Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 57
1.
Circ Res ; 134(10): e93-e111, 2024 May 10.
Article En | MEDLINE | ID: mdl-38563147

BACKGROUND: Endothelial activation promotes the release of procoagulant extracellular vesicles and inflammatory mediators from specialized storage granules. Endothelial membrane exocytosis is controlled by phosphorylation. We hypothesized that the absence of PTP1B (protein tyrosine phosphatase 1B) in endothelial cells promotes venous thromboinflammation by triggering endothelial membrane fusion and exocytosis. METHODS: Mice with inducible endothelial deletion of PTP1B (End.PTP1B-KO) underwent inferior vena cava ligation to induce stenosis and venous thrombosis. Primary endothelial cells from transgenic mice and human umbilical vein endothelial cells were used for mechanistic studies. RESULTS: Vascular ultrasound and histology showed significantly larger venous thrombi containing higher numbers of Ly6G (lymphocyte antigen 6 family member G)-positive neutrophils in mice with endothelial PTP1B deletion, and intravital microscopy confirmed the more pronounced neutrophil recruitment following inferior vena cava ligation. RT2 PCR profiler array and immunocytochemistry analysis revealed increased endothelial activation and adhesion molecule expression in primary End.PTP1B-KO endothelial cells, including CD62P (P-selectin) and VWF (von Willebrand factor). Pretreatment with the NF-κB (nuclear factor kappa B) kinase inhibitor BAY11-7082, antibodies neutralizing CD162 (P-selectin glycoprotein ligand-1) or VWF, or arginylglycylaspartic acid integrin-blocking peptides abolished the neutrophil adhesion to End.PTP1B-KO endothelial cells in vitro. Circulating levels of annexin V+ procoagulant endothelial CD62E+ (E-selectin) and neutrophil (Ly6G+) extracellular vesicles were also elevated in End.PTP1B-KO mice after inferior vena cava ligation. Higher plasma MPO (myeloperoxidase) and Cit-H3 (citrullinated histone-3) levels and neutrophil elastase activity indicated neutrophil activation and extracellular trap formation. Infusion of End.PTP1B-KO extracellular vesicles into C57BL/6J wild-type mice most prominently enhanced the recruitment of endogenous neutrophils, and this response was blunted in VWF-deficient mice or by VWF-blocking antibodies. Reduced PTP1B binding and tyrosine dephosphorylation of SNAP23 (synaptosome-associated protein 23) resulting in increased VWF exocytosis and neutrophil adhesion were identified as mechanisms, all of which could be restored by NF-κB kinase inhibition using BAY11-7082. CONCLUSIONS: Our findings show that endothelial PTP1B deletion promotes venous thromboinflammation by enhancing SNAP23 phosphorylation, endothelial VWF exocytosis, and neutrophil recruitment.


Exocytosis , Mice, Knockout , Protein Tyrosine Phosphatase, Non-Receptor Type 1 , Venous Thrombosis , von Willebrand Factor , Animals , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Humans , Mice , von Willebrand Factor/metabolism , von Willebrand Factor/genetics , Venous Thrombosis/metabolism , Venous Thrombosis/genetics , Venous Thrombosis/pathology , Human Umbilical Vein Endothelial Cells/metabolism , Inflammation/metabolism , Inflammation/genetics , Mice, Inbred C57BL , Neutrophils/metabolism , Endothelial Cells/metabolism , Cells, Cultured , Vena Cava, Inferior/metabolism , Vena Cava, Inferior/pathology , Male , Neutrophil Infiltration , NF-kappa B/metabolism
2.
Endocrinology ; 163(2)2022 02 01.
Article En | MEDLINE | ID: mdl-34967909

Reproductive dysfunction in women has been linked to high caloric diet (HCD)-feeding and obesity. Central resistance to leptin and insulin have been shown to accompany diet-induced infertility in rodent studies, and we have previously shown that deleting suppressor of cytokine signaling 3, which is a negative regulator of leptin signaling, from all forebrain neurons partially protects mice from HCD-induced infertility. In this study, we were interested in exploring the role of protein tyrosine phosphatase 1B (PTP1B), which is a negative regulator of both leptin and insulin signaling, in the pathophysiology of HCD-induced obesity and infertility. To this end, we generated male and female neuron-specific PTP1B knockout mice and compared their body weight gain, food intake, glucose tolerance, and fertility relative to control littermates under both normal calorie diet and HCD feeding conditions. Both male and female mice with neuronal PTP1B deletion exhibited slower body weight gain in response to HCD feeding, yet only male knockout mice exhibited improved glucose tolerance compared with controls. Neuronal PTP1B deletion improved the time to first litter in HCD-fed mice but did not protect female mice from eventual HCD-induced infertility. While the mice fed a normal caloric diet remained fertile throughout the 150-day period of assessment, HCD-fed females became infertile after producing only a single litter, regardless of their genotype. These data show that neuronal PTP1B deletion is able to partially protect mice from HCD-induced obesity but is not a critical mediator of HCD-induced infertility.


Brain/enzymology , Infertility, Female/prevention & control , Neurons/enzymology , Obesity/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/physiology , Animals , Crosses, Genetic , Energy Intake , Female , Infertility, Female/etiology , Male , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , Obesity/enzymology , Obesity/etiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Sexual Maturation
3.
Biochem Biophys Res Commun ; 533(3): 296-303, 2020 12 10.
Article En | MEDLINE | ID: mdl-32958258

Pseudomonas aeruginosa is the main conditional pathogen of immunodeficiency individuals. The mechanisms governing immune response to P. aeruginosa infection by macrophages remain incompletely defined. Herein, we demonstrate that protein tyrosine phosphatase-1B (PTP1B) is a critical negative regulator of P. aeruginosa infection response by macrophages. PTP1B-deficient macrophages display greatly enhanced bacterial phagocytosis and killing, accompanied by increased lysosome formation during P. aeruginosa infection. We also found that PTP1B repressed nitric oxide (NO) production and nitric oxide synthase (iNOS) induction following P. aeruginosa infection. PTP1B deficiency tended to upregulate the production of TRIF-interferon (IFN) pathway cytokines and chemokines, including IFN-ß and interferon γ-inducible protein 10 (CXCL10, IP-10). Unexpectedly, the phosphorylation level of STAT1 was not regulated by PTP1B. In vivo experiments also confirmed that the regulatory function of PTP1B was not dependent on STAT1. These findings demonstrate that STAT1 is dispensable for negative regulation of P. aeruginosa clearance by macrophages.


Host-Pathogen Interactions/genetics , Macrophages/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Pseudomonas Infections/genetics , Pseudomonas aeruginosa/immunology , STAT1 Transcription Factor/genetics , Animals , Chemokine CXCL10/genetics , Chemokine CXCL10/immunology , Chemokine CXCL2/genetics , Chemokine CXCL2/immunology , Gene Expression Regulation , Host-Pathogen Interactions/immunology , Humans , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-1beta/genetics , Interleukin-1beta/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Macrophages/microbiology , Mice , Mice, Knockout , Nitric Oxide/immunology , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/genetics , Nitric Oxide Synthase Type II/immunology , Phagocytosis , Primary Cell Culture , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/immunology , Pseudomonas Infections/immunology , Pseudomonas Infections/microbiology , Pseudomonas aeruginosa/pathogenicity , STAT1 Transcription Factor/immunology , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
4.
Mol Metab ; 35: 100954, 2020 05.
Article En | MEDLINE | ID: mdl-32244182

OBJECTIVE: Non-alcoholic steatohepatitis (NASH) is characterized by a robust pro-inflammatory component at both hepatic and systemic levels together with a disease-specific gut microbiome signature. Protein tyrosine phosphatase 1 B (PTP1B) plays distinct roles in non-immune and immune cells, in the latter inhibiting pro-inflammatory signaling cascades. In this study, we have explored the role of PTP1B in the composition of gut microbiota and gut barrier dynamics in methionine and choline-deficient (MCD) diet-induced NASH in mice. METHODS: Gut features and barrier permeability were characterized in wild-type (PTP1B WT) and PTP1B-deficient knockout (PTP1B KO) mice fed a chow or methionine/choline-deficient (MCD) diet for 4 weeks. The impact of inflammation was studied in intestinal epithelial and enteroendocrine cells. The secretion of GLP-1 was evaluated in primary colonic cultures and plasma of mice. RESULTS: We found that a shift in the gut microbiota shape, disruption of gut barrier function, higher levels of serum bile acids, and decreased circulating glucagon-like peptide (GLP)-1 are features during NASH. Surprisingly, despite the pro-inflammatory phenotype of global PTP1B-deficient mice, they were partly protected against the alterations in gut microbiota composition during NASH and presented better gut barrier integrity and less permeability under this pathological condition. These effects concurred with higher colonic mucosal inflammation, decreased serum bile acids, and protection against the decrease in circulating GLP-1 levels during NASH compared with their WT counterparts together with increased expression of GLP-2-sensitive genes in the gut. At the molecular level, stimulation of enteroendocrine STC-1 cells with a pro-inflammatory conditioned medium (CM) from lipopolysaccharide (LPS)-stimulated macrophages triggered pro-inflammatory signaling cascades that were further exacerbated by a PTP1B inhibitor. Likewise, the pro-inflammatory CM induced GLP-1 secretion in primary colonic cultures, an effect augmented by PTP1B inhibition. CONCLUSION: Altogether our results have unraveled a potential role of PTP1B in the gut-liver axis during NASH, likely mediated by increased sensitivity to GLPs, with potential therapeutic value.


Gastrointestinal Microbiome/genetics , Intestinal Mucosa/metabolism , Non-alcoholic Fatty Liver Disease/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Animals , Choline Deficiency/complications , Diet/adverse effects , Disease Models, Animal , Gene Expression , Gene Knockout Techniques , Glucagon-Like Peptide 1/blood , Inflammation/metabolism , Liver/metabolism , Male , Methionine/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/etiology , Permeability , RAW 264.7 Cells
5.
Redox Biol ; 26: 101263, 2019 09.
Article En | MEDLINE | ID: mdl-31299613

Inflammation is typically associated with the development of fibrosis, cirrhosis and hepatocellular carcinoma. The key role of protein tyrosine phosphatase 1B (PTP1B) in inflammatory responses has focused this study in understanding its implication in liver fibrosis. Here we show that hepatic PTP1B mRNA expression increased after bile duct ligation (BDL), while BDL-induced liver fibrosis was markedly reduced in mice lacking Ptpn1 (PTP1B-/-) as assessed by decreased collagen deposition and α-smooth muscle actin (α-SMA) expression. PTP1B-/- mice also showed a significant increase in mRNA levels of key markers of monocytes recruitment (Cd68, Adgre1 and Ccl2) compared to their wild-type (PTP1B+/+) littermates at early stages of injury after BDL. Interestingly, the lack of PTP1B strongly increased the NADPH oxidase (NOX) subunits Nox1/Nox4 ratio and downregulated Cybb expression after BDL, revealing a pro-survival pattern of NADPH oxidase induction in response to liver injury. Chimeric mice generated by transplantation of PTP1B-/- bone marrow (BM) into irradiated PTP1B+/+ mice revealed similar hepatic expression profile of NOX subunits than PTP1B-/- mice while these animals did not show differences in infiltration of myeloid cells at 7 days post-BDL, suggesting that PTP1B deletion in other liver cells is necessary for boosting the early inflammatory response to the BDL. PTP1B-/- BM transplantation into PTP1B+/+ mice also led to a blockade of TGF-ß and α-SMA induction after BDL. In vitro experiments demonstrated that deficiency of PTP1B in hepatocytes protects against bile acid-induced apoptosis and abrogates hepatic stellate cells (HSC) activation, an effect ameliorated by NOX1 inhibition. In conclusion, our results have revealed that the lack of PTP1B switches NOX expression pattern in response to liver injury after BDL and reduces HSC activation and liver fibrosis.


Liver Cirrhosis/etiology , Liver Cirrhosis/metabolism , NADPH Oxidases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Animals , Apoptosis/genetics , Bile Acids and Salts/metabolism , Biomarkers , Cell Line , Culture Media, Conditioned/metabolism , Culture Media, Conditioned/pharmacology , Disease Models, Animal , Female , Gene Expression , Hepatic Stellate Cells/metabolism , Hepatocytes/metabolism , Immunohistochemistry , Kupffer Cells/metabolism , Liver Cirrhosis/pathology , Male , Mice , NADPH Oxidases/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , RNA, Small Interfering/genetics , Reactive Oxygen Species/metabolism , Transforming Growth Factor beta/metabolism
6.
Vascul Pharmacol ; 109: 36-44, 2018 10.
Article En | MEDLINE | ID: mdl-29894845

Protein tyrosine phosphatase 1B (PTP1B) impairs nitric oxide (NO) production and induces endothelial dysfunction in various diseases, including diabetes, septic shock and heart failure. In non-cardiovascular tissues, PTP1B modulates endoplasmic reticulum stress (ERS) however this role has never been assessed in endothelial cells. We evaluated the link between PTP1B, ERS and endothelial dysfunction in mice. Induction of ERS (Tunicamycin) in vivo in mice or ex vivo in mouse arteries led to severe arterial endothelial dysfunction (i.e. reduced flow-dependent, NO mediated dilatation in isolated small mesenteric arteries), and this was prevented by the PTP1B inhibitor trodusquemine and absent in PTP1B-/- mice. Trodusquemine also prevented the Tunicamycin -induced increased arterial levels of the molecular ERS actors 78 kDa glucose-regulated protein (GRP78) and Activating Transcription Factor 6 (ATF6α). Tunicamycin strongly increased the interactions of PTP1B with GRP78 and the activated forms of protein kinase RNA-like endoplasmic reticulum kinase (PERK) and IRE1α (proximity Ligation Assay). Thus, PTP1B plays a central role in the regulation of ERS in the endothelium, and the endothelial protective effect of PTP1B inhibition appears likely due at least in part to reduction of endothelial ERS, notably by promoting PERK protective pathway. Modulation of ER stress via PTP1B inhibitors may be a promising approach to protect the endothelium in cardiovascular diseases.


Endoplasmic Reticulum Stress/drug effects , Endothelium, Vascular/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Tunicamycin/pharmacology , Vasodilation/drug effects , Activating Transcription Factor 6/metabolism , Animals , Cholestanes/pharmacology , Endoplasmic Reticulum Chaperone BiP , Endoribonucleases/metabolism , Endothelium, Vascular/enzymology , Endothelium, Vascular/physiopathology , Enzyme Inhibitors/pharmacology , Heat-Shock Proteins/metabolism , Male , Mice, Inbred BALB C , Mice, Knockout , Nitric Oxide/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Signal Transduction/drug effects , Spermine/analogs & derivatives , Spermine/pharmacology , eIF-2 Kinase/metabolism
7.
Redox Biol ; 17: 213-223, 2018 07.
Article En | MEDLINE | ID: mdl-29705509

Protein tyrosine phosphatase 1B (PTP1B) is widely expressed in mammalian tissues, in particular in immune cells, and plays a pleiotropic role in dephosphorylating many substrates. Moreover, PTP1B expression is enhanced in response to pro-inflammatory stimuli and to different cell stressors. Taking advantage of the use of mice deficient in PTP1B we have investigated the effect of γ-radiation in these animals and found enhanced lethality and decreased respiratory exchange ratio vs. the corresponding wild type animals. Using bone-marrow derived macrophages and mouse embryonic fibroblasts (MEFs) from wild-type and PTP1B-deficient mice, we observed a differential response to various cell stressors. PTP1B-deficient macrophages exhibited an enhanced response to γ-radiation, UV-light, LPS and S-nitroso-glutathione. Macrophages exposed to γ-radiation show DNA damage and fragmentation, increased ROS production, a lack in GSH elevation and enhanced acidic ß-galactosidase activity. Interestingly, these differences were not observed in MEFs. Differential gene expression analysis of WT and KO macrophages revealed that the main pathways affected after irradiation were an up-regulation of protein secretion, TGF-ß signaling and angiogenesis among other, and downregulation of Myc targets and Hedgehog signaling. These results demonstrate a key role for PTP1B in the protection against the cytotoxicity of irradiation in intact animal and in macrophages, which might be therapeutically relevant.


Cell Proliferation/radiation effects , Macrophages/radiation effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Radiation Injuries/genetics , Animals , DNA Damage/radiation effects , Fibroblasts/radiation effects , Gamma Rays/adverse effects , Gene Expression Regulation/radiation effects , Glutathione/genetics , Glutathione/metabolism , Mice , Mice, Knockout , Phosphorylation/radiation effects , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , RNA Interference , Radiation Injuries/pathology , Radiation Injuries/prevention & control , Reactive Oxygen Species/metabolism , beta-Galactosidase/genetics
8.
Am J Physiol Heart Circ Physiol ; 314(6): H1279-H1288, 2018 06 01.
Article En | MEDLINE | ID: mdl-29569957

We have previously shown that protein tyrosine phosphatase 1B (PTP1B) inactivation in mice [PTP1B-deficient (PTP1B-/-) mice] improves left ventricular (LV) angiogenesis, perfusion, remodeling, and function and limits endothelial dysfunction after myocardial infarction. However, whether PTP1B inactivation slows aging-associated cardiovascular dysfunction remains unknown. Wild-type (WT) and PTP1B-/- mice were allowed to age until 18 mo. Compared with old WT mice, in which aging increased the LV mRNA expression of PTP1B, old PTP1B-/- mice had 1) reduced cardiac hypertrophy with decreased LV mRNA levels of hypertrophic markers and atrial and brain natriuretic peptides, 2) lower LV fibrosis (collagen: 16 ± 3% in WT mice and 5 ± 3% in PTP1B-/- mice, P < 0.001) with decreased mRNA levels of transforming growth-factor-ß1 and matrix metalloproteinase-2, and 3) higher LV capillary density and lower LV mRNA level of hypoxic inducible factor-1α, which was associated over time with a higher rate of proangiogenic M2 type macrophages and a stable LV mRNA level of VEGF receptor-2. Echocardiography revealed an age-dependent LV increase in end-diastolic volume in WT mice together with alterations of fractional shortening and diastole (transmitral Doppler E-to-A wave ratio). Invasive hemodynamics showed better LV systolic contractility and better diastolic compliance in old PTP1B-/- mice (LV end-systolic pressure-volume relation: 13.9 ± 0.9 in WT mice and 18.4 ± 1.6 in PTP1B-/- mice; LV end-diastolic pressure-volume relation: 5.1 ± 0.8 mmHg/relative volume unit in WT mice and 1.2 ± 0.3 mmHg/relative volume unit in PTP1B-/- mice, P < 0.05). In addition, old PTP1B-/- mice displayed a reduced amount of LV reactive oxygen species. Finally, in isolated resistance mesenteric arteries, PTP1B inactivation reduced aging-associated endothelial dysfunction (flow-mediated dilatation: -0.4 ± 2.1% in WT mice and 8.2 ± 2.8% in PTP1B-/- mice, P < 0.05). We conclude that PTP1B inactivation slows aging-associated LV remodeling and dysfunction and reduces endothelial dysfunction in mesenteric arteries. NEW & NOTEWORTHY The present study shows that protein tyrosine phosphatase 1B inactivation in aged mice improves left ventricular systolic and diastolic function associated with reduced adverse cardiac remodeling (hypertrophy, fibrosis, and capillary rarefaction) and limits vascular endothelial dysfunction. This suggests that protein tyrosine phosphatase 1B inhibition could be an interesting treatment approach in age-related cardiovascular dysfunction.


Heart Failure/prevention & control , Heart Ventricles/enzymology , Hypertrophy, Left Ventricular/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left , Ventricular Remodeling , Age Factors , Aging/genetics , Aging/metabolism , Animals , Disease Models, Animal , Fibrosis , Gene Expression Regulation, Enzymologic , Heart Failure/enzymology , Heart Failure/genetics , Heart Failure/physiopathology , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Hemodynamics , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/genetics , Hypertrophy, Left Ventricular/physiopathology , Male , Mesenteric Arteries/enzymology , Mesenteric Arteries/physiopathology , Mice, Inbred BALB C , Mice, Knockout , Neovascularization, Physiologic , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology
9.
Am J Physiol Regul Integr Comp Physiol ; 314(3): R478-R488, 2018 03 01.
Article En | MEDLINE | ID: mdl-29351427

Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of leptin receptor signaling and may contribute to leptin resistance in diet-induced obesity. Although PTP1B inhibition has been suggested as a potential weight loss therapy, the role of specific neuronal PTP1B signaling in cardiovascular and metabolic regulation and the importance of sex differences in this regulation are still unclear. In this study, we investigated the impact of proopiomelanocortin (POMC) neuronal PTP1B deficiency in cardiometabolic regulation in male and female mice fed a high-fat diet (HFD). When compared with control mice (PTP1B flox/flox), male and female mice deficient in POMC neuronal PTP1B (PTP1B flox/flox/POMC-Cre) had attenuated body weight gain (males: -18%; females: -16%) and fat mass (males: -33%; female: -29%) in response to HFD. Glucose tolerance was improved by 40%, and liver lipid accumulation was reduced by 40% in PTP1B/POMC-Cre males but not in females. When compared with control mice, deficiency of POMC neuronal PTP1B did not alter mean arterial pressure (MAP) in male or female mice (males: 112 ± 1 vs. 112 ± 1 mmHg in controls; females: 106 ± 3 vs. 109 ± 3 mmHg in controls). Deficiency of POMC neuronal PTP1B also did not alter MAP response to acute stress in males or females compared with control mice (males: Δ32 ± 0 vs. Δ29 ± 4 mmHg; females: Δ22 ± 2 vs. Δ27 ± 4 mmHg). These data demonstrate that POMC-specific PTP1B deficiency improved glucose tolerance and attenuated diet-induced fatty liver only in male mice and attenuated weight gain in males and females but did not enhance the MAP and HR responses to a HFD or to acute stress.


Arcuate Nucleus of Hypothalamus/enzymology , Blood Glucose/metabolism , Glucose Intolerance/enzymology , Lipid Metabolism , Liver/metabolism , Neurons/enzymology , Non-alcoholic Fatty Liver Disease/enzymology , Obesity/enzymology , Pro-Opiomelanocortin/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Solitary Nucleus/enzymology , Animals , Arcuate Nucleus of Hypothalamus/physiopathology , Biomarkers/blood , Diet, High-Fat , Disease Models, Animal , Female , Glucose Intolerance/blood , Glucose Intolerance/physiopathology , Glucose Intolerance/prevention & control , Liver/pathology , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Non-alcoholic Fatty Liver Disease/blood , Non-alcoholic Fatty Liver Disease/physiopathology , Non-alcoholic Fatty Liver Disease/prevention & control , Obesity/etiology , Obesity/physiopathology , Obesity/prevention & control , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Sex Factors , Solitary Nucleus/physiopathology , Weight Gain
10.
Biochim Biophys Acta Mol Basis Dis ; 1863(12): 3060-3074, 2017 12.
Article En | MEDLINE | ID: mdl-28941626

Endoplasmic reticulum (ER) stress has been demonstrated to prompt various cardiovascular risks although the underlying mechanism remains elusive. Protein tyrosine phosphatase-1B (PTP1B) serves as an essential negative regulator for insulin signaling. This study examined the role of PTP1B in ER stress-induced myocardial anomalies and underlying mechanism involved with a focus on autophagy. WT and PTP1B knockout mice were subjected to the ER stress inducer tunicamycin (1mg/kg). Cardiac function was evaluated with echocardiography and an Ion-Optix MyoCam system. Western blot analysis was used to monitor the levels of ER stress, autophagy and insulin signaling including insulin receptor substrate (IRS), tribbles homolog 3 (TRIB3), Atg5/7, p62 and LC3-II. Our results showed that ER stress resulted in compromised echocardiographic and cardiomyocyte contractile function, intracellular Ca2+ mishandling, ER stress, O2- production, apoptosis, the effects of which (with the exception of ER stress) were significantly attenuated or negated by PTP1B ablation. Levels of serine phosphorylation of IRS-1, TRIB3, Atg5/7, LC3B and the autophagy adaptor p62 were significantly upregulated while IRS-1 tyrosine phosphorylation was reduced by tunicamycin, the effect of which were obliterated by PTP1B ablation. In vitro study revealed that the autophagy inducer rapamycin and TRIB3 overexpression cancelled PTP1B ablation-offered beneficial effects on cardiomyocyte function or O2- production in murine cardiomyocytes or H9C2 myoblasts. Antioxidant or gene silencing of TRIB3 mimicked PTP1B ablation-induced protective effects. These findings collectively suggested that PTP1B ablation protects against ER stress-induced cardiac anomalies through regulation of autophagy.


Cardiomyopathies/enzymology , Endoplasmic Reticulum Stress/physiology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Animals , Apoptosis/drug effects , Autophagy/drug effects , Autophagy/physiology , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 7/metabolism , Calcium/metabolism , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Cell Cycle Proteins/metabolism , Cell Line , Endoplasmic Reticulum Stress/drug effects , Gene Knockout Techniques , Insulin Receptor Substrate Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/metabolism , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Oxygen/metabolism , Peptides/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Signal Transduction , Sirolimus/pharmacology , Tunicamycin/pharmacology , Tyrosine/metabolism
11.
Mol Metab ; 6(8): 845-853, 2017 08.
Article En | MEDLINE | ID: mdl-28752048

OBJECTIVE: Cardiovascular disease (CVD) is the most prevalent cause of mortality among patients with Type 1 or Type 2 diabetes, due to accelerated atherosclerosis. Recent evidence suggests a strong link between atherosclerosis and insulin resistance due to impaired insulin receptor (IR) signaling. Moreover, inflammatory cells, in particular macrophages, play a key role in pathogenesis of atherosclerosis and insulin resistance in humans. We hypothesized that inhibiting the activity of protein tyrosine phosphatase 1B (PTP1B), the major negative regulator of the IR, specifically in macrophages, would have beneficial anti-inflammatory effects and lead to protection against atherosclerosis and CVD. METHODS: We generated novel macrophage-specific PTP1B knockout mice on atherogenic background (ApoE-/-/LysM-PTP1B). Mice were fed standard or pro-atherogenic diet, and body weight, adiposity (echoMRI), glucose homeostasis, atherosclerotic plaque development, and molecular, biochemical and targeted lipidomic eicosanoid analyses were performed. RESULTS: Myeloid-PTP1B knockout mice on atherogenic background (ApoE-/-/LysM-PTP1B) exhibited a striking improvement in glucose homeostasis, decreased circulating lipids and decreased atherosclerotic plaque lesions, in the absence of body weight/adiposity differences. This was associated with enhanced phosphorylation of aortic Akt, AMPKα and increased secretion of circulating anti-inflammatory cytokine interleukin-10 (IL-10) and prostaglandin E2 (PGE2), without measurable alterations in IR phosphorylation, suggesting a direct beneficial effect of myeloid-PTP1B targeting. CONCLUSIONS: Here we demonstrate that inhibiting the activity of PTP1B specifically in myeloid lineage cells protects against atherosclerotic plaque formation, under atherogenic conditions, in an ApoE-/- mouse model of atherosclerosis. Our findings suggest for the first time that macrophage PTP1B targeting could be a therapeutic target for atherosclerosis treatment and reduction of CVD risk.


Atherosclerosis/metabolism , Macrophages/metabolism , Plaque, Atherosclerotic/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , AMP-Activated Protein Kinase Kinases , Animals , Apolipoproteins E/genetics , Atherosclerosis/pathology , Cells, Cultured , Dinoprostone/metabolism , Female , Interleukin-10/metabolism , Male , Mice , Protein Kinases/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
12.
Biochem Biophys Res Commun ; 488(1): 116-121, 2017 06 17.
Article En | MEDLINE | ID: mdl-28479249

Hypothalamic insulin receptor signaling regulates energy balance and glucose homeostasis via agouti-related protein (AgRP). While protein tyrosine phosphatase 1B (PTP1B) is classically known to be a negative regulator of peripheral insulin signaling by dephosphorylating both insulin receptor ß (IRß) and insulin receptor substrate, the role of PTP1B in hypothalamic insulin signaling remains to be fully elucidated. In the present study, we investigated the role of PTP1B in hypothalamic insulin signaling using PTP1B deficient (KO) mice in vivo and ex vivo. For the in vivo study, hypothalamic insulin resistance induced by a high-fat diet (HFD) improved in KO mice compared to wild-type (WT) mice. Hypothalamic AgRP mRNA expression levels were also significantly decreased in KO mice independent of body weight changes. In an ex vivo study using hypothalamic organotypic cultures, insulin treatment significantly increased the phosphorylation of both IRß and Akt in the hypothalamus of KO mice compared to WT mice, and also significantly decreased AgRP mRNA expression levels in KO mice. While incubation with inhibitors of phosphatidylinositol-3 kinase (PI3K) had no effect on basal levels of Akt phosphorylation, these suppressed insulin induction of Akt phosphorylation to almost basal levels in WT and KO mice. The inhibition of the PI3K-Akt pathway blocked the downregulation of AgRP mRNA expression in KO mice treated with insulin. These data suggest that PTP1B acts on the hypothalamic insulin signaling via the PI3K-Akt pathway. Together, our results suggest a deficiency of PTP1B improves hypothalamic insulin sensitivity resulting in the attenuation of AgRP mRNA expression under HFD conditions.


Agouti-Related Protein/genetics , Diet, High-Fat , Hypothalamus/metabolism , Insulin Resistance/genetics , Insulin/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , RNA, Messenger/genetics , Agouti-Related Protein/metabolism , Animals , Gene Expression Profiling , Insulin/blood , Mice , Mice, Knockout , RNA, Messenger/metabolism
13.
Shock ; 48(3): 355-363, 2017 09.
Article En | MEDLINE | ID: mdl-28272165

Hyperglycemia is a common feature of septic patients and has been associated with poor outcome and high mortality. In contrast, insulin has been shown to decrease mortality and to prevent the incidence of multiorgan failure but is often associated with deleterious hypoglycemia. Protein Tyrosine Phosphatase 1B (PTP1B) is a negative regulator of both insulin signaling and NO production, and has been shown to be an aggravating factor in septic shock. To evaluate the potential therapeutic effect of PTP1B blockade on glucose metabolism and insulin resistance in an experimental model of sepsis, we assessed the effect of PTP1B gene deletion in a cecal ligation and puncture (CLP) model of sepsis. PTP1B gene deletion significantly limited CLP-induced insulin resistance, improved AMP-activated protein kinase signaling pathway and Glucose Transporter 4 translocation, and decreased inflammation. These effects were associated with a reduction of sepsis-induced endothelial dysfunction/impaired NO production and especially of insulin-mediated dilatation. This modulation of insulin resistance may contribute to the beneficial effect of PTP1B blockade in septic shock, especially in terms of inflammation and cardiac metabolism.


Endothelium, Vascular/metabolism , Insulin Resistance , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Sepsis/metabolism , Sepsis/prevention & control , Signal Transduction , Vasodilation , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/metabolism , Animals , Cell Line , Disease Models, Animal , Endothelium, Vascular/pathology , Gene Deletion , Mice , Mice, Knockout , Nitric Oxide/genetics , Nitric Oxide/metabolism , Sepsis/genetics , Sepsis/pathology
14.
EBioMedicine ; 16: 172-183, 2017 Feb.
Article En | MEDLINE | ID: mdl-28094236

Protein tyrosine phosphatase 1B (PTP1B) regulates leptin signaling in hypothalamic neurons via the JAK2-STAT3 pathway. PTP1B has also been implicated in the regulation of inflammation in the periphery. However, the role of PTP1B in hypothalamic inflammation, which is induced by a high-fat diet (HFD), remains to be elucidated. Here, we showed that STAT3 phosphorylation (p-STAT3) was increased in microglia in the hypothalamic arcuate nucleus of PTP1B knock-out mice (KO) on a HFD, accompanied by decreased Tnf and increased Il10 mRNA expression in the hypothalamus compared to wild-type mice (WT). In hypothalamic organotypic cultures, incubation with TNFα led to increased p-STAT3, accompanied by decreased Tnf and increased Il10 mRNA expression, in KO compared to WT. Incubation with p-STAT3 inhibitors or microglial depletion eliminated the differences in inflammation between genotypes. These data indicate an important role of JAK2-STAT3 signaling negatively regulated by PTP1B in microglia, which attenuates hypothalamic inflammation under HFD conditions.


Hypothalamus/metabolism , Inflammation/metabolism , Janus Kinase 2/metabolism , Microglia/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , STAT3 Transcription Factor/metabolism , Animals , Blotting, Western , Diet, High-Fat/adverse effects , Enzyme Activation , Female , Gene Expression , Hypothalamus/pathology , Inflammation/etiology , Inflammation/genetics , Interleukin-10/genetics , Interleukin-10/metabolism , Male , Mice, Knockout , Microscopy, Confocal , Organ Culture Techniques , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
15.
Cardiovasc Res ; 111(3): 204-16, 2016 08 01.
Article En | MEDLINE | ID: mdl-27207947

AIMS: Cardiac angiogenesis is an important determinant of heart failure. We examined the hypothesis that protein tyrosine phosphatase (PTP)-1B, a negative regulator of vascular endothelial growth factor (VEGF) receptor-2 activation, is causally involved in the cardiac microvasculature rarefaction during hypertrophy and that deletion of PTP1B in endothelial cells prevents the development of heart failure. METHODS AND RESULTS: Cardiac hypertrophy was induced by transverse aortic constriction (TAC) in mice with endothelial-specific deletion of PTP1B (End.PTP1B-KO) and controls (End.PTP1B-WT). Survival up to 20 weeks after TAC was significantly improved in mice lacking endothelial PTP1B. Serial echocardiography revealed a better systolic pump function, less pronounced cardiac hypertrophy, and left ventricular dilation compared with End.PTP1B-WT controls. Histologically, banded hearts from End.PTP1B-KO mice exhibited increased numbers of PCNA-positive, proliferating endothelial cells resulting in preserved cardiac capillary density and improved perfusion as well as reduced hypoxia, apoptotic cell death, and fibrosis. Increased relative VEGFR2 and ERK1/2 phosphorylation and greater eNOS expression were present in the hearts of End.PTP1B-KO mice. The absence of PTP1B in endothelial cells also promoted neovascularization following peripheral ischaemia, and bone marrow transplantation excluded a major contribution of Tie2-positive haematopoietic cells to the improved angiogenesis in End.PTP1B-KO mice. Increased expression of caveolin-1 as well as reduced NADPH oxidase-4 expression, ROS generation and TGFß signalling were observed and may have mediated the cardioprotective effects of endothelial PTP1B deletion. CONCLUSIONS: Endothelial PTP1B deletion improves cardiac VEGF signalling and angiogenesis and protects against chronic afterload-induced heart failure. PTP1B may represent a useful target to preserve cardiac function during hypertrophy.


Endothelial Cells/enzymology , Heart Failure/prevention & control , Ischemia/enzymology , Muscle, Skeletal/blood supply , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Animals , Aorta/physiopathology , Aorta/surgery , Apoptosis , Arterial Pressure , Caveolin 1/metabolism , Constriction , Disease Models, Animal , Endothelial Cells/pathology , Fibrosis , Genetic Predisposition to Disease , Heart Failure/enzymology , Heart Failure/etiology , Heart Failure/physiopathology , Hindlimb , Hypertrophy, Left Ventricular/enzymology , Hypertrophy, Left Ventricular/physiopathology , Hypertrophy, Left Ventricular/prevention & control , Ischemia/genetics , Ischemia/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , NADPH Oxidase 4 , NADPH Oxidases/metabolism , Neovascularization, Physiologic , Nitric Oxide Synthase Type III/metabolism , Phenotype , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Receptor, TIE-2/metabolism , Signal Transduction , Time Factors , Vascular Endothelial Growth Factor Receptor-2/metabolism , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/physiopathology , Ventricular Dysfunction, Left/prevention & control , Ventricular Function, Left , Ventricular Remodeling
16.
Circ Heart Fail ; 9(4): e002895, 2016 Apr.
Article En | MEDLINE | ID: mdl-27059805

BACKGROUND: Chronic heart failure (CHF) induces endothelial dysfunction in part because of decreased nitric oxide (NO(·)) production, but the direct link between endothelial dysfunction and aggravation of CHF is not directly established. We previously reported that increased NO production via inhibition of protein tyrosine phosphatase 1B (PTP1B) is associated with reduced cardiac dysfunction in CHF. Investigation of the role of endothelial PTP1B in these effects may provide direct evidence of the link between endothelial dysfunction and CHF. METHODS AND RESULTS: Endothelial deletion of PTP1B was obtained by crossing LoxP-PTP1B with Tie2-Cre mice. CHF was assessed 4 months after myocardial infarction. In some experiments, to exclude gene extinction in hematopoietic cells, Tie2-Cre/LoxP-PTP1B mice were lethally irradiated and reconstituted with bone marrow from wild-type mice, to obtain mouse with endothelial-specific deletion of PTP1B. Vascular function evaluated ex vivo in mesenteric arteries showed that in wild-type mice, CHF markedly impaired NO-dependent flow-mediated dilatation. CHF-induced endothelial dysfunction was less marked in endoPTP1B(-/-) mice, suggesting restored NO production. Echocardiographic, hemodynamic, and histological evaluations demonstrated that the selectively improved endothelial function was associated with reduced left ventricular dysfunction and remodeling, as well as increased survival, in the absence of signs of stimulated angiogenesis or increased cardiac perfusion. CONCLUSIONS: Prevention of endothelial dysfunction, by endothelial PTP1B deficiency, is sufficient to reduce cardiac dysfunction post myocardial infarction. Our results provide for the first time a direct demonstration that endothelial protection per se reduces CHF and further suggest a causal role for endothelial dysfunction in CHF development.


Endothelium, Vascular/enzymology , Heart Failure/prevention & control , Mesenteric Arteries/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Vasodilation , Ventricular Dysfunction, Left/prevention & control , Animals , Bone Marrow Transplantation , Chronic Disease , Disease Models, Animal , Endothelium, Vascular/physiopathology , Heart Failure/enzymology , Heart Failure/genetics , Heart Failure/physiopathology , Mesenteric Arteries/physiopathology , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/enzymology , Myocardial Infarction/genetics , Myocardial Infarction/physiopathology , Nitric Oxide/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Signal Transduction , Time Factors , Ventricular Dysfunction, Left/enzymology , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/physiopathology , Ventricular Function, Left , Ventricular Remodeling
17.
Am J Pathol ; 186(5): 1234-44, 2016 05.
Article En | MEDLINE | ID: mdl-27105736

Pseudomonas aeruginosa is a major opportunistic pathogen in immune-compromised individuals. Mechanisms governing immune responses to P. aeruginosa infection remain incompletely defined. Herein, we demonstrate that protein tyrosine phosphatase-1B (PTP1B) is a critical negative regulator in P. aeruginosa infection. PTP1B-deficient mice display greatly enhanced bacterial clearance and reduced disease scores, which are accompanied by increased neutrophil infiltration and cytokine production. Interestingly, PTP1B deficiency mainly up-regulates the production of interferon-stimulated response elements-regulated cytokines and chemokines, including chemokine ligand 5 (regulated on activation normal T cell expressed and secreted), CXCL10 (interferon γ-inducible protein 10), and interferon-ß production. Further studies reveal that PTP1B deficiency leads to increased interferon regulatory factor 7 (IRF7) expression and activation. These findings demonstrate a novel regulatory mechanism of the immune response to P. aeruginosa infection through PTP1B-IRF7 interaction. This novel PTP1B-IRF7-interferon-stimulated response elements pathway may have broader implications in Toll-like receptor-mediated innate immunity.


Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Pseudomonas Infections/enzymology , Pseudomonas aeruginosa/enzymology , Animals , Antibodies, Bacterial/immunology , Bronchoalveolar Lavage Fluid/immunology , Bronchoalveolar Lavage Fluid/microbiology , Chemokines/biosynthesis , Cytokines/biosynthesis , Dendritic Cells/immunology , In Vitro Techniques , Interferon Regulatory Factor-7/metabolism , Lung Diseases/enzymology , Lung Diseases/immunology , Lung Diseases/microbiology , Mice , NF-kappa B/immunology , Neutrophils/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/immunology , Pseudomonas Infections/immunology , Pseudomonas aeruginosa/immunology , Signal Transduction/immunology
18.
Mucosal Immunol ; 9(5): 1317-29, 2016 09.
Article En | MEDLINE | ID: mdl-26813343

Protein tyrosine phosphatase 1B (PTP1B) has anti-inflammatory potential but PTP1B responses are desensitized in the lung by prolonged cigarette smoke exposure. Here we investigate whether PTP1B expression affects lung disease severity during respiratory syncytial viral (RSV) exacerbations of chronic obstructive pulmonary disease (COPD). Ptp1b(-/-) mice infected with RSV exhibit exaggerated immune cell infiltration, damaged epithelial cell barriers, cytokine production, and increased apoptosis. Elevated expression of S100A9, a damage-associated molecular pattern molecule, was observed in the lungs of Ptp1b(-/-) mice during RSV infection. Utilizing a neutralizing anti-S100A9 IgG antibody, it was determined that extracellular S100A9 signaling significantly affects lung damage during RSV infection. Preexposure to cigarette smoke desensitized PTP1B activity that coincided with enhanced S100A9 secretion and inflammation in wild-type animals during RSV infection. S100A9 levels in human bronchoalveolar lavage fluid had an inverse relationship with lung function in healthy subjects, smokers, and COPD subjects. Fully differentiated human bronchial epithelial cells isolated from COPD donors cultured at the air liquid interface secreted more S100A9 than cells from healthy donors or smokers following RSV infection. Together, these findings show that reduced PTP1B responses contribute to disease symptoms in part by enhancing S100A9 expression during viral-associated COPD exacerbations.


Calgranulin B/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/immunology , Pulmonary Disease, Chronic Obstructive/immunology , Respiratory Syncytial Virus Infections/immunology , Smoking/immunology , Animals , Antibodies, Neutralizing/pharmacology , Bronchoalveolar Lavage Fluid/chemistry , Bronchoalveolar Lavage Fluid/cytology , Calgranulin B/genetics , Case-Control Studies , Disease Models, Animal , Female , Gene Expression Regulation , Humans , Macrophages, Alveolar/immunology , Macrophages, Alveolar/pathology , Mice , Mice, Knockout , Primary Cell Culture , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Pulmonary Disease, Chronic Obstructive/complications , Pulmonary Disease, Chronic Obstructive/genetics , Pulmonary Disease, Chronic Obstructive/virology , Respiratory Syncytial Virus Infections/complications , Respiratory Syncytial Virus Infections/genetics , Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Viruses/growth & development , Respiratory Syncytial Viruses/immunology , Signal Transduction , Smoking/genetics , Smoking/pathology , Tobacco Smoke Pollution
19.
Arterioscler Thromb Vasc Biol ; 35(1): 163-74, 2015 Jan.
Article En | MEDLINE | ID: mdl-25395617

OBJECTIVE: Impaired wound healing is a major complication of diabetes mellitus. The mechanisms that govern wound healing, however, are complex and incompletely understood. In the present study, we determined the inhibitory role of protein tyrosine phosphatase 1B (PTP1B) in the process of diabetic wound healing. APPROACH AND RESULTS: First, by comparing the wound healing process in PTP1B knockout (PTP1B(-/-)) mice, ob/ob mice and their wild-type littermates in the presence or absence of streptozotocin treatment, we showed that the inhibition of mouse wound healing in streptozotocin-induced diabetic conditions is because of the upregulation and activation of PTP1B. Second, the impaired wound healing in ob/ob mice and streptozotocin-treated wild-type mice was rescued by a PTP1B inhibitor. Third, PTP1B, which is upregulated under hyperglycemic condition, inhibited the tube formation, proliferation, and migration of human microvascular endothelial cells induced by vascular endothelial growth factor, whereas this inhibition was largely abolished by the PTP1B inhibitor. Finally, mechanism study further indicated that PTP1B likely suppressed the proliferation, migration, and tube formation of vascular endothelial cells through dephosphorylation of vascular endothelial growth factor receptor 2. CONCLUSIONS: Our study demonstrated that PTP1B negatively modulated the diabetic wound healing process by dephosphorylating the endothelial cell vascular endothelial growth factor receptor 2 and that the specific inhibitor of PTP1B might serve as a potential novel therapeutic tool for diabetic wound healing.


Diabetes Mellitus, Experimental/enzymology , Diabetic Angiopathies/enzymology , Endothelial Cells/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , Skin Ulcer/enzymology , Vascular Endothelial Growth Factor Receptor-2/metabolism , Wound Healing , Animals , Cell Line , Cell Movement , Cell Proliferation , Diabetes Mellitus, Experimental/drug therapy , Diabetes Mellitus, Experimental/genetics , Diabetes Mellitus, Experimental/pathology , Diabetes Mellitus, Experimental/physiopathology , Diabetic Angiopathies/drug therapy , Diabetic Angiopathies/genetics , Diabetic Angiopathies/pathology , Diabetic Angiopathies/physiopathology , Endothelial Cells/drug effects , Enzyme Activation , Enzyme Inhibitors/pharmacology , Humans , Mice, Inbred C57BL , Mice, Knockout , Neovascularization, Physiologic , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , Signal Transduction , Skin Ulcer/drug therapy , Skin Ulcer/genetics , Skin Ulcer/pathology , Skin Ulcer/physiopathology , Time Factors , Transfection , Up-Regulation , Wound Healing/drug effects
20.
Biochim Biophys Acta ; 1852(2): 299-309, 2015 Feb.
Article En | MEDLINE | ID: mdl-25018087

Obesity-induced cardiomyopathy may be mediated by alterations in multiple signaling cascades involved in glucose and lipid metabolism. Protein tyrosine phosphatase-1B (PTP1B) is an important negative regulator of insulin signaling. This study was designed to evaluate the role of PTP1B in high fat diet-induced cardiac contractile anomalies. Wild-type and PTP1B knockout mice were fed normal (10%) or high (45%) fat diet for 5months prior to evaluation of cardiac function. Myocardial function was assessed using echocardiography and an Ion-Optix MyoCam system. Western blot analysis was employed to evaluate levels of AMPK, mTOR, raptor, Beclin-1, p62 and LC3-II. RT-PCR technique was employed to assess genes involved in hypertrophy and lipid metabolism. Our data revealed increased LV thickness and LV chamber size as well as decreased fractional shortening following high fat diet intake, the effect was nullified by PTP1B knockout. High fat diet intake compromised cardiomyocyte contractile function as evidenced by decreased peak shortening, maximal velocity of shortening/relengthening, intracellular Ca²âº release as well as prolonged duration of relengthening and intracellular Ca²âº decay, the effects of which were alleviated by PTP1B knockout. High fat diet resulted in enlarged cardiomyocyte area and increased lipid accumulation, which were attenuated by PTP1B knockout. High fat diet intake dampened myocardial autophagy as evidenced by decreased LC3-II conversion and Beclin-1, increased p62 levels as well as decreased phosphorylation of AMPK and raptor, the effects of which were significantly alleviated by PTP1B knockout. Pharmacological inhibition of AMPK using compound C disengaged PTP1B knockout-conferred protection against fatty acid-induced cardiomyocyte contractile anomalies. Taken together, our results suggest that PTP1B knockout offers cardioprotection against high fat diet intake through activation of AMPK. This article is part of a Special Issue entitled: Autophagy and protein quality control in cardiometabolic diseases.


AMP-Activated Protein Kinases/metabolism , Autophagy , Diet, High-Fat , Gene Deletion , Myocardium/pathology , Obesity/enzymology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism , AMP-Activated Protein Kinases/antagonists & inhibitors , Animals , Blotting, Western , Cardiomegaly/complications , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Feeding Behavior , Lipid Metabolism , Male , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Obesity/complications , Obesity/pathology , Obesity/physiopathology , Protein Tyrosine Phosphatase, Non-Receptor Type 1/deficiency , TOR Serine-Threonine Kinases/metabolism
...