Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Oncol Lett ; 27(3): 123, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38348384

RESUMO

New therapeutic approaches are needed for osteosarcoma, which is the most common malignancy of the bone, especially for metastatic cases. Nintedanib is a potent, oral tyrosine kinase inhibitor approved for treating idiopathic pulmonary fibrosis, which blocks a variety of receptor signals, including fibroblast growth factor receptors, vascular endothelial growth factor receptors and platelet-derived growth factor receptors. The present study assessed the effect of nintedanib on previously developed mouse AXT osteosarcoma cells, and on AXT-derived osteosarcoma developed in C57BL/6 mice, which displays lethal tumors with osteoid formation and lung metastatic lesions that mimics human disease. In vitro analysis, including flow cytometry and immunoblotting, revealed that nintedanib inhibited AXT cell proliferation and cell cycle progression, induced apoptosis, and inactivated AKT and ERK1/2. Immunoblot analysis using tumor lysates demonstrated that nintedanib inhibited its target molecules in vivo. As a single agent, nintedanib decreased the size of primary AXT-derived osteosarcoma, and reduced circulating tumor cells and lung metastasis. Immunohistochemical findings indicated that nintedanib exerted antitumor activity mainly by inhibiting the formation of CD31-positive tumor vasculature, while αSMA-positive cells were still enriched in tumors after nintedanib treatment. In addition, nintedanib exhibited an anti-osteosarcoma effect on C57BL/6 severe combined immunodeficient mice in which T- and B-cell function is obsolete, suggesting that the antitumor effect of nintedanib was not attributable to antitumor immunity. Collectively, these findings indicated that nintedanib holds potential for treating osteosarcoma.

2.
Mol Neurobiol ; 2024 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-38308664

RESUMO

While patients with cancer show a higher prevalence of psychiatric disorders than the general population, the mechanism underlying this interaction remains unclear. The present study examined whether tumor-bearing (TB) mice show psychological changes using the conditioned fear paradigm and the role of cytokines in these changes. TB mice were established by transplantation with mouse osteosarcoma AXT cells. These TB mice were then found to exhibit disruption in extinction of conditioned fear memory. Eighteen cytokines in serum were increased in TB mice, among which i.c.v. injection of interleukin (IL)-1ß and IL-6 strengthened fear memory in normal mice. Contents of IL-17 and keratinocyte-derived cytokine (KC) in the amygdala and KC in the hippocampus were increased in TB mice. KC mRNA in both the amygdala and hippocampus was also increased in TB mice, and i.c.v. injection of KC dose-dependently strengthened fear memory in normal mice. In addition, injection of IL-1ß, but not IL-6, increased KC mRNA in the amygdala and hippocampus. In TB mice KC mRNA was increased in both astrocytes and microglia of the amygdala and hippocampus. The microglia inhibitor minocycline, but not the astrocyte inhibitor fluorocitrate, alleviated disruption in extinction of conditioned fear memory in TB mice. Microinjection of KC into the hippocampus, but not into the amygdala, increased fear memory in normal mice. These findings indicate that TB mice show an increase in serum cytokines, including IL-1ß, that increases KC production in microglia of the hippocampus, which then disrupts extinction of fear memory.

3.
Cells ; 11(22)2022 11 15.
Artigo em Inglês | MEDLINE | ID: mdl-36429043

RESUMO

Novel therapeutic targets are needed to better treat osteosarcoma, which is the most common bone malignancy. We previously developed mouse osteosarcoma cells, designated AX (accelerated bone formation) cells from bone marrow stromal cells. AX cells harbor both wild-type and mutant forms of p53 (R270C in the DNA-binding domain, which is equivalent to human R273C). In this study, we showed that mutant p53 did not suppress the transcriptional activation function of wild-type p53 in AX cells. Notably, AXT cells, which are cells derived from tumors originating from AX cells, lost wild-type p53 expression, were devoid of the intact transcription activation function, and were resistant to doxorubicin. ChIP-seq analyses revealed that this mutant form of p53 bound to chromatin in the vicinity of the transcription start sites of various genes but exhibited a different binding profile from wild-type p53. The knockout of mutant p53 in AX and AXT cells by CRISPR-Cas9 attenuated tumor growth but did not affect the invasion of these cells. In addition, depletion of mutant p53 did not prevent metastasis in vivo. Therefore, the therapeutic potency targeting R270C (equivalent to human R273C) mutant p53 is limited in osteosarcoma. However, considering the heterogeneous nature of osteosarcoma, it is important to further evaluate the biological and clinical significance of mutant p53 in various cases.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Camundongos , Animais , Humanos , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Linhagem Celular Tumoral , Osteossarcoma/metabolismo , Processos Neoplásicos , Neoplasias Ósseas/metabolismo
4.
J Orthop Res ; 39(12): 2732-2743, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-33751653

RESUMO

Osteosarcoma is the most common high-grade malignancy of bone, and novel therapeutic options are urgently required. Previously, we developed mouse osteosarcoma AXT cells that can proliferate both under adherent and nonadherent conditions. Based on metabolite levels, nonadherent conditions were more similar to the in vivo environment than adherent conditions. A drug screen identified MEK inhibitors, including trametinib, that preferentially decreased the viability of nonadherent AXT cells. Trametinib inhibited the cell cycle and induced apoptosis in AXT cells, and both effects were stronger under nonadherent conditions. Trametinib also potently decreased viability in U2OS cells, but its effects were less prominent in MG63 or Saos2 cells. By contrast, MG63 and Saos2 cells were more sensitive to PI3K inhibition than AXT or U2OS cells. Notably, the combination of MAPK/ERK kinase (MEK) and PI3K inhibition synergistically decreased viability in U2OS and AXT cells, but this effect was less pronounced in MG63 or Saos2 cells. Therefore, signal dependence for cell survival and crosstalk between MEK-ERK and PI3K-AKT pathways in osteosarcoma are cell context-dependent. The activation status of other kinases including CREB varied in a cell context-dependent manner, which might determine the response to MEK inhibition. A single dose of trametinib was sufficient to decrease the size of the primary tumor and circulating tumor cells in vivo. Moreover, combined administration of trametinib and rapamycin or conventional anticancer drugs further increased antitumor activity. Thus, given optimal biomarkers for predicting its effects, trametinib holds therapeutic potential for the treatment of osteosarcoma.


Assuntos
Neoplasias Ósseas , Osteossarcoma , Animais , Apoptose , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Camundongos , Quinases de Proteína Quinase Ativadas por Mitógeno/metabolismo , Quinases de Proteína Quinase Ativadas por Mitógeno/farmacologia , Osteossarcoma/tratamento farmacológico , Osteossarcoma/metabolismo , Fosfatidilinositol 3-Quinases
5.
Mol Pain ; 14: 1744806918756406, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29357732

RESUMO

Chronic pain induced by nerve damage due to trauma or invasion of cancer to the bone elicits severe ongoing pain as well as hyperalgesia and allodynia likely reflecting adaptive changes within central circuits that amplify nociceptive signals. The present study explored the possible contribution of the mesolimbic dopaminergic circuit in promoting allodynia related to neuropathic and cancer pain. Mice with ligation of the sciatic nerve or treated with intrafemoral osteosarcoma cells showed allodynia to a thermal stimulus applied to the paw on the injured side. Patch clamp electrophysiology revealed that the intrinsic neuronal excitability of ventral tegmental area (VTA) dopamine neurons projecting to the nucleus accumbens (N.Acc.) was significantly reduced in those mice. We used tyrosine hydroxylase (TH)-cre mice that were microinjected with adeno-associated virus (AAV) to express channelrhodopsin-2 (ChR2) to allow optogenetic stimulation of VTA dopaminergic neurons in the VTA or in their N.Acc. terminals. Optogenetic activation of these cells produced a significant but transient anti-allodynic effect in nerve injured or tumor-bearing mice without increasing response thresholds to thermal stimulation in sham-operated animals. Suppressed activity of mesolimbic dopaminergic neurons is likely to contribute to decreased inhibition of N.Acc. output neurons and to neuropathic or cancer pain-induced allodynia suggesting strategies for modulation of pathological pain states.


Assuntos
Neoplasias Ósseas/complicações , Neurônios Dopaminérgicos/patologia , Hiperalgesia/etiologia , Hiperalgesia/patologia , Nervo Isquiático/lesões , Área Tegmentar Ventral/patologia , Animais , Neoplasias Ósseas/fisiopatologia , Dor do Câncer/etiologia , Dor do Câncer/patologia , Dor do Câncer/fisiopatologia , Linhagem Celular Tumoral , Neurônios Dopaminérgicos/metabolismo , Hiperalgesia/fisiopatologia , Ligadura , Masculino , Camundongos Endogâmicos C57BL , Neuralgia/patologia , Núcleo Accumbens/patologia , Núcleo Accumbens/fisiopatologia , Nervo Isquiático/patologia , Nervo Isquiático/fisiopatologia , Área Tegmentar Ventral/fisiopatologia
6.
Cancer Sci ; 108(9): 1793-1802, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28643892

RESUMO

Osteosarcoma is the most common type of primary bone tumor, and novel therapeutic approaches for this disease are urgently required. To identify effective agents, we screened a panel of Food and Drug Administration (FDA)-approved drugs in AXT cells, our newly established mouse osteosarcoma line, and identified calcitriol as a candidate compound with therapeutic efficacy for this disease. Calcitriol inhibited cell proliferation in AXT cells by blocking cell cycle progression. From a mechanistic standpoint, calcitriol induced endoplasmic reticulum (ER) stress, which was potentially responsible for downregulation of cyclin D1, activation of p38 MAPK, and intracellular production of reactive oxygen species (ROS). Knockdown of Atf4 or Ddit3 restored cell viability after calcitriol treatment, indicating that the ER stress response was indeed responsible for the anti-proliferative effect in AXT cells. Notably, the ER stress response was induced to a lesser extent in human osteosarcoma than in AXT cells, consistent with the weaker suppressive effect on cell growth in the human cells. Thus, the magnitude of ER stress induced by calcitriol might be an index of its anti-osteosarcoma effect. Although mice treated with calcitriol exhibited weight loss and elevated serum calcium levels, a single dose was sufficient to decrease osteosarcoma tumor size in vivo. Our findings suggest that calcitriol holds therapeutic potential for treatment of osteosarcoma, assuming that techniques to diminish its toxicity could be established. In addition, our results show that calcitriol could still be safely administered to osteosarcoma patients for its original purposes, including treatment of osteoporosis.


Assuntos
Antineoplásicos/administração & dosagem , Neoplasias Ósseas/tratamento farmacológico , Calcitriol/administração & dosagem , Retículo Endoplasmático/metabolismo , Osteossarcoma/tratamento farmacológico , Animais , Neoplasias Ósseas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Retículo Endoplasmático/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Feminino , Humanos , Injeções Intraperitoneais , Camundongos Endogâmicos C57BL , Osteossarcoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Mol Cancer Ther ; 16(1): 182-192, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27799356

RESUMO

Osteosarcoma is the most common type of primary bone tumor, novel therapeutic agents for which are urgently needed. To identify such agents, we screened a panel of approved drugs with a mouse model of osteosarcoma. The screen identified simvastatin, which inhibited the proliferation and migration of osteosarcoma cells in vitro Simvastatin also induced apoptosis in osteosarcoma cells in a manner dependent on inhibition of the mevalonate biosynthetic pathway. It also disrupted the function of the small GTPase RhoA and induced activation of AMP-activated protein kinase (AMPK) and p38 MAPK, with AMPK functioning upstream of p38 MAPK. Inhibitors of AMPK or p38 MAPK attenuated the induction of apoptosis by simvastatin, whereas metformin enhanced this effect of simvastatin by further activation of AMPK. Although treatment with simvastatin alone did not inhibit osteosarcoma tumor growth in vivo, its combination with a fat-free diet induced a significant antitumor effect that was enhanced further by metformin administration. Our findings suggest that simvastatin induces apoptosis in osteosarcoma cells via activation of AMPK and p38 MAPK, and that, in combination with other approaches, it holds therapeutic potential for osteosarcoma. Mol Cancer Ther; 16(1); 182-92. ©2016 AACR.


Assuntos
Apoptose/efeitos dos fármacos , Neoplasias Ósseas/metabolismo , Osteossarcoma/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Neoplasias Ósseas/genética , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Humanos , Metformina/farmacologia , Camundongos , Osteossarcoma/genética , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteína rhoA de Ligação ao GTP/genética , Proteína rhoA de Ligação ao GTP/metabolismo
8.
Cancer Sci ; 106(7): 875-82, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25940371

RESUMO

Osteosarcoma (OS) is the most frequent primary solid malignant tumor of bone. Its prognosis remains poor in the substantial proportion of patients who do not respond to chemotherapy and novel therapeutic options are therefore needed. We previously established a mouse model that mimics the aggressive behavior of human OS. Enzyme-linked immunosorbent assay-based screening of such mouse tumor lysates identified platelet-derived growth factor-BB (PDGF-BB) as an abundant soluble factor, the gene for which was expressed dominantly in surrounding non-malignant cells of the tumor, whereas that for the cognate receptor (PDGF receptor ß) was highly expressed in OS cells. Platelet-derived growth factor-BB induced activation of both MEK-ERK and phosphatidylinositol 3-kinase-protein kinase B signaling pathways and promoted survival in OS cells deprived of serum, and these effects were blocked by the PDGF receptor inhibitor imatinib. However, these actions of PDGF-BB and imatinib were mostly masked in the presence of serum. Whereas imatinib alone did not manifest an antitumor effect in mice harboring OS tumors, combined treatment with imatinib and adriamycin exerted a synergistic antiproliferative effect on OS cells in vivo. These results suggest that treatment of OS with imatinib is effective only when cell survival is dependent on PDGF signaling or when imatinib is combined with another therapeutic intervention that renders the tumor cells susceptible to imatinib action, such as by inducing cellular stress.


Assuntos
Antineoplásicos/farmacologia , Benzamidas/farmacologia , Doxorrubicina/farmacologia , Piperazinas/farmacologia , Pirimidinas/farmacologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Becaplermina , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sinergismo Farmacológico , Feminino , Humanos , Mesilato de Imatinib , Camundongos Endogâmicos C57BL , Osteossarcoma , Proteínas Proto-Oncogênicas c-sis/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
9.
Cancer Res ; 74(22): 6531-41, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25273088

RESUMO

Osteosarcoma is a malignant bone tumor in children and adolescents characterized by intrinsic therapeutic resistance. The IGF2 is expressed at elevated levels in osteosarcoma after treatment with chemotherapy, prompting an examination of its functional contributions to resistance. We found that continuous exposure to IGF2 or insulin in the absence of serum created a dormant growth state in osteosarcoma cells that conferred resistance to various chemotherapeutic drugs in vitro. Mechanistic investigations revealed that this dormant state correlated with downregulation of downstream signaling by the IGF1 receptor, heightened cell survival, enhanced autophagy, and the presence of extracellular glutamine. Notably, inhibiting autophagy or depleting glutamine was sufficient to increase chemotherapeutic sensitivity in osteosarcoma xenografts in mice. Clinically, we confirmed that IGF expression levels were elevated in human osteosarcoma specimens from patients who received chemotherapy. Together, our results suggest that activation of IGF or insulin signaling preserves the survival of osteosarcoma cells under chemotherapeutic stress, providing a drug-resistant population that may engender minimal residual disease. Attenuating this survival mechanism may help overcome therapeutic resistance in osteosarcoma.


Assuntos
Autofagia/fisiologia , Neoplasias Ósseas/tratamento farmacológico , Fator de Crescimento Insulin-Like II/farmacologia , Osteossarcoma/tratamento farmacológico , Adolescente , Adulto , Animais , Neoplasias Ósseas/patologia , Linhagem Celular Tumoral , Sobrevivência Celular , Criança , Citoproteção , Resistencia a Medicamentos Antineoplásicos , Feminino , Glutamina/metabolismo , Humanos , Insulina/farmacologia , Fator de Crescimento Insulin-Like II/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Osteossarcoma/patologia
10.
PLoS One ; 7(11): e50621, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226335

RESUMO

Osteosarcoma is a high-grade malignant bone tumor that manifests ingravescent clinical behavior. The intrinsic events that confer malignant properties on osteosarcoma cells have remained unclear, however. We previously established two lines of mouse osteosarcoma cells: AX cells, which are able to form tumors in syngeneic mice, and AXT cells, which were derived from such tumors and acquired an increased tumorigenic capacity during tumor development. We have now identified Igf2 mRNA-binding protein3 (Imp3) as a key molecule responsible for this increased tumorigenicity of AXT cells in vivo. Imp3 is consistently up-regulated in tumors formed by AX cells, and its expression in these cells was found to confer malignant properties such as anchorage-independent growth, loss of contact inhibition, and escape from anoikis in vitro. The expression level of Imp3 also appeared directly related to tumorigenic ability in vivo which is the critical determination for tumor-initiating cells. The effect of Imp3 on tumorigenicity of osteosarcoma cells did not appear to be mediated through Igf2-dependent mechanism. Our results implicate Imp3 as a key regulator of stem-like tumorigenic characteristics in osteosarcoma cells and as a potential therapeutic target for this malignancy.


Assuntos
Osteossarcoma/patologia , Proteínas de Ligação a RNA/genética , Regulação para Cima , Animais , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Terapia de Alvo Molecular , Osteossarcoma/tratamento farmacológico , Fenótipo
11.
Leuk Res ; 35(2): 243-9, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20542334

RESUMO

Constitutive activation of NF-κB and STAT3 plays an important role in the cellular proliferation and survival of multiple myeloma cells. We first found that auranofin (AF), a coordinated gold compound, induced a significant level of cell cycle arrest at G1 phase and subsequent apoptosis of myeloma cells. Further, AF inhibited constitutive and IL-6-induced activation of JAK2 and phosphorylation of STAT3 followed by the decreased expression of Mcl-1. AF down-regulated the activation of NF-κB, and the combination of AF and a specific NF-κB inhibitor resulted in a marked decrease of Mcl-1 expression. These results suggest that AF inhibits both IL-6 induced-JAK/STAT pathway and NF-κB activation in myeloma cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Auranofina/farmacologia , Mieloma Múltiplo/metabolismo , Transdução de Sinais/efeitos dos fármacos , Western Blotting , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Expressão Gênica/efeitos dos fármacos , Humanos , Proteína de Sequência 1 de Leucemia de Células Mieloides , NF-kappa B/efeitos dos fármacos , NF-kappa B/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Fator de Transcrição STAT3/efeitos dos fármacos , Fator de Transcrição STAT3/metabolismo , Transfecção
12.
J Biol Chem ; 284(30): 20175-83, 2009 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-19447888

RESUMO

Micro opioid receptor (MOR) agonists such as morphine are applied widely in clinical practice as pain therapy. The effects of morphine through MOR, such as analgesia and development of tolerance and dependence, are influenced by individual specificity. Recently, we analyzed single nucleotide polymorphisms on the human MOR gene to investigate the factors that contribute to individual specificity. In process of single nucleotide polymorphisms analysis, we found that specific nuclear proteins bound to G(-172) --> T region in exon 1 in MOR gene, and its affinity to DNA was increased by base substitution from G(-172) to T(-172). The isolated protein was identified by mass spectrometry and was confirmed by Western blotting to be poly(ADP-ribose) polymerase-1 (PARP-1). The overexpressed PARP-1 bound to G(-172) --> T and enhanced the transcription of reporter vectors containing G(-172) and T(-172). Furthermore, PARP-1 inhibitor (benzamide) decreased PARP-1 binding to G(-172) --> T without affecting mRNA or protein expression level of PARP-1 and down-regulated the subsequent MOR gene expression in SH-SY5Y cells. Moreover, we found that tumor necrosis factor-alpha enhanced MOR gene expression as well as increased PARP-1 binding to the G(-172) --> T region and G(-172) --> T-dependent transcription in SH-SY5Y cells. These effects were also inhibited by benzamide. In this study, our data suggest that PARP-1 positively regulates MOR gene transcription via G(-172) --> T, which might influence individual specificity in therapeutic opioid effects.


Assuntos
Poli Adenosina Difosfato Ribose/análise , Poli Adenosina Difosfato Ribose/metabolismo , Polimorfismo de Nucleotídeo Único , Receptores Opioides mu/genética , Receptores Opioides mu/metabolismo , Fator de Necrose Tumoral alfa/metabolismo , Sequência de Bases , Benzamidas/farmacologia , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , DNA/genética , DNA/metabolismo , Éxons , Regulação da Expressão Gênica , Humanos , Rim/citologia , Neuroblastoma/genética , Neuroblastoma/metabolismo , Poli Adenosina Difosfato Ribose/antagonistas & inibidores , Poli Adenosina Difosfato Ribose/genética , Regiões Promotoras Genéticas , Ligação Proteica , RNA Mensageiro/genética , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Ativação Transcricional
13.
Biol Pharm Bull ; 32(4): 721-3, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19336912

RESUMO

micro-Opioid receptor agonists, such as morphine, are widely applied in pain therapy clinical practice. However, the effects exerted by morphine via receptor are influenced by individual specificity. Single nucleotide polymorphisms (SNPs) in micro-opioid receptor gene (OPRM1) have been reported to influence receptor expression and function. Subsequently, we analyzed SNPs frequency and linkage disequilibrium associated with OPRM1 transcriptional region and 4 exons among healthy Japanese individuals. Consequently, we detected 10 SNPs (-1748G/A, -1565T/C, -1045A/G, -172G/T, -38C/A, 118A/G, ivs2+31 G/A, ivs2+691 C/G, ivs4+274 A/G, and ivs4+435 G/A). Moreover, linkage analysis revealed novel linkage between -1748G/A and -172G/T, which was not observed in studies performed in other nations. In contrast, SNPs frequency detected in this study was similar to previously reported results on Asians; however, linkage disequilibrium reports from different nations differed. These results possibly provide useful information for OPRM1 genotyping in the Japanese population.


Assuntos
Receptores Opioides mu/genética , Primers do DNA , Genótipo , Haplótipos , Humanos , Japão/epidemiologia , Desequilíbrio de Ligação , Polimorfismo de Nucleotídeo Único , Sequências Reguladoras de Ácido Nucleico/genética , Transcrição Gênica/genética
15.
Mol Cancer Ther ; 6(3): 987-94, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17363492

RESUMO

Emodin is an active component of a traditional Chinese and Japanese medicine isolated from the root and rhizomes of Rheum palmatum L. Here, we show that emodin significantly induces cytotoxicity in the human myeloma cells through the elimination of myeloid cell leukemia 1 (Mcl-1). Emodin inhibited interleukin-6-induced activation of Janus-activated kinase 2 (JAK2) and phosphorylation of signal transducer and activator of transcription 3 (STAT3), followed by the decreased expression of Mcl-1. Activation of caspase-3 and caspase-9 was triggered by emodin, but the expression of other antiapoptotic Bcl-2 family members, except Mcl-1, did not change in the presence of emodin. To clarify the importance of Mcl-1 in emodin-induced apoptosis, the Mcl-1 expression vector was introduced into the human myeloma cells by electroporation. Induction of apoptosis by emodin was almost abrogated in Mcl-1-overexpressing myeloma cells as the same level as in parental cells, which were not treated with emodin. In conclusion, emodin inhibits interleukin-6-induced JAK2/STAT3 pathway selectively and induces apoptosis in myeloma cells via down-regulation of Mcl-1, which is a good target for treating myeloma. Taken together, our results show emodin as a new potent anticancer agent for the treatment of multiple myeloma patients.


Assuntos
Apoptose/efeitos dos fármacos , Emodina/farmacologia , Inibidores Enzimáticos/farmacologia , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Janus Quinase 2/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Proliferação de Células/efeitos dos fármacos , Humanos , Interleucina-6/farmacologia , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Mieloma Múltiplo/metabolismo , Mieloma Múltiplo/patologia , Proteína de Sequência 1 de Leucemia de Células Mieloides , Proteínas de Neoplasias/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas/efeitos dos fármacos
16.
Endocrinology ; 148(5): 2335-44, 2007 May.
Artigo em Inglês | MEDLINE | ID: mdl-17255207

RESUMO

In Graves' disease, the overstimulation of the thyroid gland and hyperthyroidism are caused by autoantibodies directed against the TSH receptor (TSHR) that mimics the action of TSH. The establishment of an animal model is an important step to study the pathophysiology of autoimmune hyperthyroidism and for immunological analysis. In this study, we adopted the technique of electroporation (EP) for genetic immunization to achieve considerable enhancement of in vivo human TSHR (hTSHR) expression and efficient induction of hyperthyroidism in mice. In a preliminary study using beta-galactosidase (beta-gal) expression vectors, beta-gal introduced into the muscle by EP showed over 40-fold higher enzymatic activity than that introduced via previous direct gene transfer methods. The sustained hTSHR mRNA expression derived from cDNA transferred by EP was detectable in muscle tissue for at least 2 wk by RT-PCR. Based on these results, we induced hyperthyroidism via two expression vectors inserted with hTSHR or hTSHR289His cDNA. Consequently, 12.0-31.8% BALB/c mice immunized with hTSHR and 79.2-95.7% immunized with hTSHR289His showed high total T(4) levels due to the TSHR-stimulating antibody after three to four times repeated immunization by EP, and thyroid follicles of which were hyperplastic and had highly irregular epithelium. Moreover, TSHR-stimulating antibody surprisingly persisted more than 8 months after the last immunization. These results demonstrate that genetic immunization by in vivo EP is more efficient than previous procedures, and that it is useful for delineating the pathophysiology of Graves' disease.


Assuntos
Modelos Animais de Doenças , Eletroporação/métodos , Doença de Graves/imunologia , Hipertireoidismo/imunologia , Camundongos Endogâmicos BALB C , Animais , Autoanticorpos/sangue , Autoanticorpos/imunologia , Células CHO , Linhagem Celular , Doença Crônica , Cricetinae , Cricetulus , Ensaio de Imunoadsorção Enzimática , Epitopos/imunologia , Feminino , Doença de Graves/patologia , Doença de Graves/fisiopatologia , Hipertireoidismo/patologia , Hipertireoidismo/fisiopatologia , Imunização/métodos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Receptores da Tireotropina/genética , Receptores da Tireotropina/imunologia , Glândula Tireoide/imunologia , Glândula Tireoide/patologia , Tiroxina/sangue
17.
J Biol Chem ; 277(35): 31871-6, 2002 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-12091384

RESUMO

Protein kinase C-associated kinase (PKK) is a recently described kinase of unknown function that was identified on the basis of its specific interaction with PKC beta. PKK contains N-terminal kinase and C-terminal ankyrin repeats domains linked to an intermediate region. Here we report that the kinase domain of PKK is highly homologous to that of two mediators of nuclear factor-kappa B (NF-kappa B) activation, RICK and RIP, but these related kinases have different C-terminal domains for binding to upstream factors. We find that expression of PKK, like RICK and RIP, induces NF-kappa B activation. Mutational analysis revealed that the kinase domain of PKK is essential for NF-kappa B activation, whereas replacement of serine residues in the putative activation loop did not affect the ability of PKK to activate NF-kappa B. A catalytic inactive PKK mutant inhibited NF-kappa B activation induced by phorbol ester and Ca(2+)-ionophore, but it did not block that mediated by tumor necrosis factor alpha, interleukin-1 beta, or Nod1. Inhibition of NF-kappa B activation by dominant negative PKK was reverted by co-expression of PKC beta I, suggesting a functional association between PKK and PKC beta I. PKK-mediated NF-kappa B activation required IKK alpha and IKK beta but not IKK gamma, the regulatory subunit of the IKK complex. Moreover, NF-kappa B activation induced by PKK was not inhibited by dominant negative Bimp1 and proceeded in the absence of Bcl10, two components of a recently described PKC signaling pathway. These results suggest that PKK is a member of the RICK/RIP family of kinases, which is involved in a PKC-activated NF-kappa B signaling pathway that is independent of Bcl10 and IKK gamma.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal , NF-kappa B/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Quinases/metabolismo , Acetato de Tetradecanoilforbol/farmacologia , Animais , Proteína 10 de Linfoma CCL de Células B , Sítios de Ligação , Calcimicina/farmacologia , Linhagem Celular , Fibroblastos/enzimologia , Deleção de Genes , Humanos , Camundongos , Plasmídeos , Proteínas Quinases/genética , Proteínas Serina-Treonina Quinases , Fator de Transcrição AP-1/genética , Fator de Transcrição AP-1/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA