Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Oxid Med Cell Longev ; 2022: 5397733, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35047106

RESUMO

The infection of coronavirus disease (COVID-19) seriously threatens human life. It is urgent to generate effective and safe specific antibodies (Abs) against the pathogenic elements of COVID-19. Mice were immunized with SARS-CoV-2 spike protein antigens: S ectodomain-1 (CoV, in short) mixed in Alum adjuvant for 2 times and boosted with CoV weekly for 6 times. A portion of mice were treated with Maotai liquor (MTL, in short) or/and heat stress (HS) together with CoV boosting. We observed that the anti-CoV Ab was successfully induced in mice that received the CoV/Alum immunization for 2 times. However, upon boosting with CoV, the CoV Ab production diminished progressively; spleen CoV Ab-producing plasma cell counts reduced, in which substantial CoV-specific Ab-producing plasma cells (sPC) were apoptotic. Apparent oxidative stress signs were observed in sPCs; the results were reproduced by exposing sPCs to CoV in the culture. The presence of MTL or/and HS prevented the CoV-induced oxidative stress in sPCs and promoted and stabilized the CoV Ab production in mice in re-exposure to CoV. In summary, CoV/Alum immunization can successfully induce CoV Ab production in mice that declines upon reexposure to CoV. Concurrent administration of MTL/HS stabilizes and promotes the CoV Ab production in mice.


Assuntos
Anticorpos Neutralizantes/biossíntese , Anticorpos Antivirais/biossíntese , Apoptose , COVID-19/imunologia , Plasmócitos/imunologia , SARS-CoV-2/fisiologia , Superóxido Dismutase-1/fisiologia , Adjuvantes Imunológicos , Bebidas Alcoólicas , Compostos de Alúmen , Enzima de Conversão de Angiotensina 2/fisiologia , Animais , Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , COVID-19/enzimologia , Vacinas contra COVID-19/imunologia , Resposta ao Choque Térmico , Imunização Secundária , Imunogenicidade da Vacina , Janus Quinase 2/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Estresse Oxidativo , Plasmócitos/efeitos dos fármacos , Plasmócitos/patologia , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT1/fisiologia , Transdução de Sinais , Organismos Livres de Patógenos Específicos , Glicoproteína da Espícula de Coronavírus/imunologia , Vacinação
2.
J BUON ; 26(4): 1635-1641, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34565029

RESUMO

PURPOSE: The purpose of this study was to analyze the function of curcumin to suppress the proliferative and invasive abilities of papillary thyroid carcinoma (PTC) through inhibiting the JAK2/STAT3 pathway. METHODS: After treatment of different doses of curcumin in TPC-1 and SW1736 cells, changes in viability, clonality, cell cycle, apoptosis, wound healing and invasion were determined. Western blot analyses were performed to detect protein levels of apoptosis-associated genes, JAK2 and STAT3 in TPC-1 and SW1736 cells treated with different doses of curcumin. RESULTS: Curcumin treatment dose-dependently reduced viability, clonality and metastatic ability in TPC-1 and SW1736 cells. After treatment of 10 µM or 20 µM curcumin, PTC cells were blocked in G2/M phase, and their apoptotic rate increased. Curcumin treatment downregulated Bcl-2 and upregulated Bax in PTC cells. In addition, curcumin treatment downregulated p-JAK2 and p-STAT3 in TPC-1 and SW1736 cells. CONCLUSIONS: Curcumin treatment blocks PTC cells to proliferate and invade via inhibiting the JAK2/STAT3 pathway.


Assuntos
Curcumina/farmacologia , Curcumina/uso terapêutico , Janus Quinase 2/efeitos dos fármacos , Janus Quinase 2/fisiologia , Fator de Transcrição STAT3/efeitos dos fármacos , Fator de Transcrição STAT3/fisiologia , Câncer Papilífero da Tireoide/tratamento farmacológico , Câncer Papilífero da Tireoide/patologia , Neoplasias da Glândula Tireoide/tratamento farmacológico , Neoplasias da Glândula Tireoide/patologia , Proliferação de Células/efeitos dos fármacos , Humanos , Invasividade Neoplásica , Células Tumorais Cultivadas
3.
Exp Hematol ; 91: 10-21, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32910996

RESUMO

Erythropoiesis is one of the most demanding processes in the body, with more than 2 million red blood cells produced every second. Multiple hereditary and acquired red blood cell disorders arise from this complex system, with existing treatments effective in managing some of these conditions but few offering a long-term cure. Finding new treatments relies on the full understanding of the cellular and molecular interactions associated with the production and maturation of red blood cells, which take place within the erythroblastic island niche. The elucidation of processes associated within the erythroblastic island niche in health and during stress erythropoiesis has relied on in vivo modeling in mice, with complexities dissected using simple in vitro systems. Recent progress using state-of-the-art stem cell technology and gene editing has enabled a more detailed study of the human niche. Here, we review these different models and describe how they have been used to identify and characterize the cellular and molecular pathways associated with red blood cell production and maturation. We speculate that these systems could be applied to modeling red blood cell diseases and finding new druggable targets, which would prove especially useful for patients resistant to existing treatments. These models could also aid in research into the manufacture of red blood cells in vitro to replace donor blood transfusions, which is the most common treatment of blood disorders.


Assuntos
Modelos Animais de Doenças , Eritroblastos/citologia , Eritropoese/fisiologia , Modelos Biológicos , Nicho de Células-Tronco/fisiologia , Estresse Fisiológico/fisiologia , Animais , Moléculas de Adesão Celular/deficiência , Comunicação Celular , Células Cultivadas , Técnicas de Cocultura , Avaliação Pré-Clínica de Medicamentos , Eritropoese/efeitos dos fármacos , Eritropoese/genética , Hematínicos/uso terapêutico , Doenças Hematológicas/tratamento farmacológico , Doenças Hematológicas/fisiopatologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Células-Tronco Pluripotentes Induzidas/efeitos dos fármacos , Janus Quinase 2/genética , Janus Quinase 2/fisiologia , Macrófagos/classificação , Macrófagos/fisiologia , Camundongos , Camundongos Transgênicos , Nicho de Células-Tronco/efeitos dos fármacos , Estresse Fisiológico/genética
4.
Int Immunopharmacol ; 64: 24-32, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30145467

RESUMO

Despite remarkable advances in multiple myeloma (MM) therapy, this condition remains incurable. BF211 is an active compound derived from bufalin, which is isolated from the Traditional Chinese Medicine, Chansu. In this study, we explored the cytotoxicity of BF211 in 20 tumor cell lines and discovered that the MM cell lines, ARP-1 and CAG, exhibited greater sensitivity to BF211. Compared with bufalin, BF211 induced a greater apoptotic effect and lower acute toxicity at nanomolar concentration. The IL-6/JAK2/STAT3 signaling pathway is essential to the progression and development of MM. We showed that exogenous IL-6 promoted MM cell proliferation in a dose-dependent manner and this effect was blocked by BF211. Furthermore, BF211 suppressed the phosphorylation of JAK2 and STAT3 both in vivo and in vitro. In a mouse MM xenograft model, BF211 significantly inhibited tumor growth and did not affect body weight. In conclusion, the anti-MM activity of BF211 is mediated mainly by suppressing the IL-6/JAK2/STAT3 signaling pathway. Thus, we suggest that BF211 warrants further investigation in clinical trials in MM.


Assuntos
Antineoplásicos/farmacologia , Bufanolídeos/farmacologia , Interleucina-6/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Mieloma Múltiplo/tratamento farmacológico , Piperazinas/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Humanos , Interleucina-6/fisiologia , Janus Quinase 2/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Mieloma Múltiplo/patologia , Fator de Transcrição STAT3/fisiologia
5.
Planta Med ; 84(1): 20-25, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-28651290

RESUMO

The anti-inflammatory effects of boldine and reticuline isolated from Litsea cubeba were evaluated by using xylene-induced ear edema and carrageenan-induced paw edema in mice and rats. Our results demonstrated that intragastric administration with boldine and reticuline significantly mitigated ear weight in mice and decreased paw volume in rats. A combination administration of boldine (0.5 mg/kg) + reticuline (0.25 mg/kg) resulted in a potentiated inhibition in these two models. In parallel, boldine or reticuline reduce the infiltration of neutrophil leukocytes in rat paw tissue, respectively, and the combination of the two groups performed a better anti-inflammatory activity as shown in histopathologies. Boldine, reticuline, and their combination notably inhibited mRNA expressions of TNF-α, and IL-6 and reduced the phosphorylation levels of Janus kinase 2 (JAK2) and signal transducer and activator of transcription 3 (STAT3). Beyond that, their combination also can reduce the phosphorylation levels of p65 and IκBα in the pathological tissues of animals, as observed by real-time PCR and western blot analyses, respectively. These findings indicate for the first time that boldine and reticuline have not only anti-inflammatory activity but also potential synergistic effects in vivo. The underlying mechanism may relate to the inhibition on the expression of pro-inflammatory cytokines, such as TNF-α and IL-6, which may be a consequence of JAK2/STAT3 and NF-κB pathway involvements. This study provides useful data for further exploration and application of boldine and reticuline as potential anti-inflammatory medicines.


Assuntos
Anti-Inflamatórios/farmacologia , Aporfinas/farmacologia , Janus Quinase 2/antagonistas & inibidores , Litsea , NF-kappa B/antagonistas & inibidores , Extratos Vegetais/farmacologia , Fator de Transcrição STAT3/antagonistas & inibidores , Transdução de Sinais/efeitos dos fármacos , Animais , Benzilisoquinolinas/farmacologia , Western Blotting , Edema/tratamento farmacológico , Interleucina-6/metabolismo , Janus Quinase 2/fisiologia , Litsea/química , Masculino , Camundongos , Camundongos Endogâmicos ICR , NF-kappa B/fisiologia , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição STAT3/fisiologia , Fator de Necrose Tumoral alfa/metabolismo
6.
Zhongguo Zhong Yao Za Zhi ; 40(15): 3081-7, 2015 Aug.
Artigo em Chinês | MEDLINE | ID: mdl-26677715

RESUMO

To study the preventive effect of sophocarpine (Soc) on dextran sulfate sodium (DSS)-induced colitis in mice, in order to analyze the influence of Soc on toll like receptor 4 (TLR4)/mitogen-activated protein kinases (MAPKs) and janus tyrosine kinase 2 signal transducer and activator of transcription 3 (JAK2/STAT3) signal pathways in mice intestinal tissues. The mice was given 2.5% DSS for 6 days to induce the acute colitis model. The Soc-treated group was intraperitoneally injected with sophocarpine 30 mg · kg(-1) · d(-1) since the day before the experiment to the end. The disease activity index (DAI) was assessed everyday, and the colonic morphology and histological damage were observed with HE staining. The mRNA expressions of tumor necrosis factor-α (TNF-α), interleukin-1ß (IL-1ß) and interleukin-6 (IL-6) were detected by real-time RT-PCR. The changes in key protein kinase p38 mitogen-activated protein kinase (p38MAPK), c-Jun NH2-terminal protein kinase1/2 (JNK1/2), extracellular signal-regulated kinase1/2 (ERK1/2), JAK2, STAT3 in TLR4/MAPKs and JAK2/STAT3 signaling pathways were detected by western blot. The result showed that the model group showed statistical significance in body weight, DAI, colon length and histopathological changes compared with the normal group (P <0.05); however, the Soc-treated group showed significant improvements in the above indexes compared with the model group (P <0.05). TNF-α, IL-1ß and IL-6 in the model group was significantly higher than that in the normal group (P <0.05), but lowered in the Soc-treated group to varying degrees (P <0.05). In the normal group, the expressions of TLR4 and the phosphorylation of P38, JNK1/2, JAK2, STAT3 were at low levels; in the model group, the phosphorylation of P38, JNK1/2, JAK2, STAT3 increased; the Soc-treated group showed a decrease in TLR4 expression compared with the model group, with notable declines in the phosphorylation of TLR4, P38, JNK1/2, JAK2, STAT3. These findings indicate that Soc can inhibit TLR4/MAPKs, K2/STAT3 signaling pathway activation, reduce the expression of proinflammatory cytokines TNF-α, IL-1ß and IL-6 and relieve inflammatory reactions, so as to effectively prevent experimental colitis.


Assuntos
Alcaloides/uso terapêutico , Colite/tratamento farmacológico , Alcaloides/farmacologia , Animais , Colite/imunologia , Colite/patologia , Citocinas/genética , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Fosforilação , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/fisiologia , Receptor 4 Toll-Like/antagonistas & inibidores , Receptor 4 Toll-Like/fisiologia
7.
Br J Haematol ; 160(2): 177-87, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23157224

RESUMO

Aberrant JAK2 signalling plays an important role in the aetiology of myeloproliferative neoplasms (MPNs). JAK2 inhibitors, however, do not readily eliminate neoplastic MPN cells and thus do not induce patient remission. Further understanding JAK2 signalling in MPNs may uncover novel avenues for therapeutic intervention. Recent work has suggested a potential role for cellular cholesterol in the activation of JAK2 by the erythropoietin receptor and in the development of an MPN-like disorder in mice. Our study demonstrates for the first time that the MPN-associated JAK2-V617F kinase localizes to lipid rafts and that JAK2-V617F-dependent signalling is inhibited by lipid raft disrupting agents, which target membrane cholesterol, a critical component of rafts. We also show for the first time that statins, 3-hydroxy-3-methyl-glutaryl coenzyme A (HMG-CoA) reductase inhibitors, widely used to treat hypercholesterolaemia, induce apoptosis and inhibit JAK2-V617F-dependent cell growth. These cells are more sensitive to statin treatment than non-JAK2-V617F-dependent cells. Importantly, statin treatment inhibited erythropoietin-independent erythroid colony formation of primary cells from MPN patients, but had no effect on erythroid colony formation from healthy individuals. Our study is the first to demonstrate that JAK2-V617F signalling is dependent on lipid rafts and that statins may be effective in a potential therapeutic approach for MPNs.


Assuntos
Janus Quinase 2/fisiologia , Microdomínios da Membrana/fisiologia , Mutação de Sentido Incorreto , Transtornos Mieloproliferativos/enzimologia , Mutação Puntual , Transdução de Sinais/efeitos dos fármacos , Sinvastatina/farmacologia , beta-Ciclodextrinas/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/enzimologia , Células Cultivadas/efeitos dos fármacos , Células Cultivadas/enzimologia , Colesterol/análise , Colesterol/fisiologia , Ensaio de Unidades Formadoras de Colônias , Avaliação Pré-Clínica de Medicamentos , Células Precursoras Eritroides/efeitos dos fármacos , Células Precursoras Eritroides/enzimologia , Humanos , Janus Quinase 2/genética , Células K562/efeitos dos fármacos , Células K562/enzimologia , Leucemia Eritroblástica Aguda/enzimologia , Leucemia Eritroblástica Aguda/patologia , Leucemia Megacarioblástica Aguda/enzimologia , Leucemia Megacarioblástica Aguda/patologia , Células Progenitoras de Megacariócitos/efeitos dos fármacos , Células Progenitoras de Megacariócitos/enzimologia , Lipídeos de Membrana/fisiologia , Microdomínios da Membrana/efeitos dos fármacos , Transtornos Mieloproliferativos/sangue , Fosforilação/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Fator de Transcrição STAT5/metabolismo
8.
Blood ; 119(20): 4625-35, 2012 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-22378845

RESUMO

The constitutively active JAK2 V617F mutant is the major determinant of human myeloproliferative neoplasms (MPNs). We show that coexpression of murine JAK2 V617F and the murine thrombopoietin (Tpo) receptor (TpoR, c-MPL) in hematopoietic cell lines or heterozygous knock-in of JAK2 V617F in mice leads to down-modulation of TpoR levels. Enhanced TpoR ubiquitinylation, proteasomal degradation, reduced recycling, and maturation are induced by the constitutive JAK2 V617F activity. These effects can be prevented in cell lines by JAK2 and proteasome inhibitors. Restoration of TpoR levels by inhibitors could be detected in platelets from JAK2 inhibitor-treated myelofibrosis patients that express the JAK2 V617F mutant, and in platelets from JAK2 V617F knock-in mice that were treated in vivo with JAK2 or proteasome inhibitors. In addition, we show that Tpo can induce both proliferative and antiproliferative effects via TpoR at low and high JAK2 activation levels, respectively, or on expression of JAK2 V617F. The antiproliferative signaling and receptor down-modulation by JAK2 V617F were dependent on signaling via TpoR cytosolic tyrosine 626. We propose that selection against TpoR antiproliferative signaling occurs by TpoR down-modulation and that restoration of down-modulated TpoR levels could become a biomarker for the treatment of MPNs.


Assuntos
Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/fisiologia , Inibidores de Proteassoma , Inibidores de Proteínas Quinases/farmacologia , Receptores de Trombopoetina/genética , Substituição de Aminoácidos/genética , Substituição de Aminoácidos/fisiologia , Animais , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Células Cultivadas , Regulação para Baixo/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Mutação de Sentido Incorreto/fisiologia , Fenilalanina/genética , Receptor Cross-Talk/efeitos dos fármacos , Receptor Cross-Talk/fisiologia , Receptores de Trombopoetina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Valina/genética
9.
Reprod Fertil Dev ; 24(3): 482-9, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22401280

RESUMO

Endometrial cysteine-rich protein 61 (CYR61, CCN1) is a growth factor-inducible gene whose expression is elevated during the proliferative phase of the menstrual cycle and which has been implicated in the pathogenesis of endometriosis. This study aimed to define the mediators of epidermal growth factor (EGF) signalling on CYR61 expression in spontaneously immortalised human endometrial epithelial cells (HES) as a model system. After 30 min of EGF treatment, the receptor was phosphorylated and internalised as well as mRNA CYR61 increased in HES cells. However, neither inhibition of C-terminal EGF receptor (EGFR)-phosphorylation nor blockage of the mitogen-activated proteinkinase/extracellular signal-regulated kinase (MAPK/ERK) pathway was able to reduce CYR61 levels. Surprisingly, the HES cells showed upregulation of CYR61 mRNA expression after inhibition of the MAPK/ERK pathway when treated with EGF. Specific inhibitor studies identified the contribution of Janus kinase 2 (JAK2) and the signal transducer and activator of transcription protein STAT3 to the regulation of CYR61 expression. The JAK2/STAT3 interaction contributed to the basal expression of CYR61 and mediated EGF-driven regulation of CYR61 after 30 and 120 min of treatment. In summary, EGF-mediated CYR61 upregulation in HES cells involves STAT3 and is counter-regulated by the EGFR/MAPK/ERK pathway.


Assuntos
Proteína Rica em Cisteína 61/genética , Endométrio/efeitos dos fármacos , Fator de Crescimento Epidérmico/farmacologia , Janus Quinase 2/metabolismo , Fator de Transcrição STAT3/metabolismo , Antineoplásicos/farmacologia , Células Cultivadas , Proteína Rica em Cisteína 61/metabolismo , Avaliação Pré-Clínica de Medicamentos , Endométrio/metabolismo , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/fisiologia , Fator de Crescimento Epidérmico/fisiologia , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Receptores ErbB/fisiologia , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Janus Quinase 2/fisiologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Quinazolinas/farmacologia , Fator de Transcrição STAT3/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Tirfostinas/farmacologia , Regulação para Cima/efeitos dos fármacos
10.
J Med Chem ; 55(6): 2623-40, 2012 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-22339472

RESUMO

Herein, we describe the synthesis and SAR of a series of small molecule macrocycles that selectively inhibit JAK2 kinase within the JAK family and FLT3 kinase. Following a multiparameter optimization of a key aryl ring of the previously described SB1518 (pacritinib), the highly soluble 14l was selected as the optimal compound. Oral efficacy in the murine collagen-induced arthritis (CIA) model for rheumatoid arthritis (RA) supported 14l as a potential treatment for autoimmune diseases and inflammatory disorders such as psoriasis and RA. Compound 14l (SB1578) was progressed into development and is currently undergoing phase 1 clinical trials in healthy volunteers.


Assuntos
Antirreumáticos/síntese química , Artrite Reumatoide/tratamento farmacológico , Compostos Heterocíclicos de 4 ou mais Anéis/síntese química , Janus Quinase 2/antagonistas & inibidores , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Animais , Antirreumáticos/farmacocinética , Antirreumáticos/farmacologia , Artrite Experimental/induzido quimicamente , Artrite Experimental/tratamento farmacológico , Linhagem Celular , Permeabilidade da Membrana Celular , Colágeno Tipo II , Cães , Feminino , Compostos Heterocíclicos de 4 ou mais Anéis/farmacocinética , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Humanos , Janus Quinase 2/fisiologia , Macaca mulatta , Masculino , Camundongos , Camundongos Nus , Microssomos/metabolismo , Modelos Moleculares , Ratos , Transdução de Sinais/efeitos dos fármacos , Solubilidade , Estereoisomerismo , Relação Estrutura-Atividade , TYK2 Quinase/antagonistas & inibidores
11.
J Neurosci ; 31(1): 184-92, 2011 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-21209203

RESUMO

Gonadotropin-releasing hormone (GnRH) neurons represent the final common output of signals from the brain that regulates reproductive function. A wide range of environmental factors impact GnRH neuron activity including disease, stress, nutrition, and seasonal cues, as well as gonadal steroid hormones. The CNS response is thought to be mediated, at least in part, through intermediate signaling molecules that affect GnRH neuronal activity. In vitro, GnRH neuronal cell lines respond to a variety of ligands that activate the Jak (Janus-activated kinase)/STAT (signal transducers and activators of transcription) intracellular signaling pathway. To determine its biological function in reproduction, we used Cre (cAMP response element)/LoxP technology to generate GnRH neuron-specific Jak2 conditional knock-out (Jak2 G(-/-)) mice. GnRH mRNA levels were reduced in Jak2 G(-/-) mice when compared with controls, while the number of GnRH neurons was equivalent, indicating a reduction in GnRH gene expression. Secretion of GnRH is also reduced as basal serum luteinizing hormone (LH) levels were significantly lower in female Jak2 G(-/-) mice while the pituitary responded normally to exogenous GnRH. Preovulatory LH surge levels were blunted in Jak2 G(-/-) mice, which was correlated with reduced GnRH neuronal activation as assessed by c-Fos. However, the activation of GnRH neurons following release from estrogen-negative feedback is retained. Female Jak2 G(-/-) mice exhibited significantly delayed puberty and first estrus, abnormal estrous cyclicity, and impaired fertility. These results demonstrate an essential role for Jak2 signaling in GnRH neurons for normal reproductive development and fertility in female mice.


Assuntos
Regulação para Baixo/fisiologia , Hormônio Liberador de Gonadotropina/metabolismo , Janus Quinase 2/fisiologia , Reprodução/fisiologia , Animais , Contagem de Células/métodos , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Ciclo Estral/genética , Éxons/genética , Feminino , Fertilidade/genética , Hormônio Liberador de Gonadotropina/genética , Hormônio Liberador de Gonadotropina/farmacologia , Proteínas de Fluorescência Verde/genética , Hipotálamo/citologia , Janus Quinase 2/deficiência , Hormônio Luteinizante/sangue , Camundongos , Camundongos Knockout , Neurônios/metabolismo , Ovariectomia , Ovário/patologia , Proteínas Proto-Oncogênicas c-fos/metabolismo , Puberdade Tardia/genética , RNA Mensageiro/metabolismo , Reprodução/efeitos dos fármacos , Reprodução/genética
12.
Free Radic Biol Med ; 49(11): 1815-21, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20875851

RESUMO

Most neuroprotection studies with nicotinic agonists have shown efficacy when given before the stressor. Here we have investigated whether the α7 nicotinic acetylcholine receptor (nAChR) agonist PNU282987 can prevent cell death once the cells have already undergone an oxidative stress. The combination of rotenone (30 µM) plus oligomycin A (10 µM) (rot/oligo) has been used as an in vitro model of mitochondrial ROS production. SH-SY5Y cells incubated with rot/oligo for 8h and left for another 16 h in MEM/F-12 experienced 30% apoptotic cell death. Under these experimental conditions, PNU282987 administered after rot/oligo (PST/PNU) prevented cell death in a concentration-dependent manner. Co-incubation of PNU282987 with 100 nM methyllycaconitine (a selective α7 nAChR antagonist), 10 µM mecamylamine (a nonselective nAChR antagonist), 3 µM LY294002 (a PI3K inhibitor), or 10 µM AG490 (a Jak2 inhibitor) prevented the protection afforded by PST/PNU. Moreover, the increase in ROS, active caspase-3, and apoptosis caused by rot/oligo was also prevented by PST/PNU. Furthermore, PNU282987 increased the expression of heme oxygenase-1 (HO-1), a critical cell defense enzyme against oxidative stress; this increase was prevented by AG490 or LY294002. The HO-1 inhibitor Sn(IV) protoporphyrin-IX also inhibited the PST/PNU protecting effects. These results suggest that activation of α7 nAChR linked to the Jak2/PI3K/Akt cascade induces the antioxidant enzyme HO-1 to provide neuroprotection.


Assuntos
Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Compostos Bicíclicos com Pontes/farmacologia , Heme Oxigenase-1/fisiologia , Janus Quinase 2/fisiologia , Proteína Oncogênica v-akt/fisiologia , Estresse Oxidativo/fisiologia , Receptores Nicotínicos/fisiologia , Aconitina/análogos & derivados , Aconitina/farmacologia , Células Cultivadas , Citoproteção/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Heme Oxigenase-1/metabolismo , Humanos , Janus Quinase 2/metabolismo , Agonistas Nicotínicos/farmacologia , Antagonistas Nicotínicos/farmacologia , Proteína Oncogênica v-akt/metabolismo , Estresse Oxidativo/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Receptores Nicotínicos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Fatores de Tempo , Receptor Nicotínico de Acetilcolina alfa7
13.
Carcinogenesis ; 31(12): 2097-104, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-20732905

RESUMO

Cucurbitacin E (CuE, α-elaterin), a tetracyclic triterpenes compound from folk traditional Chinese medicine plants, has been shown to inhibit cancer cell growth, inflammatory response and bilirubin-albumin binding. However, the effects of CuE on tumor angiogenesis and its potential molecular mechanism are still unknown. Here, we demonstrated that CuE significantly inhibited human umbilical vascular endothelial cell (HUVEC) proliferation, migration and tubulogenesis in vitro and blocked angiogenesis in chick embryo chorioallantoic membrane assay and mouse corneal angiogenesis model in vivo. Furthermore, we found that CuE remarkably induced HUVEC apoptosis, inhibited tumor angiogenesis and suppressed human prostate tumor growth in xenograft tumor model. Finally, we showed that CuE blocked vascular endothelial growth factor receptor (VEGFR) 2-mediated Janus kinase (Jak) 2-signal transducer and activator of transcription (STAT) 3 signaling pathway in endothelial cells and suppressed the downstream protein kinases, such as extracellular signal-regulated kinase and p38 mitogen-activated protein kinases. Therefore, our studies provided the first evidence that CuE inhibited tumor angiogenesis by inhibiting VEGFR2-mediated Jak-STAT3 and mitogen-activated protein kinases signaling pathways and CuE is a potential candidate in angiogenesis-related disease therapy.


Assuntos
Inibidores da Angiogênese/farmacologia , Janus Quinase 2/fisiologia , Neoplasias Experimentais/irrigação sanguínea , Neovascularização Patológica/prevenção & controle , Fator de Transcrição STAT3/fisiologia , Triterpenos/farmacologia , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/fisiologia , Transporte Ativo do Núcleo Celular , Animais , Apoptose/efeitos dos fármacos , Células Cultivadas , Embrião de Galinha , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Fosforilação , Transdução de Sinais/fisiologia , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
14.
Br J Haematol ; 150(1): 46-57, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20507304

RESUMO

Constitutive activation of Janus kinase (Jak) 2 is the most prevalent pathogenic event observed in the myeloproliferative disorders (MPD), suggesting that inhibitors of Jak2 may prove valuable in their management. Inhibition of the Aurora kinases has also proven to be an effective therapeutic strategy in a number of haematological malignancies. AT9283 is a multi-targeted kinase inhibitor with potent activity against Jak2 and Aurora kinases A and B, and is currently being evaluated in clinical trials. To investigate the therapeutic potential of AT9283 in the MPD we studied its activity in a number of Jak2-dependent systems. AT9283 potently inhibited proliferation and Jak2-related signalling in Jak2-dependent cell lines as well as inhibiting the formation of erythroid colonies from haematopoietic progenitors isolated from MPD patients with Jak2 mutations. The compound also demonstrated significant therapeutic potential in vivo in an ETV6-JAK2 (TEL-JAK2) murine leukaemia model. Inhibition of both Jak2 and Aurora B was observed in the model systems used, indicating a dual mechanism of action. Our results suggest that AT9283 may be a valuable therapy in patients with MPD and that the dual inhibition of Jak2 and the Aurora kinases may potentially offer combinatorial efficacy in the treatment of these diseases.


Assuntos
Antineoplásicos/farmacologia , Benzimidazóis/farmacologia , Janus Quinase 2/antagonistas & inibidores , Transtornos Mieloproliferativos/tratamento farmacológico , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Ureia/análogos & derivados , Animais , Antineoplásicos/uso terapêutico , Aurora Quinase B , Aurora Quinases , Benzimidazóis/uso terapêutico , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Avaliação Pré-Clínica de Medicamentos/métodos , Células Precursoras Eritroides/efeitos dos fármacos , Humanos , Janus Quinase 2/genética , Janus Quinase 2/fisiologia , Leucemia Experimental/tratamento farmacológico , Leucemia Experimental/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Mutação , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/patologia , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas , Ureia/farmacologia , Ureia/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Immunol ; 184(9): 5298-307, 2010 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-20363976

RESUMO

Inhibiting signal transduction induced by inflammatory cytokines offers a new approach for the treatment of autoimmune diseases such as rheumatoid arthritis. Kinase inhibitors have shown promising oral disease-modifying antirheumatic drug potential with efficacy similar to anti-TNF biologics. Direct and indirect inhibition of the JAKs, with small molecule inhibitors like CP-690,550 and INCB018424 or neutralizing Abs, such as the anti-IL6 receptor Ab tocilizumab, have demonstrated rapid and sustained improvement in clinical measures of disease, consistent with their respective preclinical experiments. Therefore, it is of interest to identify optimized JAK inhibitors with unique profiles to maximize therapeutic opportunities. INCB028050 is a selective orally bioavailable JAK1/JAK2 inhibitor with nanomolar potency against JAK1 (5.9 nM) and JAK2 (5.7 nM). INCB028050 inhibits intracellular signaling of multiple proinflammatory cytokines including IL-6 and IL-23 at concentrations <50 nM. Significant efficacy, as assessed by improvements in clinical, histologic and radiographic signs of disease, was achieved in the rat adjuvant arthritis model with doses of INCB028050 providing partial and/or periodic inhibition of JAK1/JAK2 and no inhibition of JAK3. Diminution of inflammatory Th1 and Th17 associated cytokine mRNA levels was observed in the draining lymph nodes of treated rats. INCB028050 was also effective in multiple murine models of arthritis, with no evidence of suppression of humoral immunity or adverse hematologic effects. These data suggest that fractional inhibition of JAK1 and JAK2 is sufficient for significant activity in autoimmune disease models. Clinical evaluation of INCB028050 in RA is ongoing.


Assuntos
Artrite Experimental/tratamento farmacológico , Artrite Experimental/enzimologia , Janus Quinase 1/antagonistas & inibidores , Janus Quinase 2/antagonistas & inibidores , Inibidores de Proteínas Quinases/administração & dosagem , Animais , Artrite Experimental/imunologia , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/enzimologia , Doenças Autoimunes/imunologia , Linhagem Celular , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Humanos , Mediadores da Inflamação/antagonistas & inibidores , Mediadores da Inflamação/fisiologia , Janus Quinase 1/fisiologia , Janus Quinase 2/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos DBA , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Distribuição Aleatória , Ratos , Ratos Endogâmicos Lew , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
16.
Leukemia ; 21(3): 505-10, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17230231

RESUMO

An acquired autoactivating mutation with a V617F amino-acid substitution in the JAK2 tyrosine kinase is frequently found in BCR/ABL-negative myeloproliferative disorders (MPD). Hypermethylation of CpG islands within gene promoter regions is associated with transcriptional inactivation and represents an important mechanism of gene silencing in the pathogenesis of hematopoietic malignancies. In this study, we determined the DNA methylation status of 13 cancer-related genes in the context of JAK2 mutations in 39 patients with MPD. Genes analyzed for hypermethylation were SOCS-1, SHP-1, E-cadherin, MGMT, TIMP-2, TIMP-3, p15, p16, p73, DAPK1, RASSF1A, RARbeta2 and hMLH1. We found at least one hypermethylated gene in 15/39 MPD patient specimens, and in 6/39 samples aberrant methylation of the negative cytokine regulator SOCS-1 was present. The JAK2V617F mutation was found in 21/39 patients as determined by allele-specific polymerase chain reaction. Hypermethylation of SOCS-1 was observed in 3/21 patients with an autoactivating JAK2 mutation and in 3/18 patients with wild-type JAK2. Our results suggest that epigenetic inactivation of SOCS-1 may be a complementary mechanism to the JAK2V617F mutation in the pathogenesis of MPD that leads to dysregulation of JAK-STAT signal transduction and thus contributes to growth factor hypersensitivity.


Assuntos
Ilhas de CpG , Metilação de DNA , Epigênese Genética , Inativação Gênica , Janus Quinase 2/genética , Transtornos Mieloproliferativos/genética , Proteínas Supressoras da Sinalização de Citocina/genética , Substituição de Aminoácidos , Análise Mutacional de DNA , Progressão da Doença , Feminino , Proteínas de Fusão bcr-abl , Regulação da Expressão Gênica , Genes Neoplásicos , Humanos , Janus Quinase 2/fisiologia , Leucemia Mieloide/genética , Masculino , Mutação de Sentido Incorreto , Transtornos Mieloproliferativos/patologia , Mutação Puntual , Receptores do Ácido Retinoico/genética , Transdução de Sinais/genética , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA