Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Artigo em Inglês | MEDLINE | ID: mdl-38814596

RESUMO

Background: Infectious bone defect refers to severe bone tissue damage caused by skeletal infection, often resulting in impaired skeletal function and intense inflammatory responses. Treating infectious bone defects is a challenging task, as conventional treatment methods often fail to completely eliminate the infection focus and may easily lead to inflammatory responses in the bone defect area. Objective: To examine the impacts of bone transport (BT) in conjunction with drug-loaded calcium sulfate (DLCS) on the expression of inflammatory factors and vascular endothelial growth factor (VEGF) in rats with infectious bone defects. Methods: A total of 40 rats were randomly allocated to 4 groups-the sham, model, BT, and BT + DLCS groups-with 10 rats in each group. Interleukin 10 (IL-10), tumor necrosis factor (TNF), nuclear factor-κB (NF-κB), insulinlike growth factor 1 (IGF 1), and recombinant human basic fibroblast growth factor (rhbFGF) concentrations in serum were measured using enzyme-linked immunosorbent assay. In bone tissue, histopathological changes in defective bone were assessed through hematoxylin-eosin staining, CD34 expression was examined by immunohistochemistry, and VEGF expression was examined by Western blot. Results: In comparison with the sham group, the model group had significant increases in serum IL-10, TNF, and NF-κB concentrations as well as notable decreases in IGF-1 and rhbFGF serum concentrations and CD34 and VEGF expression in the bone tissue (P < .05). In contrast to the model group, both the BT and BT + DLCS groups had significant reductions in serum concentrations of IL-10, TNF, and NF-κB. Additionally, the BT and BT + DLCS groups had significant increases in serum concentrations of IGF-1 and rhbFGF as well as expression of CD34 and VEGF in the bone tissue (P < .05). The BT + DLCS group had significantly lower serum concentrations of IL-10, TNF, and NF-κB compared with the BT group. Furthermore, the BT + DLCS group had significantly elevated serum concentrations of IGF-1 and rhbFGF as well as increased expression of CD34 and VEGF in the bone tissue compared with the BT group (P < .05). Conclusion: The promotion of infected bone defect healing in rats through the combination of BT and DLCS may be attributed to the suppression of inflammatory responses and the elevation of VEGF expression to facilitate vascular regeneration.

2.
Front Cell Dev Biol ; 9: 724059, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34820369

RESUMO

Hypoxia is a universal pathological feature of solid tumors. Hypoxic tumor cells acquire metastatic and lethal phenotypes primarily through the activities of hypoxia-inducible factor 1 alpha (HIF1α). Therefore, HIF1α is considered as a promising therapeutic target. However, HIF inhibitors have not proven to be effective in clinical testing. The underlying mechanism is unclear. We report that oncogenic protein ID1 is upregulated in hypoxia by HIF1α shRNA or pharmacological inhibitors. In turn, ID1 supports tumor growth in hypoxia in vitro and in xenografts in vivo, conferring adaptive survival response and resistance. Mechanistically, ID1 proteins interfere HIF1-mediated gene transcription activation, thus ID1 protein degradation is accelerated by HIF1α-dependent mechanisms in hypoxia. Inhibitions of HIF1α rescues ID1, which compensates the loss of HIF1α by the upregulation of GLS2 and glutamine metabolism, thereby switching the metabolic dependency of HIF1α -inhibited cells from glucose to glutamine.

3.
Cell Rep Med ; 2(5): 100267, 2021 05 18.
Artigo em Inglês | MEDLINE | ID: mdl-34095877

RESUMO

The lack of effective treatment options for advanced non-clear cell renal cell carcinoma (NCCRCC) is a critical unmet clinical need. Applying a high-throughput drug screen to multiple human kidney cancer cells, we identify the combination of the VEGFR-MET inhibitor cabozantinib and the SRC inhibitor dasatinib acts synergistically in cells to markedly reduce cell viability. Importantly, the combination is well tolerated and causes tumor regression in vivo. Transcriptional and phosphoproteomic profiling reveals that the combination converges to downregulate the MAPK-ERK signaling pathway, a result not predicted by single-agent analysis alone. Correspondingly, the addition of a MEK inhibitor synergizes with either dasatinib or cabozantinib to increase its efficacy. This study, by using approved, clinically relevant drugs, provides the rationale for the design of effective combination treatments in NCCRCC that can be rapidly translated to the clinic.


Assuntos
Anilidas/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Dasatinibe/farmacologia , Piridinas/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Linhagem Celular Tumoral , Humanos , Neoplasias Renais/tratamento farmacológico , Inibidores de Proteínas Quinases/administração & dosagem , Transdução de Sinais/efeitos dos fármacos , Quinases da Família src/metabolismo
4.
Nat Commun ; 10(1): 164, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30622254

RESUMO

The original version of this Article contained errors in Fig. 7. In panels e and f, the graph titles incorrectly read 'LNCaP-AdtNs' and 'LAPC4-AdtNs', respectively. These errors have now been corrected in both the PDF and HTML versions of the Article.

5.
Nat Commun ; 9(1): 4972, 2018 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-30478344

RESUMO

Despite recent advances, the efficacy of androgen/androgen receptor (AR)-targeted therapy remains  limited for many patients with metastatic prostate cancer. This is in part because prostate cancers adaptively switch to the androgen/AR-independent pathway for survival and growth, thereby conferring therapy resistance. Tumor hypoxia is considered as a major cause of treatment resistance. However, the exact mechanism is largely unclear. Here we report that chronic-androgen deprivation therapy (ADT) in the condition of hypoxia induces adaptive androgen/AR-independence, and therefore confers resistance to androgen/AR-targeted therapy, e.g., enzalutamide. Mechanistically, this is mediated by glucose-6-phosphate isomerase (GPI), which is transcriptionally repressed by AR in hypoxia, but restored and increased by AR inhibition. In turn, GPI maintains glucose metabolism and energy homeostasis in hypoxia by redirecting the glucose flux from androgen/AR-dependent pentose phosphate pathway (PPP) to hypoxia-induced glycolysis pathway, thereby reducing the growth inhibitory effect of enzalutamide. Inhibiting GPI overcomes the therapy resistance in hypoxia in vitro and increases enzalutamide efficacy in vivo.


Assuntos
Androgênios/farmacologia , Resistencia a Medicamentos Antineoplásicos , Terapia de Alvo Molecular , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Receptores Androgênicos/metabolismo , Hipóxia Tumoral/efeitos dos fármacos , Benzamidas , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Epigênese Genética/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glucose/metabolismo , Glucose-6-Fosfato Isomerase/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Nitrilas , Feniltioidantoína/análogos & derivados , Feniltioidantoína/farmacologia , Neoplasias da Próstata/genética , Transcrição Gênica/efeitos dos fármacos , Hipóxia Tumoral/genética , Regulação para Cima/efeitos dos fármacos
6.
Genes Dev ; 31(20): 2067-2084, 2017 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-29138276

RESUMO

There is limited knowledge about the metabolic reprogramming induced by cancer therapies and how this contributes to therapeutic resistance. Here we show that although inhibition of PI3K-AKT-mTOR signaling markedly decreased glycolysis and restrained tumor growth, these signaling and metabolic restrictions triggered autophagy, which supplied the metabolites required for the maintenance of mitochondrial respiration and redox homeostasis. Specifically, we found that survival of cancer cells was critically dependent on phospholipase A2 (PLA2) to mobilize lysophospholipids and free fatty acids to sustain fatty acid oxidation and oxidative phosphorylation. Consistent with this, we observed significantly increased lipid droplets, with subsequent mobilization to mitochondria. These changes were abrogated in cells deficient for the essential autophagy gene ATG5 Accordingly, inhibition of PLA2 significantly decreased lipid droplets, decreased oxidative phosphorylation, and increased apoptosis. Together, these results describe how treatment-induced autophagy provides nutrients for cancer cell survival and identifies novel cotreatment strategies to override this survival advantage.


Assuntos
Antineoplásicos/farmacologia , Neoplasias/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose , Autofagia , Benzamidas/farmacologia , Linhagem Celular Tumoral , Respiração Celular/efeitos dos fármacos , Sobrevivência Celular , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Gotículas Lipídicas/metabolismo , Camundongos , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Neoplasias/enzimologia , Neoplasias/patologia , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Inibidores de Fosfolipase A2/farmacologia , Fosfolipídeos/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Pirimidinas/farmacologia , Células Tumorais Cultivadas
7.
ACS Appl Mater Interfaces ; 9(41): 35721-35728, 2017 Oct 18.
Artigo em Inglês | MEDLINE | ID: mdl-28948777

RESUMO

Enzymatic fuel cell (EFC)-based self-powered biosensors have attracted considerable attention because of their unique feature of no need for extra power sources during the entire detection process, which endows them with the merits of simplicity, rapidness, low cost, anti-interference, and ease of use. Herein, we proposed, for the first time, an EFC-based self-powered homogeneous immunosensing platform by integrating the target-induced biofuel release and bioconjugate immunoassay for ultrasensitive melamine (ME) detection. In this design, the biofuel, i.e., glucose molecules, was entrapped in the pores of positively charged mesoporous silica nanoparticles and capped by the biogate AuNPs-labeled anti-ME antibody (AuNPs-Ab). The presence of the target ME triggered the entrapped glucose release due to the removal of the biogate via immunoreaction, which resulted in the transfer of electrons produced by glucose oxidation at the bioanode to the biocathode, and thus, the open-circuit voltage of the EFC-based self-powered immunosensor dramatically increased, realizing the ultrasensitive turn-on assay for ME. The limit of detection for ME assay was down to 2.1 pM (S/N = 3), superior to those previously reported in the literature. Notably, real milk samples need no special sample pretreatment for the detection of ME because of the good anti-interference ability of EFC-based self-powered biosensors and the excellent selectivity of the homogeneous immunoassay. Therefore, this appealing self-powered homogeneous immunosensing platform holds great promise as a successful prototype of portable and on-site bioassay in the field of food safety.


Assuntos
Glucose/química , Técnicas Biossensoriais , Imunoensaio , Limite de Detecção , Triazinas
8.
Sci Rep ; 6: 34513, 2016 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-27694829

RESUMO

cAMP-response element binding protein (CREB) is a nuclear transcription factor activated by multiple extracellular signals including growth factors and hormones. These extracellular cues activate CREB through phosphorylation at Ser133 by various protein serine/threonine kinases. Once phosphorylated, it promotes its association with transcription coactivators CREB-binding protein (CBP) and its paralog p300 to activate CREB-dependent gene transcription. Tumor tissues of different origins have been shown to present overexpression and/or overactivation of CREB, indicating CREB as a potential cancer drug target. We previously identified 666-15 as a potent inhibitor of CREB with efficacious anti-cancer activity both in vitro and in vivo. Herein, we investigated the specificity of 666-15 and evaluated its potential in vivo toxicity. We found that 666-15 was fairly selective in inhibiting CREB. 666-15 was also found to be readily bioavailable to achieve pharmacologically relevant concentrations for CREB inhibition. Furthermore, the mice treated with 666-15 showed no evidence of changes in body weight, complete blood count, blood chemistry profile, cardiac contractility and tissue histologies from liver, kidney and heart. For the first time, these results demonstrate that pharmacological inhibition of CREB is well-tolerated in vivo and indicate that such inhibitors should be promising cancer therapeutics.


Assuntos
Anilidas , Antineoplásicos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/antagonistas & inibidores , Naftalenos , Proteínas de Neoplasias/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Anilidas/efeitos adversos , Anilidas/farmacocinética , Anilidas/farmacologia , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacocinética , Antineoplásicos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Células HEK293 , Humanos , Camundongos , Naftalenos/efeitos adversos , Naftalenos/farmacocinética , Naftalenos/farmacologia , Proteínas de Neoplasias/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia
9.
Ecotoxicol Environ Saf ; 131: 7-13, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27162129

RESUMO

Nonylphenol polyethoxylates (NPEOs) are a group of surfactants that are widely used in industrial and household products and often detected in the environment. The metabolite of NPEOs, named nonylphenol (NP), has proven to be an endocrine disruptor, and its environmental behavior and eco-toxicity have been widely investigated in previous studies. However, to the best of our knowledge, insight into the toxicity differences of NP and NPEOs on important crops remains limited. Therefore, this study investigated the comparative toxicity of NP, nonylphenol-4-ethoxylate (NP4EO), and nonylphenol-10-ethoxylate (NP10EO) on wheat seedlings using hydroponic experiments. The results indicated that NP is most toxic to wheat followed by NP4EO, and NP10EO is the least toxic to wheat. The adverse effects of NP on wheat were observed for all the tested parameters including germination, shoot length, root length, chlorophyll, lipid peroxidation, and enzymatic activities. To gain insight into the molecular response, we analyzed the transcript abundance of SOD-Cu/Zn and CAT with NP, NP4EO, and NP10EO exposure using quantitative real-time PCR. The data revealed that both genes exhibited up- or down-regulated expression patterns that were consistent with the activities of the two enzymes. This result further conformed that NP is most toxic to wheat plants.


Assuntos
Fenóis/toxicidade , Éteres Fenílicos/toxicidade , Polietilenoglicóis/toxicidade , Plântula/efeitos dos fármacos , Tensoativos/toxicidade , Triticum/efeitos dos fármacos , Catalase/genética , Catalase/metabolismo , Clorofila/metabolismo , Disruptores Endócrinos/toxicidade , Germinação/efeitos dos fármacos , Hidroponia , Peroxidação de Lipídeos/efeitos dos fármacos , Raízes de Plantas/efeitos dos fármacos , Brotos de Planta/efeitos dos fármacos , Plântula/crescimento & desenvolvimento , Plântula/metabolismo , Superóxido Dismutase/genética , Superóxido Dismutase/metabolismo , Triticum/crescimento & desenvolvimento , Triticum/metabolismo
10.
Oncotarget ; 6(42): 44675-87, 2015 Dec 29.
Artigo em Inglês | MEDLINE | ID: mdl-26625308

RESUMO

The intracytoplasmic tyrosine kinase Src serves both as a conduit and a regulator for multiple processes required for the proliferation and survival cancer cells. In some cancers, Src engages with receptor tyrosine kinases to mediate downstream signaling and in other cancers, it regulates gene expression. Src therefore represents a viable oncologic target. However, clinical responses to Src inhibitors, such as dasatinib have been disappointing to date. We identified Stat3 signaling as a potential bypass mechanism that enables renal cell carcinoma (RCC) cells to escape dasatinib treatment. Combined Src-Stat3 inhibition using dasatinib and CYT387 (a JAK/STAT inhibitor) synergistically reduced cell proliferation and increased apoptosis in RCC cells. Moreover, dasatinib and CYT387 combine to suppress YAP1, a transcriptional co-activator that promotes cell proliferation, survival and organ size. Importantly, this combination was well tolerated, and caused marked tumor inhibition in RCC xenografts. These results suggest that combination therapy with inhibitors of Stat3 signaling may be a useful therapeutic approach to increase the efficacy of Src inhibitors.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Benzamidas/farmacologia , Carcinoma de Células Renais/tratamento farmacológico , Dasatinibe/farmacologia , Neoplasias Renais/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Pirimidinas/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Quinases da Família src/antagonistas & inibidores , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Apoptose/efeitos dos fármacos , Carcinoma de Células Renais/enzimologia , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Regulação Neoplásica da Expressão Gênica , Neoplasias Renais/enzimologia , Neoplasias Renais/genética , Neoplasias Renais/patologia , Camundongos , Terapia de Alvo Molecular , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Fatores de Tempo , Fatores de Transcrição , Transcrição Gênica , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas de Sinalização YAP , Quinases da Família src/metabolismo
11.
Environ Sci Pollut Res Int ; 22(24): 19667-75, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26278900

RESUMO

The effects of imidacloprid in the soil environment are a worldwide concern. However, the impact of imidacloprid on soil microorganisms under salt stress is almost unknown. Therefore, an indoor incubation test was performed, and the denaturing gradient gel electrophoresis (DGGE) approach was used to determine the response of different saline soil bacterial and fungal community structures to the presence of imidacloprid (0.4, 2, 10 mg kg(-1)). The results showed that the soil bacterial diversity slightly declined with increasing imidacloprid concentration in soils with low salinity. In moderately saline soils, a new band in the DGGE profile suggested that imidacloprid could improve the soil bacterial diversity to some degree. An analysis of variance indicated that the measured soil bacterial diversity parameters were significantly affected by dose and incubation time. Compared with the control, the soil fungal community structure showed no obvious changes in low and moderately saline soils treated with imidacloprid. The results of these observations provide a basic understanding of the potential ecological effects of imidacloprid on different microorganisms in saline soils.


Assuntos
Imidazóis/farmacologia , Inseticidas/farmacologia , Nitrocompostos/farmacologia , Microbiologia do Solo , Bactérias/efeitos dos fármacos , Fungos/efeitos dos fármacos , Neonicotinoides , Salinidade , Solo/química , Poluentes do Solo/farmacologia
12.
J Med Chem ; 58(12): 5075-87, 2015 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-26023867

RESUMO

Recent studies have shown that nuclear transcription factor cyclic adenosine monophosphate response element binding protein (CREB) is overexpressed in many different types of cancers. Therefore, CREB has been pursued as a novel cancer therapeutic target. Naphthol AS-E and its closely related derivatives have been shown to inhibit CREB-mediated gene transcription and cancer cell growth. Previously, we identified naphthamide 3a as a different chemotype to inhibit CREB's transcription activity. In a continuing effort to discover more potent CREB inhibitors, a series of structural congeners of 3a was designed and synthesized. Biological evaluations of these compounds uncovered compound 3i (666-15) as a potent and selective inhibitor of CREB-mediated gene transcription (IC50 = 0.081 ± 0.04 µM). 666-15 also potently inhibited cancer cell growth without harming normal cells. In an in vivo MDA-MB-468 xenograft model, 666-15 completely suppressed the tumor growth without overt toxicity. These results further support the potential of CREB as a valuable cancer drug target.


Assuntos
Antineoplásicos/química , Antineoplásicos/uso terapêutico , Neoplasias da Mama/tratamento farmacológico , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/antagonistas & inibidores , Naftalenos/química , Naftalenos/uso terapêutico , Ativação Transcricional/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Mama/efeitos dos fármacos , Mama/metabolismo , Mama/patologia , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Células HEK293 , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Naftalenos/farmacologia
13.
Biochim Biophys Acta ; 1853(5): 881-91, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25637186

RESUMO

HIF-1α is degraded by oxygen-dependent mechanisms but stabilized in hypoxia to form transcriptional complex HIF-1, which transactivates genes promoting cancer hallmarks. However, how HIF-1α is specifically regulated in hypoxia is poorly understood. Here, we report that the histone methyltransferase SET9 promotes HIF-1α protein stability in hypoxia and enhances HIF-1 mediated glycolytic gene transcription, thereby playing an important role in mediating cancer cell adaptation and survival to hypoxic stress. Specifically, SET9 interacts with HIF-1α and promotes HIF-1α protein stability in hypoxia. Silencing SET9 by siRNA reduces HIF-1α protein stability in hypoxia, and attenuates the hypoxic induction of HIF-1 target genes mediating hypoxic glycolysis. Mechanistically, we find that SET9 is enriched at the hypoxia response elements (HRE) within promoters of the HIF-1-responsive glycolytic genes. Silencing SET9 reduces HIF-1α levels at these HREs in hypoxia, thereby attenuating HIF-1-mediated gene transcription. Further, silencing SET9 by siRNA reduces hypoxia-induced glycolysis and inhibits cell viability of hypoxic cancer cells. Our findings suggest that SET9 enriches at HRE sites of HIF-1 responsive glycolytic genes and stabilizes HIF-1α at these sites in hypoxia, thus establishes an epigenetic mechanism of the metabolic adaptation in hypoxic cancer cells.


Assuntos
Histona-Lisina N-Metiltransferase/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Lisina/metabolismo , Hipóxia Celular , Linhagem Celular Tumoral , Sobrevivência Celular , Cromatina/metabolismo , Citoproteção , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes , Glicólise/genética , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Regiões Promotoras Genéticas/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Ligação Proteica , Estabilidade Proteica , Proteólise , Elementos de Resposta/genética , Ativação Transcricional/genética
14.
Prostate ; 73(10): 1028-37, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23389923

RESUMO

BACKGROUND: Resistance to chemotherapy represents a significant obstacle in prostate cancer therapeutics. Novel mechanistic understandings in cancer cell chemotherapeutic sensitivity and resistance can optimize treatment and improve patient outcome. Molecular alterations in the metabolic pathways are associated with cancer development; however, the role of these alterations in chemotherapy efficacy is largely unknown. METHODS: In a bed-side to bench-side reverse translational approach, we used cDNA microarray and qRT-PCR to identify genes that are associated with biochemical relapse after chemotherapy. Further, we tested the function of these genes in cell proliferation, metabolism, and chemosensitivity in prostate cancer cell lines. RESULTS: We report that the gene encoding mitochondrial malate dehydrogenase 2 (MDH2) is overexpressed in clinical prostate cancer specimens. Patients with MDH2 overexpression had a significantly shorter period of relapse-free survival (RFS) after undergoing neoadjuvant chemotherapy. To understand the molecular mechanism underlying this clinical observation, we observed that MDH2 expression was elevated in prostate cancer cell lines compared to benign prostate epithelial cells. Stable knockdown of MDH2 via shRNA in prostate cancer cell lines decreased cell proliferation and increased docetaxel sensitivity. Further, MDH2 shRNA enhanced docetaxel-induced activations of JNK signaling and induced metabolic inefficiency. CONCLUSION: Taken together, these data suggest a novel function for MDH2 in prostate cancer development and chemotherapy resistance, in which MDH2 regulates chemotherapy-induced signal transduction and oxidative metabolism.


Assuntos
Antineoplásicos/uso terapêutico , Resistência a Medicamentos/genética , Metabolismo Energético/genética , Sistema de Sinalização das MAP Quinases/genética , Malato Desidrogenase/metabolismo , Neoplasias da Próstata/metabolismo , Taxoides/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células , Intervalo Livre de Doença , Docetaxel , Humanos , Malato Desidrogenase/genética , Masculino , Consumo de Oxigênio/genética , Neoplasias da Próstata/tratamento farmacológico , Neoplasias da Próstata/genética , Espécies Reativas de Oxigênio/metabolismo
15.
J Biol Chem ; 287(42): 35496-35505, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-22908229

RESUMO

Lysine acetylation regulates protein stability and function. p300 is a component of the HIF-1 transcriptional complex and positively regulates the transactivation of HIF-1. Here, we show a novel molecular mechanism by which p300 facilitates HIF-1 activity. p300 increases HIF-1α (HIF1α) protein acetylation and stability. The regulation can be opposed by HDAC1, but not by HDAC3, and is abrogated by disrupting HIF1α-p300 interaction. Mechanistically, p300 specifically acetylates HIF1α at Lys-709, which increases the protein stability and decreases polyubiquitination in both normoxia and hypoxia. Compared with the wild-type protein, a HIF1α K709A mutant protein is more stable, less polyubiquitinated, and less dependent on p300. Overexpression of the HIF1α wild-type or K709A mutant in cancer cells lacking the endogenous HIF1α shows that the K709A mutant is transcriptionally more active toward the HIF-1 reporter and some endogenous target genes. Cancer cells containing the K709A mutant are less sensitive to hypoxia-induced growth arrest than the cells containing the HIF1α wild-type. Taken together, these data demonstrate a novel biological consequence upon HIF1α-p300 interaction, in which HIF1α can be stabilized by p300 via Lys-709 acetylation.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo , Acetilação , Substituição de Aminoácidos , Pontos de Checagem do Ciclo Celular/fisiologia , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Células HEK293 , Histona Desacetilase 1/genética , Histona Desacetilase 1/metabolismo , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Lisina/genética , Lisina/metabolismo , Mutação de Sentido Incorreto , Estabilidade Proteica , Fatores de Transcrição de p300-CBP/genética
16.
J Comp Neurol ; 520(10): 2143-62, 2012 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-22173890

RESUMO

Kisspeptin is essential for reproductive functions in humans. As a model for the human we have used the female guinea pig, which has a long ovulatory cycle similar to that of primates. Initially, we cloned a guinea pig kisspeptin cDNA sequence and subsequently explored the distribution and 17ß-estradiol (E2) regulation of kisspeptin mRNA (Kiss1) and protein (kisspeptin) by using in situ hybridization, real-time PCR and immunocytochemistry. In ovariectomized females, Kiss1 neurons were scattered throughout the preoptic periventricular areas (PV), but the vast majority of Kiss1 neurons were localized in the arcuate nucleus (Arc). An E2 treatment that first inhibits (negative feedback) and then augments (positive feedback) serum luteinizing hormone (LH) increased Kiss1 mRNA density and number of cells expressing Kiss1 in the PV at both time points. Within the Arc, Kiss1 mRNA density was reduced at both time points. Quantitative real-time PCR confirmed the in situ hybridization results during positive feedback. E2 reduced the number of immunoreactive kisspeptin cells in the PV at both time points, perhaps an indication of increased release. Within the Arc, the kisspeptin immunoreactivity was decreased during negative feedback but increased during positive feedback. Therefore, it appears that in guinea pig both the PV and the Arc kisspeptin neurons act cooperatively to excite gonadotropin-releasing hormone (GnRH) neurons during positive feedback. We conclude that E2 regulation of negative and positive feedback may reflect a complex interaction of the kisspeptin circuitry, and both the PV and the Arc respond to hormone signals to encode excitation of GnRH neurons during the ovulatory cycle.


Assuntos
Estradiol/farmacologia , Estrogênios/farmacologia , Expressão Gênica/efeitos dos fármacos , Hipotálamo/efeitos dos fármacos , Kisspeptinas/metabolismo , Análise de Variância , Animais , Clonagem Molecular , Diencéfalo/efeitos dos fármacos , Diencéfalo/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Cobaias , Hipotálamo/anatomia & histologia , Hipotálamo/metabolismo , Kisspeptinas/genética , Ovariectomia , RNA Mensageiro/metabolismo
17.
Pharmacol Biochem Behav ; 100(3): 566-74, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22079349

RESUMO

Anaplastic Lymphoma Kinase (Alk) is a receptor tyrosine kinase expressed throughout the adult mammalian hippocampus. Recent studies in Drosophila and prior studies in Caenorhabditis elegans have implicated Alk signaling in learning and neurogenesis. We have studied the roles of Alk and the closely related receptor Leukocyte Tyrosine Kinase (Ltk) in learning, behavior and neurogenesis. In the hippocampus, both receptors are expressed throughout the dentate gyrus, CA1 and CA3. To assess the functional roles of Alk and Ltk in the mammalian brain, we analyzed phenotypes in Alk mutant, Ltk mutant and Alk/Ltk double-mutant mice compared to wild-type littermates. Similar to Drosophila, we found enhanced performance in spatial memory in Alk mutant mice. Also similar to Drosophila, we observed reduced neurogenesis associated with loss of Alk function. We also report genetic interactions between Alk and Ltk with respect to neurogenesis and behavioral measures such as activity, anxiety levels, and retention of spatial memory.


Assuntos
Hipocampo/enzimologia , Aprendizagem , Memória , Neurogênese , Neurônios/enzimologia , Receptores Proteína Tirosina Quinases/metabolismo , Quinase do Linfoma Anaplásico , Animais , Ansiedade/genética , Comportamento Animal , Regulação Enzimológica da Expressão Gênica , Hipocampo/citologia , Hipocampo/metabolismo , Masculino , Camundongos , Camundongos Knockout , Camundongos Mutantes , Atividade Motora , Mutação , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Neurônios/citologia , Neurônios/metabolismo , Especificidade de Órgãos , RNA Mensageiro/metabolismo , Receptores Proteína Tirosina Quinases/genética , Comportamento Espacial
18.
J Biol Chem ; 286(44): 38095-38102, 2011 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-21917920

RESUMO

Hypoxia-inducible factor 1 α (HIF1α) is an essential part of the HIF-1 transcriptional complex that regulates angiogenesis, cellular metabolism, and cancer development. In von Hippel-Lindau (VHL)-null kidney cancer cell lines, we reported previously that HIF1α proteins can be acetylated and inhibited by histone deacetylase (HDAC) inhibitors or specific siRNA against HDAC4. To investigate the mechanism and biological consequence of the inhibition, we have generated stable HDAC4 knockdown via shRNA in VHL-positive normal and cancer cell lines. We report that HDAC4 regulates HIF1α protein acetylation and stability. Specifically, the HIF1α protein acetylation can be increased by HDAC4 shRNA and decreased by HDAC4 overexpression. HDAC4 shRNA inhibits HIF1α protein stability. In contrast, HDAC1 or HDAC3 shRNA has no such inhibitory effect. Mutations of the first five lysine residues (lysine 10, 11, 12, 19, and 21) to arginine within the HIF1α N terminus reduce protein acetylation but render the mutant HIF1α protein resistant to HDAC4 and HDACi-mediated inhibition. Functionally, in VHL-positive cancer cell lines, stable inhibition of HDAC4 decreases both the HIF-1 transcriptional activity and a subset of HIF-1 hypoxia target gene expression. On the cellular level, HDAC4 inhibition reduces the hypoxia-related increase of glycolysis and resistance to docetaxel chemotherapy. Taken together, the novel biological relationship between HDAC4 and HIF1α presented here suggests a potential role for the deacetylase enzyme in regulating HIF-1 cancer cell response to hypoxia and presents a more specific molecular target of inhibition.


Assuntos
Regulação Neoplásica da Expressão Gênica , Histona Desacetilases/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia , Lisina/química , Mutação , Proteínas Repressoras/metabolismo , Acetilação , Linhagem Celular Tumoral , Cicloeximida/farmacologia , Genes Reporter , Glicólise , Células HEK293 , Humanos , Inibidores da Síntese de Proteínas/farmacologia , RNA Interferente Pequeno/metabolismo
19.
Mol Pharmacol ; 72(4): 885-96, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17622577

RESUMO

Hypothalamic proopiomelanocortin (POMC) neurons play a critical role in the regulation of energy balance, and there is a convergence of critical synaptic input including GABA and serotonin on POMC neurons to regulate their output. We found previously that 17beta-estradiol (E(2)) reduced the potency of the GABA(B) receptor agonist baclofen to activate G protein-coupled inwardly rectifying potassium (GIRK) channels in hypothalamic POMC neurons through a membrane estrogen receptor (mER) via a Galpha(q) phospholipase C (PLC)-protein kinase Cdelta-protein kinase A pathway. We hypothesized that the mER and neurotransmitter receptor signaling pathways converge to control energy homeostasis. Because 5-HT(2C) receptors mediate many of the effects of serotonin in POMC neurons, we elucidated the common signaling pathways of E(2) and 5-HT in guinea pigs using single-cell reverse transcription-polymerase chain reaction (RT-PCR), real time RT-PCR, and whole-cell patch recording. Both 5-hydroxytryptamine(2C) (5-HT(2C)) and 5-HT(2A) receptors were coexpressed in POMC neurons. The 5-HT(2A/C) agonist (+/-)-1-(2,5-dimethoxy-4-iodophenyl)-2-aminopropane (DOI) desensitized the GABA(B) response in a dose-dependent manner, which was antagonized by the selective 5-HT(2C) receptor antagonists 8-[5-(2,4-dimethoxy-5-(4-trifluoromethylphenylsulphonamido) phenyl-5-oxopentyl]1,3,8-triazaspiro[4.5] decane-2,4-dione hydrochloride (RS102221) and 1,2,3, 4,10,14b-hexahydro-2-methyldibenzo [c,f]pyrazino[1,2-a]-azepine hydrochloride (ORG 3363). The 5-HT(2C) receptor was Galpha(q)-coupled to PLC activation and hydrolysis of plasma membrane phosphatidylinositol bisphosphate to directly inhibit GIRK channel activity. Coapplication of the two agonists at their EC(50) concentrations (DOI, 20 muM, and E(2), 50 nM) produced additive effects. Although there was a significant gender difference in the effects of E(2) on baclofen responses, there was no gender difference in 5-HT(2C) receptor-mediated effects. Finally, both DOI and estrogen (intracerebroventricular) inhibited feeding in ovariectomized female mice. Therefore, the Galpha(q) signaling pathways of the mER and 5-HT(2C) receptors may converge to enhance synaptic efficacy in brain circuits that are critical for maintaining homeostatic functions.


Assuntos
Metabolismo Energético , Homeostase , Hipotálamo/metabolismo , Neurônios/metabolismo , Pró-Opiomelanocortina/metabolismo , Receptor 5-HT2C de Serotonina/metabolismo , Transdução de Sinais , Sequência de Aminoácidos , Animais , Sequência de Bases , Primers do DNA , Estradiol/farmacologia , Feminino , Cobaias , Hipotálamo/citologia , Hipotálamo/efeitos dos fármacos , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Técnicas de Patch-Clamp , Proteína Quinase C-delta/metabolismo , RNA Mensageiro/genética , Receptor 5-HT2C de Serotonina/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fosfolipases Tipo C/metabolismo
20.
J Neurosci ; 26(43): 11072-82, 2006 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-17065449

RESUMO

Low voltage-activated (T-type) Ca2+ channels are responsible for generating low-threshold spikes (LTS) that facilitate burst firing and transmitter release in neurons. The T-type Ca2+ channels contain a regulatory alpha1 subunit, and several isoforms of the alpha1 subunit (Cav3.1, 3.2, 3.3) have been cloned. The Cav 3.1 alpha1 subunit is abundantly expressed in the hypothalamus. Previously, we found that 17 beta-estradiol (E2) increased the number of arcuate neurons expressing LTS. Therefore, we used an ovariectomized female guinea pig model to measure the distribution and regulation of Cav3.1 mRNA expression by E2. Guinea pig Cav3.1 alpha1 subunit sequences, which were cloned by PCR, were used in ribonuclease protection (RPA) and in situ hybridization assays to evaluate mRNA expression. Based on a RPA, E2 significantly increased the mRNA expression of Cav3.1 alpha1 subunit in the mediobasal hypothalamus and the pituitary. In situ hybridization analysis revealed that E2 significantly increased Cav 3.1 mRNA expression in medial preoptic nuclei, bed nuclei stria terminalis, and the arcuate nucleus. Whole-cell patch recordings in arcuate neurons revealed that E2 treatment significantly increased the peak T-type Ca2+ current density by twofold without affecting the activation/inactivation characteristics and augmented the rebound excitation by threefold to fourfold. These results suggest that estrogen regulates the mRNA expression of T-type calcium channels, which leads to increased functional expression of the channel. Increased expression of T-type channels could be one mechanism by which estrogen augments burst firing and transmitter release in hypothalamic neurons.


Assuntos
Canais de Cálcio Tipo T/biossíntese , Estradiol/farmacologia , Hipotálamo/metabolismo , Hipófise/metabolismo , Regulação para Cima/fisiologia , Animais , Canais de Cálcio Tipo T/genética , Estradiol/metabolismo , Feminino , Cobaias , Hipotálamo/efeitos dos fármacos , Ovariectomia , Hipófise/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA