Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Gut ; 66(1): 145-155, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-26385087

RESUMO

OBJECTIVE: Angiotensin II (AngII) activates via angiotensin-II-type-I receptor (AT1R) Janus-kinase-2 (JAK2)/Arhgef1 pathway and subsequently RHOA/Rho-kinase (ROCK), which induces experimental and probably human liver fibrosis. This study investigated the relationship of JAK2 to experimental and human portal hypertension. DESIGN: The mRNA and protein levels of JAK2/ARHGEF1 signalling components were analysed in 49 human liver samples and correlated with clinical parameters of portal hypertension in these patients. Correspondingly, liver fibrosis (bile duct ligation (BDL), carbon tetrachloride (CCl4)) was induced in floxed-Jak2 knock-out mice with SM22-promotor (SM22Cre+-Jak2f/f). Transcription and contraction of primary myofibroblasts from healthy and fibrotic mice and rats were analysed. In two different cirrhosis models (BDL, CCl4) in rats, the acute haemodynamic effect of the JAK2 inhibitor AG490 was assessed using microsphere technique and isolated liver perfusion experiments. RESULTS: Hepatic transcription of JAK2/ARHGEF1 pathway components was upregulated in liver cirrhosis dependent on aetiology, severity and complications of human liver cirrhosis (Model for End-stage Liver disease (MELD) score, Child score as well as ascites, high-risk varices, spontaneous bacterial peritonitis). SM22Cre+- Jak2f/f mice lacking Jak2 developed less fibrosis and lower portal pressure (PP) than SM22Cre--Jak2f/f upon fibrosis induction. Myofibroblasts from SM22Cre+-Jak2f/f mice expressed less collagen and profibrotic markers upon activation. AG490 relaxed activated hepatic stellate cells in vitro. In cirrhotic rats, AG490 decreased hepatic vascular resistance and consequently the PP in vivo and in situ. CONCLUSIONS: Hepatic JAK2/ARHGEF1/ROCK expression is associated with portal hypertension and decompensation in human cirrhosis. The deletion of Jak2 in myofibroblasts attenuated experimental fibrosis and acute inhibition of JAK2 decreased PP. Thus, JAK2 inhibitors, already in clinical use for other indications, might be a new approach to treat cirrhosis with portal hypertension.


Assuntos
Hipertensão Portal/genética , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Cirrose Hepática/genética , Fatores de Troca de Nucleotídeo Guanina Rho/genética , Fatores de Troca de Nucleotídeo Guanina Rho/metabolismo , Adulto , Animais , Tetracloreto de Carbono , Colágeno/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Células Estreladas do Fígado/efeitos dos fármacos , Humanos , Hipertensão Portal/metabolismo , Hipertensão Portal/fisiopatologia , Janus Quinase 2/antagonistas & inibidores , Ligadura , Cirrose Hepática/etiologia , Cirrose Hepática/metabolismo , Cirrose Hepática/fisiopatologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/genética , Pessoa de Meia-Idade , Proteínas Musculares/genética , Miofibroblastos/fisiologia , Pressão na Veia Porta/efeitos dos fármacos , RNA Mensageiro/metabolismo , Ratos/fisiologia , Ratos Sprague-Dawley , Índice de Gravidade de Doença , Transdução de Sinais , Transcrição Gênica , Tirfostinas/farmacologia , Regulação para Cima , Resistência Vascular/efeitos dos fármacos , Adulto Jovem , Quinases Associadas a rho/genética , Proteína rhoA de Ligação ao GTP/genética
2.
J Immunol ; 193(2): 477-84, 2014 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-24920845

RESUMO

Pruritus occurs frequently in patients with polycythemia vera (PV), and the pathophysiology of PV-associated pruritus is unclear. We have previously demonstrated that transgenic mice expressing JAK2V617F displayed clear PV-like phenotypes. In the current study, we found frequent occurrence of pruritus with aged JAK2V617F transgenic mice and further investigated the underlying mechanisms by studying mast cells, key players in allergic reactions and anaphylaxis. Massive accumulations of mast cells were observed in the skin of pruritic JAK2V617F transgenic mice. In vitro culture yielded much higher mast cell counts from the bone marrow, spleen, peripheral blood, and peritoneal cavity of JAK2V617F transgenic mice than from controls. Cultured mast cells from JAK2V617F transgenic mice exhibited enhanced proliferative signals, relative resistance to cell death upon growth factor deprivation, and a growth advantage over control cells under suboptimal growth conditions. However, these mast cells displayed normal morphology and contained normal levels of mast cell proteases before and after degranulation. Finally, the JAK2 inhibitor G6 effectively reduced mast cell numbers and alleviated pruritus in JAK2V617F transgenic mice. Collectively, these data demonstrate that mast cells are involved in PV-associated pruritogenesis and that JAK2 inhibitors are potential antipruritus drugs.


Assuntos
Janus Quinase 2/metabolismo , Mastócitos/metabolismo , Policitemia Vera/metabolismo , Prurido/metabolismo , Substituição de Aminoácidos , Animais , Western Blotting , Células da Medula Óssea/metabolismo , Células CHO , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Cricetinae , Cricetulus , Feminino , Humanos , Interleucina-3/farmacologia , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/genética , Masculino , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Policitemia Vera/genética , Inibidores de Proteínas Quinases/farmacologia , Prurido/genética , Prurido/prevenção & controle , Pele/metabolismo , Pele/patologia , Fator de Células-Tronco/farmacologia , Estilbenos/farmacologia , Fatores de Tempo
3.
Arch Pharm (Weinheim) ; 349(12): 925-933, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-27862215

RESUMO

Janus kinases (JAKs) and their gain-of-function mutants have been implicated in a range of oncological, inflammatory, and autoimmune conditions, which has sparked great research interest in the discovery and development of small-molecule JAK inhibitors. Two molecules are currently marketed as JAK inhibitors, but due to the displayed side effects (owing to their suboptimal selectivities among the various JAK subtypes) new JAK inhibitors are still sought after. We present the results of an extensive virtual screening campaign based on a multi-step screening protocol involving ligand docking. The screening yielded five new, experimentally validated inhibitors of JAK1 with 8-hydroxyquinoline as a novel hinge-binding scaffold. The compounds did not only display favorable potencies in a JAK1V658F -driven cell-based assay but were also shown to be non-cytotoxic on rat liver cells.


Assuntos
Janus Quinase 1/antagonistas & inibidores , Oxiquinolina/análogos & derivados , Oxiquinolina/farmacologia , Animais , Morte Celular/efeitos dos fármacos , Células Cultivadas , Camundongos , Simulação de Acoplamento Molecular , Mutação , Oxiquinolina/síntese química , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacologia , Ratos , Relação Estrutura-Atividade
4.
Am J Pathol ; 181(3): 858-65, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22796437

RESUMO

Philadelphia chromosome-negative myeloproliferative neoplasms, including polycythemia vera, essential thrombocytosis, and myelofibrosis, are disorders characterized by abnormal hematopoiesis. Among these myeloproliferative neoplasms, myelofibrosis has the most unfavorable prognosis. Furthermore, currently available therapies for myelofibrosis have little to no efficacy in the bone marrow and hence, are palliative. We recently developed a Janus kinase 2 (Jak2) small molecule inhibitor called G6 and found that it exhibits marked efficacy in a xenograft model of Jak2-V617F-mediated hyperplasia and a transgenic mouse model of Jak2-V617F-mediated polycythemia vera/essential thrombocytosis. However, its efficacy in Jak2-mediated myelofibrosis has not previously been examined. Here, we hypothesized that G6 would be efficacious in Jak2-V617F-mediated myelofibrosis. To test this, mice expressing the human Jak2-V617F cDNA under the control of the vav promoter were administered G6 or vehicle control solution, and efficacy was determined by measuring parameters within the peripheral blood, liver, spleen, and bone marrow. We found that G6 significantly reduced extramedullary hematopoiesis in the liver and splenomegaly. In the bone marrow, G6 significantly reduced pathogenic Jak/STAT signaling by 53%, megakaryocytic hyperplasia by 70%, and the Jak2 mutant burden by 68%. Furthermore, G6 significantly improved the myeloid to erythroid ratio and significantly reversed the myelofibrosis. Collectively, these results indicate that G6 is efficacious in Jak2-V617F-mediated myelofibrosis, and given its bone marrow efficacy, it may alter the natural history of this disease.


Assuntos
Janus Quinase 2/metabolismo , Mielofibrose Primária/tratamento farmacológico , Mielofibrose Primária/enzimologia , Inibidores de Proteínas Quinases/uso terapêutico , Bibliotecas de Moléculas Pequenas/uso terapêutico , Estilbenos/uso terapêutico , Substituição de Aminoácidos/genética , Animais , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Modelos Animais de Doenças , Hematopoese Extramedular/efeitos dos fármacos , Humanos , Hiperplasia , Janus Quinase 2/antagonistas & inibidores , Megacariócitos/efeitos dos fármacos , Megacariócitos/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Células Mieloides/efeitos dos fármacos , Células Mieloides/patologia , Fosforilação/efeitos dos fármacos , Mielofibrose Primária/sangue , Mielofibrose Primária/fisiopatologia , Inibidores de Proteínas Quinases/farmacologia , Reticulina/efeitos dos fármacos , Reticulina/metabolismo , Fator de Transcrição STAT5/metabolismo , Bibliotecas de Moléculas Pequenas/farmacologia , Baço/efeitos dos fármacos , Baço/patologia , Baço/fisiopatologia , Esplenomegalia/complicações , Esplenomegalia/tratamento farmacológico , Esplenomegalia/patologia , Esplenomegalia/fisiopatologia , Estilbenos/farmacologia
5.
Compr Physiol ; 13(2): 4409-4491, 2023 03 30.
Artigo em Inglês | MEDLINE | ID: mdl-36994769

RESUMO

Aldosterone exerts profound effects on renal and cardiovascular physiology. In the kidney, aldosterone acts to preserve electrolyte and acid-base balance in response to changes in dietary sodium (Na+ ) or potassium (K+ ) intake. These physiological actions, principally through activation of mineralocorticoid receptors (MRs), have important effects particularly in patients with renal and cardiovascular disease as demonstrated by multiple clinical trials. Multiple factors, be they genetic, humoral, dietary, or otherwise, can play a role in influencing the rate of aldosterone synthesis and secretion from the adrenal cortex. Normally, aldosterone secretion and action respond to dietary Na+ intake. In the kidney, the distal nephron and collecting duct are the main targets of aldosterone and MR action, which stimulates Na+ absorption in part via the epithelial Na+ channel (ENaC), the principal channel responsible for the fine-tuning of Na+ balance. Our understanding of the regulatory factors that allow aldosterone, via multiple signaling pathways, to function properly clearly implicates this hormone as central to many pathophysiological effects that become dysfunctional in disease states. Numerous pathologies that affect blood pressure (BP), electrolyte balance, and overall cardiovascular health are due to abnormal secretion of aldosterone, mutations in MR, ENaC, or effectors and modulators of their action. Study of the mechanisms of these pathologies has allowed researchers and clinicians to create novel dietary and pharmacological targets to improve human health. This article covers the regulation of aldosterone synthesis and secretion, receptors, effector molecules, and signaling pathways that modulate its action in the kidney. We also consider the role of aldosterone in disease and the benefit of mineralocorticoid antagonists. © 2023 American Physiological Society. Compr Physiol 13:4409-4491, 2023.


Assuntos
Aldosterona , Rim , Humanos , Aldosterona/metabolismo , Aldosterona/farmacologia , Rim/metabolismo , Néfrons/metabolismo , Sódio/metabolismo , Pressão Sanguínea
6.
J Biol Chem ; 286(6): 4280-91, 2011 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-21127060

RESUMO

Using structure-based virtual screening, we previously identified a novel stilbenoid inhibitor of Jak2 tyrosine kinase named G6. Here, we hypothesized that G6 suppresses Jak2-V617F-mediated human pathological cell growth in vitro and in vivo. We found that G6 inhibited proliferation of the Jak2-V617F expressing human erythroleukemia (HEL) cell line by promoting marked cell cycle arrest and inducing apoptosis. The G6-dependent increase in apoptosis levels was concomitant with increased caspase 3/7 activity and cleavage of PARP. G6 also selectively inhibited phosphorylation of STAT5, a downstream signaling target of Jak2. Using a mouse model of Jak2-V617F-mediated hyperplasia, we found that G6 significantly decreased the percentage of blast cells in the peripheral blood, reduced splenomegaly, and corrected a pathologically low myeloid to erythroid ratio in the bone marrow by eliminating HEL cell engraftment in this tissue. In addition, drug efficacy correlated with the presence of G6 in the plasma, marrow, and spleen. Collectively, these data demonstrate that the stilbenoid compound, G6, suppresses Jak2-V617F-mediated aberrant cell growth. As such, G6 may be a potential therapeutic lead candidate against Jak2-mediated, human disease.


Assuntos
Ciclo Celular/efeitos dos fármacos , Janus Quinase 2/metabolismo , Leucemia Eritroblástica Aguda/enzimologia , Mutação de Sentido Incorreto , Inibidores de Proteínas Quinases/farmacologia , Estilbenos/farmacologia , Substituição de Aminoácidos , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Ciclo Celular/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Janus Quinase 2/genética , Leucemia Eritroblástica Aguda/tratamento farmacológico , Leucemia Eritroblástica Aguda/genética , Camundongos , Camundongos Mutantes , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
7.
Mol Cell Biochem ; 367(1-2): 125-40, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22584586

RESUMO

Jak2 mutations in the exon 14 and exon 12 regions that cause constitutive activation have been associated with myeloproliferative neoplasms. We have previously shown that a pi stacking interaction between F617 and F595 is important for the constitutive activation of Jak2-V617F (Gnanasambandan et al., Biochemistry 49:9972-9984, 2010). Here, using a combination of molecular dynamics (MD) simulations and in vitro mutagenesis, we studied the molecular mechanism for the constitutive activation of the Jak2 exon 12 mutation, H538Q/K539L. The activation levels of Jak2-H538Q/K539L were found to be similar to that of Jak2-V617F, and Jak2-H538Q/K539L/V617F. Data from MD simulations indicated a shift in the salt bridge interactions of D620 and E621 with K539 in Jak2-WT to R541 in Jak2-H538Q/K539L. When compared to Jak2-WT, K539A mutation resulted in increased activation, while K539D or K539E mutations diminished Jak2 activation by 50 %. In the context of Jak2-H538Q/K539L, R541A mutation reduced its activation by 50 %, while R541D and R541E mutations returned its activation levels to that of Jak2-WT. Collectively, these results indicate that a shift in the salt bridge interaction of D620 and E621 with K539 in Jak2-WT to R541 in Jak2-H538Q/K539L is critical for constitutive activation of this Jak2 exon 12 mutant.


Assuntos
Ácido Aspártico/química , Ácido Glutâmico/química , Janus Quinase 2/química , Motivos de Aminoácidos , Substituição de Aminoácidos , Animais , Arginina/química , Células COS , Domínio Catalítico , Chlorocebus aethiops , Ativação Enzimática , Regulação da Expressão Gênica , Genes Reporter , Humanos , Janus Quinase 2/genética , Luciferases de Vaga-Lume/biossíntese , Luciferases de Vaga-Lume/genética , Lisina/química , Simulação de Dinâmica Molecular , Mutagênese Sítio-Dirigida , Transdução de Sinais , Termodinâmica
8.
Bioorg Med Chem Lett ; 22(3): 1402-7, 2012 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-22227213

RESUMO

In this study, we analyzed the structure-activity relationship properties of the small molecule Jak2 inhibitor G6. We synthesized a set of derivatives containing the native para-hydroxyl structure or an alternative meta-hydroxyl structure and examined their Jak2 inhibitory properties. We found that the para-hydroxyl derivative known as NB15 had excellent Jak2 inhibitory properties in silico, in vitro, and ex vivo when compared with meta-hydroxyl derivatives. These results indicate that NB15 is a potent derivative of the Jak2 inhibitor G6, and that maintaining the para-hydroxyl orientation of G6 is critical for its Jak2 inhibitory potential.


Assuntos
Benzilaminas/química , Benzilaminas/farmacologia , Janus Quinase 2/antagonistas & inibidores , Modelos Moleculares , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Animais , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Hidroxilação , Camundongos , Camundongos Transgênicos , Estrutura Molecular , Estilbenos/química , Estilbenos/farmacologia , Relação Estrutura-Atividade
9.
J Mol Cell Cardiol ; 50(6): 1026-34, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21420414

RESUMO

The in vitro treatment of vascular smooth muscle cells (VSMC) with angiotensin II (Ang II) causes Janus kinase 2 (Jak2) to interact with the Ang II type 1 receptor (AT(1)-R) resulting in enhanced cell growth. However, the role that Jak2 plays in AT(1)-R-mediated vascular cell growth and remodeling in vivo is less clear. We hypothesized that in vivo, Jak2 plays a rate-limiting role in Ang II-mediated neointima formation following vascular injury. Using the Cre-loxP system, we conditionally ablated Jak2 from the VSMC of mice. We found that these mice are protected from Ang II-mediated neointima formation following iron chloride-induced vascular injury. In addition, the VSMC Jak2 null mice were protected from injury-induced vascular fibrosis and the pathological loss of the contractile marker, smooth muscle α-actin. Finally, when compared to controls, the VSMC Jak2 null mice exhibited significantly less Ang II-induced VSMC proliferation and migration in vitro and in vivo and more apoptosis. These results suggest that Jak2 plays a central role in the causation of Ang II-induced neointima formation following vascular injury and may provide a novel target for the prevention of neointima formation.


Assuntos
Angiotensina II/farmacologia , Angiotensina II/uso terapêutico , Janus Quinase 2/genética , Músculo Liso Vascular/efeitos dos fármacos , Músculo Liso Vascular/metabolismo , Neointima , Lesões do Sistema Vascular/tratamento farmacológico , Actinas/metabolismo , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Modelos Animais de Doenças , Feminino , Fibrose/tratamento farmacológico , Fibrose/genética , Masculino , Camundongos , Camundongos Knockout , Neointima/induzido quimicamente , Neointima/tratamento farmacológico , Neointima/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Lesões do Sistema Vascular/patologia
10.
Biochemistry ; 50(36): 7774-86, 2011 Sep 13.
Artigo em Inglês | MEDLINE | ID: mdl-21823612

RESUMO

Hyperkinetic Jak2 tyrosine kinase signaling has been implicated in several human diseases including leukemia, lymphoma, myeloma, and the myeloproliferative neoplasms. Using structure-based virtual screening, we previously identified a novel Jak2 inhibitor named G6. We showed that G6 specifically inhibits Jak2 kinase activity and suppresses Jak2-mediated cellular proliferation. To elucidate the molecular and biochemical mechanisms by which G6 inhibits Jak2-mediated cellular proliferation, we treated Jak2-V617F expressing human erythroleukemia (HEL) cells for 12 h with either vehicle control or 25 µM of the drug and compared protein expression profiles using two-dimensional gel electrophoresis. One differentially expressed protein identified by electrospray mass spectroscopy was the intermediate filament protein, vimentin. It was present in DMSO treated cells but absent in G6 treated cells. HEL cells treated with G6 showed both time- and dose-dependent cleavage of vimentin as well as a marked reorganization of vimentin intermediate filaments within intact cells. In a mouse model of Jak2-V617F mediated human erythroleukemia, G6 also decreased the levels of vimentin protein, in vivo. The G6-induced cleavage of vimentin was found to be Jak2-dependent and calpain-mediated. Furthermore, we found that intracellular calcium mobilization is essential and sufficient for the cleavage of vimentin. Finally, we show that the cleavage of vimentin intermediate filaments, per se, is sufficient to reduce HEL cell viability. Collectively, these results suggest that G6-induced inhibition of Jak2-mediated pathogenic cell growth is concomitant with the disruption of intracellular vimentin filaments. As such, this work describes a novel pathway for the targeting of Jak2-mediated pathological cell growth.


Assuntos
Janus Quinase 2/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Estilbenos/farmacologia , Vimentina/metabolismo , Animais , Calpaína/metabolismo , Morte Celular , Linhagem Celular Tumoral , Humanos , Janus Quinase 2/metabolismo , Leucemia Eritroblástica Aguda/metabolismo , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Espectrometria de Massas por Ionização por Electrospray , Vimentina/química
11.
J Biol Chem ; 285(41): 31399-407, 2010 Oct 08.
Artigo em Inglês | MEDLINE | ID: mdl-20667821

RESUMO

Somatic mutations in the Jak2 protein, such as V617F, cause aberrant Jak/STAT signaling and can lead to the development of myeloproliferative neoplasms. This discovery has led to the search for small molecule inhibitors that target Jak2. Using structure-based virtual screening, our group recently identified a novel small molecule inhibitor of Jak2 named G6. Here, we identified a structure-function correlation of this compound. Specifically, five derivative compounds of G6 having structural similarity to the original lead compound were obtained and analyzed for their ability to (i) inhibit Jak2-V617F-mediated cell growth, (ii) inhibit the levels of phospho-Jak2, phospho-STAT3, and phospho-STAT5; (iii) induce apoptosis in human erythroleukemia cells; and (iv) suppress pathologic cell growth of Jak2-V617F-expressing human bone marrow cells ex vivo. Additionally, we computationally examined the interactions of these compounds with the ATP-binding pocket of the Jak2 kinase domain. We found that the stilbenoid core-containing derivatives of G6 significantly inhibited Jak2-V617F-mediated cell proliferation in a time- and dose-dependent manner. They also inhibited phosphorylation of Jak2, STAT3, and STAT5 proteins within cells, resulting in higher levels of apoptosis via the intrinsic apoptotic pathway. Finally, the stilbenoid derivatives inhibited the pathologic growth of Jak2-V617F-expressing human bone marrow cells ex vivo. Collectively, our data demonstrate that G6 has a stilbenoid core that is indispensable for maintaining its Jak2 inhibitory potential.


Assuntos
Janus Quinase 2/antagonistas & inibidores , Policitemia Vera/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Estilbenos/farmacologia , Substituição de Aminoácidos , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Mutação de Sentido Incorreto , Policitemia Vera/enzimologia , Policitemia Vera/genética , Inibidores de Proteínas Quinases/química , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/genética , Fator de Transcrição STAT5/metabolismo , Estilbenos/química , Relação Estrutura-Atividade
12.
Biochemistry ; 49(46): 9972-84, 2010 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-20958061

RESUMO

Somatic mutations in the Jak2 allele that lead to constitutive kinase activation of the protein have been identified in human disease conditions such as the myeloproliferative neoplasms (MPNs). The most common mutation in these patients is a V617F substitution mutation, which is believed to play a causative role in the MPN pathogenesis. As such, identifying the molecular basis for the constitutive activation of Jak2-V617F is important for understanding its clinical implications and potential treatment. Here, we hypothesized that conversion of residue 617 from Val to Phe resulted in the formation of novel π stacking interactions with neighboring Phe residues. To test this, we first examined the Jak2 structure via molecular modeling and identified a potential π stacking interaction between F594, F595, and F617. Disruption of this interaction through site-directed mutagenesis impaired Jak2 autophosphorylation, Jak2-dependent gene transcription, and in vitro kinase activity of the Jak2-V617F protein. Further, substitution of F594 and F595 with Trp did not affect Jak2 function significantly, but replacement with charged residues did, showing the importance of aromaticity and hydropathy index conservation at these positions. Using molecular dynamics (MD) simulations, we found that the π stacking interaction between residues 595 and 617 in the Jak2-V617F protein was of much greater energy and conformed to the properties of π stacking, relative to the Jak2-WT or Jak2-V617F/F594A/F595A. In summary, we have identified a π stacking interaction between F595 and F617 that is specific to and is critical for the constitutive activation of Jak2-V617F.


Assuntos
Janus Quinase 2/química , Fenilalanina/química , Animais , Células COS , Chlorocebus aethiops , Simulação por Computador , Humanos , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Modelos Moleculares , Mutação de Sentido Incorreto , Fenilalanina/genética , Conformação Proteica , Transfecção
13.
Bioorg Med Chem Lett ; 19(13): 3598-601, 2009 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-19447617

RESUMO

Janus kinase 2 (JAK2) plays a crucial role in the pathomechanism of myeloproliferative disorders and hematologic malignancies. A somatic mutation of JAK2 (Val617Phe) was previously shown to occur in 98% of patients with polycythemia vera and 50% of patients with essential thrombocythemia and primary myelofibrosis. Thus, effective JAK2 kinase inhibitors may be of significant therapeutic importance. Here, we applied a structure-based virtual screen to identify novel JAK2 inhibitors. One JAK2 inhibitor in particular, G6, demonstrated remarkable potency as well as specificity, which makes it as a potential lead candidate against diseases related to elevated JAK2 tyrosine kinase activity.


Assuntos
Alcenos/química , Janus Quinase 2/antagonistas & inibidores , Fenóis/química , Inibidores de Proteínas Quinases/química , Alcenos/farmacologia , Domínio Catalítico , Linhagem Celular Tumoral , Simulação por Computador , Cristalografia por Raios X , Avaliação Pré-Clínica de Medicamentos , Humanos , Janus Quinase 2/metabolismo , Proteínas Mutantes/antagonistas & inibidores , Proteínas Mutantes/metabolismo , Fenóis/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Relação Estrutura-Atividade
14.
Curr Oncol Rep ; 11(2): 117-24, 2009 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19216843

RESUMO

Although the Jak2-V617F mutation has generated strong awareness because of its causative role in myeloproliferative disorders, reports of Jak2 gene aberrations linked to hematologic malignancies have preceded those of V617F by nearly a decade. These malignant mutations include Jak2 amino acid substitutions, deletions, insertions, and chromosomal translocations. As a consequence, researchers are increasingly focused on identifying Jak2 inhibitors that suppress aberrant Jak2 kinase activity. Some of these inhibitors may one day become therapeutically beneficial for individuals with Jak2-related hematologic malignancies. This review summarizes various Jak2 mutations associated with hematologic malignancies and assesses some of the Jak2 inhibitors in the preclinical phase or in clinical trials. By reviewing these specific areas, we hope to have a better understanding of Jak2's role in hematologic malignancies and to shed light on the utility of Jak2 inhibitors.


Assuntos
Neoplasias Hematológicas/tratamento farmacológico , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/fisiologia , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Neoplasias Hematológicas/etiologia , Humanos , Janus Quinase 2/genética , Mutação , Transtornos Mieloproliferativos/etiologia
15.
Mol Cancer Ther ; 7(8): 2308-18, 2008 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-18723478

RESUMO

Jak2 tyrosine kinase is essential for animal development and hyperkinetic Jak2 function has been linked to a host of human diseases. Control of this pathway using Jak2-specific inhibitors would therefore potentially serve as a useful research tool and/or therapeutic agent. Here, we used a high-throughput program called DOCK to predict the ability of 20,000 small molecules to interact with a structural pocket adjacent to the ATP-binding site of murine Jak2. One small molecule, 2-methyl-1-phenyl-4-pyridin-2-yl-2-(2-pyridin-2-ylethyl)butan-1-one (herein designated as Z3), bound to Jak2 with a favorable energy score. Z3 inhibited Jak2-V617F and Jak2-WT autophosphorylation in a dose-dependent manner but was not cytotoxic to cells at concentrations that inhibited kinase activity. Z3 selectively inhibited Jak2 kinase function with no effect on Tyk2 or c-Src kinase function. Z3 significantly inhibited proliferation of the Jak2-V617F-expressing, human erythroleukemia cell line, HEL 92.1.7. The Z3-mediated reduction in cell proliferation correlated with reduced Jak2 and STAT3 tyrosine phosphorylation levels as well as marked cell cycle arrest. Finally, Z3 inhibited the growth of hematopoietic progenitor cells isolated from the bone marrow of an essential thrombocythemia patient harboring the Jak2-V617F mutation and a polycythemia vera patient carrying a Jak2-F537I mutation. Collectively, the data suggest that Z3 is a novel specific inhibitor of Jak2 tyrosine kinase.


Assuntos
Butanonas/farmacologia , Divisão Celular/efeitos dos fármacos , Janus Quinase 2/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Trifosfato de Adenosina/metabolismo , Animais , Células COS , Linhagem Celular Tumoral , Chlorocebus aethiops , Humanos , Imunoprecipitação , Janus Quinase 2/metabolismo , Fosforilação
16.
Biochemistry ; 47(32): 8326-34, 2008 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-18636744

RESUMO

Jak2 is a 130 kDa tyrosine kinase that is important in a number of cellular signaling pathways. Its function is intrinsically regulated by the phosphorylation of a handful of its 49 tyrosines. Here, we report that tyrosine 972 (Y972) is a novel site of Jak2 phosphorylation and, hence, autoregulation. Specifically, we found that Y972 is phosphorylated and confirmed that this residue resides on the surface of the protein. Using expression plasmids that expressed either wild-type Jak2 or a full-length Jak2 cDNA containing a single Y972F substitution mutation, we investigated the consequences of losing Y972 phosphorylation on Jak2 function. We determined that the loss of Y972 phosphorylation significantly reduced the levels of both Jak2 total tyrosine phosphorylation and phosphorylation of Y1007/Y1008. Additionally, Y972 phosphorylation was shown to be important for maximal kinase function. Interestingly, in response to classical cytokine activation, the Jak2 Y972F mutant exhibited a moderately impaired level of activation when compared to the wild-type protein. However, when Jak2 was activated via a GPCR ligand, the ability of the Y972F mutant to be activated was completely lost, therefore suggesting a differential role of Y972 in Jak2 activation. Finally, we found that phosphorylation of Y972 enhances Jak2 kinase function via a mechanism that appears to stabilize the active conformation of the protein. Collectively, our results suggest that Y972 is a novel site of Jak2 phosphorylation and plays an important differential role in ligand-dependent Jak2 activation via a mechanism that involves stabilization of the Jak2 active conformation.


Assuntos
Janus Quinase 2/metabolismo , Tirosina/metabolismo , Animais , Sítios de Ligação/genética , Células COS , Chlorocebus aethiops , Ativação Enzimática/genética , Humanos , Janus Quinase 2/genética , Janus Quinase 2/fisiologia , Camundongos , Fosforilação , Tirosina/genética
17.
Cell Signal ; 19(3): 600-9, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17027227

RESUMO

Previous work has suggested that the protein tyrosine phosphatase, SHP-2, may act to facilitate angiotensin II (Ang II)-mediated, Jak2-dependent signaling. However, the mechanisms by which this occurs are not known. Here, Ang II-mediated, Jak2-dependent signaling was analyzed in a fibroblast cell line lacking the N-terminal, SH2 domain of SHP-2 (SHP-2(Delta46-110)). While the SHP-2(Delta46-110) cells were capable of activating Jak2 tyrosine kinase, they were unable to facilitate AT1 receptor/Jak2 co-association, STAT activation and subsequent Ang II-mediated gene transcription when compared to wild type control cells. These data therefore suggested that the N-terminal SH2 domain of SHP-2 was acting to recruit Jak2 to the AT1 receptor signaling complex. We found that the N-terminal SH2 domain of SHP-2 binds Jak2 predominantly, but not exclusively at tyrosine 201. Mass spectrometry analysis confirmed that this tyrosine residue is in fact phosphorylated. When this tyrosine was converted to phenylalanine, the ability of Jak2 to activate subsequent downstream signaling events was reduced. In summary, we have identified a novel site of Jak2 tyrosine autophosphorylation; namely, tyrosine 201. Our data suggest that the N-terminal SH2 domain of SHP-2 binds this amino acid residue. The functional consequence of this interaction is to recruit Jak2 to the AT1 receptor signaling complex and in turn promote downstream Jak2-dependent signaling.


Assuntos
Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Janus Quinase 2/metabolismo , Proteínas Tirosina Fosfatases/metabolismo , Receptor Tipo 1 de Angiotensina/fisiologia , Transdução de Sinais , Domínios de Homologia de src/fisiologia , Animais , Western Blotting , Linhagem Celular , Ativação Enzimática , Fibroblastos/metabolismo , Genes Reporter , Glutationa Transferase/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/química , Janus Quinase 2/genética , Luciferases/metabolismo , Camundongos , Modelos Biológicos , Modelos Moleculares , Testes de Precipitina , Estrutura Terciária de Proteína , Proteína Tirosina Fosfatase não Receptora Tipo 1 , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteínas Tirosina Fosfatases/química , Proteínas Recombinantes de Fusão/metabolismo , Proteínas Tirosina Fosfatases Contendo o Domínio SH2 , Tirosina/metabolismo
18.
Blood Adv ; 1(16): 1224-1237, 2017 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-29296762

RESUMO

Genetics play a significant role in venous thromboembolism (VTE), yet current clinical laboratory-based testing identifies a known heritable thrombophilia (factor V Leiden, prothrombin gene mutation G20210A, or a deficiency of protein C, protein S, or antithrombin) in only a minority of VTE patients. We hypothesized that a substantial number of VTE patients could have lesser-known thrombophilia mutations. To test this hypothesis, we performed whole-exome sequencing (WES) in 64 patients with VTE, focusing our analysis on a novel 55-gene extended thrombophilia panel that we compiled. Our extended thrombophilia panel identified a probable disease-causing genetic variant or variant of unknown significance in 39 of 64 study patients (60.9%), compared with 6 of 237 control patients without VTE (2.5%) (P < .0001). Clinical laboratory-based thrombophilia testing identified a heritable thrombophilia in only 14 of 54 study patients (25.9%). The majority of WES variants were either associated with thrombosis based on prior reports in the literature or predicted to affect protein structure based on protein modeling performed as part of this study. Variants were found in major thrombophilia genes, various SERPIN genes, and highly conserved areas of other genes with established or potential roles in coagulation or fibrinolysis. Ten patients (15.6%) had >1 variant. Sanger sequencing performed in family members of 4 study patients with and without VTE showed generally concordant results with thrombotic history. WES and extended thrombophilia testing are promising tools for improving our understanding of VTE pathogenesis and identifying inherited thrombophilias.

19.
Cell Biochem Biophys ; 44(2): 213-22, 2006.
Artigo em Inglês | MEDLINE | ID: mdl-16456223

RESUMO

Jak2 is a nonreceptor tyrosine kinase that is essential for proper animal development and physiology. It is activated by ligand-occupied cell-surface receptors. Once activated, it then tyrosine phosphorylates the latent cytoplasmic transcription factors, termed the signal transducers and activators of transcription (STAT) proteins. Thus Jak2 is viewed as a classic mediator of ligand-dependent signal transduction. Recent studies, however, suggest that Jak2 may mediate cellular gene expression outside of the classically defined, ligand-activated, Jak/STAT-signaling paradigm. Here we review these studies, provide additional data, and discuss whether Jak2 is a mediator of ligand-independent gene transcription, and, in turn, whether our current understanding of the Jak/STAT signaling paradigm should be modified to incorporate these observations.


Assuntos
Proteínas Tirosina Quinases , Proteínas Proto-Oncogênicas , Transdução de Sinais , Animais , Regulação da Expressão Gênica , Humanos , Janus Quinase 2 , Ligantes , Transdução de Sinais/genética , Transcrição Gênica
20.
Oncotarget ; 7(24): 35989-36001, 2016 Jun 14.
Artigo em Inglês | MEDLINE | ID: mdl-27056884

RESUMO

Previous studies have shown that the bone marrow micro-environment supports the myeloproliferative neoplasms (MPN) phenotype including via the production of cytokines that can induce resistance to frontline MPN therapies. However, the mechanisms by which this occurs are poorly understood. Moreover, the ability to rapidly identify drug agents that can act as adjuvants to existing MPN frontline therapies is virtually non-existent. Here, using a novel predictive simulation approach, we sought to determine the effect of various drug agents on MPN cell lines, both with and without the micro-environment derived inflammatory cytokines. We first created individual simulation models for two representative MPN cell lines; HEL and SET-2, based on their genomic mutation and copy number variation (CNV) data. Running computational simulations on these virtual cell line models, we identified a synergistic effect of two drug agents on cell proliferation and viability; namely, the Jak2 kinase inhibitor, G6, and the Bcl-2 inhibitor, ABT737. IL-6 did not show any impact on the cells due to the predicted lack of IL-6 signaling within these cells. Interestingly, TNFα increased the sensitivity of the single drug agents and their use in combination while IFNγ decreased the sensitivity. In summary, this study predictively identified two drug agents that reduce MPN cell viability via independent mechanisms that was prospectively validated. Moreover, their efficacy is either potentiated or inhibited, by some of the micro-environment derived cytokines. Lastly, this study has validated the use of this simulation based technology to prospectively determine such responses.


Assuntos
Simulação por Computador , Modelos Biológicos , Inibidores de Proteínas Quinases/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Sinergismo Farmacológico , Humanos , Interleucina-6/farmacologia , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Mutação , Transtornos Mieloproliferativos/genética , Transtornos Mieloproliferativos/metabolismo , Transtornos Mieloproliferativos/patologia , Proteínas Proto-Oncogênicas c-bcl-2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Reprodutibilidade dos Testes , Microambiente Tumoral , Fator de Necrose Tumoral alfa/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA