζ-mediated NUMB phosphorylation and dissociation from the cortical membrane to direct asymmetric division of mammary stem cells, where fused mitochondria are tethered by MFN1-PKCζ to the cortical membrane and asymmetrically segregated to the stem cell-like progeny with enhanced glutathione synthesis and reactive oxygen species scavenging capacities, allowing sustaining of a self-renewing stem cell pool. Suppression of MFN1 expression leads to equal distribution of the fragmented mitochondria in both progenies that undergo symmetric luminal cell differentiation. Together, this study elucidates an essential role of mitofusin in stem cell fate determination to mediate EMT-associated stemness.


Assuntos
Polaridade Celular , Transição Epitelial-Mesenquimal , GTP Fosfo-Hidrolases/metabolismo , Proteínas de Transporte da Membrana Mitocondrial/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Animais , Linhagem Celular , Feminino , Humanos , Camundongos , Camundongos Knockout , MicroRNAs/metabolismo , Mitocôndrias/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo/metabolismo
14.
Oncotarget ; 9(34): 23494-23504, 2018 May 04.
Artigo em Inglês | MEDLINE | ID: mdl-29805750

RESUMO

Malignant peripheral nerve sheath tumors (MPNSTs) are a type of sarcoma with poor prognosis due to their complex genetic changes, invasive growth, and insensitivity to chemo- and radiotherapies. One of the most frequently lost chromosome arms in human MPNSTs is chromosome 9p. However, the cancer driver genes located on it remain largely unknown, except the tumor suppressor gene, p16 (INK4)/CDKN2A. Previously, we identified RECK as a tumor suppressor gene candidate on chromosome 9p using zebrafish-human comparative oncogenomics. In this study, we investigated the tumorigenesis of the reck gene using zebrafish genetic models in both tp53 and ribosomal protein gene mutation background. We also examined the biological effects of RECK gene restoration in human MPNST cell lines. These results provide the first genetic evidence that reck is a bona fide tumor suppressor gene for MPNSTs in zebrafish. In addition, restoration of the RECK gene in human MPNST cells leads to growth inhibition suggesting that the reactivation of RECK could serve as a potential therapeutic strategy for MPNSTs.

15.
Sci Total Environ ; 599-600: 94-97, 2017 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-28467913

RESUMO

UV (Ultraviolet)-based treatment has been demonstrated to be effective for removal of some disinfection byproducts (DBPs) and to be beneficial for reduction of genotoxicity and cytotoxicity in chlorinated water. However, to a large extent, UV-induced effects on chemistry and toxicology have been treated as a black box, in the sense that little or no UV dose-dependent behavior has been reported. To address this issue, the effects of UV254 irradiation on 1,4-dibenzoquinone (BQ), 2,6-dichloro-1,4-benzoquinone (DCBQ), and chlorocreatinine (Cl-Cre) as model DBPs were examined, both in terms of photodegradation and cytotoxicity. These compounds have been identified as DBPs that are relevant in swimming pool settings; however, these compounds will be relevant in other water treatment settings, including drinking water production and wastewater reuse. UV254 irradiation was shown to promote photodecay of all three compounds. However, for BQ and DCBQ, the corresponding cytotoxicity of the UV-irradiated samples remained essentially unchanged, even when the compound was completely photodegraded. These results indicate that the photodegradation products of BQ and DCBQ carry similar cytotoxicity as their respective parent compounds. On the other hand, UV254-irradiation of Cl-Cre yielded a decrease in cytotoxicity that correlated with photodechlorination of Cl-Cre. These experiments also demonstrated a reduction in cytotoxicity in connection with photodechlorination of an N-chlorinated organic compound. Overall, the results of these experiments indicate the importance of defining products of UV photodecay processes, both in terms of chemistry and toxicity; these attributes are expected to be important in many UV-based applications, including potable water production, water reuse, and recreational water settings.

