1
|
Ali S, Vidal-Gómez X, Piquet M, Vergori L, Simard G, Dubois S, Ducluzeau PH, Pomiès P, Kamli-Salino S, Delibégovic M, Henni S, Gagnadoux F, Andriantsitohaina R, Martínez MC. Circulating extracellular vesicle-carried PTP1B and PP2A phosphatases as regulators of insulin resistance. Diabetologia 2025; 68:231-242. [PMID: 39422717 DOI: 10.1007/s00125-024-06288-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/20/2024] [Indexed: 10/19/2024]
Abstract
AIMS/HYPOTHESIS Metabolic disorders associated with abdominal obesity, dyslipidaemia, arterial hypertension and hyperglycaemia are risk factors for the development of insulin resistance. Extracellular vesicles (EVs) may play an important role in the regulation of metabolic signalling pathways in insulin resistance and associated complications. METHODS Circulating large EVs (lEVs) and small EVs (sEVs) from individuals with (IR group) and without insulin resistance (n-IR group) were isolated and characterised. lEVs and sEVs were administered by i.v. injection to mice and systemic, adipose tissue and liver insulin signalling were analysed. The role of phosphatases was analysed in target tissues and cells. RESULTS Injection of lEVs and sEVs from IR participants impaired systemic, adipose tissue and liver insulin signalling in mice, while EVs from n-IR participants had no effect. Moreover, lEVs and sEVs from IR participants brought about a twofold increase in adipocyte size and adipogenic gene expression. EVs from IR participants expressed two types of phosphatases, phosphotyrosine 1 phosphatase (PTP1B) and protein phosphatase 2 (PP2A), IR lEVs being enriched with the active form of PTP1B while IR sEVs mainly carried active PP2A. Blockade of PTP1B activity in IR lEVs fully restored IRS1 and Akt phosphorylation in adipocytes and blunted insulin-induced Akt phosphorylation by inhibition of the macrophage secretome in hepatocytes. Conversely, blockade of PP2A activity in IR sEVs completely prevented insulin resistance in adipocytes and hepatocytes. CONCLUSIONS/INTERPRETATION These data demonstrate that inhibition of phosphatases carried by EVs from IR participants rescues insulin signalling in adipocytes and hepatocytes and point towards PTP1B and PP2A carried by IR EVs as being novel potential therapeutic targets against insulin resistance in adipose tissue and liver and the development of obesity.
Collapse
Affiliation(s)
- Sakina Ali
- SOPAM, U1063, Inserm, UNIV Angers, SFR ICAT, Angers, France
| | - Xavier Vidal-Gómez
- SOPAM, U1063, Inserm, UNIV Angers, SFR ICAT, Angers, France
- University of Montpellier, PhyMedExp, Inserm, CNRS UMR, Montpellier, France
| | - Megan Piquet
- SOPAM, U1063, Inserm, UNIV Angers, SFR ICAT, Angers, France
- University of Montpellier, PhyMedExp, Inserm, CNRS UMR, Montpellier, France
| | - Luisa Vergori
- SOPAM, U1063, Inserm, UNIV Angers, SFR ICAT, Angers, France
| | - Gilles Simard
- SOPAM, U1063, Inserm, UNIV Angers, SFR ICAT, Angers, France
- Centre Hospitalo-Universitaire d'Angers (CHU), Angers, France
| | - Séverine Dubois
- SOPAM, U1063, Inserm, UNIV Angers, SFR ICAT, Angers, France
- Centre Hospitalo-Universitaire d'Angers (CHU), Angers, France
| | - Pierre-Henri Ducluzeau
- Service de Médecine Interne, Unité d'Endocrinologie Diabétologie et Nutrition, Centre Hospitalier Universitaire et Faculté de Médecine, Université de Tours, Tours, France
| | - Pascal Pomiès
- University of Montpellier, PhyMedExp, Inserm, CNRS UMR, Montpellier, France
| | - Sarah Kamli-Salino
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Mirela Delibégovic
- Aberdeen Cardiovascular and Diabetes Centre, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | - Samir Henni
- Centre Hospitalo-Universitaire d'Angers (CHU), Angers, France
| | - Frédéric Gagnadoux
- SOPAM, U1063, Inserm, UNIV Angers, SFR ICAT, Angers, France
- Département de Pneumologie et Médecine du Sommeil, CHU d'Angers, Angers, France
| | - Ramaroson Andriantsitohaina
- SOPAM, U1063, Inserm, UNIV Angers, SFR ICAT, Angers, France
- University of Montpellier, PhyMedExp, Inserm, CNRS UMR, Montpellier, France
| | - M Carmen Martínez
- SOPAM, U1063, Inserm, UNIV Angers, SFR ICAT, Angers, France.
- University of Montpellier, PhyMedExp, Inserm, CNRS UMR, Montpellier, France.
| |
Collapse
|
2
|
Sun Y, Dinenno FA, Tang P, Kontaridis MI. Protein tyrosine phosphatase 1B in metabolic and cardiovascular diseases: from mechanisms to therapeutics. Front Cardiovasc Med 2024; 11:1445739. [PMID: 39238503 PMCID: PMC11374623 DOI: 10.3389/fcvm.2024.1445739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 08/05/2024] [Indexed: 09/07/2024] Open
Abstract
Protein Tyrosine Phosphatase 1B (PTP1B) has emerged as a significant regulator of metabolic and cardiovascular disease. It is a non-transmembrane protein tyrosine phosphatase that negatively regulates multiple signaling pathways integral to the regulation of growth, survival, and differentiation of cells, including leptin and insulin signaling, which are critical for development of obesity, insulin resistance, type 2 diabetes, and cardiovascular disease. Given PTP1B's central role in glucose homeostasis, energy balance, and vascular function, targeted inhibition of PTP1B represents a promising strategy for treating these diseases. However, challenges, such as off-target effects, necessitate a focus on tissue-specific approaches, to maximize therapeutic benefits while minimizing adverse outcomes. In this review, we discuss molecular mechanisms by which PTP1B influences metabolic and cardiovascular functions, summarize the latest research on tissue-specific roles of PTP1B, and discuss the potential for PTP1B inhibitors as future therapeutic agents.
Collapse
Affiliation(s)
- Yan Sun
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY, United States
| | - Frank A Dinenno
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY, United States
| | - Peiyang Tang
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY, United States
| | - Maria I Kontaridis
- Department of Biomedical Research and Translational Medicine, Masonic Medical Research Institute, Utica, NY, United States
- Department of Medicine, Division of Cardiology, Beth Israel Deaconess Medical Center, Boston, MA, United States
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
3
|
Delibegović M, Dall'Angelo S, Dekeryte R. Protein tyrosine phosphatase 1B in metabolic diseases and drug development. Nat Rev Endocrinol 2024; 20:366-378. [PMID: 38519567 DOI: 10.1038/s41574-024-00965-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/16/2024] [Indexed: 03/25/2024]
Abstract
Protein tyrosine phosphatase 1B (PTP1B), a non-transmembrane phosphatase, has a major role in a variety of signalling pathways, including direct negative regulation of classic insulin and leptin signalling pathways, and is implicated in the pathogenesis of several cardiometabolic diseases and cancers. As such, PTP1B has been a therapeutic target for over two decades, with PTP1B inhibitors identified either from natural sources or developed throughout the years. Some of these inhibitors have reached phase I and/or II clinical trials in humans for the treatment of type 2 diabetes mellitus, obesity and/or metastatic breast cancer. In this Review, we summarize the cellular processes and regulation of PTP1B, discuss evidence from in vivo preclinical and human studies of the association between PTP1B and different disorders, and discuss outcomes of clinical trials. We outline challenges associated with the targeting of this phosphatase (which was, until the past few years, viewed as difficult to target), the current state of the field of PTP1B inhibitors (and dual phosphatase inhibitors) and future directions for manipulating the activity of this key metabolic enzyme.
Collapse
Affiliation(s)
- Mirela Delibegović
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Institute of Medical Sciences, Aberdeen, UK.
| | - Sergio Dall'Angelo
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Institute of Medical Sciences, Aberdeen, UK
| | - Ruta Dekeryte
- Aberdeen Cardiovascular and Diabetes Centre, University of Aberdeen, Institute of Medical Sciences, Aberdeen, UK
| |
Collapse
|
4
|
Engin A. Endothelial Dysfunction in Obesity and Therapeutic Targets. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1460:489-538. [PMID: 39287863 DOI: 10.1007/978-3-031-63657-8_17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Parallel to the increasing prevalence of obesity in the world, the mortality from cardiovascular disease has also increased. Low-grade chronic inflammation in obesity disrupts vascular homeostasis, and the dysregulation of adipocyte-derived endocrine and paracrine effects contributes to endothelial dysfunction. Besides the adipose tissue inflammation, decreased nitric oxide (NO)-bioavailability, insulin resistance (IR), and oxidized low-density lipoproteins (oxLDLs) are the main factors contributing to endothelial dysfunction in obesity and the development of cardiorenal metabolic syndrome. While normal healthy perivascular adipose tissue (PVAT) ensures the dilation of blood vessels, obesity-associated PVAT leads to a change in the profile of the released adipo-cytokines, resulting in a decreased vasorelaxing effect. Higher stiffness parameter β, increased oxidative stress, upregulation of pro-inflammatory cytokines, and nicotinamide adenine dinucleotide phosphate (NADP) oxidase in PVAT turn the macrophages into pro-atherogenic phenotypes by oxLDL-induced adipocyte-derived exosome-macrophage crosstalk and contribute to the endothelial dysfunction. In clinical practice, carotid ultrasound, higher leptin levels correlate with irisin over-secretion by human visceral and subcutaneous adipose tissues, and remnant cholesterol (RC) levels predict atherosclerotic disease in obesity. As a novel therapeutic strategy for cardiovascular protection, liraglutide improves vascular dysfunction by modulating a cyclic adenosine monophosphate (cAMP)-independent protein kinase A (PKA)-AMP-activated protein kinase (AMPK) pathway in PVAT in obese individuals. Because the renin-angiotensin-aldosterone system (RAAS) activity, hyperinsulinemia, and the resultant IR play key roles in the progression of cardiovascular disease in obesity, RAAS-targeted therapies contribute to improving endothelial dysfunction. By contrast, arginase reciprocally inhibits NO formation and promotes oxidative stress. Thus, targeting arginase activity as a key mediator in endothelial dysfunction has therapeutic potential in obesity-related vascular comorbidities. Obesity-related endothelial dysfunction plays a pivotal role in the progression of type 2 diabetes (T2D). The peroxisome proliferator-activated receptor gamma (PPARγ) agonist, rosiglitazone (thiazolidinedione), is a popular drug for treating diabetes; however, it leads to increased cardiovascular risk. Selective sodium-glucose co-transporter-2 (SGLT-2) inhibitor empagliflozin (EMPA) significantly improves endothelial dysfunction and mortality occurring through redox-dependent mechanisms. Although endothelial dysfunction and oxidative stress are alleviated by either metformin or EMPA, currently used drugs to treat obesity-related diabetes neither possess the same anti-inflammatory potential nor simultaneously target endothelial cell dysfunction and obesity equally. While therapeutic interventions with glucagon-like peptide-1 (GLP-1) receptor agonist liraglutide or bariatric surgery reverse regenerative cell exhaustion, support vascular repair mechanisms, and improve cardiometabolic risk in individuals with T2D and obesity, the GLP-1 analog exendin-4 attenuates endothelial endoplasmic reticulum stress.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
5
|
Cornejo PJ, Vergoni B, Ohanna M, Angot B, Gonzalez T, Jager J, Tanti JF, Cormont M. The Stress-Responsive microRNA-34a Alters Insulin Signaling and Actions in Adipocytes through Induction of the Tyrosine Phosphatase PTP1B. Cells 2022; 11:cells11162581. [PMID: 36010657 PMCID: PMC9406349 DOI: 10.3390/cells11162581] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 08/02/2022] [Accepted: 08/05/2022] [Indexed: 11/18/2022] Open
Abstract
Metabolic stresses alter the signaling and actions of insulin in adipocytes during obesity, but the molecular links remain incompletely understood. Members of the microRNA-34 (miR-34 family play a pivotal role in stress response, and previous studies showed an upregulation of miR-34a in adipose tissue during obesity. Here, we identified miR-34a as a new mediator of adipocyte insulin resistance. We confirmed the upregulation of miR-34a in adipose tissues of obese mice, which was observed in the adipocyte fraction exclusively. Overexpression of miR-34a in 3T3-L1 adipocytes or in fat pads of lean mice markedly reduced Akt activation by insulin and the insulin-induced glucose transport. This was accompanied by a decreased expression of VAMP2, a target of miR-34a, and an increased expression of the tyrosine phosphatase PTP1B. Importantly, PTP1B silencing prevented the inhibitory effect of miR-34a on insulin signaling. Mechanistically, miR-34a decreased the NAD+ level through inhibition of Naprt and Nampt, resulting in an inhibition of Sirtuin-1, which promoted an upregulation of PTP1B. Furthermore, the mRNA expression of Nampt and Naprt was decreased in adipose tissue of obese mice. Collectively, our results identify miR-34a as a new inhibitor of insulin signaling in adipocytes, providing a potential pathway to target to fight insulin resistance.
Collapse
Affiliation(s)
- Pierre-Jean Cornejo
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
| | - Bastien Vergoni
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
| | - Mickaël Ohanna
- Université Côte d’Azur, Inserm, C3M, “Team Microenvironnement, Signalisation et Cancer”, 06204 Nice, France
| | - Brice Angot
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
| | - Teresa Gonzalez
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
- Aix Marseille Université, Inserm, INRAE, C2VN, 13385 Marseille, France
| | - Jennifer Jager
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
| | - Jean-François Tanti
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
| | - Mireille Cormont
- Université Côte d’Azur, Inserm, C3M, Team “Molecular and Cellular Physiopathology of Obesity and Diabetes”, 06204 Nice, France
- Correspondence: ; Tel.: +33-4-89-15-38-31
| |
Collapse
|
6
|
Coulis G, Londhe AD, Sagabala RS, Shi Y, Labbé DP, Bergeron A, Sahadevan P, Nawaito SA, Sahmi F, Josse M, Vinette V, Guertin MC, Karsenty G, Tremblay ML, Tardif JC, Allen BG, Boivin B. Protein tyrosine phosphatase 1B regulates miR-208b-argonaute 2 association and thyroid hormone responsiveness in cardiac hypertrophy. Sci Signal 2022; 15:eabn6875. [PMID: 35439023 DOI: 10.1126/scisignal.abn6875] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Increased production of reactive oxygen species plays an essential role in the pathogenesis of several diseases, including cardiac hypertrophy. In our search to identify redox-sensitive targets that contribute to redox signaling, we found that protein tyrosine phosphatase 1B (PTP1B) was reversibly oxidized and inactivated in hearts undergoing hypertrophy. Cardiomyocyte-specific deletion of PTP1B in mice (PTP1B cKO mice) caused a hypertrophic phenotype that was exacerbated by pressure overload. Furthermore, we showed that argonaute 2 (AGO2), a key component of the RNA-induced silencing complex, was a substrate of PTP1B in cardiomyocytes and in the heart. Our results revealed that phosphorylation at Tyr393 and inactivation of AGO2 in PTP1B cKO mice prevented miR-208b-mediated repression of thyroid hormone receptor-associated protein 1 (THRAP1; also known as MED13) and contributed to thyroid hormone-mediated cardiac hypertrophy. In support of this conclusion, inhibiting the synthesis of triiodothyronine (T3) with propylthiouracil rescued pressure overload-induced hypertrophy and improved myocardial contractility and systolic function in PTP1B cKO mice. Together, our data illustrate that PTP1B activity is cardioprotective and that redox signaling is linked to thyroid hormone responsiveness and microRNA-mediated gene silencing in pathological hypertrophy.
Collapse
Affiliation(s)
- Gérald Coulis
- Department of Nanobioscience, College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA.,Montreal Heart Institute, Montreal, QC H1T 1C8, Canada
| | - Avinash D Londhe
- Department of Nanobioscience, College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA
| | - R Sudheer Sagabala
- Department of Nanobioscience, College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA
| | - Yanfen Shi
- Montreal Heart Institute, Montreal, QC H1T 1C8, Canada
| | - David P Labbé
- Department of Medicine, Division of Experimental Medicine, McGill University, Montreal, QC H3G 1Y6, Canada.,Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada.,Department of Surgery, Division of Urology, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Alexandre Bergeron
- Montreal Heart Institute, Montreal, QC H1T 1C8, Canada.,Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Pramod Sahadevan
- Montreal Heart Institute, Montreal, QC H1T 1C8, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Sherin A Nawaito
- Montreal Heart Institute, Montreal, QC H1T 1C8, Canada.,Pharmacology and Physiology, Université de Montréal, Montréal, QC H3C 3J7, Canada.,Department of Physiology, Faculty of Medicine, Suez Canal University, Ismailia, Egypt
| | - Fatiha Sahmi
- Montreal Heart Institute, Montreal, QC H1T 1C8, Canada
| | - Marie Josse
- Department of Nanobioscience, College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA
| | - Valérie Vinette
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada.,Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | | | - Gérard Karsenty
- Department of Genetics and Development, Columbia University, New York, NY 10032, USA
| | - Michel L Tremblay
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, QC H3A 1A3, Canada.,Department of Biochemistry, McGill University, Montreal, QC H3G 1Y6, Canada
| | - Jean-Claude Tardif
- Montreal Heart Institute, Montreal, QC H1T 1C8, Canada.,Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| | - Bruce G Allen
- Montreal Heart Institute, Montreal, QC H1T 1C8, Canada.,Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada.,Department of Biochemistry and Molecular Medicine, Université de Montréal, Montréal, QC H3C 3J7, Canada.,Pharmacology and Physiology, Université de Montréal, Montréal, QC H3C 3J7, Canada
| | - Benoit Boivin
- Department of Nanobioscience, College of Nanoscale Science and Engineering, SUNY Polytechnic Institute, Albany, NY 12203, USA.,Montreal Heart Institute, Montreal, QC H1T 1C8, Canada.,Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
| |
Collapse
|
7
|
Teimouri M, Hosseini H, ArabSadeghabadi Z, Babaei-Khorzoughi R, Gorgani-Firuzjaee S, Meshkani R. The role of protein tyrosine phosphatase 1B (PTP1B) in the pathogenesis of type 2 diabetes mellitus and its complications. J Physiol Biochem 2022; 78:307-322. [PMID: 34988903 DOI: 10.1007/s13105-021-00860-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 11/16/2021] [Indexed: 01/16/2023]
Abstract
Insulin resistance, the most important characteristic of the type 2 diabetes mellitus (T2DM), is mostly caused by impairment in the insulin receptor (IR) signal transduction pathway. Protein tyrosine phosphatase 1B (PTP1B), one of the main negative regulators of the IR signaling pathway, is broadly expressed in various cells and tissues. PTP1B decreases the phosphorylation of the IR resulting in insulin resistance in various tissues. The evidence for the physiological role of PTP1B in regulation of metabolic pathways came from whole-body PTP1B-knockout mice. Whole-body and tissue-specific PTP1B-knockout mice showed improvement in adiposity, insulin resistance, and glucose tolerance. In addition, the key role of PTP1B in the pathogenesis of T2DM and its complications was further investigated in mice models of PTP1B deficient/overexpression. In recent years, targeting PTP1B using PTP1B inhibitors is being considered an attractive target to treat T2DM. PTP1B inhibitors improve the sensitivity of the insulin receptor and have the ability to cure insulin resistance-related diseases. We herein summarized the biological functions of PTP1B in different tissues in vivo and in vitro. We also describe the effectiveness of potent PTP1B inhibitors as pharmaceutical agents to treat T2DM.
Collapse
Affiliation(s)
- Maryam Teimouri
- Department of Clinical Biochemistry, School of Allied Medical Sciences, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Hossein Hosseini
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra ArabSadeghabadi
- Department of Clinical Sciences, Faculty of Veterinary Science, Bu-Ali Sina University, Hamedan, Iran
| | - Reyhaneh Babaei-Khorzoughi
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sattar Gorgani-Firuzjaee
- Department of Medical Laboratory Sciences, School of Allied Health Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Reza Meshkani
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Stanford SM, Collins M, Diaz MA, Holmes ZJ, Gries P, Bliss MR, Lodi A, Zhang V, Tiziani S, Bottini N. The low molecular weight protein tyrosine phosphatase promotes adipogenesis and subcutaneous adipocyte hypertrophy. J Cell Physiol 2021; 236:6630-6642. [PMID: 33615467 DOI: 10.1002/jcp.30307] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/29/2020] [Accepted: 01/15/2021] [Indexed: 12/28/2022]
Abstract
Obesity is a major contributing factor to the pathogenesis of Type 2 diabetes. Multiple human genetics studies suggest that high activity of the low molecular weight protein tyrosine phosphatase (LMPTP) promotes metabolic syndrome in obesity. We reported that LMPTP is a critical promoter of insulin resistance in obesity by regulating liver insulin receptor signaling and that inhibition of LMPTP reverses obesity-associated diabetes in mice. Since LMPTP is expressed in adipose tissue but little is known about its function, here we examined the role of LMPTP in adipocyte biology. Using conditional knockout mice, we found that selective deletion of LMPTP in adipocytes impaired obesity-induced subcutaneous adipocyte hypertrophy. We assessed the role of LMPTP in adipogenesis in vitro, and found that LMPTP deletion or knockdown substantially impaired differentiation of primary preadipocytes and 3T3-L1 cells into adipocytes, respectively. Inhibition of LMPTP in 3T3-L1 preadipocytes also reduced adipogenesis and expression of proadipogenic transcription factors peroxisome proliferator activated receptor gamma (PPARγ) and CCAAT/enhancer-binding protein alpha. Inhibition of LMPTP increased basal phosphorylation of platelet-derived growth factor receptor alpha (PDGFRα) on activation motif residue Y849 in 3T3-L1, resulting in increased activation of the mitogen-associated protein kinases p38 and c-Jun N-terminal kinase and increased PPARγ phosphorylation on inhibitory residue S82. Analysis of the metabolome of differentiating 3T3-L1 cells suggested that LMPTP inhibition decreased cell glucose utilization while enhancing mitochondrial respiration and nucleotide synthesis. In summary, we report a novel role for LMPTP as a key driver of adipocyte differentiation via control of PDGFRα signaling.
Collapse
Affiliation(s)
- Stephanie M Stanford
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Meghan Collins
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, Texas, USA.,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Michael A Diaz
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Zachary J Holmes
- Department of Medicine, University of California, San Diego, La Jolla, California, USA
| | - Paul Gries
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, Texas, USA.,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Matthew R Bliss
- Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Alessia Lodi
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, Texas, USA.,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Vida Zhang
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| | - Stefano Tiziani
- Department of Nutritional Sciences, The University of Texas at Austin, Austin, Texas, USA.,Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA.,Department of Oncology, Dell Medical School, The University of Texas at Austin, Austin, Texas, USA
| | - Nunzio Bottini
- Department of Medicine, University of California, San Diego, La Jolla, California, USA.,Division of Cellular Biology, La Jolla Institute for Allergy and Immunology, La Jolla, California, USA
| |
Collapse
|
9
|
Bourebaba L, Kornicka-Garbowska K, Al Naem M, Röcken M, Łyczko J, Marycz K. MSI-1436 improves EMS adipose derived progenitor stem cells in the course of adipogenic differentiation through modulation of ER stress, apoptosis, and oxidative stress. Stem Cell Res Ther 2021; 12:97. [PMID: 33536069 PMCID: PMC7860037 DOI: 10.1186/s13287-020-02102-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 12/14/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Protein tyrosine phosphatase 1B (PTP1B) is one of the major negative regulators of leptin and insulin signaling, and has been strongly implicated in insulin resistance development in the course of obesity and metabolic syndrome conditions; however, its exact role in controlling adipose tissue biogenesis is still poorly understood. OBJECTIVES This investigation aimed to elucidate whether selective inhibition of PTP1B using MSI-1436 compound may improve and restore the defective adipogenicity of ASCs isolated from EMS-affected horses. METHODS Equine ASC EMS cells were cultured under adipogenic conditions in the presence of PTP1B inhibitor and were subsequently tested for expression of the main adipogenic-related genes using RT-qPCR, changes in free fatty acid profiles by means of GC-MS technique, and for mitochondrial dynamics improvement through the analysis of mitochondrial transmembrane potential and oxidative stress. RESULTS Selective inhibition of PTP1B in equine ASC EMS cells improved substantially adipogenic differentiation by promoting cellular proliferation and normalizing expression of C/EBPalpha, PPARγ, and Adipoq markers that are critical for proper adipogenesis. Levels of secreted adiponectin and PPARγ were also shown to be increased in MSI-1436-conditioned cells, while total leptin levels markedly dropped under the same conditions. Moreover, MSI-1436 treatment enabled the regulation of metabolic-related transcripts that are crosslink to adipogenesis, namely Akt1, Akt2, and SHBG. The obtained results demonstrated also an obvious reduction in intracellular accumulated ROS and NO, as well as mitigated ER stress through the downregulation of Chop, Perk, Atf6, Ire1, and Xbp1 transcripts upon PTP1B inhibition. Furthermore, general fluctuations in FFA composition of all differentiated groups have been highlighted, where palmitic acid, palmitoleic acid, stearic acid, and linolelaidic acid that are known to be associated with the development of metabolic disorders were found to be normalized upon PTP1B inhibition during adipogenic differentiation. CONCLUSION The presented data provides the evidence that the use of PTP1B inhibitor may be successful in controlling and enhancing adipogenic differentiation of impaired equine ASCs affected by metabolic syndrome, and thus offers new insights for the management of obesity through the regulation of adipose tissue dynamics.
Collapse
Affiliation(s)
- Lynda Bourebaba
- Department of Experimental Biology, Wrocław University of Environmental and Life Sciences, Norwida 27B Street, A7 Building, 50-375, Wrocław, Poland
- International Institute of Translational Medicine, Malin, Jesionowa 11, 55-114, Wisznia Mała, Poland
| | - Katarzyna Kornicka-Garbowska
- Department of Experimental Biology, Wrocław University of Environmental and Life Sciences, Norwida 27B Street, A7 Building, 50-375, Wrocław, Poland
- International Institute of Translational Medicine, Malin, Jesionowa 11, 55-114, Wisznia Mała, Poland
| | - Mohamad Al Naem
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392, Giessen, Germany
| | - Michael Röcken
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392, Giessen, Germany
| | - Jacek Łyczko
- Department of Chemistry, Faculty of Biotechnology and Food Science, Wrocław University of Environmental and Life Sciences, Norwida 25, 50-375, Wrocław, Poland
| | - Krzysztof Marycz
- Department of Experimental Biology, Wrocław University of Environmental and Life Sciences, Norwida 27B Street, A7 Building, 50-375, Wrocław, Poland.
- International Institute of Translational Medicine, Malin, Jesionowa 11, 55-114, Wisznia Mała, Poland.
- Faculty of Veterinary Medicine, Equine Clinic-Equine Surgery, Justus-Liebig-University, 35392, Giessen, Germany.
| |
Collapse
|
10
|
Bscl2 Deficiency Does Not Directly Impair the Innate Immune Response in a Murine Model of Generalized Lipodystrophy. J Clin Med 2021; 10:jcm10030441. [PMID: 33498782 PMCID: PMC7865406 DOI: 10.3390/jcm10030441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/13/2021] [Accepted: 01/15/2021] [Indexed: 11/28/2022] Open
Abstract
Congenital Generalized Lipodystrophy type 2 (CGL2) is the most severe form of lipodystrophy and is caused by mutations in the BSCL2 gene. Affected patients exhibit a near complete lack of adipose tissue and suffer severe metabolic disease. A recent study identified infection as a major cause of death in CGL2 patients, leading us to examine whether Bscl2 loss could directly affect the innate immune response. We generated a novel mouse model selectively lacking Bscl2 in the myeloid lineage (LysM-B2KO) and also examined the function of bone-marrow-derived macrophages (BMDM) isolated from global Bscl2 knockout (SKO) mice. LysM-B2KO mice failed to develop lipodystrophy and metabolic disease, providing a model to study the direct role of Bscl2 in myeloid lineage cells. Lipopolysaccharide-mediated stimulation of inflammatory cytokines was not impaired in LysM-B2KO mice or in BMDM isolated from either LysM-B2KO or SKO mice. Additionally, intracellular fate and clearance of bacteria in SKO BMDM challenged with Staphylococcus aureus was indistinguishable from that in BMDM isolated from littermate controls. Overall, our findings reveal that selective Bscl2 deficiency in macrophages does not critically impact the innate immune response to infection. Instead, an increased susceptibility to infection in CGL2 patients is likely to result from severe metabolic disease.
Collapse
|
11
|
Adipocyte G i signaling is essential for maintaining whole-body glucose homeostasis and insulin sensitivity. Nat Commun 2020; 11:2995. [PMID: 32532984 PMCID: PMC7293267 DOI: 10.1038/s41467-020-16756-x] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Accepted: 05/18/2020] [Indexed: 12/13/2022] Open
Abstract
Adipocyte dysfunction links obesity to insulin resistance and type 2 diabetes. Adipocyte function is regulated by receptor-mediated activation of heterotrimeric G proteins. Little is known about the potential in vivo metabolic roles of Gi-type G proteins expressed by adipocytes, primarily due to the lack of suitable animal models. To address this question, we generated mice lacking functional Gi proteins selectively in adipocytes. Here we report that these mutant mice displayed significantly impaired glucose tolerance and reduced insulin sensitivity when maintained on an obesogenic diet. In contrast, using a chemogenetic strategy, we demonstrated that activation of Gi signaling selectively in adipocytes greatly improved glucose homeostasis and insulin signaling. We also elucidated the cellular mechanisms underlying the observed metabolic phenotypes. Our data support the concept that adipocyte Gi signaling is essential for maintaining euglycemia. Drug-mediated activation of adipocyte Gi signaling may prove beneficial for restoring proper glucose homeostasis in type 2 diabetes. Gs-coupled receptor signaling is well known to modulate adipocyte metabolism, but the role of Gi-coupled receptors in adipose tissue is less well understood. Here the authors show that signaling via Gi-type G proteins expressed by adipocytes is essential for maintaining proper blood glucose homeostasis.
Collapse
|
12
|
The Role of Protein Tyrosine Phosphatase (PTP)-1B in Cardiovascular Disease and Its Interplay with Insulin Resistance. Biomolecules 2019; 9:biom9070286. [PMID: 31319588 PMCID: PMC6680919 DOI: 10.3390/biom9070286] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 07/06/2019] [Accepted: 07/12/2019] [Indexed: 12/19/2022] Open
Abstract
Endothelial dysfunction is a key feature of cardiovascular disorders associated with obesity and diabetes. Several studies identified protein tyrosine phosphatase (PTP)-1B, a member of the PTP superfamily, as a major negative regulator for insulin receptor signaling and a novel molecular player in endothelial dysfunction and cardiovascular disease. Unlike other anti-diabetic approaches, genetic deletion or pharmacological inhibition of PTP1B was found to improve glucose homeostasis and insulin signaling without causing lipid buildup in the liver, which represents an advantage over existing therapies. Furthermore, PTP1B was reported to contribute to cardiovascular disturbances, at various molecular levels, which places this enzyme as a unique single therapeutic target for both diabetes and cardiovascular disorders. Synthesizing selective small molecule inhibitors for PTP1B is faced with multiple challenges linked to its similarity of sequence with other PTPs; however, overcoming these challenges would pave the way for novel approaches to treat diabetes and its concurrent cardiovascular complications. In this review article, we summarized the major roles of PTP1B in cardiovascular disease with special emphasis on endothelial dysfunction and its interplay with insulin resistance. Furthermore, we discussed some of the major challenges hindering the synthesis of selective inhibitors for PTP1B.
Collapse
|
13
|
Gozal D, Khalyfa A, Qiao Z, Akbarpour M, Maccari R, Ottanà R. Protein-Tyrosine Phosphatase-1B Mediates Sleep Fragmentation-Induced Insulin Resistance and Visceral Adipose Tissue Inflammation in Mice. Sleep 2018. [PMID: 28651353 DOI: 10.1093/sleep/zsx111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Study Objectives Sleep fragmentation (SF) is highly prevalent and has emerged as an important contributing factor to obesity and metabolic syndrome. We hypothesized that SF-induced increases in protein tyrosine phosphatase-1B (PTP-1B) expression and activity underlie increased food intake, inflammation, and leptin and insulin resistance. Methods Wild-type (WT) and ObR-PTP-1b-/- mice (Tg) were exposed to SF and control sleep (SC), and food intake was monitored. WT mice received a PTP-1B inhibitor (RO-7d; Tx) or vehicle (Veh). Upon completion of exposures, systemic insulin and leptin sensitivity tests were performed as well as assessment of visceral white adipose tissue (vWAT) insulin receptor sensitivity and macrophages (ATM) polarity. Results SF increased food intake in either untreated or Veh-treated WT mice. Leptin-induced hypothalamic STAT3 phosphorylation was decreased, PTP-1B activity was increased, and reduced insulin sensitivity emerged both systemic and in vWAT, with the latter displaying proinflammatory ATM polarity changes. All of the SF-induced effects were abrogated following PTP-1B inhibitor treatment and in Tg mice. Conclusions SF induces increased food intake, reduced leptin signaling in hypothalamus, systemic insulin resistance, and reduced vWAT insulin sensitivity and inflammation that are mediated by increased PTP-1B activity. Thus, PTP-1B may represent a viable therapeutic target in the context of SF-induced weight gain and metabolic dysfunction.
Collapse
Affiliation(s)
- David Gozal
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Abdelnaby Khalyfa
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Zhuanghong Qiao
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Mahzad Akbarpour
- Section of Pediatric Sleep Medicine, Department of Pediatrics, Comer Children's Hospital, Biological Sciences Division, The University of Chicago, Chicago, IL
| | - Rosanna Maccari
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, PoloAnnunziata, V.le SS. Annunziata, Messina, Italy
| | - Rosaria Ottanà
- Dipartimento di Scienze Chimiche, Biologiche, Farmaceutiche ed Ambientali, PoloAnnunziata, V.le SS. Annunziata, Messina, Italy
| |
Collapse
|
14
|
Le Sommer S, Morrice N, Pesaresi M, Thompson D, Vickers MA, Murray GI, Mody N, Neel BG, Bence KK, Wilson HM, Delibegović M. Deficiency in Protein Tyrosine Phosphatase PTP1B Shortens Lifespan and Leads to Development of Acute Leukemia. Cancer Res 2018; 78:75-87. [PMID: 29122767 PMCID: PMC5756472 DOI: 10.1158/0008-5472.can-17-0946] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 06/29/2017] [Accepted: 10/25/2017] [Indexed: 01/05/2023]
Abstract
Protein tyrosine phosphatase PTP1B is a critical regulator of signaling pathways controlling metabolic homeostasis, cell proliferation, and immunity. In this study, we report that global or myeloid-specific deficiency of PTP1B in mice decreases lifespan. We demonstrate that myeloid-specific deficiency of PTP1B is sufficient to promote the development of acute myeloid leukemia. LysM-PTP1B-/- mice lacking PTP1B in the innate myeloid cell lineage displayed a dysregulation of bone marrow cells with a rapid decline in population at midlife and a concomitant increase in peripheral blood blast cells. This phenotype manifested further with extramedullary tumors, hepatic macrophage infiltration, and metabolic reprogramming, suggesting increased hepatic lipid metabolism prior to overt tumor development. Mechanistic investigations revealed an increase in anti-inflammatory M2 macrophage responses in liver and spleen, as associated with increased expression of arginase I and the cytokines IL10 and IL4. We also documented STAT3 hypersphosphorylation and signaling along with JAK-dependent upregulation of antiapoptotic proteins Bcl2 and BclXL. Our results establish a tumor suppressor role for PTP1B in the myeloid lineage cells, with evidence that its genetic inactivation in mice is sufficient to drive acute myeloid leukemia.Significance: This study defines a tumor suppressor function for the protein tyrosine phosphatase PTP1B in myeloid lineage cells, with evidence that its genetic inactivation in mice is sufficient to drive acute myeloid leukemia. Cancer Res; 78(1); 75-87. ©2017 AACR.
Collapse
Affiliation(s)
| | - Nicola Morrice
- Institute of Medical Sciences, University of Aberdeen, United Kingdom
| | - Martina Pesaresi
- Institute of Medical Sciences, University of Aberdeen, United Kingdom
| | - Dawn Thompson
- Institute of Medical Sciences, University of Aberdeen, United Kingdom
| | - Mark A Vickers
- Institute of Medical Sciences, University of Aberdeen, United Kingdom
| | - Graeme I Murray
- Institute of Medical Sciences, University of Aberdeen, United Kingdom
| | - Nimesh Mody
- Institute of Medical Sciences, University of Aberdeen, United Kingdom
| | - Benjamin G Neel
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Medical Center, New York University, New York, New York
| | - Kendra K Bence
- Department of Biomedical Sciences, University of Pennsylvania School of Veterinary Medicine, Philadelphia
| | - Heather M Wilson
- Institute of Medical Sciences, University of Aberdeen, United Kingdom.
| | | |
Collapse
|
15
|
Pharmacological inhibition of protein tyrosine phosphatase 1B protects against atherosclerotic plaque formation in the LDLR -/- mouse model of atherosclerosis. Clin Sci (Lond) 2017; 131:2489-2501. [PMID: 28899902 PMCID: PMC6365594 DOI: 10.1042/cs20171066] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 09/04/2017] [Accepted: 09/06/2017] [Indexed: 02/03/2023]
Abstract
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) signalling. Here, we demonstrate that inhibiting the activity of protein tyrosine phosphatase 1B (PTP1B), the major negative regulator of the IR prevents and reverses atherosclerotic plaque formation in an LDLR−/− mouse model of atherosclerosis. Acute (single dose) or chronic PTP1B inhibitor (trodusquemine) treatment of LDLR−/− mice decreased weight gain and adiposity, improved glucose homeostasis and attenuated atherosclerotic plaque formation. This was accompanied by a reduction in both, circulating total cholesterol and triglycerides, a decrease in aortic monocyte chemoattractant protein-1 (MCP-1) expression levels and hyperphosphorylation of aortic Akt/PKB and AMPKα. Our findings are the first to demonstrate that PTP1B inhibitors could be used in prevention and reversal of atherosclerosis development and reduction in CVD risk.
Collapse
|
16
|
Thompson D, Morrice N, Grant L, Le Sommer S, Ziegler K, Whitfield P, Mody N, Wilson HM, Delibegović M. Myeloid protein tyrosine phosphatase 1B (PTP1B) deficiency protects against atherosclerotic plaque formation in the ApoE -/- mouse model of atherosclerosis with alterations in IL10/AMPKα pathway. Mol Metab 2017; 6:845-853. [PMID: 28752048 PMCID: PMC5518727 DOI: 10.1016/j.molmet.2017.06.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 06/02/2017] [Accepted: 06/06/2017] [Indexed: 12/21/2022] Open
Abstract
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. PTP1B inhibition as therapy for atherosclerosis/cardiovascular disease is proposed. Myeloid-PTP1B mice on ApoE−/− background (ApoE−/−/LysM-PTP1B) were generated. ApoE−/−/LysM-PTP1B had improved glucose homeostasis with no body weight differences. ApoE−/−/LysM-PTP1B had lower lipids and protection against atherosclerotic plaques. Protection was via a PGE2/IL-10/AMPKα mechanism.
Collapse
Affiliation(s)
- D Thompson
- Institute of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK.
| | - N Morrice
- Institute of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - L Grant
- Institute of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - S Le Sommer
- Institute of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - K Ziegler
- Department of Diabetes and Cardiovascular Science, University of the Highlands and Islands, Centre for Health Science, Inverness, UK
| | - P Whitfield
- Department of Diabetes and Cardiovascular Science, University of the Highlands and Islands, Centre for Health Science, Inverness, UK
| | - N Mody
- Institute of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - H M Wilson
- Institute of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK
| | - M Delibegović
- Institute of Medicine, Medical Sciences and Nutrition, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
17
|
Abstract
Chronic inflammatory state in obesity causes dysregulation of the endocrine and paracrine actions of adipocyte-derived factors, which disrupt vascular homeostasis and contribute to endothelial vasodilator dysfunction and subsequent hypertension. While normal healthy perivascular adipose tissue (PVAT) ensures the dilation of blood vessels, obesity-associated PVAT leads to a change in profile of the released adipo-cytokines, resulting in a decreased vasorelaxing effect. Adipose tissue inflammation, nitric oxide (NO)-bioavailability, insulin resistance and oxidized low-density lipoprotein (oxLDL) are main participating factors in endothelial dysfunction of obesity. In this chapter, disruption of inter-endothelial junctions between endothelial cells, significant increase in the production of reactive oxygen species (ROS), inflammation mediators, which are originated from inflamed endothelial cells, the balance between NO synthesis and ROS , insulin signaling and NO production, and decrease in L-arginine/endogenous asymmetric dimethyl-L-arginine (ADMA) ratio are discussed in connection with endothelial dysfunction in obesity.
Collapse
Affiliation(s)
- Atilla Engin
- Faculty of Medicine, Department of General Surgery, Gazi University, Besevler, Ankara, Turkey.
- , Mustafa Kemal Mah. 2137. Sok. 8/14, 06520, Cankaya, Ankara, Turkey.
| |
Collapse
|
18
|
Zhang X, Tian J, Li J, Huang L, Wu S, Liang W, Zhong L, Ye J, Ye F. A novel protein tyrosine phosphatase 1B inhibitor with therapeutic potential for insulin resistance. Br J Pharmacol 2016; 173:1939-49. [PMID: 26990621 DOI: 10.1111/bph.13483] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Revised: 03/05/2016] [Accepted: 03/09/2016] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Insulin-sensitizing drugs are currently limited, and identifying new candidates is a challenge. Protein tyrosine phosphatase 1B (PTP1B) negatively regulates insulin signalling, and its inhibition is anticipated to improve insulin resistance. Here, the pharmacological properties of CX08005, a novel PTP1B inhibitor, were investigated. EXPERIMENTAL APPROACH Recombinant hPTP1B protein was used to study enzyme activity and mode of inhibition. Docking simulation explored the interactions between CX08005 and PTP1B. Insulin sensitivity was evaluated by glucose tolerance test (GTT) in diet-induced obese (DIO) and KKAy mice; glucose-stimulated insulin secretion (GSIS), homeostasis model assessment of insulin resistance index (HOMA-IR) and whole-body insulin sensitivity (ISWB ) were also determined. A hyperinsulinaemic-euglycaemic clamp was performed to evaluate insulin-stimulated glucose disposal in both whole-body and insulin-sensitive tissues. Furthermore, CX08005's effects on muscle, fat and liver cells were determined in vitro. KEY RESULTS CX08005 competitively inhibited PTP1B by binding to the catalytic P-loop through hydrogen bonds. In DIO mice, CX08005 ameliorated glucose intolerance dose-dependently (50-200 mg·kg(-1) ·day(-1) ) and decreased the HOMA-IR. In KKAy mice, CX08005 (50 mg·kg(-1) ·day(-1) ) improved glucose intolerance, GSIS, ISWB and hyperglycaemia. CX08005 also enhanced insulin-stimulated glucose disposal, increased glucose infusion rate and glucose uptake in muscle and fat in DIO mice (hyperinsulinaemic-euglycaemic clamp test). CX08005 enhanced insulin-induced glucose uptake in 3T3-L1 adipocytes and C2C12 myotubes, and increased phosphorylation of IRβ/IRS1 and downstream molecules in hepatocytes in a dose- and insulin-dependent manner respectively. CONCLUSIONS AND IMPLICATIONS Our results strongly suggest that CX08005 directly enhances insulin action in vitro and in vivo through competitive inhibition of PTP1B.
Collapse
Affiliation(s)
- Xiaolin Zhang
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Jinying Tian
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Juan Li
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Liwei Huang
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Song Wu
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Wei Liang
- University of Chinese Academy of Sciences, Beijing, China
| | - Liangwei Zhong
- University of Chinese Academy of Sciences, Beijing, China
| | - Jianping Ye
- Pennington Biomedical Research Center, Louisiana State University, Louisiana, LA, USA
| | - Fei Ye
- Beijing Key Laboratory of New Drug Mechanisms and Pharmacological Evaluation Study, Institute of Materia Medica, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
19
|
Mcilroy GD, Tammireddy SR, Maskrey BH, Grant L, Doherty MK, Watson DG, Delibegović M, Whitfield PD, Mody N. Fenretinide mediated retinoic acid receptor signalling and inhibition of ceramide biosynthesis regulates adipogenesis, lipid accumulation, mitochondrial function and nutrient stress signalling in adipocytes and adipose tissue. Biochem Pharmacol 2015; 100:86-97. [PMID: 26592777 PMCID: PMC4762576 DOI: 10.1016/j.bcp.2015.11.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2015] [Accepted: 11/16/2015] [Indexed: 11/29/2022]
Abstract
Fenretinide (FEN) is a synthetic retinoid that inhibits obesity and insulin resistance in high-fat diet (HFD)-fed mice and completely prevents 3T3-L1 pre-adipocyte differentiation. The aim of this study was to determine the mechanism(s) of FEN action in 3T3-L1 adipocytes and in mice. We used the 3T3-L1 model of adipogenesis, fully differentiated 3T3-L1 adipocytes and adipose tissue from HFD-induced obese mice to investigate the mechanisms of FEN action. We measured expression of adipogenic and retinoid genes by qPCR and activation of nutrient-signalling pathways by western blotting. Global lipid and metabolite analysis was performed and specific ceramide lipid species measured by liquid chromatography-mass spectrometry. We provide direct evidence that FEN inhibits 3T3-L1 adipogenesis via RA-receptor (RAR)-dependent signaling. However, RARα antagonism did not prevent FEN-induced decreases in lipid levels in mature 3T3-L1 adipocytes, suggesting an RAR-independent mechanism. Lipidomics analysis revealed that FEN increased dihydroceramide lipid species 5- to 16-fold in adipocytes, indicating an inhibition of the final step of ceramide biosynthesis. A similar blockade in adipose tissue from FEN-treated obese mice was associated with a complete normalisation of impaired mitochondrial β-oxidation and tricarboxylic acid cycle flux. The FEN catabolite, 4-oxo-N-(4-hydroxyphenyl)retinamide (4-OXO), also decreased lipid accumulation without affecting adipogenesis. FEN and 4-OXO (but not RA) treatment additionally led to the activation of p38-MAPK, peIF2α and autophagy markers in adipocytes. Overall our data reveals FEN utilises both RAR-dependent and -independent pathways to regulate adipocyte biology, both of which may be required for FEN to prevent obesity and insulin resistance in vivo.
Collapse
Affiliation(s)
- George D Mcilroy
- Institute of Medical Sciences, College of Life Sciences & Medicine, University of Aberdeen, Aberdeen, UK.
| | - Seshu R Tammireddy
- Lipidomics Research Facility, Department of Diabetes and Cardiovascular Science, University of the Highlands and Islands, Inverness, UK.
| | - Benjamin H Maskrey
- Lipidomics Research Facility, Department of Diabetes and Cardiovascular Science, University of the Highlands and Islands, Inverness, UK.
| | - Louise Grant
- Institute of Medical Sciences, College of Life Sciences & Medicine, University of Aberdeen, Aberdeen, UK.
| | - Mary K Doherty
- Lipidomics Research Facility, Department of Diabetes and Cardiovascular Science, University of the Highlands and Islands, Inverness, UK.
| | - David G Watson
- Metabolomics Group, Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK.
| | - Mirela Delibegović
- Institute of Medical Sciences, College of Life Sciences & Medicine, University of Aberdeen, Aberdeen, UK.
| | - Phillip D Whitfield
- Lipidomics Research Facility, Department of Diabetes and Cardiovascular Science, University of the Highlands and Islands, Inverness, UK.
| | - Nimesh Mody
- Institute of Medical Sciences, College of Life Sciences & Medicine, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
20
|
Krüger J, Brachs S, Trappiel M, Kintscher U, Meyborg H, Wellnhofer E, Thöne-Reineke C, Stawowy P, Östman A, Birkenfeld AL, Böhmer FD, Kappert K. Enhanced insulin signaling in density-enhanced phosphatase-1 (DEP-1) knockout mice. Mol Metab 2015; 4:325-36. [PMID: 25830095 PMCID: PMC4354926 DOI: 10.1016/j.molmet.2015.02.001] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2015] [Revised: 01/30/2015] [Accepted: 02/04/2015] [Indexed: 01/06/2023] Open
Abstract
Objective Insulin resistance can be triggered by enhanced dephosphorylation of the insulin receptor or downstream components in the insulin signaling cascade through protein tyrosine phosphatases (PTPs). Downregulating density-enhanced phosphatase-1 (DEP-1) resulted in an improved metabolic status in previous analyses. This phenotype was primarily caused by hepatic DEP-1 reduction. Methods Here we further elucidated the role of DEP-1 in glucose homeostasis by employing a conventional knockout model to explore the specific contribution of DEP-1 in metabolic tissues. Ptprj−/− (DEP-1 deficient) and wild-type C57BL/6 mice were fed a low-fat or high-fat diet. Metabolic phenotyping was combined with analyses of phosphorylation patterns of insulin signaling components. Additionally, experiments with skeletal muscle cells and muscle tissue were performed to assess the role of DEP-1 for glucose uptake. Results High-fat diet fed-Ptprj−/− mice displayed enhanced insulin sensitivity and improved glucose tolerance. Furthermore, leptin levels and blood pressure were reduced in Ptprj−/− mice. DEP-1 deficiency resulted in increased phosphorylation of components of the insulin signaling cascade in liver, skeletal muscle and adipose tissue after insulin challenge. The beneficial effect on glucose homeostasis in vivo was corroborated by increased glucose uptake in skeletal muscle cells in which DEP-1 was downregulated, and in skeletal muscle of Ptprj−/− mice. Conclusion Together, these data establish DEP-1 as novel negative regulator of insulin signaling.
Collapse
Key Words
- DEP-1, density-enhanced phosphatase-1
- Density-enhanced phosphatase-1
- GTT, glucose tolerance test
- Glucose homeostasis
- HFD, high-fat diet
- IL-6, interleukin 6
- IR, insulin receptor
- ITT, insulin tolerance test
- Insulin resistance
- Insulin signaling
- KO, knockout
- LFD, low-fat diet
- MCP-1, monocyte chemotactic protein-1
- PTP, protein tyrosine phosphatase
- Phosphorylation
- RER, respiratory exchange ratio
- RTK, receptor tyrosine kinase
- WT, wild-type
Collapse
Affiliation(s)
- Janine Krüger
- Center for Cardiovascular Research/CCR, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Hessische Str. 3-4, 10115 Berlin, Charité - Universitätsmedizin Berlin, Germany
| | - Sebastian Brachs
- Center for Cardiovascular Research/CCR, Department of Endocrinology, Diabetes and Nutrition, Hessische Str. 3-4, 10115 Berlin, Charité - Universitätsmedizin Berlin, Germany
| | - Manuela Trappiel
- Center for Cardiovascular Research/CCR, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Hessische Str. 3-4, 10115 Berlin, Charité - Universitätsmedizin Berlin, Germany
| | - Ulrich Kintscher
- Center for Cardiovascular Research/CCR, Institute of Pharmacology, Hessische Str. 3-4, 10115 Berlin, Charité - Universitätsmedizin Berlin, Germany
| | - Heike Meyborg
- Department of Medicine/Cardiology, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Ernst Wellnhofer
- Department of Medicine/Cardiology, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Christa Thöne-Reineke
- Center for Cardiovascular Research/CCR, Department of Experimental Medicine, Hessische Str. 3-4, 10115 Berlin, Charité - Universitätsmedizin Berlin, Germany
| | - Philipp Stawowy
- Department of Medicine/Cardiology, Deutsches Herzzentrum Berlin, Augustenburger Platz 1, 13353 Berlin, Germany
| | - Arne Östman
- Cancer Center Karolinska, R8:03, Department of Oncology-Pathology, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Andreas L Birkenfeld
- Center for Cardiovascular Research/CCR, Department of Endocrinology, Diabetes and Nutrition, Hessische Str. 3-4, 10115 Berlin, Charité - Universitätsmedizin Berlin, Germany
| | - Frank D Böhmer
- Center for Molecular Biomedicine, Institute of Molecular Cell Biology, Universitätsklinikum Jena, Hans-Knöll-Str. 2, 07745 Jena, Germany
| | - Kai Kappert
- Center for Cardiovascular Research/CCR, Institute of Laboratory Medicine, Clinical Chemistry and Pathobiochemistry, Hessische Str. 3-4, 10115 Berlin, Charité - Universitätsmedizin Berlin, Germany
| |
Collapse
|
21
|
Kandadi MR, Panzhinskiy E, Roe ND, Nair S, Hu D, Sun A. Deletion of protein tyrosine phosphatase 1B rescues against myocardial anomalies in high fat diet-induced obesity: Role of AMPK-dependent autophagy. Biochim Biophys Acta Mol Basis Dis 2015; 1852:299-309. [DOI: 10.1016/j.bbadis.2014.07.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Revised: 06/20/2014] [Accepted: 07/03/2014] [Indexed: 01/11/2023]
|
22
|
Lees EK, Krol E, Shearer K, Mody N, Gettys TW, Delibegovic M. Effects of hepatic protein tyrosine phosphatase 1B and methionine restriction on hepatic and whole-body glucose and lipid metabolism in mice. Metabolism 2015; 64:305-14. [PMID: 25468142 PMCID: PMC4390031 DOI: 10.1016/j.metabol.2014.10.038] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2014] [Revised: 10/15/2014] [Accepted: 10/29/2014] [Indexed: 02/06/2023]
Abstract
AIMS Methionine restriction (MR) and hepatic protein tyrosine phosphatase 1B (PTP1B) knockdown both improve hepatic insulin sensitivity by targeting different proteins within the insulin signaling pathway, as well as diminishing hepatic triglyceride content through decreasing hepatic lipogenesis. We hypothesized that a combined approach of hepatic PTP1B inhibition and methionine restriction could lead to a synergistic effect on improvements in glucose homeostasis and lipid metabolism. METHODS Male and female hepatic PTP1B knockout (Alb-Ptp1b(-/-)) and control wild-type (Ptp1b(fl/fl)) mice were maintained on control diet (0.86% methionine) or MR diet (0.172% methionine) for 8weeks. Body weight and food intake were recorded and physiological tests for whole-body glucose homeostasis were performed. Serum and tissues were analyzed biochemically. RESULTS MR decreased body weight and increased food intake in Ptp1b(fl/fl) mice as expected, without changing PTP1B protein expression levels or activity. In females, MR treatment alone improved glucose tolerance in Ptp1b(fl/fl) mice, which was further amplified with hepatic PTP1B deficiency. However, other markers of glucose homeostasis were similar between MR-fed groups. In males, MR improved glucose homeostasis in both, Alb-Ptp1b(-/-) and wild-type Ptp1b(fl/fl) mice to a similar extent. Hepatic PTP1B inhibition in combination with MR could not further enhance insulin-stimulated hepatic protein kinase B/Akt phosphorylation compared to MR treatment alone and therefore led to no further increase in hepatic insulin signaling. The combined treatment did not further improve lipid metabolism relative to MR diet alone. CONCLUSIONS Methionine restriction improves glucose and lipid homeostasis; however, adding hepatic PTP1B inhibition to MR is unlikely to yield any additional protective effects.
Collapse
Affiliation(s)
- Emma Katherine Lees
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Aberdeen, UK.
| | - Elzbieta Krol
- School of Biological Sciences, University of Aberdeen, Aberdeen, UK.
| | - Kirsty Shearer
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Aberdeen, UK.
| | - Nimesh Mody
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Aberdeen, UK.
| | - Thomas W Gettys
- Nutrient Sensing and Adipocyte Signaling Department, Pennington Biomedical Research Center, Baton Rouge, LA 70808, USA.
| | - Mirela Delibegovic
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Aberdeen, UK.
| |
Collapse
|
23
|
Gurzov EN, Stanley WJ, Brodnicki TC, Thomas HE. Protein tyrosine phosphatases: molecular switches in metabolism and diabetes. Trends Endocrinol Metab 2015; 26:30-9. [PMID: 25432462 DOI: 10.1016/j.tem.2014.10.004] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 10/28/2014] [Accepted: 10/30/2014] [Indexed: 02/06/2023]
Abstract
Protein tyrosine phosphatases (PTPs) are a large family of enzymes that generally oppose the actions of protein tyrosine kinases (PTKs). Genetic polymorphisms for particular PTPs are associated with altered risk of both type 1 diabetes (T1D) and type 2 diabetes (T2D). Moreover, recent evidence suggests that PTPs play crucial roles in metabolism. They can act as regulators of liver homeostasis, food intake, or immune-mediated pancreatic b cell death. In this review we describe the mechanisms by which different members of the non-receptor PTP (PTPN) family influence metabolic physiology. This 'metabolic job' of PTPs is discussed in depth and the role of these proteins in different cell types compared. Understanding the pathways regulated by PTPs will provide novel therapeutic strategies for the treatment of diabetes.
Collapse
|
24
|
Lucas E, Cruces-Sande M, Briones AM, Salaices M, Mayor F, Murga C, Vila-Bedmar R. Molecular physiopathology of obesity-related diseases: multi-organ integration by GRK2. Arch Physiol Biochem 2015; 121:163-77. [PMID: 26643283 DOI: 10.3109/13813455.2015.1107589] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Obesity is a worldwide problem that has reached epidemic proportions both in developed and developing countries. The excessive accumulation of fat poses a risk to health since it favours the development of metabolic alterations including insulin resistance and tissue inflammation, which further contribute to the progress of the complex pathological scenario observed in the obese. In this review we put together the different outcomes of fat accumulation and insulin resistance in the main insulin-responsive tissues, and discuss the role of some of the key molecular routes that control disease progression both in an organ-specific and also in a more systemic manner. In particular, we focus on the importance of studying the integrated regulation of different organs and pathways that contribute to the global pathophysiology of this condition with a specific emphasis on the role of emerging key molecular nodes such as the G protein-coupled receptor kinase 2 (GRK2) signalling hub.
Collapse
Affiliation(s)
- Elisa Lucas
- a Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC) , Universidad Autónoma de Madrid , Madrid , Spain
- b Instituto de Investigación Sanitaria La Princesa , Madrid , Spain
| | - Marta Cruces-Sande
- a Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC) , Universidad Autónoma de Madrid , Madrid , Spain
- b Instituto de Investigación Sanitaria La Princesa , Madrid , Spain
| | - Ana M Briones
- c Departamento de Farmacología , Universidad Autónoma de Madrid (UAM) Madrid , Spain , and
- d Instituto de Investigación Hospital Universitario La Paz (IdiPAZ) Madrid , Spain
| | - Mercedes Salaices
- c Departamento de Farmacología , Universidad Autónoma de Madrid (UAM) Madrid , Spain , and
- d Instituto de Investigación Hospital Universitario La Paz (IdiPAZ) Madrid , Spain
| | - Federico Mayor
- a Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC) , Universidad Autónoma de Madrid , Madrid , Spain
- b Instituto de Investigación Sanitaria La Princesa , Madrid , Spain
| | - Cristina Murga
- a Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC) , Universidad Autónoma de Madrid , Madrid , Spain
- b Instituto de Investigación Sanitaria La Princesa , Madrid , Spain
| | - Rocio Vila-Bedmar
- a Departamento de Biología Molecular and Centro de Biología Molecular Severo Ochoa (UAM-CSIC) , Universidad Autónoma de Madrid , Madrid , Spain
- b Instituto de Investigación Sanitaria La Princesa , Madrid , Spain
| |
Collapse
|
25
|
Li H, Dusseault J, Larose L. Nck1 depletion induces activation of the PI3K/Akt pathway by attenuating PTP1B protein expression. Cell Commun Signal 2014; 12:71. [PMID: 25398386 PMCID: PMC4236421 DOI: 10.1186/s12964-014-0071-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2014] [Accepted: 10/19/2014] [Indexed: 12/17/2022] Open
Abstract
Background Activation of the PI3K/Akt pathway mediates crucial cellular functions regulated by receptor tyrosine kinases, such as cell growth, proliferation, survival and metabolism. Previously, we reported that the whole-body knockout of the Src homology domain-containing adaptor protein Nck1 improves overall glucose homeostasis and insulin-induced activation of the PI3K/Akt pathway in liver of obese mice. The aim of the current study is to elucidate the mechanism by which Nck1 depletion regulates hepatic insulin signaling. Results Here, we demonstrate that Nck1 regulates the activation of the PI3K/Akt pathway in a protein tyrosine phosphatase 1B (PTP1B)-dependent mechanism. Indeed, depletion of Nck1 by siRNA in HepG2 cells enhances PI3K-dependent basal and growth factor-induced Akt activation. In accordance, primary hepatocytes isolated from Nck1−/− mice also display enhanced Akt activation in response to insulin. Activation of the PI3K/Akt pathway in Nck1-depleted HepG2 cells relies on higher levels of tyrosine-phosphorylated proteins and correlates with decreased PTP1B levels. Interestingly, Nck1 and PTP1B in cells are found in a common molecular complex and their interaction is dependent on the SH3 domains of Nck1. Finally, Nck1 depletion in HepG2 cells neither affects PTP1B gene transcription nor PTP1B protein stability, suggesting that Nck1 modulates PTP1B expression at the translational level. Conclusion Our study provides strong evidence supporting that the adaptor protein Nck1 interacts with PTP1B and also regulates PTP1B expression. In this manner, Nck1 plays a role in regulating the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Hui Li
- Department of Medicine, Polypeptide Laboratory, McGill University and The Research Institute of McGill University Health Centre, Montreal, QC, Canada.
| | - Julie Dusseault
- Department of Medicine, Polypeptide Laboratory, McGill University and The Research Institute of McGill University Health Centre, Montreal, QC, Canada.
| | - Louise Larose
- Department of Medicine, Polypeptide Laboratory, McGill University and The Research Institute of McGill University Health Centre, Montreal, QC, Canada.
| |
Collapse
|
26
|
Agouni A, Tual-Chalot S, Chalopin M, Duluc L, Mody N, Martinez MC, Andriantsitohaina R, Delibegović M. Hepatic protein tyrosine phosphatase 1B (PTP1B) deficiency protects against obesity-induced endothelial dysfunction. Biochem Pharmacol 2014; 92:607-17. [PMID: 25451690 DOI: 10.1016/j.bcp.2014.10.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Revised: 10/14/2014] [Accepted: 10/21/2014] [Indexed: 11/26/2022]
Abstract
Growing evidence suggests that hepatic-insulin resistance is sufficient to promote progression to cardiovascular disease. We have shown previously that liver-specific protein-tyrosine-phosphatase 1B (PTP1B) deficiency improves hepatic-insulin sensitivity and whole-body glucose homeostasis. The aim of this study was to investigate the impact of liver-specific PTP1B-deficiency (L-PTP1B-/-) on cardiac and peripheral vascular function, with special emphasis on endothelial function in the context of high-fat diet (HFD)-induced obesity. L-PTP1B-/- mice exhibited an improved glucose and lipid homeostasis and increased insulin sensitivity, without changes in body weight. HFD-feeding increased systolic blood pressure (BP) in both L-PTP1B-/- and control littermates; however, this was significantly lower in L-PTP1B-/- mice. HFD-feeding increased diastolic BP in control mice only, whilst the L-PTP1B-/- mice were completely protected. The analysis of the function of the left ventricle (LV) revealed that HFD-feeding decreased LV fractional shortening in control animals, which was not observed in L-PTP1B-/- mice. Importantly, HFD feeding significantly impaired endothelium-dependent vasorelaxation in response to acetylcholine in aortas from control mice, whilst L-PTP1B-/- mice were fully protected. This was associated with alterations in eNOS phosphorylation. Selective inhibition of COX-2, using NS-398, decreased the contractile response in response to serotonin (5-HT) only in vessels from control mice. HFD-fed control mice released enhanced levels of prostaglandin E, a vasoconstrictor metabolite; whilst both chow- and HFD-fed L-PTP1B-/- mice released higher levels of prostacylin, a vasorelaxant metabolite. Our data indicate that hepatic-PTP1B inhibition protects against HFD-induced endothelial dysfunction, underscoring the potential of peripheral PTP1B inhibitors in reduction of obesity-associated cardiovascular risk in addition to its anti-diabetic effects.
Collapse
Affiliation(s)
- Abdelali Agouni
- University of Aberdeen, Institute of Medical Sciences, School of Medical Sciences, Foresterhill Health Campus, Aberdeen AB25 2ZD, United Kingdom; University of Surrey, Faculty of Health and Medical Sciences, Department of Biochemistry and Physiology, Guildford GU2 7XH, United Kingdom
| | - Simon Tual-Chalot
- LUNAM Université, Angers, France; INSERM U1063, Stress Oxydant et Pathologies Métaboliques, Angers, France
| | - Matthieu Chalopin
- LUNAM Université, Angers, France; INSERM U1063, Stress Oxydant et Pathologies Métaboliques, Angers, France
| | - Lucie Duluc
- LUNAM Université, Angers, France; INSERM U1063, Stress Oxydant et Pathologies Métaboliques, Angers, France
| | - Nimesh Mody
- University of Aberdeen, Institute of Medical Sciences, School of Medical Sciences, Foresterhill Health Campus, Aberdeen AB25 2ZD, United Kingdom
| | - M Carmen Martinez
- LUNAM Université, Angers, France; INSERM U1063, Stress Oxydant et Pathologies Métaboliques, Angers, France; CHU Angers, France
| | - Ramaroson Andriantsitohaina
- LUNAM Université, Angers, France; INSERM U1063, Stress Oxydant et Pathologies Métaboliques, Angers, France; CHU Angers, France.
| | - Mirela Delibegović
- University of Aberdeen, Institute of Medical Sciences, School of Medical Sciences, Foresterhill Health Campus, Aberdeen AB25 2ZD, United Kingdom.
| |
Collapse
|
27
|
Ding H, Zhang Y, Xu C, Hou D, Li J, Zhang Y, Peng W, Zen K, Zhang CY, Jiang X. Norathyriol reverses obesity- and high-fat-diet-induced insulin resistance in mice through inhibition of PTP1B. Diabetologia 2014; 57:2145-54. [PMID: 24985145 DOI: 10.1007/s00125-014-3315-8] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/06/2014] [Indexed: 12/17/2022]
Abstract
AIM/HYPOTHESIS Protein tyrosine phosphatase 1B (PTP1B) negatively regulates insulin signalling. PTP1B deficiency improves obesity-induced insulin resistance and consequently improves type 2 diabetes in mice. Here, the small molecule norathyriol reversed obesity- and high-fat-diet-induced insulin resistance by inhibiting PTP1B. METHODS The inhibitory mode of PTP1B was evaluated by using the double-reciprocal substrate in the presence of norathyriol. Primary cultured hepatocytes, myoblasts and white adipocytes were used to investigate the effect of norathyriol on insulin signalling. Glucose homeostasis and insulin sensitivity were characterised by glucose and insulin tolerance tests. RESULTS Norathyriol was identified as a competitive inhibitor of PTP1B, with an IC50 of 9.59 ± 0.39 μmol/l. In cultured hepatocytes and myoblasts, norathyriol treatment blocked the PTP1B-mediated dephosphorylation of the insulin receptor. Intraperitoneal injection of norathyriol inhibited liver and muscle PTP1B activity in mice, thus contributing to the improved glucose homeostasis and insulin sensitivity. However, these beneficial effects were abolished in PTP1B-deficient mice. Notably, oral administration of norathyriol protected mice from diet-induced obesity and insulin resistance through inhibition of hypothalamic PTP1B activity. CONCLUSIONS/INTERPRETATION Our results indicate that the small molecule norathyriol is a potent PTP1B inhibitor with good cell permeability and oral availability.
Collapse
Affiliation(s)
- Hanying Ding
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, 210093, Nanjing, Jiangsu, People's Republic of China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Liu S, Xi Y, Bettaieb A, Matsuo K, Matsuo I, Kulkarni RN, Haj FG. Disruption of protein-tyrosine phosphatase 1B expression in the pancreas affects β-cell function. Endocrinology 2014; 155:3329-38. [PMID: 24956127 PMCID: PMC4138572 DOI: 10.1210/en.2013-2004] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Protein-tyrosine phosphatase 1B (PTP1B) is a physiological regulator of glucose homeostasis and energy balance. However, the role of PTP1B in pancreatic endocrine function remains largely unknown. To investigate the metabolic role of pancreatic PTP1B, we generated mice with pancreas PTP1B deletion (panc-PTP1B KO). Mice were fed regular chow or a high-fat diet, and metabolic parameters, insulin secretion and glucose tolerance were determined. On regular chow, panc-PTP1B KO and control mice exhibited comparable glucose tolerance whereas aged panc-PTP1B KO exhibited mild glucose intolerance. Furthermore, high-fat feeding promoted earlier impairment of glucose tolerance and attenuated glucose-stimulated insulin secretion in panc-PTP1B KO mice. The secretory defect in glucose-stimulated insulin secretion was recapitulated in primary islets ex vivo, suggesting that the effects were likely cell-autonomous. At the molecular level, PTP1B deficiency in vivo enhanced basal and glucose-stimulated tyrosyl phosphorylation of EphA5 in islets. Consistently, PTP1B overexpression in the glucose-responsive MIN6 β-cell line attenuated EphA5 tyrosyl phosphorylation, and substrate trapping identified EphA5 as a PTP1B substrate. In summary, these studies identify a novel role for PTP1B in pancreatic endocrine function.
Collapse
Affiliation(s)
- Siming Liu
- Nutrition Department (S.L., Y.X., A.B., K.M., I.M., F.G.H.), University of California Davis, Davis, California 95616; Joslin Diabetes Center (R.N.K.), Department of Medicine, Harvard Medical School, Boston, Massachusetts 02215; and Division of Endocrinology, Diabetes and Metabolism (F.G.H.), Department of Internal Medicine, and Comprehensive Cancer Center, University of California Davis, Sacramento, California 95817
| | | | | | | | | | | | | |
Collapse
|
29
|
Gouranton E, Romier B, Marcotorchino J, Tourniaire F, Astier J, Peiretti F, Landrier JF. Visfatin is involved in TNFα-mediated insulin resistance via an NAD(+)/Sirt1/PTP1B pathway in 3T3-L1 adipocytes. Adipocyte 2014; 3:180-9. [PMID: 25068084 PMCID: PMC4110094 DOI: 10.4161/adip.28729] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Revised: 03/21/2014] [Accepted: 03/31/2014] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor α (TNFα) is a well-known mediator of inflammation in the context of obesity in adipose tissue. Its action appears to be directly linked to perturbations of the insulin pathway, leading to the development of insulin resistance. Visfatin has been suspected to be linked to insulin sensitivity, but the mechanism involved is still partly unknown. The aim of this study was to evaluate the role of visfatin in the impairment of the insulin pathway by TNFα activity in 3T3-L1 adipocytes and to unveil the mechanisms involved in such impairment.
We demonstrated in 3T3-L1 adipocytes that visfatin was involved in TNFα-mediated insulin resistance in adipocytes. Indeed, after TNFα treatment in 3T3-L1 cells, visfatin was downregulated, leading to decreased nicotinamide adenine dinucleotide (NAD+) concentrations in cells. This decrease was followed by a decrease in Sirt1 activity, which was linked to an increase in PTP1B expression. The modulation of PTP1B by visfatin was likely responsible for the observed decreases in glucose uptake and Akt phosphorylation in 3T3-L1 adipocytes.
Here, we demonstrated a complete pathway involving visfatin, NAD+, Sirt1, and PTP1B that led to the perturbation of insulin signaling by TNFα in 3T3-L1 adipocytes.
Collapse
|
30
|
Owen C, Lees EK, Mody N, Delibegović M. Regulation of growth hormone induced JAK2 and mTOR signalling by hepatic protein tyrosine phosphatase 1B. DIABETES & METABOLISM 2014; 41:95-101. [PMID: 24948418 DOI: 10.1016/j.diabet.2014.02.008] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2013] [Revised: 02/13/2014] [Accepted: 02/18/2014] [Indexed: 01/25/2023]
Abstract
Protein tyrosine phosphatase 1B (PTP1B) regulates various signalling pathways including insulin, leptin, IGF-1 and growth hormone (GH) signalling. Transmission of the GH signal depends on Janus kinase 2 (JAK2), which is how PTP1B is thought to modulate GH signalling in the liver, based on studies utilising global PTP1B knockout mice (Ptp1b(-/-)). Here, we investigated the liver-specific role of PTP1B in GH signalling, using liver-specific Ptp1b(-/-) mice (alb-crePtp1b(-/-)), under physiological (chow) or insulin resistant (high-fat diet [HFD]) feeding conditions. Body weight and adiposity were comparable between female alb-crePtp1b(-/-) and Ptp1b(fl/fl) control mice. On chow diet, under 48-hour fasting GH-resistant conditions, GH stimulation in vivo led to a robust stimulation of the JAK-STAT signalling pathway. Alb-crePtp1b(-/-) mice exhibited significantly higher GH-induced JAK2 phosphorylation and SOCS3 gene expression post-GH stimulation. However, STAT3, STAT5 and ERK1/2 phosphorylation and SOCS2 gene expression were similar between groups. Interestingly, GH-induced mTOR phosphorylation was significantly higher in alb-crePtp1b(-/-) mice 5-min post-GH stimulation compared to controls, revealing this part of the pathway under direct control of PTP1B. Under ad lib HFD-fed conditions, GH-induced STAT5 phosphorylation significantly increased in alb-crePtp1b(-/-) mice only, with no alterations in the controls. Overall, our data demonstrate that liver-specific PTP1B deletion leads to significant alterations in GH signalling with increased JAK2, STAT5 and mTOR phosphorylation and SOCS3 gene expression.
Collapse
Affiliation(s)
- C Owen
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - E K Lees
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - N Mody
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom
| | - M Delibegović
- Institute of Medical Sciences, School of Medical Sciences, University of Aberdeen, Foresterhill, Aberdeen, AB25 2ZD, United Kingdom.
| |
Collapse
|
31
|
Gealekman O, Gurav K, Chouinard M, Straubhaar J, Thompson M, Malkani S, Hartigan C, Corvera S. Control of adipose tissue expandability in response to high fat diet by the insulin-like growth factor-binding protein-4. J Biol Chem 2014; 289:18327-38. [PMID: 24778188 DOI: 10.1074/jbc.m113.545798] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Adipose tissue expansion requires growth and proliferation of adipocytes and the concomitant expansion of their stromovascular network. We have used an ex vivo angiogenesis assay to study the mechanisms involved in adipose tissue expansion. In this assay, adipose tissue fragments placed under pro-angiogenic conditions form sprouts composed of endothelial, perivascular, and other proliferative cells. We find that sprouting was directly stimulated by insulin and was enhanced by prior treatment of mice with the insulin sensitizer rosiglitazone. Moreover, basal and insulin-stimulated sprouting increased progressively over 30 weeks of high fat diet feeding, correlating with tissue expansion during this period. cDNA microarrays analyzed to identify genes correlating with insulin-stimulated sprouting surprisingly revealed only four positively correlating (Fads3, Tmsb10, Depdc6, and Rasl12) and four negatively correlating (Asph, IGFbp4, Ppm1b, and Adcyap1r1) genes. Among the proteins encoded by these genes, IGFbp4, which suppresses IGF-1 signaling, has been previously implicated in angiogenesis, suggesting a role for IGF-1 in adipose tissue expandability. Indeed, IGF-1 potently stimulated sprouting, and the presence of activated IGF-1 receptors in the vasculature was revealed by immunostaining. Recombinant IGFbp4 blocked the effects of insulin and IGF-1 on mouse adipose tissue sprouting and also suppressed sprouting from human subcutaneous adipose tissue. These results reveal an important role of IGF-1/IGFbp4 signaling in post-developmental adipose tissue expansion.
Collapse
Affiliation(s)
| | | | | | | | - Michael Thompson
- Department of Medicine, and Diabetes Center of Excellence, University of Massachusetts Medical School, Worcester, Massachusetts 01605
| | | | | | | |
Collapse
|
32
|
Xu E, Schwab M, Marette A. Role of protein tyrosine phosphatases in the modulation of insulin signaling and their implication in the pathogenesis of obesity-linked insulin resistance. Rev Endocr Metab Disord 2014; 15:79-97. [PMID: 24264858 DOI: 10.1007/s11154-013-9282-4] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Insulin resistance is a major disorder that links obesity to type 2 diabetes mellitus (T2D). It involves defects in the insulin actions owing to a reduced ability of insulin to trigger key signaling pathways in major metabolic tissues. The pathogenesis of insulin resistance involves several inhibitory molecules that interfere with the tyrosine phosphorylation of the insulin receptor and its downstream effectors. Among those, growing interest has been developed toward the protein tyrosine phosphatases (PTPs), a large family of enzymes that can inactivate crucial signaling effectors in the insulin signaling cascade by dephosphorylating their tyrosine residues. Herein we briefly review the role of several PTPs that have been shown to be implicated in the regulation of insulin action, and then focus on the Src homology 2 (SH2) domain-containing SHP1 and SHP2 enzymes, since recent reports have indicated major roles for these PTPs in the control of insulin action and glucose metabolism. Finally, the therapeutic potential of targeting PTPs for combating insulin resistance and alleviating T2D will be discussed.
Collapse
Affiliation(s)
- Elaine Xu
- Department of Medicine, Cardiology Axis of the Institut Universitaire de Cardiologie et de Pneumologie de Québec (Hôpital Laval), Ste-Foy, Québec, Canada, G1V 4G2
| | | | | |
Collapse
|
33
|
Knobler H, Elson A. Metabolic regulation by protein tyrosine phosphatases. J Biomed Res 2014; 28:157-68. [PMID: 25013399 PMCID: PMC4085553 DOI: 10.7555/jbr.28.20140012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2014] [Accepted: 01/28/2014] [Indexed: 01/14/2023] Open
Abstract
Obesity and the metabolic syndrome and their associated morbidities are major public health issues, whose prevalence will continue to increase in the foreseeable future. Aberrant signaling by the receptors for leptin and insulin plays a pivotal role in development of the metabolic syndrome. More complete molecular-level understanding of how both of these key signaling pathways are regulated is essential for full characterization of obesity, the metabolic syndrome, and type II diabetes, and for developing novel treatments for these diseases. Phosphorylation of proteins on tyrosine residues plays a key role in mediating the effects of leptin and insulin on their target cells. Here, we discuss the molecular methods by which protein tyrosine phosphatases, which are key physiological regulators of protein phosphorylation in vivo, affect signaling by the leptin and insulin receptors in their major target tissues.
Collapse
Affiliation(s)
- Hilla Knobler
- Diabetes and Metabolic Disease Unit, Kaplan Medical Center, Rehovot 76100, Israel
| | - Ari Elson
- Department of Molecular Genetics, the Weizmann Institute of Science, Rehovot 76100, Israel
| |
Collapse
|
34
|
Fernandez-Ruiz R, Vieira E, Garcia-Roves PM, Gomis R. Protein tyrosine phosphatase-1B modulates pancreatic β-cell mass. PLoS One 2014; 9:e90344. [PMID: 24587334 PMCID: PMC3938680 DOI: 10.1371/journal.pone.0090344] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 02/02/2014] [Indexed: 12/31/2022] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of the insulin signalling pathway. It has been demonstrated that PTP1B deletion protects against the development of obesity and Type 2 Diabetes, mainly through its action on peripheral tissues. However, little attention has been paid to the role of PTP1B in β-cells. Therefore, our aim was to study the role of PTP1B in pancreatic β-cells. Silencing of PTP1B expression in a pancreatic β-cell line (MIN6 cells) reveals the significance of this endoplasmic reticulum bound phosphatase in the regulation of cell proliferation and apoptosis. Furthermore, the ablation of PTP1B is able to regulate key proteins involved in the proliferation and/or apoptosis pathways, such as STAT3, AKT, ERK1/2 and p53 in isolated islets from PTP1B knockout (PTP1B −/−) mice. Morphometric analysis of pancreatic islets from PTP1B −/− mice showed a higher β-cell area, concomitantly with higher β-cell proliferation and a lower β-cell apoptosis when compared to islets from their respective wild type (WT) littermates. At a functional level, isolated islets from 8 weeks old PTP1B −/− mice exhibit enhanced glucose-stimulated insulin secretion. Moreover, PTP1B −/− mice were able to partially reverse streptozotocin-induced β-cell loss. Together, our data highlight for the first time the involvement of PTP1B in β-cell physiology, reinforcing the potential of this phosphatase as a therapeutical target for the treatment of β-cell failure, a central aspect in the pathogenesis of Type 2 Diabetes.
Collapse
MESH Headings
- Animals
- Apoptosis
- Cell Count
- Cell Line
- Cell Proliferation
- Diabetes Mellitus, Experimental/chemically induced
- Diabetes Mellitus, Experimental/enzymology
- Diabetes Mellitus, Experimental/genetics
- Diabetes Mellitus, Experimental/pathology
- Endoplasmic Reticulum/chemistry
- Endoplasmic Reticulum/enzymology
- Gene Expression Regulation
- Glucose/metabolism
- Glucose/pharmacology
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells/drug effects
- Insulin-Secreting Cells/enzymology
- Insulin-Secreting Cells/pathology
- Male
- Mice
- Mice, Knockout
- Mitogen-Activated Protein Kinase 1/genetics
- Mitogen-Activated Protein Kinase 1/metabolism
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/antagonists & inhibitors
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics
- Protein Tyrosine Phosphatase, Non-Receptor Type 1/metabolism
- Proto-Oncogene Proteins c-akt/genetics
- Proto-Oncogene Proteins c-akt/metabolism
- RNA, Small Interfering/genetics
- RNA, Small Interfering/metabolism
- STAT3 Transcription Factor/genetics
- STAT3 Transcription Factor/metabolism
- Signal Transduction
- Streptozocin
- Tumor Suppressor Protein p53/genetics
- Tumor Suppressor Protein p53/metabolism
Collapse
Affiliation(s)
- Rebeca Fernandez-Ruiz
- Diabetes and Obesity Research Laboratory, Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Elaine Vieira
- Diabetes and Obesity Research Laboratory, Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Pablo M. Garcia-Roves
- Diabetes and Obesity Research Laboratory, Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
| | - Ramon Gomis
- Diabetes and Obesity Research Laboratory, Institut d’Investigacions Biomediques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain
- Universitat de Barcelona, Barcelona, Spain
- Hospital Clinic de Barcelona, Barcelona, Spain
- * E-mail:
| |
Collapse
|
35
|
|
36
|
Grant L, Shearer KD, Czopek A, Lees EK, Owen C, Agouni A, Workman J, Martin-Granados C, Forrester JV, Wilson HM, Mody N, Delibegovic M. Myeloid-cell protein tyrosine phosphatase-1B deficiency in mice protects against high-fat diet and lipopolysaccharide-induced inflammation, hyperinsulinemia, and endotoxemia through an IL-10 STAT3-dependent mechanism. Diabetes 2014; 63:456-70. [PMID: 24186864 DOI: 10.2337/db13-0885] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Protein tyrosine phosphatase-1B (PTP1B) negatively regulates insulin and leptin signaling, rendering it an attractive drug target for treatment of obesity-induced insulin resistance. However, some studies suggest caution when targeting macrophage PTP1B, due to its potential anti-inflammatory role. We assessed the role of macrophage PTP1B in inflammation and whole-body metabolism using myeloid-cell (LysM) PTP1B knockout mice (LysM PTP1B). LysM PTP1B mice were protected against lipopolysaccharide (LPS)-induced endotoxemia and hepatic damage associated with decreased proinflammatory cytokine secretion in vivo. In vitro, LPS-treated LysM PTP1B bone marrow-derived macrophages (BMDMs) displayed increased interleukin (IL)-10 mRNA expression, with a concomitant decrease in TNF-α mRNA levels. These anti-inflammatory effects were associated with increased LPS- and IL-10-induced STAT3 phosphorylation in LysM PTP1B BMDMs. Chronic inflammation induced by high-fat (HF) feeding led to equally beneficial effects of macrophage PTP1B deficiency; LysM PTP1B mice exhibited improved glucose and insulin tolerance, protection against LPS-induced hyperinsulinemia, decreased macrophage infiltration into adipose tissue, and decreased liver damage. HF-fed LysM PTP1B mice had increased basal and LPS-induced IL-10 levels, associated with elevated STAT3 phosphorylation in splenic cells, IL-10 mRNA expression, and expansion of cells expressing myeloid markers. These increased IL-10 levels negatively correlated with circulating insulin and alanine transferase levels. Our studies implicate myeloid PTP1B in negative regulation of STAT3/IL-10-mediated signaling, highlighting its inhibition as a potential anti-inflammatory and antidiabetic target in obesity.
Collapse
Affiliation(s)
- Louise Grant
- Institute of Medical Sciences, University of Aberdeen, College of Life Sciences and Medicine, Foresterhill Health Campus, Aberdeen, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Tsou RC, Rak KS, Zimmer DJ, Bence KK. Improved metabolic phenotype of hypothalamic PTP1B-deficiency is dependent upon the leptin receptor. Mol Metab 2014; 3:301-12. [PMID: 24749060 PMCID: PMC3986631 DOI: 10.1016/j.molmet.2014.01.008] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 01/05/2014] [Accepted: 01/11/2014] [Indexed: 12/14/2022] Open
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a known regulator of central metabolic signaling, and mice with whole brain-, leptin receptor (LepRb) expressing cell-, or proopiomelanocortin neuron-specific PTP1B-deficiency are lean, leptin hypersensitive, and display improved glucose homeostasis. However, whether the metabolic effects of central PTP1B-deficiency are due to action within the hypothalamus remains unclear. Moreover, whether or not these effects are exclusively due to enhanced leptin signaling is unknown. Here we report that mice with hypothalamic PTP1B-deficiency (Nkx2.1-PTP1B(-/-)) display decreased body weight and adiposity on high-fat diet with no associated improvements in glucose tolerance. Consistent with previous reports, we find that hypothalamic deletion of the LepRb in mice (Nkx2.1-LepRb(-/-)) results in extreme hyperphagia and obesity. Interestingly, deletion of hypothalamic PTP1B and LepRb (Nkx2.1-PTP1B(-/-):LepRb(-/-)) does not rescue the hyperphagia or obesity of Nkx2.1-LepRb(-/-) mice, suggesting that hypothalamic PTP1B contributes to the central control of energy balance through a leptin receptor-dependent pathway.
Collapse
Key Words
- BAT, Brown adipose tissue
- CNTF, Ciliary neurotrophic factor
- Cre, Cre recombinase
- GTT, Glucose tolerance test
- HFD, High-fat diet
- HPA, hypothalamus–pituitary–adrenal
- Hypothalamus
- IL-6, Interleukin-6
- ITT, Insulin tolerance test
- JAK2, Janus kinase 2
- LepRb, Leptin receptor long form
- Leptin
- Nkx2.1, NK2 homeobox 1 protein or thyroid transcription factor-1
- Obesity
- PI3K, Phosphatidylinositol 3-kinase
- POMC, Proopiomelanocortin
- PTP1B, Protein tyrosine phosphatase 1B
- PTPs, Protein tyrosine phosphatases
- Phosphatase
- Prdm16, PR domain containing 16
- SHP2, Src homology 2 domain-containing protein tyrosine phosphatase
- STAT3, Signal transducer and activator of transcription 3
- UCP1, Uncoupling protein 1
- WAT, White adipose tissue
- db/db, Leptin receptor-deficient mice
- ob/ob, leptin-deficient mice
Collapse
Affiliation(s)
- Ryan C Tsou
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kimberly S Rak
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Derek J Zimmer
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kendra K Bence
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
38
|
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of the leptin and insulin signaling pathways. The important roles of PTP1B related to obesity and diabetes were confirmed by a deletion of PTP1B gene in mice. Mice with the whole body deletion of PTP1B were protected against the development of obesity and diabetes. When PTP1B gene was deleted selectively in the brain of mice, the major effects on weight and glucose control were consistent with the whole body deletion of PTP1B. This is in contrast to the muscle-, liver-, and adipocyte-specific deletion, which had no beneficial effects on obesity. While these results indicate the importance of neuronal PTP1B in maintaining energy homeostasis, the peripheral PTP1B is also being investigated for their potential roles in the control of energy balance. Validation of PTP1B as a therapeutic target for obesity and diabetes prompted efforts to develop potent and selective inhibitors of PTP1B. Among the small molecule inhibitors investigated, trodusquemine, which acts both centrally and peripherally, is currently in phase 2 clinical trials. An approach using PTP1B-directed antisense oligonucleotides is also in phase 2 clinical trials.
Collapse
Affiliation(s)
- Hyeongjin Cho
- Department of Chemistry, Inha University, Incheon, Korea.
| |
Collapse
|
39
|
Bakke J, Bettaieb A, Nagata N, Matsuo K, Haj FG. Regulation of the SNARE-interacting protein Munc18c tyrosine phosphorylation in adipocytes by protein-tyrosine phosphatase 1B. Cell Commun Signal 2013; 11:57. [PMID: 23937695 PMCID: PMC3751566 DOI: 10.1186/1478-811x-11-57] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 08/06/2013] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Protein-tyrosine phosphatase 1B (PTP1B) is a physiological regulator of insulin signaling and adiposity and is a drug target for the treatment of obesity and diabetes. The molecular mechanisms underlying PTP1B metabolic actions require additional investigation. RESULTS Herein, we identify Munc18c as a novel PTP1B substrate in adipocytes and in vivo. We demonstrate nutritional regulation of Munc18c in adipose tissue revealing decreased expression upon high fat feeding. In addition, PTP1B deficiency leads to elevated Munc18c tyrosine phosphorylation and dissociation from syntaxin4. At the molecular level, we identify Munc18c Tyr218/219 and Tyr521 as key residues that mediate Munc18c interaction with PTP1B. Further, we uncover an essential role of Munc18c total tyrosine phosphorylation in general, and Tyr218/219 and Tyr521 in particular, in regulating its interactions and glucose uptake in adipocytes. CONCLUSION In conclusion, our findings identify PTP1B as the first known tyrosine phosphatase for Munc18c and a regulator of its phosphorylation and function in adipocytes.
Collapse
Affiliation(s)
- Jesse Bakke
- Nutrition Department, University of California Davis, One Shields Ave, 3135 Meyer Hall, Davis, CA 95616, USA
| | | | | | | | | |
Collapse
|
40
|
Bettaieb A, Bakke J, Nagata N, Matsuo K, Xi Y, Liu S, AbouBechara D, Melhem R, Stanhope K, Cummings B, Graham J, Bremer A, Zhang S, Lyssiotis CA, Zhang ZY, Cantley LC, Havel PJ, Haj FG. Protein tyrosine phosphatase 1B regulates pyruvate kinase M2 tyrosine phosphorylation. J Biol Chem 2013; 288:17360-71. [PMID: 23640882 PMCID: PMC3682537 DOI: 10.1074/jbc.m112.441469] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 04/24/2013] [Indexed: 11/06/2022] Open
Abstract
Protein-tyrosine phosphatase 1B (PTP1B) is a physiological regulator of glucose homeostasis and adiposity and is a drug target for the treatment of obesity and diabetes. Here we identify pyruvate kinase M2 (PKM2) as a novel PTP1B substrate in adipocytes. PTP1B deficiency leads to increased PKM2 total tyrosine and Tyr(105) phosphorylation in cultured adipocytes and in vivo. Substrate trapping and mutagenesis studies identify PKM2 Tyr-105 and Tyr-148 as key sites that mediate PTP1B-PKM2 interaction. In addition, in vitro analyses illustrate a direct effect of Tyr-105 phosphorylation on PKM2 activity in adipocytes. Importantly, PTP1B pharmacological inhibition increased PKM2 Tyr-105 phosphorylation and decreased PKM2 activity. Moreover, PKM2 Tyr-105 phosphorylation is regulated nutritionally, decreasing in adipose tissue depots after high-fat feeding. Further, decreased PKM2 Tyr-105 phosphorylation correlates with the development of glucose intolerance and insulin resistance in rodents, non-human primates, and humans. Together, these findings identify PKM2 as a novel substrate of PTP1B and provide new insights into the regulation of adipose PKM2 activity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Kimber Stanhope
- From the Nutrition Department and
- Department of Molecular Biosciences, University of California Davis, Davis, California 95616
| | - Bethany Cummings
- From the Nutrition Department and
- Department of Molecular Biosciences, University of California Davis, Davis, California 95616
| | - James Graham
- From the Nutrition Department and
- Department of Molecular Biosciences, University of California Davis, Davis, California 95616
| | - Andrew Bremer
- the Department of Pediatrics, Vanderbilt University, Nashville, Tennessee 37232
| | - Sheng Zhang
- the Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, Indiana 46202
| | - Costas A. Lyssiotis
- the Beth Israel Deaconess Medical Center, Department of Medicine, and
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Zhong-Yin Zhang
- the Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, Indiana 46202
| | - Lewis C. Cantley
- the Beth Israel Deaconess Medical Center, Department of Medicine, and
- Department of Systems Biology, Harvard Medical School, Boston, Massachusetts 02115, and
| | - Peter J. Havel
- From the Nutrition Department and
- Department of Molecular Biosciences, University of California Davis, Davis, California 95616
| | - Fawaz G. Haj
- From the Nutrition Department and
- the Department of Internal Medicine and
- Comprehensive Cancer Center, University of California Davis, Sacramento, California 95817
| |
Collapse
|
41
|
Bae KH, Kim WK, Lee SC. Involvement of protein tyrosine phosphatases in adipogenesis: new anti-obesity targets? BMB Rep 2013; 45:700-6. [PMID: 23261055 PMCID: PMC4133817 DOI: 10.5483/bmbrep.2012.45.12.235] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Obesity is a worldwide epidemic as well as being a major risk factor for diabetes, cardiovascular diseases and several types of cancers. Obesity is mainly due to the overgrowth of adipose tissue arising from an imbalance between energy intake and energy expenditure. Adipose tissue, primarily composed of adipocytes, plays a key role in maintaining whole body energy homeostasis. In view of the treatment of obesity and obesity-related diseases, it is critical to understand the detailed signal transduction mechanisms of adipogenic differentiation. Adipogenic differentiation is tightly regulated by many key signal cascades, including insulin signaling. These signal cascades generally transfer or amplify the signal by using serial tyrosine phosphorylations. Thus, protein tyrosine kinases and protein tyrosine phosphatases are closely related to adipogenic differentiation. Compared to protein tyrosine kinases, protein tyrosine phosphatases have received little attention in adipogenic differentiation. This review aims to highlight the involvement of protein tyrosine phosphatases in adipogenic differentiation and the possibility of protein tyrosine phosphatases as drugs to target obesity. [BMB Reports 2012; 45(12): 700-706]
Collapse
Affiliation(s)
- Kwang-Hee Bae
- Medical Proteomics Research Center, KRIBB, Daejeon 305-806, Korea.
| | | | | |
Collapse
|
42
|
Song DD, Chen Y, Li ZY, Guan YF, Zou DJ, Miao CY. Protein tyrosine phosphatase 1B inhibits adipocyte differentiation and mediates TNFα action in obesity. Biochim Biophys Acta Mol Cell Biol Lipids 2013; 1831:1368-76. [PMID: 23711960 DOI: 10.1016/j.bbalip.2013.05.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2013] [Revised: 04/29/2013] [Accepted: 05/16/2013] [Indexed: 01/06/2023]
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a negative regulator of systemic glucose and insulin homeostasis; however, its exact role in adipocytes is poorly understood. This study was to elucidate the role of PTP1B in adipocyte differentiation and its implication in obesity. During differentiation of 3T3-L1 white preadipocytes, PTP1B decreased progressively with adipocyte maturation. Lentivirus-mediated PTP1B overexpression in preadipocytes delayed adipocyte differentiation, shown as lack of mature adipocytes, low level of lipid accumulation, and down-regulation of main markers (PPARγ2, SREBP-1c, FAS and LPL). In contrast, lentivirus-mediated PTP1B knockdown accelerated adipocyte differentiation, demonstrated as full of mature adipocytes, high level of lipid accumulation, and up-regulation of main markers. Dominant-negative inhibition on endogenous PTP1B by lentivirus-mediated overexpression of PTP1B double mutant in Tyr-46 and Asp-181 residues (LV-D/A-Y/F) also stimulated adipogenesis, more efficient than PTP1B knockdown. Diet-induced obesity mice exhibited an up-regulation of PTP1B and TNFα accompanied by a down-regulation of PPARγ2 in white adipose tissue. TNFα recombinant protein impeded PTP1B reduction and inhibited adipocyte differentiation in vitro; this inhibitory effect was prevented by LV-D/A-Y/F. Moreover, PTP1B inhibitor treatment improved adipogenesis and suppressed TNFα in adipose tissue of obese mice. All together, PTP1B negatively regulates adipocyte development and may mediate TNFα action to impair adipocyte differentiation in obesity. Our study provides novel evidence for the importance of PTP1B in obesity and for the potential application of PTP1B inhibitors.
Collapse
Affiliation(s)
- Dan-Dan Song
- Department of Endocrinology, Second Military Medical University, Shanghai, China
| | | | | | | | | | | |
Collapse
|
43
|
Mcilroy GD, Delibegovic M, Owen C, Stoney PN, Shearer KD, McCaffery PJ, Mody N. Fenretinide treatment prevents diet-induced obesity in association with major alterations in retinoid homeostatic gene expression in adipose, liver, and hypothalamus. Diabetes 2013. [PMID: 23193184 PMCID: PMC3581207 DOI: 10.2337/db12-0458] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
The synthetic retinoid, Fenretinide (FEN), inhibits obesity and insulin resistance in mice and is in early clinical trials for treatment of insulin resistance in obese humans. We aimed to determine whether alterations in retinoic acid (RA)-responsive genes contribute to the beneficial effects of FEN. We examined the effect of FEN on 3T3-L1 adipocyte differentiation and alterations in gene expression in C57Bl/6 and retinaldehyde dehydrogenase (RALDH) 1 knockout (KO) mice fed a high-fat (HF) diet. FEN completely inhibited adipocyte differentiation by blocking CCAAT/enhancer-binding protein (C/EBP) α/peroxisome proliferator-activated receptor (PPAR) γ-mediated induction of downstream genes and upregulating RA-responsive genes like cellular retinol-binding protein-1. In mice fed an HF diet, RA-responsive genes were markedly increased in adipose, liver, and hypothalamus, with short-term and long-term FEN treatment. In adipose, FEN inhibited the downregulation of PPARγ and improved insulin sensitivity and the levels of adiponectin, resistin, and serum RBP (RBP4). FEN inhibited hyperleptinemia in vivo and leptin expression in adipocytes. Surprisingly, hypothalamic neuropeptide Y expression was completely suppressed, suggesting a central effect of FEN to normalize hyperglycemia. Moreover, FEN induced RA-responsive genes in RALDH1 KO mice, demonstrating that FEN can augment RA signaling when RA synthesis is impaired. We show that FEN-mediated beneficial effects are through alterations in retinoid homeostasis genes, and these are strong candidates as therapeutic targets for the treatment of obesity and insulin resistance.
Collapse
Affiliation(s)
- George D Mcilroy
- University of Aberdeen, College of Life Sciences & Medicine, Institute of Medical Sciences, Foresterhill, Aberdeen, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
44
|
Lee KY, Russell SJ, Ussar S, Boucher J, Vernochet C, Mori MA, Smyth G, Rourk M, Cederquist C, Rosen ED, Kahn BB, Kahn CR. Lessons on conditional gene targeting in mouse adipose tissue. Diabetes 2013; 62:864-74. [PMID: 23321074 PMCID: PMC3581196 DOI: 10.2337/db12-1089] [Citation(s) in RCA: 281] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Conditional gene targeting has been extensively used for in vivo analysis of gene function in adipocyte cell biology but often with debate over the tissue specificity and the efficacy of inactivation. To directly compare the specificity and efficacy of different Cre lines in mediating adipocyte specific recombination, transgenic Cre lines driven by the adipocyte protein 2 (aP2) and adiponectin (Adipoq) gene promoters, as well as a tamoxifen-inducible Cre driven by the aP2 gene promoter (iaP2), were bred to the Rosa26R (R26R) reporter. All three Cre lines demonstrated recombination in the brown and white fat pads. Using different floxed loci, the individual Cre lines displayed a range of efficacy to Cre-mediated recombination that ranged from no observable recombination to complete recombination within the fat. The Adipoq-Cre exhibited no observable recombination in any other tissues examined, whereas both aP2-Cre lines resulted in recombination in endothelial cells of the heart and nonendothelial, nonmyocyte cells in the skeletal muscle. In addition, the aP2-Cre line can lead to germline recombination of floxed alleles in ~2% of spermatozoa. Thus, different "adipocyte-specific" Cre lines display different degrees of efficiency and specificity, illustrating important differences that must be taken into account in their use for studying adipose biology.
Collapse
Affiliation(s)
- Kevin Y. Lee
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Steven J. Russell
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Siegfried Ussar
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Jeremie Boucher
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Cecile Vernochet
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Marcelo A. Mori
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Graham Smyth
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Michael Rourk
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Carly Cederquist
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
| | - Evan D. Rosen
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - Barbara B. Kahn
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, Massachusetts
| | - C. Ronald Kahn
- Section on Integrative Physiology and Metabolism, Joslin Diabetes Center, Harvard Medical School, Boston, Massachusetts
- Corresponding author: C. Ronald Kahn,
| |
Collapse
|
45
|
Johnson AR, Milner JJ, Makowski L. The inflammation highway: metabolism accelerates inflammatory traffic in obesity. Immunol Rev 2013; 249:218-38. [PMID: 22889225 DOI: 10.1111/j.1600-065x.2012.01151.x] [Citation(s) in RCA: 424] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
As humans evolved, perhaps the two strongest selection determinants of survival were a robust immune response able to clear bacterial, viral, and parasitic infection and an ability to efficiently store nutrients to survive times when food sources were scarce. These traits are not mutually exclusive. It is now apparent that critical proteins necessary for regulating energy metabolism, such as peroxisome proliferator-activated receptors, Toll-like receptors, and fatty acid-binding proteins, also act as links between nutrient metabolism and inflammatory pathway activation in immune cells. Obesity in humans is a symptom of energy imbalance: the scale has been tipped such that energy intake exceeds energy output and may be a result, in part, of evolutionary selection toward a phenotype characterized by efficient energy storage. As discussed in this review, obesity is a state of low-grade, chronic inflammation that promotes the development of insulin resistance and diabetes. Ironically, the formation of systemic and/or local, tissue-specific insulin resistance upon inflammatory cell activation may actually be a protective mechanism that co-evolved to repartition energy sources within the body during times of stress during infection. However, the point has been reached where a once beneficial adaptive trait has become detrimental to the health of the individual and an immense public health and economic burden. This article reviews the complex relationship between obesity, insulin resistance/diabetes, and inflammation, and although the liver, brain, pancreas, muscle, and other tissues are relevant, we focus specifically on how the obese adipose microenvironment can promote immune cell influx and sustain damaging inflammation that can lead to the onset of insulin resistance and diabetes. Finally, we address how substrate metabolism may regulate the immune response and discuss how fuel uptake and metabolism may be a targetable approach to limit or abrogate obesity-induced inflammation.
Collapse
Affiliation(s)
- Amy R Johnson
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
46
|
Tsou RC, Bence KK. Central regulation of metabolism by protein tyrosine phosphatases. Front Neurosci 2013; 6:192. [PMID: 23308070 PMCID: PMC3538333 DOI: 10.3389/fnins.2012.00192] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Accepted: 12/17/2012] [Indexed: 11/13/2022] Open
Abstract
Protein tyrosine phosphatases (PTPs) are important regulators of intracellular signaling pathways via the dephosphorylation of phosphotyrosyl residues on various receptor and non-receptor substrates. The phosphorylation state of central nervous system (CNS) signaling components underlies the molecular mechanisms of a variety of physiological functions including the control of energy balance and glucose homeostasis. In this review, we summarize the current evidence implicating PTPs as central regulators of metabolism, specifically highlighting their interactions with the neuronal leptin and insulin signaling pathways. We discuss the role of a number of PTPs (PTP1B, SHP2, TCPTP, RPTPe, and PTEN), reviewing the findings from genetic mouse models and in vitro studies which highlight these phosphatases as key central regulators of energy homeostasis.
Collapse
Affiliation(s)
- Ryan C Tsou
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania Philadelphia, PA, USA
| | | |
Collapse
|
47
|
Tsou RC, Zimmer DJ, De Jonghe BC, Bence KK. Deficiency of PTP1B in leptin receptor-expressing neurons leads to decreased body weight and adiposity in mice. Endocrinology 2012; 153:4227-37. [PMID: 22802463 PMCID: PMC3423620 DOI: 10.1210/en.2012-1548] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Accepted: 07/02/2012] [Indexed: 01/19/2023]
Abstract
Protein tyrosine phosphatase 1B (PTP1B) is a ubiquitously expressed tyrosine phosphatase implicated in the negative regulation of leptin and insulin receptor signaling. PTP1B(-/-) mice possess a lean metabolic phenotype attributed at least partially to improved hypothalamic leptin sensitivity. Interestingly, mice lacking both leptin and PTP1B (ob/ob:PTP1B(-/-)) have reduced body weight compared with mice lacking leptin only, suggesting that PTP1B may have important leptin-independent metabolic effects. We generated mice with PTP1B deficiency specifically in leptin receptor (LepRb)-expressing neurons (LepRb-PTP1B(-/-)) and compared them with LepRb-Cre-only wild-type (WT) controls and global PTP1B(-/-) mice. Consistent with PTP1B's role as a negative regulator of leptin signaling, our results show that LepRb-PTP1B(-/-) mice are leptin hypersensitive and have significantly reduced body weight when maintained on chow or high-fat diet (HFD) compared with WT controls. LepRb-PTP1B(-/-) mice have a significant decrease in adiposity on HFD compared with controls. Notably, the extent of attenuated body weight gain on HFD, as well as the extent of leptin hypersensitivity, is similar between LepRb-PTP1B(-/-) mice and global PTP1B(-/-) mice. Overall, these results demonstrate that PTP1B deficiency in LepRb-expressing neurons results in reduced body weight and adiposity compared with WT controls and likely underlies the improved metabolic phenotype of global and brain-specific PTP1B-deficient models. Subtle phenotypic differences between LepRb-PTP1B(-/-) and global PTP1B(-/-) mice, however, suggest that PTP1B independent of leptin signaling may also contribute to energy balance in mice.
Collapse
Affiliation(s)
- Ryan C Tsou
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104-6046, USA
| | | | | | | |
Collapse
|
48
|
Hoggard N, Agouni A, Mody N, Delibegovic M. Serum levels of RBP4 and adipose tissue levels of PTP1B are increased in obese men resident in northeast Scotland without associated changes in ER stress response genes. Int J Gen Med 2012; 5:403-11. [PMID: 22615536 PMCID: PMC3355846 DOI: 10.2147/ijgm.s25879] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Background Retinol-binding protein 4 (RBP4) is an adipokine identified as a marker of insulin resistance in mice and humans. Protein tyrosine phosphatase 1B (PTP1B) expression levels as well as other genes involved in the endoplasmic reticulum (ER) stress response are increased in adipose tissue of obese, high-fat-diet-fed mice. In this study we investigated if serum and/or adipose tissue RBP4 protein levels and expression levels of PTP1B and other ER stress-response genes are altered in obese and obese/diabetic men resident in northeast Scotland. Methods We studied three groups of male volunteers: (1) normal/overweight (body mass index [BMI] < 30), (2) obese (BMI > 30), and (3) obese/diabetic (BMI > 30) controlling their diabetes either by diet or the antidiabetic drug metformin. We analyzed their serum and adipose tissue RBP4 protein levels as well as adipose tissue mRNA expression of PTP1B, binding immunoglobulin protein (BIP), activated transcription factor 4 (ATF4), and glucose-regulated protein 94 (GRP94) alongside other markers of adiposity (percentage body fat, leptin, cholesterol, triglycerides) and insulin resistance (oral glucose tolerance tests, insulin, homeostatic model assessment–insulin resistance, C-reactive protein, and adiponectin). Results We found that obese Scottish subjects had significantly higher serum RBP4 protein levels in comparison to the normal/overweight subjects (P < 0.01). Serum RBP4 levels were normalized in obese/diabetic subjects treated with diet or metformin (P < 0.05). Adipose tissue RBP4 protein levels were comparable between all three groups of subjects as were serum and adipose transthyretin levels. Adipose tissue PTP1B mRNA levels were increased in obese subjects in comparison to normal/overweight subjects (P < 0.05); however diet and/or metformin treatment did not reverse this effect. Adipose tissue BIP, ATF4, and GRP94 expression levels were unchanged in obese and obese/diabetic subjects. Conclusions Human obesity results in an increase in serum but not adipose tissue RBP4 protein levels, and these are normalized in obese/diabetic subjects, which exhibit improvements in insulin sensitivity through diet or metformin treatment. However, while adipose tissue PTP1B mRNA levels increase in obese Scottish subjects, these remain high in obese/diabetics on diet or metformin treatment.
Collapse
Affiliation(s)
- Nigel Hoggard
- Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, UK
| | | | | | | |
Collapse
|
49
|
Tsou RC, Bence KK. The Genetics of PTPN1 and Obesity: Insights from Mouse Models of Tissue-Specific PTP1B Deficiency. J Obes 2012; 2012:926857. [PMID: 22811891 PMCID: PMC3395189 DOI: 10.1155/2012/926857] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 04/18/2012] [Accepted: 04/18/2012] [Indexed: 02/05/2023] Open
Abstract
The protein tyrosine phosphatase PTP1B is a negative regulator of both insulin and leptin signaling and is involved in the control of glucose homeostasis and energy expenditure. Due to its prominent role in regulating metabolism, PTP1B is a promising therapeutic target for the treatment of human obesity and type 2 diabetes. The PTP1B protein is encoded by the PTPN1 gene on human chromosome 20q13, a region that shows linkage with insulin resistance, type 2 diabetes, and obesity in human populations. In this paper, we summarize the genetics of the PTPN1 locus and associations with metabolic disease. In addition, we discuss the tissue-specific functions of PTP1B as gleaned from genetic mouse models.
Collapse
Affiliation(s)
- Ryan C. Tsou
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Vet 223E, Philadelphia, PA 19104, USA
| | - Kendra K. Bence
- Department of Animal Biology, School of Veterinary Medicine, University of Pennsylvania, 3800 Spruce Street, Vet 223E, Philadelphia, PA 19104, USA
- *Kendra K. Bence:
| |
Collapse
|