Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Commun Biol ; 5(1): 132, 2022 02 15.
Article in English | MEDLINE | ID: mdl-35169231

ABSTRACT

Atherosclerosis is a chronic inflammatory condition in which macrophages play a major role. Janus kinase 2 (JAK2) is a pivotal molecule in inflammatory and metabolic signaling, and Jak2V617F activating mutation has recently been implicated with enhancing clonal hematopoiesis and atherosclerosis. To determine the essential in vivo role of macrophage (M)-Jak2 in atherosclerosis, we generate atherosclerosis-prone ApoE-null mice deficient in M-Jak2. Contrary to our expectation, these mice exhibit increased plaque burden with no differences in macrophage proliferation, recruitment or bone marrow clonal expansion. Notably, M-Jak2-deficient bone marrow derived macrophages show a significant defect in cholesterol efflux. Pharmacologic JAK2 inhibition with ruxolitinib also leads to defects in cholesterol efflux and accelerates atherosclerosis. Liver X receptor agonist abolishes the efflux defect and attenuates the accelerated atherosclerosis that occurs with M-Jak2 deficiency. Macrophages of individuals with the Jak2V617F mutation show increased efflux which is normalized when treated with a JAK2 inhibitor. Together, M-Jak2-deficiency leads to accelerated atherosclerosis primarily through defects in cholesterol efflux from macrophages.


Subject(s)
Atherosclerosis , Cholesterol , Janus Kinase 2 , Animals , Atherosclerosis/enzymology , Atherosclerosis/genetics , Atherosclerosis/metabolism , Cholesterol/metabolism , Janus Kinase 2/deficiency , Janus Kinase 2/genetics , Janus Kinase 2/metabolism , Macrophages/metabolism , Mice , Mice, Inbred C57BL
2.
Andrology ; 9(5): 1662-1671, 2021 09.
Article in English | MEDLINE | ID: mdl-34085398

ABSTRACT

BACKGROUND: Janus kinase 2 (JAK2) is activated in diabetic mellitus (DM) conditions and may enhance oxidative stress, apoptosis and fibrosis in many tissues. Whether JAK2 activation is involved in the occurrence of diabetic erectile dysfunction (ED) is unknown. OBJECTIVES: We performed this study to investigate the effect of JAK2 deficiency on diabetic ED. MATERIALS AND METHODS: Conditional JAK2 gene knockout mice (Cre+/+ -JAK2fl/fl ) were used, in which JAK2 gene knockout could be induced by tamoxifen. Mice fell into four groups: control, JAK2 knockout (JAK2-/- ), DM, and DM with JAK2-/- . DM was induced by intraperitoneal injection of streptozotocin. Two months later, JAK2 gene knockout was induced with tamoxifen in Cre+/+ -JAK2fl/fl mice. After another 2 months, erectile function was measured by electrical stimulation of the cavernous nerve, and penile tissues were harvested. Ratio of maximal intracavernosal pressure (MIP) to mean arterial blood pressure (MAP), expression and phosphorylation of JAK2, oxidative stress level, NO/Cyclic Guanosine Monophosphate (cGMP) pathway, apoptosis, fibrosis, and transforming growth factor beta 1 (TGF-ß1)/Smad/Collagen IV pathway in corpus cavernosum, were measured. RESULTS: JAK2 expression was remarkably decreased after induction with tamoxifen. JAK2 was activated in penile tissues of diabetic mice, and JAK2 deficiency could improve the impaired erectile function caused by DM. However, in mice without DM, JAK2 deficiency had no apparent influence on erectile function. Levels of oxidative stress, apoptosis, fibrosis, and TGF-ß1/Smad/Collagen IV pathway were all elevated by DM, whereas JAK2 deficiency lessened these alterations in diabetic mice. Moreover, JAK2 deficiency improved the expression of the down-regulated NO/cGMP pathway in diabetic mice. In non-diabetic mice, no apparent changes were found in aforementioned parameters after JAK2 gene knockout. DISCUSSION AND CONCLUSION: Our study showed that JAK2 deficiency could improve erectile function in diabetic mice, which might be mediated by reduction in oxidative stress, apoptosis, and fibrosis in corpus cavernosum.


Subject(s)
Diabetes Mellitus, Experimental/genetics , Erectile Dysfunction/genetics , Janus Kinase 2/deficiency , Penis/pathology , Animals , Apoptosis/genetics , Diabetes Mellitus, Experimental/complications , Fibrosis , Male , Mice , Oxidative Stress/genetics , Streptozocin
4.
Circ Res ; 122(3): 523-532, 2018 02 02.
Article in English | MEDLINE | ID: mdl-29420212

ABSTRACT

Increasing evidence shows that conventional cardiovascular risk factors are incompletely predictive of cardiovascular disease, particularly in elderly individuals, suggesting that there may still be unidentified causal risk factors. Although the accumulation of somatic DNA mutations is a hallmark of aging, its relevance in cardiovascular disease or other age-related conditions has been, with the exception of cancer, largely unexplored. Here, we review recent clinical and preclinical studies that have identified acquired mutations in hematopoietic stem cells and subsequent clonal hematopoiesis as a new cardiovascular risk factor and a potential major driver of atherosclerosis. Understanding the mechanisms underlying the connection between somatic mutation-driven clonal hematopoiesis and cardiovascular disease will be highly relevant in the context of personalized medicine, as it may provide key information for the design of diagnostic, preventive, or therapeutic strategies tailored to the effects of specific somatic mutations.


Subject(s)
Aging/genetics , Cardiovascular Diseases/etiology , Hematopoiesis/genetics , Hematopoietic Stem Cells/pathology , Mutation , Aged , Aging/pathology , Animals , Atherosclerosis/etiology , Atherosclerosis/genetics , Atherosclerosis/pathology , Atherosclerosis/therapy , Bone Marrow Transplantation , Cardiovascular Diseases/genetics , Cardiovascular Diseases/pathology , Causality , Clone Cells/pathology , DNA (Cytosine-5-)-Methyltransferases/genetics , DNA (Cytosine-5-)-Methyltransferases/physiology , DNA Methyltransferase 3A , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Dioxygenases , Genes, Neoplasm , Genetic Association Studies , Hematologic Neoplasms/genetics , Hematologic Neoplasms/pathology , Humans , Janus Kinase 2/deficiency , Janus Kinase 2/genetics , Janus Kinase 2/physiology , Mice , Population Dynamics , Precision Medicine , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Risk Factors
5.
Sci Rep ; 7(1): 7653, 2017 08 09.
Article in English | MEDLINE | ID: mdl-28794431

ABSTRACT

During obesity, macrophages can infiltrate metabolic tissues, and contribute to chronic low-grade inflammation, and mediate insulin resistance and diabetes. Recent studies have elucidated the metabolic role of JAK2, a key mediator downstream of various cytokines and growth factors. Our study addresses the essential role of macrophage JAK2 in the pathogenesis to obesity-associated inflammation and insulin resistance. During high-fat diet (HFD) feeding, macrophage-specific JAK2 knockout (M-JAK2-/-) mice gained less body weight compared to wildtype littermate control (M-JAK2+/+) mice and were protected from HFD-induced systemic insulin resistance. Histological analysis revealed smaller adipocytes and qPCR analysis showed upregulated expression of some adipogenesis markers in visceral adipose tissue (VAT) of HFD-fed M-JAK2-/- mice. There were decreased crown-like structures in VAT along with reduced mRNA expression of some macrophage markers and chemokines in liver and VAT of HFD-fed M-JAK2-/- mice. Peritoneal macrophages from M-JAK2-/- mice and Jak2 knockdown in macrophage cell line RAW 264.7 also showed lower levels of chemokine expression and reduced phosphorylated STAT3. However, leptin-dependent effects on augmenting chemokine expression in RAW 264.7 cells did not require JAK2. Collectively, our findings show that macrophage JAK2 deficiency improves systemic insulin sensitivity and reduces inflammation in VAT and liver in response to metabolic stress.


Subject(s)
Diet, High-Fat , Inflammation/etiology , Janus Kinase 2/deficiency , Macrophages/metabolism , Adipocytes/metabolism , Adipocytes/pathology , Animals , Chemokines/genetics , Chemokines/metabolism , Diet, High-Fat/adverse effects , Disease Models, Animal , Gene Expression , Hypertrophy , Inflammation/metabolism , Inflammation/pathology , Insulin Resistance/genetics , Intra-Abdominal Fat/metabolism , Liver/metabolism , Macrophages/immunology , Male , Mice , Mice, Knockout , Myeloid Cells/immunology , Myeloid Cells/metabolism
6.
J Am Soc Nephrol ; 28(9): 2641-2653, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28424277

ABSTRACT

The nonreceptor kinase Janus kinase 2 (JAK2) has garnered attention as a promising therapeutic target for the treatment of CKD. However, being ubiquitously expressed in the adult, JAK2 is also likely to be necessary for normal organ function. Here, we investigated the phenotypic effects of JAK2 deficiency. Mice in which JAK2 had been deleted from podocytes exhibited an elevation in urine albumin excretion that was accompanied by increased podocyte autophagosome fractional volume and p62 aggregation, which are indicative of impaired autophagy completion. In cultured podocytes, knockdown of JAK2 similarly impaired autophagy and led to downregulation in the expression of lysosomal genes and decreased activity of the lysosomal enzyme, cathepsin D. Because transcription factor EB (TFEB) has recently emerged as a master regulator of autophagosome-lysosome function, controlling the expression of several of the genes downregulated by JAK2 knockdown, we questioned whether TFEB is regulated by JAK2. In immortalized mouse podocytes, JAK2 knockdown decreased TFEB promoter activity, expression, and nuclear localization. In silico analysis and chromatin immunoprecipitation assays revealed that the downstream mediator of JAK2 signaling STAT1 binds to the TFEB promoter. Finally, overexpression of TFEB in JAK2-deficient podocytes reversed lysosomal dysfunction and restored albumin permselectivity. Collectively, these observations highlight the homeostatic actions of JAK2 in podocytes and the importance of TFEB to autophagosome-lysosome function in these cells. These results also raise the possibility that therapeutically modulating TFEB activity may improve podocyte health in glomerular disease.


Subject(s)
Autophagy/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , Janus Kinase 2/genetics , Podocytes/metabolism , Albuminuria/genetics , Animals , Autophagosomes/ultrastructure , Cathepsin D/metabolism , Cells, Cultured , Computer Simulation , Down-Regulation , Gene Knockdown Techniques , Janus Kinase 2/deficiency , Janus Kinase 2/metabolism , Kidney Glomerulus/cytology , Lysosomes/ultrastructure , Male , Mice , Microtubule-Associated Proteins/metabolism , Peptides/metabolism , Phenotype , Podocytes/ultrastructure , RNA, Messenger/metabolism , STAT1 Transcription Factor/genetics , STAT1 Transcription Factor/metabolism
7.
Am J Pathol ; 185(12): 3202-10, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26475415

ABSTRACT

The Janus kinase (JAK) system is involved in numerous cell signaling processes and is highly expressed in cardiac tissue. The JAK isoform JAK2 is activated by numerous factors known to influence cardiac function and pathologic conditions. However, although abundant, the role of JAK2 in the regulation or maintenance of cardiac homeostasis remains poorly understood. Using the Cre-loxP system, we generated a cardiac-specific deletion of Jak2 in the mouse to assess the effect on cardiac function with animals followed up for a 4-month period after birth. These animals had marked mortality during this period, although at 4 months mortality in male mice (47%) was substantially higher compared with female mice (30%). Both male and female cardiac Jak2-deleted mice had hypertrophy, dilated cardiomyopathy, and severe left ventricular dysfunction, including a marked reduction in ejection fractions as assessed by serial echocardiography, although the responses in females were somewhat less severe. Defective cardiac function was associated with altered protein levels of sarcoplasmic reticulum calcium-regulatory proteins particularly in hearts from male mice that had depressed levels of SERCA2 and phosphorylated phospholamban. In contrast, SERCA2 was unchanged in hearts of female mice, whereas phosphorylated phospholamban was increased. Our findings suggest that cardiac JAK2 is critical for maintaining normal heart function, and its ablation produces a severe pathologic phenotype composed of myocardial remodeling, heart failure, and pronounced mortality.


Subject(s)
Cardiomegaly/enzymology , Janus Kinase 2/physiology , Ventricular Dysfunction, Left/enzymology , Ventricular Remodeling/physiology , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Female , Gene Deletion , Genotype , Janus Kinase 2/deficiency , Janus Kinase 2/genetics , Male , Mice, Knockout , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Remodeling/genetics
8.
Mol Endocrinol ; 27(8): 1333-42, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23782652

ABSTRACT

Nonalcoholic fatty liver disease (NAFLD) is considered the hepatic expression of the metabolic syndrome, and its prevalence is increasing. The factors that influence the development of fatty liver and its progression to steatohepatitis and cirrhosis are not well understood. The pleiotropic hormone, GH, has been associated with an increased risk of NAFLD in humans and mice. GH is known to have diverse effects on lipid metabolism including decreasing body fat in vivo, presumably through stimulation of lipolysis via an undefined mechanism. Previously we described mice with hepatocyte-specific deletion of the GH signaling mediator, Janus kinase 2 (JAK2L). JAK2L animals have elevated serum GH, reduced body fat, high liver triglyceride content, and increased serum markers of hepatocyte injury (alanine transaminase and aspartate transaminase). We aimed to determine whether the elevation of GH in JAK2L mice contributed to fatty liver by promoting lipolysis directly in adipocytes. We generated mice with adipocyte-specific disruption of JAK2 (JAK2A) and found that GH resistance in adipocytes reduced lipolysis and increased body fat. JAK2A mice were then crossed to JAK2L mice, and the resultant JAK2L/A animals had increased body fat and decreased lipolysis, despite elevated circulating GH. Furthermore, the increased triglyceride content, serum alanine transaminase, and serum aspartate transaminase observed in JAK2L mice were nearly normalized with the additional disruption of JAK2 in adipocytes (JAK2L/A mice). Our results offer novel mechanistic insights into the long-recognized effects of GH on lipid flux and suggest that GH signaling may play an important regulatory role in the development of NAFLD.


Subject(s)
Fatty Liver/metabolism , Growth Hormone/metabolism , Janus Kinase 2/genetics , Lipid Metabolism/genetics , Lipolysis/genetics , Adipocytes/cytology , Adipose Tissue/cytology , Adipose Tissue/physiology , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Body Composition , Fatty Liver/genetics , Fibrosis , Gene Expression , Growth Hormone/blood , Janus Kinase 2/deficiency , Liver/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease , Receptors, Somatotropin , Signal Transduction , Triglycerides/analysis , Triglycerides/metabolism
9.
Am J Pathol ; 183(2): 617-25, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23747947

ABSTRACT

A number of inhibitors have been used to dissect the functional relevance of Jak2 in endothelial homeostasis, with disparate results. Given that Jak2 deficiency leads to embryonic lethality, the exact role of Jak2 in the regulation of postnatal endothelial function is yet to be fully elucidated. We generated a model in which Jak2 deficiency can be induced by tamoxifen in adult mice. Loss of Jak2 significantly impaired endothelium-dependent response capacity for vasodilators. Matrigel plug assays indicated a notable decrease in endothelial angiogenic function in Jak2-deficient mice. Studies in a hindlimb ischemic model indicated that Jak2 activity is likely to be a prerequisite for prompt perfusion recovery, based on the concordance of temporal changes in Jak2 expression during the course of ischemic injury and perfusion recovery. A remarkable delay in perfusion recovery, along with reduced capillary and arteriole formation, was observed in Jak2-deficient mice. Antibody array studies indicated that loss of Jak2 led to repressed eNOS expression. In mechanistic studies, Jak2 deficiency attenuated Raf-1/MEK1 signaling, which then reduced activity of Sp-1, an essential transcription factor responsible for eNOS expression. These data are important not only for understanding the exact role that Jak2 plays in endothelial homeostasis, but also for assessing Jak2-based therapeutic strategies in a variety of clinical settings.


Subject(s)
Janus Kinase 2/deficiency , MAP Kinase Kinase 1/physiology , Nitric Oxide Synthase Type III/metabolism , Protein Kinases/physiology , Proto-Oncogene Proteins c-raf/physiology , Animals , Aorta/drug effects , Aorta/enzymology , Endothelium, Vascular/drug effects , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiology , Enzyme Inhibitors/pharmacology , Hindlimb/blood supply , Ischemia/enzymology , Janus Kinase 2/drug effects , MAP Kinase Signaling System/physiology , Male , Mice , Mice, Knockout , Mice, Transgenic , Tamoxifen/pharmacology , Vasodilator Agents/pharmacology
10.
Blood ; 121(7): 1188-99, 2013 Feb 14.
Article in English | MEDLINE | ID: mdl-23264594

ABSTRACT

To establish a preclinical animal model for testing drugs with potential effects on myeloproliferative neoplasms (MPNs), we first performed a detailed phenotypic characterization of Cre-inducible transgenic JAK2-V617F mice. Deleting the conditional mouse Jak2-knockout alleles increased erythropoiesis and accentuated the polycythemia vera phenotype, but did not alter platelet or granulocyte levels. In a transplantation assay, JAK2-V617F(+) BM cells had an advantage over wild-type competitor cells. Using this competitive repopulation assay, we compared the effects of INC424 (ruxolitinib), a dual Jak1/Jak2 inhibitor, and hydroxyurea (HU). HU led to weight loss, but did not reduce spleen weight. The hematologic parameters were lowered and a slight decrease of the mutant allele burden was noted. INC424 had little effect on body weight, but strongly decreased spleen size and rapidly normalized RBC and neutrophil parameters. No significant decrease in the mutant allele burden was observed. INC424 reduced the phospho-Stat5 levels, whereas HU strongly increased phospho-Stat5, most likely because of the elevated erythropoietin levels in response to the HU-induced anemia. This compensatory increase in JAK/STAT signaling may counteract the beneficial effects of cytoreduction at higher doses of HU and represents an adverse effect that should be avoided.


Subject(s)
Hydroxyurea/pharmacology , Janus Kinase 2/genetics , Polycythemia Vera/drug therapy , Polycythemia Vera/genetics , Pyrazoles/pharmacology , Alleles , Amino Acid Substitution , Animals , Bone Marrow Transplantation , Disease Models, Animal , Female , Hematopoiesis/drug effects , Hematopoiesis/genetics , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Mutation , Nitriles , Phenotype , Polycythemia Vera/metabolism , Polycythemia Vera/pathology , Pyrimidines , STAT5 Transcription Factor/metabolism , Signal Transduction/drug effects
11.
Nature ; 489(7414): 155-9, 2012 Sep 06.
Article in English | MEDLINE | ID: mdl-22820254

ABSTRACT

The identification of somatic activating mutations in JAK2 (refs 1­4) and in the thrombopoietin receptor gene (MPL) in most patients with myeloproliferative neoplasm (MPN) led to the clinical development of JAK2 kinase inhibitors. JAK2 inhibitor therapy improves MPN-associated splenomegaly and systemic symptoms but does not significantly decrease or eliminate the MPN clone in most patients with MPN. We therefore sought to characterize mechanisms by which MPN cells persist despite chronic inhibition of JAK2. Here we show that JAK2 inhibitor persistence is associated with reactivation of JAK­STAT signalling and with heterodimerization between activated JAK2 and JAK1 or TYK2, consistent with activation of JAK2 in trans by other JAK kinases. Further, this phenomenon is reversible: JAK2 inhibitor withdrawal is associated with resensitization to JAK2 kinase inhibitors and with reversible changes in JAK2 expression. We saw increased JAK2 heterodimerization and sustained JAK2 activation in cell lines, in murine models and in patients treated with JAK2 inhibitors. RNA interference and pharmacological studies show that JAK2-inhibitor-persistent cells remain dependent on JAK2 protein expression. Consequently, therapies that result in JAK2 degradation retain efficacy in persistent cells and may provide additional benefit to patients with JAK2-dependent malignancies treated with JAK2 inhibitors.


Subject(s)
Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Myeloproliferative Disorders/drug therapy , Protein Multimerization , STAT Transcription Factors/metabolism , Signal Transduction , Animals , Cell Line , Disease Models, Animal , Drug Resistance, Neoplasm/drug effects , Enzyme Activation/drug effects , Gene Knockdown Techniques , Granulocytes/drug effects , Granulocytes/enzymology , Granulocytes/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , HSP90 Heat-Shock Proteins/metabolism , Humans , Janus Kinase 1/biosynthesis , Janus Kinase 1/deficiency , Janus Kinase 1/genetics , Janus Kinase 1/metabolism , Janus Kinase 2/deficiency , Janus Kinase 2/genetics , Mice , Myeloproliferative Disorders/enzymology , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology , Phosphorylation , Protein Biosynthesis , RNA Interference , Signal Transduction/drug effects , TYK2 Kinase/biosynthesis , TYK2 Kinase/deficiency , TYK2 Kinase/genetics , TYK2 Kinase/metabolism
12.
Nat Chem Biol ; 8(3): 285-93, 2012 Jan 29.
Article in English | MEDLINE | ID: mdl-22286129

ABSTRACT

Constitutive activation of STAT5 is critical for the maintenance of chronic myeloid leukemia (CML) characterized by the BCR-ABL oncoprotein. Tyrosine kinase inhibitors (TKIs) for the STAT5-activating kinase JAK2 have been discussed as a treatment option for CML patients. Using murine leukemia models combined with inducible ablation of JAK2, we show JAK2 dependence for initial lymphoid transformation, which is lost once leukemia is established. In contrast, initial myeloid transformation and leukemia maintenance were independent of JAK2. Nevertheless, several JAK2 TKIs induced apoptosis in BCR-ABL(+) cells irrespective of the presence of JAK2. This is caused by the previously unknown direct 'off-target' inhibition of BCR-ABL. Cellular and enzymatic analyses suggest that BCR-ABL phosphorylates STAT5 directly. Our findings suggest uncoupling of the canonical JAK2-STAT5 module upon BCR-ABL expression, thereby making JAK2 targeting dispensable. Thus, attempts to pharmacologically target STAT5 in BCR-ABL(+) diseases need to focus on STAT5 itself.


Subject(s)
Fusion Proteins, bcr-abl/metabolism , Janus Kinase 2/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Animals , Antineoplastic Agents/pharmacology , Benzamides , Cell Line, Tumor , Fusion Proteins, bcr-abl/antagonists & inhibitors , HEK293 Cells , Humans , Imatinib Mesylate , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/deficiency , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Piperazines/pharmacology , Pyrimidines/pharmacology , STAT5 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , U937 Cells
13.
Cell Signal ; 23(11): 1806-15, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21726629

ABSTRACT

Jak2 tyrosine kinase plays an important role in cytokine mediated signal transduction. There are 49 tyrosine residues in Jak2 and phosphorylation of some of these are known to play important roles in the regulation of Jak2 kinase activity. Here, using mass spectrometry, we identified tyrosine residues Y372 and Y373 as novel sites of Jak2 phosphorylation. Mutation of Y372 to F (Y372F) significantly inhibited Jak2 phosphorylation, including that of Y1007, whereas the Jak2-Y373F mutant displayed only modest reduction in phosphorylation. Relative to Jak2-WT, the ability of Jak2-Y372F to bind to and phosphorylate STAT1 was decreased, resulting in reduced Jak2-mediated downstream gene transcription. While the Y372F mutation had no effect on receptor-independent, hydrogen peroxide-mediated Jak2 activation, it impaired interferon-gamma (IFNγ) and epidermal growth factor (EGF)-dependent Jak2 activation. Interestingly however, the Y372F mutant exhibited normal receptor binding properties. Finally, co-expression of SH2-Bß only partially restored the activation of the Jak2-Y372F mutant suggesting that the mechanism whereby phosphorylation of Y372 is important for Jak2 activation is via dimerization. As such, our results indicate that Y372 plays a critical yet differential role in Jak2 activation and function via a mechanism involving Jak2 dimerization and stabilization of the active conformation.


Subject(s)
Enzyme Activation/drug effects , Gene Expression Regulation , Janus Kinase 2 , STAT1 Transcription Factor/metabolism , Signal Transduction/genetics , Tyrosine/metabolism , Animals , COS Cells , Chlorocebus aethiops , Dimerization , Enzyme Activation/genetics , Epidermal Growth Factor/pharmacology , Hydrogen Peroxide/pharmacology , Interferon-gamma/pharmacology , Janus Kinase 2/chemistry , Janus Kinase 2/deficiency , Janus Kinase 2/genetics , Mass Spectrometry , Mice , Mice, Knockout , Mutation , Phosphorylation , Plasmids , Protein Binding/drug effects , Protein Binding/genetics , Transcription, Genetic , Transfection , Tyrosine/genetics , Vaccinia virus
14.
J Clin Invest ; 121(4): 1412-23, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21364286

ABSTRACT

Non-alcoholic fatty liver disease is associated with multiple comorbid conditions, including diabetes, obesity, infection, and malnutrition. Mice with hepatocyte-specific disruption of growth hormone (GH) signaling develop fatty liver (FL), although the precise mechanism underlying this finding remains unknown. Because GH signals through JAK2, we developed mice bearing hepatocyte-specific deletion of JAK2 (referred to herein as JAK2L mice). These mice were lean, but displayed markedly elevated levels of GH, liver triglycerides (TGs), and plasma FFAs. Because GH is known to promote lipolysis, we crossed GH-deficient little mice to JAK2L mice, and this rescued the FL phenotype. Expression of the fatty acid transporter CD36 was dramatically increased in livers of JAK2L mice, as was expression of Pparg. Since GH signaling represses PPARγ expression and Cd36 is a known transcriptional target of PPARγ, we treated JAK2L mice with the PPARγ-specific antagonist GW9662. This resulted in reduced expression of liver Cd36 and decreased liver TG content. These results provide a mechanism for the FL observed in mice with liver-specific disruption in GH signaling and suggest that the development of FL depends on both GH-dependent increases in plasma FFA and increased hepatic uptake of FFA, likely mediated by increased expression of CD36.


Subject(s)
Fatty Liver/genetics , Fatty Liver/physiopathology , Growth Hormone/metabolism , Janus Kinase 2/deficiency , Liver/physiopathology , Anilides/pharmacology , Animals , CD36 Antigens/metabolism , Disease Models, Animal , Fatty Acids, Nonesterified/metabolism , Fatty Liver/etiology , Fatty Liver/pathology , Female , Gene Deletion , Hepatocytes/physiology , Janus Kinase 2/genetics , Liver/drug effects , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Organ Specificity , PPAR gamma/antagonists & inhibitors , Signal Transduction , Triglycerides/metabolism
15.
Cardiovasc Res ; 91(1): 171-9, 2011 Jul 01.
Article in English | MEDLINE | ID: mdl-21354995

ABSTRACT

AIMS: Angiotensin II (Ang II) type AT(1) receptors expressed on vascular smooth muscle cells (VSMCs) couple to the Jak2 signalling pathway. However, the importance of this tissue-specific coupling is poorly understood. The purpose of this investigation was to determine the importance of VSMC-derived Jak2 in angiotensin II-mediated hypertension. METHODS AND RESULTS: The Cre-loxP system was used to conditionally eliminate Jak2 tyrosine kinase expression within the smooth muscle cells of mice. Following chronic Ang II infusion, the resulting increase in mean arterial pressure (MAP) was significantly attenuated in the Jak2 null mice when compared with littermate controls. The VSMC Jak2 null mice were also protected from the Ang II-induced vascular remodelling. Aortic rings from the VSMC Jak2 null mice exhibited reduced Ang II-induced contraction and enhanced endothelial-dependent relaxation via increased nitric oxide (NO) bioavailability. When compared with controls, the VSMC Jak2 nulls also had lower levels of hydrogen peroxide, Rho kinase activity, and intracellular Ca(2+) in response to Ang II. CONCLUSIONS: The data indicate that VSMC Jak2 expression is involved in the pathogenesis of Ang II-dependent hypertension due to the increased presence of reactive oxygen species (ROS). As such, VSMC-derived Jak2 tyrosine kinase modulates overall vascular tone via multiple, non-redundant mechanisms.


Subject(s)
Angiotensin II , Hypertension/enzymology , Janus Kinase 2/metabolism , Muscle, Smooth, Vascular/enzymology , Oxidative Stress , Reactive Oxygen Species/metabolism , Vasoconstriction , Animals , Aorta/enzymology , Aorta/physiopathology , Blood Pressure , Calcium/metabolism , Disease Models, Animal , Dose-Response Relationship, Drug , Endothelial Cells/metabolism , Hydrogen Peroxide/metabolism , Hypertension/chemically induced , Hypertension/genetics , Hypertension/physiopathology , Hypertension/prevention & control , Janus Kinase 2/deficiency , Janus Kinase 2/genetics , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiopathology , Nitric Oxide/metabolism , Time Factors , Up-Regulation , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation , Vasodilator Agents/pharmacology , rho-Associated Kinases/metabolism
16.
J Neurosci ; 31(1): 184-92, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21209203

ABSTRACT

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.


Subject(s)
Down-Regulation/physiology , Gonadotropin-Releasing Hormone/metabolism , Janus Kinase 2/physiology , Reproduction/physiology , Animals , Cell Count/methods , Down-Regulation/drug effects , Down-Regulation/genetics , Estrous Cycle/genetics , Exons/genetics , Female , Fertility/genetics , Gonadotropin-Releasing Hormone/genetics , Gonadotropin-Releasing Hormone/pharmacology , Green Fluorescent Proteins/genetics , Hypothalamus/cytology , Janus Kinase 2/deficiency , Luteinizing Hormone/blood , Mice , Mice, Knockout , Neurons/metabolism , Ovariectomy , Ovary/pathology , Proto-Oncogene Proteins c-fos/metabolism , Puberty, Delayed/genetics , RNA, Messenger/metabolism , Reproduction/drug effects , Reproduction/genetics
17.
Mol Cell Biol ; 30(12): 2957-70, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20385773

ABSTRACT

The signal transducer and activator of transcription 5 (Stat5) plays a pivotal role in the proliferation, secretory differentiation, and survival of mammary epithelial cells. However, there is little information about Stat5 target genes that facilitate these biological processes. We provide here experimental evidence that the prolactin-mediated phosphorylation of Stat5 regulates the transcriptional activation of the Akt1 gene. Stat5 binds to consensus sequences within the Akt1 locus in a growth factor-dependent manner to initiate transcription of a unique Akt1 mRNA from a distinct promoter, which is only active in the mammary gland. Elevating the levels of active Akt1 restores the expression of cyclin D1 and proliferation of Jak2-deficient mammary epithelial cells, which provides evidence that Akt1 acts downstream of Jak/Stat signaling. The ligand-inducible expression of Stat5 in transgenic females mediates a sustained upregulation of Akt1 in mammary epithelial cells during the onset of postlactational involution. Stat5-expressing mammary glands exhibit a delay in involution despite induction of proapoptotic signaling events. Collectively, the results of the present study elucidate an underlying mechanism by which active Stat5 mediates evasion from apoptosis and self-sufficiency in growth signals.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/enzymology , Mammary Glands, Animal/cytology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/genetics , STAT5 Transcription Factor/metabolism , Transcriptional Activation/genetics , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Consensus Sequence , Cyclin D1/metabolism , Doxycycline/pharmacology , Epithelial Cells/drug effects , Female , Gene Expression Regulation, Enzymologic/drug effects , Janus Kinase 2/deficiency , Janus Kinase 2/metabolism , Lactation/drug effects , Lactation/genetics , Mice , Models, Biological , Organ Specificity/drug effects , Organ Specificity/genetics , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Prolactin/metabolism , Signal Transduction/drug effects , Transcriptional Activation/drug effects
18.
Blood ; 114(14): 3127-30, 2009 Oct 01.
Article in English | MEDLINE | ID: mdl-19628707

ABSTRACT

Two putative types of circulating endothelial progenitor cells have been recently identified in vitro: (1) endothelial colony-forming cell (ECFC) and (2) colony-forming unit-endothelial cell (CFU-EC). Only the former is now recognized to belong to endothelial lineage. We have used the ECFC and CFU-EC assays to readdress the issue of the clonal relation between endothelial progenitor cells and hematopoietic stem cells in patients with Philadelphia-positive and Philadelphia-negative chronic myeloproliferative disorders. Both ECFCs and CFU-ECs were cultured from peripheral blood mononuclear cells, and either BCR-ABL rearrangement or JAK2-V617F mutation were assessed in both types of endothelial colonies. We found that ECFCs lack the disease-specific markers, which are otherwise present in CFU-ECs, thus reinforcing the concept that the latter belongs to the hematopoietic lineage, and showing that in chronic myeloproliferative disorders the cell that gives rise to circulating ECFC has a distinct origin from the cell of the hematopoietic malignant clone.


Subject(s)
Biomarkers, Tumor/genetics , Endothelial Cells/pathology , Fusion Proteins, bcr-abl/deficiency , Hematopoietic Stem Cells/pathology , Janus Kinase 2/deficiency , Myeloproliferative Disorders/genetics , Stem Cells/pathology , Adult , Aged , Cells, Cultured , Chronic Disease , Colony-Forming Units Assay , Female , Fusion Proteins, bcr-abl/genetics , Gene Rearrangement , Humans , Janus Kinase 2/genetics , Male , Middle Aged , Mutation/genetics , Myeloproliferative Disorders/metabolism , Myeloproliferative Disorders/pathology
19.
Haematologica ; 93(11): 1723-7, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18728027

ABSTRACT

We analyzed the effect of hydroxyurea on the JAK2V617F allelic ratio (%JAK2V617F), measured in purified blood granulocytes, of patients with polycythemia vera and essential thrombocythemia. Thirty-six patients were examined sequentially prior to and after start of hydroxy-urea therapy (8 polycythemia vera, 17 essential thrombocythemia), or while remaining untreated (2 polycythemia vera, 9 essential thrombocythemia). Hydroxyurea therapy (median duration: 15 months) reduced the %JAK2V617F by >30% in 13/25 patients (4 polycythemia vera, 9 essential thrombocythemia). For 3 patients, JAK2V617F remained undetectable for 3-27 months. In addition, a single time point study of two large cohorts of patients, examined either at the time of diagnosis (99 polycythemia vera, 178 essential thrombocythemia) or while receiving hydroxyurea (36 polycythemia vera, 98 essential thrombocythemia; median length of therapy: 32 months), confirmed reduction of %JAK2V617F in the hydroxyurea-treated group (24% vs. 33% JAK2V617F at diagnosis, p<0.01). Prospective studies are needed to determine the prognostic value of reduced JAK2V617F allele burden under cytoreductive therapy.


Subject(s)
Hydroxyurea/therapeutic use , Janus Kinase 2/genetics , Polycythemia Vera/drug therapy , Thrombocythemia, Essential/drug therapy , Aged , Aged, 80 and over , Amino Acid Substitution , Antisickling Agents/therapeutic use , Cohort Studies , DNA Primers , Female , Granulocytes/physiology , Humans , Janus Kinase 2/blood , Janus Kinase 2/deficiency , Male , Middle Aged , Mutation , Polycythemia Vera/genetics , Retrospective Studies , Thrombocythemia, Essential/genetics
20.
Leuk Res ; 32(12): 1931-5, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18555525

ABSTRACT

Atypical chronic myeloid leukemia (aCML) as defined by the WHO classification is a rare hematopoietic stem cell disorder, which shows both myeloproliferative as well as myelodysplastic features. Because of the presence of neutrophilic leukocytosis, aCML may resemble chronic myelogenous leukemia. However, in contrast with the latter, aCML lacks a Philadelphia chromosome or the BCR/ABL fusion gene. The molecular pathogenesis of aCML and its relationship to other myeloproliferative neoplasms is unknown. To clarify these points, the presence of JAK2 V617F was examined by a retrospective analysis of archival specimens obtained from two large medical institutions. Paraffin-embedded bone marrow (BM) trephines and clot sections were examined by an allele-specific TaqMan PCR suitable for use with decalcified tissue. Fifty-nine cases of Philadelphia (Ph) chromosome negative chronic myeloproliferative neoplasms (CMPN) and normal bone marrows (BM) served as controls. None of the nine amplifiable cases of aCML and none of the normal BM controls showed a JAK2 V617F mutation, in contrast to 45/59 (76%) of the Ph chromosome negative CMPN cases. Atypical CML should therefore be considered as a JAK2 negative chronic myeloid neoplasm that remains properly categorized, alongside chronic myelomonocytic leukemia and juvenile myelomonocytic leukemia, within the WHO group of myelodysplastic/myeloproliferative neoplasms.


Subject(s)
Amino Acid Substitution , Janus Kinase 2/genetics , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/classification , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/genetics , Blast Crisis/blood , Blast Crisis/pathology , Blood Cell Count , Fusion Proteins, bcr-abl/genetics , Humans , Janus Kinase 2/deficiency , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/blood , Leukemia, Myeloid, Chronic, Atypical, BCR-ABL Negative/pathology , Mastocytosis/pathology , Neutrophils/pathology , Philadelphia Chromosome , World Health Organization
SELECTION OF CITATIONS
SEARCH DETAIL
...