16.
Sci Rep ; 7: 40325, 2017 01 09.
Artigo em Inglês | MEDLINE | ID: mdl-28067315

RESUMO

Malignant peripheral nerve sheath tumors (MPNSTs) are a type of rare sarcomas with a poor prognosis due to its highly invasive nature and limited treatment options. Currently there is no targeted-cancer therapy for this type of malignancy. Thus, it is important to identify more cancer driver genes that may serve as targets of cancer therapy. Through comparative oncogenomics, we have found that KANK1 was a candidate tumor suppressor gene (TSG) for human MPNSTs. Although KANK1 is known as a cytoskeleton regulator, its tumorigenic function in MPNSTs remains largely unknown. In this study, we report that restoration of KANK1 in human MPNST cells inhibits cell growth both in human cell culture and xenograft mice by increasing apoptosis. Consistently, knockdown of KANK1 in neurofibroma cells promoted cell growth. Using RNA-seq analysis, we identified CXXC5 and other apoptosis-related genes, and demonstrated that CXXC5 is regulated by KANK1. Knockdown of CXXC5 was found to diminish KANK1-induced apoptosis in MPNST cells. Thus, KANK1 inhibits MPNST cell growth though CXXC5 mediated apoptosis. Our results suggest that KANK1 may function as a tumor suppressor in human MPNSTs, and thus it may be useful for targeted therapy.


Assuntos
Apoptose , Proteínas de Transporte/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias de Bainha Neural/genética , Neoplasias de Bainha Neural/patologia , Proteínas Supressoras de Tumor/metabolismo , Proteínas Adaptadoras de Transdução de Sinal , Animais , Apoptose/genética , Proteínas de Transporte/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Proteínas do Citoesqueleto , Proteínas de Ligação a DNA , Dosagem de Genes , Técnicas de Silenciamento de Genes , Humanos , Camundongos Endogâmicos NOD , Camundongos SCID , Fatores de Transcrição , Ensaios Antitumorais Modelo de Xenoenxerto , Peixe-Zebra
17.
Oncotarget ; 8(30): 49869-49881, 2017 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-28562331

RESUMO

Overexpression of the GLI1 gene has frequently been found in various cancer types, particularly in brain tumors, in which aberrant GLI1 induction promotes cancer cell growth. Therefore, identifying the molecular players controlling GLI1 expression is of clinical importance. Previously, we reported that AMPK directly phosphorylated and destabilized GLI1, resulting in the suppression of the Hedgehog signaling pathway. The current study not only demonstrates that AMPK inhibits GLI1 nuclear localization, but further reveals that ß-TrCP plays an essential role in AMPK-induced GLI1 degradation. We found that activation of AMPK promotes the interaction between ß-TrCP and GLI1, and induces ß-TrCP-mediated GLI1-ubiquitination and degradation. Inhibiting AMPK activity results in the dissociation of the ß-TrCP and GLI1 interaction, and diminishes ß-TrCP-mediated-GLI1 ubiquitination and degradation. On GLI1, substitution of AMPK phosphorylation sites to aspartic acid (GLI13E) results in stronger binding affinity of GLI1 with ß-TrCP, accompanied by enhanced GLI1 ubiquitination and later degradation. In contrast, the GLI1 alanine mutant (GLI13A) shows weaker binding with ß-TrCP, which is accompanied by reduced ß-TrCP-mediated ubiquitination and degradation. Together, these results demonstrate that AMPK regulates GLI1 interaction with ß-TrCP by phosphorylating GLI1 and thus both post-translational modifications by AMPK and ß-TrCP ultimately impact GLI1 degradation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Transdução de Sinais , Proteína GLI1 em Dedos de Zinco/metabolismo , Proteínas Contendo Repetições de beta-Transducina/metabolismo , Transporte Ativo do Núcleo Celular , Animais , Linhagem Celular Tumoral , Proliferação de Células , Expressão Gênica , Humanos , Camundongos , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fosforilação , Transporte Proteico , Proteólise , Ubiquitinação , Proteína GLI1 em Dedos de Zinco/genética
18.
Int J Oncol ; 29(3): 643-8, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16865280

RESUMO

Genotoxic stress such as ionizing radiation can induce DNA damage and promote cell-cycle arrest or apoptosis through either a p53-dependent or -independent pathway. Recently, members of the FOXO Forkhead transcription factor family have been implicated in playing a role in both DNA repair and apoptosis in mammalian cells that promoted us to examine the role of FOXO transcription factors in ionizing radiation-induced apoptosis. Here, we show that ionizing radiation can promote FOXO3a (FKHRL1) transcriptional activity and protein expression level, and induce nuclear translocation of FOXO3a in Saos2, a p53-null osteosarcoma cell line. Ionizing radiation stimulates expression of apoptosis-inducing proteins such as Fas ligand and the Bcl-2 interacting mediator of cell death (Bim) leading to cellular apoptosis. The observed upregulation of proapoptotic genes and apoptosis in cells without p53 in response to ionizing radiation suggests a novel p53-independent mechanism underlying ionizing radiation-induced apoptosis in cancer cells.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Apoptose/efeitos da radiação , Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/efeitos da radiação , Glicoproteínas de Membrana/metabolismo , Proteínas de Membrana/metabolismo , Osteossarcoma/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Fatores de Necrose Tumoral/metabolismo , Proteína 11 Semelhante a Bcl-2 , Neoplasias Ósseas/metabolismo , Neoplasias Ósseas/radioterapia , Núcleo Celular/metabolismo , Núcleo Celular/efeitos da radiação , Células Cultivadas , Proteína Ligante Fas , Proteína Forkhead Box O3 , Humanos , Rim/metabolismo , Rim/efeitos da radiação , Osteossarcoma/radioterapia , Regiões Promotoras Genéticas , Transporte Proteico/efeitos da radiação , Radiação Ionizante , Transcrição Gênica/efeitos da radiação , Proteína Supressora de Tumor p53/metabolismo , Regulação para Cima
19.
Cancer Res ; 64(24): 9080-5, 2004 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-15604276

RESUMO

The adenoviral protein E1A associates with multiple anticancer activities, including stabilization of p53 tumor suppressor, and has been tested through gene therapy approaches in clinical trials. To identify potential E1A-binding proteins involved in E1A's anticancer activities, we screened a yeast two-hybrid library and identified Mdm4, an Mdm2-related p53-binding protein, as a novel E1A-binding protein. The NH(2)-terminal region of Mdm4 and the CR1 domain of E1A were required for the interaction between E1A and Mdm4. E1A preferentially bound to Mdm4 rather than Mdm2 and formed a complex with p53 in the presence of Mdm4, resulting in the stabilization of p53 in a p14(ARF)-independent manner. E1A failed to stabilize p53 in the absence of Mdm4, showing that Mdm4 was required for p53 stabilization by E1A. Moreover, E1A-mediated stabilization of p53 occurred in nucleus. Although it had no effect on the p53-Mdm2 interaction, E1A facilitated Mdm4 binding to p53 and inhibited Mdm2 binding to Mdm4, resulting in decreased nuclear exportation of p53. Thus, our findings highlighted a novel mechanism, whereby E1A stabilized the p53 tumor suppressor through Mdm4.


Assuntos
Proteínas E1A de Adenovirus/metabolismo , Proteínas Proto-Oncogênicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Linhagem Celular Tumoral , Núcleo Celular/metabolismo , Humanos , Camundongos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Ligação Proteica , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-mdm2 , Proteína Supressora de Tumor p14ARF/metabolismo , Técnicas do Sistema de Duplo-Híbrido , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligases
20.
PLoS One ; 11(9): e0163229, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27658201

RESUMO

BACKGROUND: The mouse double minute 1 (Mdm1) gene was first reported and cloned in mouse tumor cell lines as an oncogene candidate. Later, it was found that mutation of Mdm1 might cause age-related retinal degeneration 2 in mice by genetic linkage analysis. Additionally, the MDM1 protein was found to be expressed in the centrosomes, cilia, and the nucleus of multiciliated tracheal epithelial cells in mice. These observations suggest that MDM1 may have some basal functions in cell physiology. However, the evolutionary history of this gene and its expression during embryonic development remain largely unexplored. RESULTS: Using molecular phylogenetic analysis, we found that the MDM1 gene encoded an evolutionarily conserved protein across all metazoans. We also found that the MDM1 gene was in a conserved synteny in vertebrates. In almost all the species that were analyzed, there was only one MDM1 gene based on current genome annotations. Since vertebrate genomes underwent two to three rounds of whole-genome duplications around the origin of the vertebrates, it is interesting that only one MDM1 ohnolog was retained. This observation implies that other MDM1 ohnologs were lost after the whole-genome duplications. Furthermore, using whole-mount in situ hybridization, we found that mdm1 was expressed in the forebrain, nephric ducts, and tail buds during zebrafish early embryonic development. CONCLUSION: MDM1 is an evolutionary conserved gene, and its homologous genes can be traced back to basal metazoan lineages. In vertebrates, the MDM1 gene is in a conserved synteny and there is only one MDM1 ohnolog suggesting it is a "duplication-resistant" gene. Its expression patterns in early zebrafish embryos indicate that mdm1 may play important roles in the development of the central nervous system, kidneys, and hematopoietic system.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA