101
|
Pasarín M, Abraldes JG, Liguori E, Kok B, La Mura V. Intrahepatic vascular changes in non-alcoholic fatty liver disease: Potential role of insulin-resistance and endothelial dysfunction. World J Gastroenterol 2017; 23:6777-6787. [PMID: 29085222 PMCID: PMC5645612 DOI: 10.3748/wjg.v23.i37.6777] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 09/06/2017] [Accepted: 09/19/2017] [Indexed: 02/06/2023] Open
Abstract
Metabolic syndrome is a cluster of several clinical conditions characterized by insulin-resistance and high cardiovascular risk. Non-alcoholic fatty liver disease is the liver expression of the metabolic syndrome, and insulin resistance can be a frequent comorbidity in several chronic liver diseases, in particular hepatitis C virus infection and/or cirrhosis. Several studies have demonstrated that insulin action is not only relevant for glucose control, but also for vascular homeostasis. Insulin regulates nitric oxide production, which mediates to a large degree the vasodilating, anti-inflammatory and antithrombotic properties of a healthy endothelium, guaranteeing organ perfusion. The effects of insulin on the liver microvasculature and the effects of IR on sinusoidal endothelial cells have been studied in animal models of non-alcoholic fatty liver disease. The hypotheses derived from these studies and the potential translation of these results into humans are critically discussed in this review.
Collapse
Affiliation(s)
- Marcos Pasarín
- Hepatic Hemodynamic Laboratory, Liver Unit, Hospital Clinic, IDIBAPS (Institut d’Investigacions Biomèdiques August Pi i Sunyer), University of Barcelona, 08036 Barcelona, Spain
| | - Juan G Abraldes
- Cirrhosis Care Clinic, Division of Gastroenterology (Liver Unit), CEGIIR, University of Alberta, AB T6G 2R3 Edmonton, Canada
| | - Eleonora Liguori
- Internal Medicine, IRCCS San Donato, Department of Biomedical Sciences for Health, University of Milan, 20097 San Donato Milanese, Italy
| | - Beverley Kok
- Cirrhosis Care Clinic, Division of Gastroenterology (Liver Unit), CEGIIR, University of Alberta, AB T6G 2R3 Edmonton, Canada
| | - Vincenzo La Mura
- Internal Medicine, IRCCS San Donato, Department of Biomedical Sciences for Health, University of Milan, 20097 San Donato Milanese, Italy
| |
Collapse
|
102
|
Katashima CK, Silva VRR, Lenhare L, Marin RM, Carvalheira JBC. iNOS promotes hypothalamic insulin resistance associated with deregulation of energy balance and obesity in rodents. Sci Rep 2017; 7:9265. [PMID: 28835706 PMCID: PMC5569114 DOI: 10.1038/s41598-017-08920-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2016] [Accepted: 07/20/2017] [Indexed: 02/07/2023] Open
Abstract
Inducible nitric oxide (iNOS)-mediated S-nitrosation of the metabolic signaling pathway has emerged as a post-translational modification that triggers insulin resistance in obesity and aging. However, the effects of S-nitrosation in controlling energy homeostasis are unknown. Thus, in the present study we aimed to evaluate the effects of S-nitrosation in insulin signaling pathway in the hypothalamus of rodents. Herein, we demonstrated that the intracerebroventricular infusion of the nitric oxide (NO) donor S-nitrosoglutathione (GSNO) promoted hypothalamic insulin signaling resistance and replicated the food intake pattern of obese individuals. Indeed, obesity induced S-nitrosation of hypothalamic IR and Akt, whereas inhibition of iNOS or S-nitrosation of insulin signaling pathway protected against hypothalamic insulin resistance and normalized energy homeostasis. Overall, these findings indicated that S-nitrosation of insulin signaling pathway is required to sustain hypothalamic insulin resistance in obesity.
Collapse
Affiliation(s)
| | | | - Luciene Lenhare
- Department of Internal Medicine, University of Campinas, UNICAMP, Campinas, SP, Brazil
| | - Rodrigo Miguel Marin
- Department of Internal Medicine, University of Campinas, UNICAMP, Campinas, SP, Brazil
| | | |
Collapse
|
103
|
Ghasemi A, Jeddi S. Anti-obesity and anti-diabetic effects of nitrate and nitrite. Nitric Oxide 2017; 70:9-24. [PMID: 28804022 DOI: 10.1016/j.niox.2017.08.003] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Revised: 07/02/2017] [Accepted: 08/08/2017] [Indexed: 02/06/2023]
Abstract
Prevalence of obesity is increasing worldwide and type 2 diabetes to date is the most devastating complication of obesity. Decreased nitric oxide bioavailability is a feature of obesity and diabetes that links these two pathologies. Nitric oxide is synthesized both by nitric oxide synthase enzymes from l-arginine and nitric oxide synthase-independent from nitrate/nitrite. Nitric oxide production from nitrate/nitrite could potentially be used for nutrition-based therapy in obesity and diabetes. Nitric oxide deficiency also contributes to pathogeneses of cardiovascular disease and hypertension, which are associated with obesity and diabetes. This review summarizes pathways for nitric oxide production and focuses on the anti-diabetic and anti-obesity effects of the nitrate-nitrite-nitric oxide pathway. In addition to increasing nitric oxide production, nitrate and nitrite reduce oxidative stress, increase adipose tissue browning, have favorable effects on nitric oxide synthase expression, and increase insulin secretion, all effects that are potentially promising for management of obesity and diabetes. Based on current data, it could be suggested that amplifying the nitrate-nitrite-nitric oxide pathway is a diet-based strategy for increasing nitric oxide bioavailability and the management of these two interlinked conditions. Adding nitrate/nitrite to drugs that are currently used for managing diabetes (e.g. metformin) and possibly anti-obesity drugs may also enhance their efficacy.
Collapse
Affiliation(s)
- Asghar Ghasemi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Sajad Jeddi
- Endocrine Physiology Research Center, Research Institute for Endocrine Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
104
|
Mitochondrial form, function and signalling in aging. Biochem J 2017; 473:3421-3449. [PMID: 27729586 DOI: 10.1042/bcj20160451] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 06/17/2016] [Indexed: 02/07/2023]
Abstract
Aging is often accompanied by a decline in mitochondrial mass and function in different tissues. Additionally, cell resistance to stress is frequently found to be prevented by higher mitochondrial respiratory capacity. These correlations strongly suggest mitochondria are key players in aging and senescence, acting by regulating energy homeostasis, redox balance and signalling pathways central in these processes. However, mitochondria display a wide array of functions and signalling properties, and the roles of these different characteristics are still widely unexplored. Furthermore, differences in mitochondrial properties and responses between tissues and cell types, and how these affect whole body metabolism are also still poorly understood. This review uncovers aspects of mitochondrial biology that have an impact upon aging in model organisms and selected mammalian cells and tissues.
Collapse
|
105
|
Jankovic A, Korac A, Buzadzic B, Stancic A, Otasevic V, Ferdinandy P, Daiber A, Korac B. Targeting the NO/superoxide ratio in adipose tissue: relevance to obesity and diabetes management. Br J Pharmacol 2017; 174:1570-1590. [PMID: 27079449 PMCID: PMC5446578 DOI: 10.1111/bph.13498] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 03/31/2016] [Accepted: 04/04/2016] [Indexed: 12/21/2022] Open
Abstract
Insulin sensitivity and metabolic homeostasis depend on the capacity of adipose tissue to take up and utilize excess glucose and fatty acids. The key aspects that determine the fuel-buffering capacity of adipose tissue depend on the physiological levels of the small redox molecule, nitric oxide (NO). In addition to impairment of NO synthesis, excessive formation of the superoxide anion (О2•- ) in adipose tissue may be an important interfering factor diverting the signalling of NO and other reactive oxygen and nitrogen species in obesity, resulting in metabolic dysfunction of adipose tissue over time. Besides its role in relief from superoxide burst, enhanced NO signalling may be responsible for the therapeutic benefits of different superoxide dismutase mimetics, in obesity and experimental diabetes models. This review summarizes the role of NO in adipose tissue and highlights the effects of NO/О2•- ratio 'teetering' as a promising pharmacological target in the metabolic syndrome. LINKED ARTICLES This article is part of a themed section on Redox Biology and Oxidative Stress in Health and Disease. To view the other articles in this section visit http://onlinelibrary.wiley.com/doi/10.1111/bph.v174.12/issuetoc.
Collapse
Affiliation(s)
- Aleksandra Jankovic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Aleksandra Korac
- Faculty of Biology, Center for Electron MicroscopyUniversity of BelgradeBelgradeSerbia
| | - Biljana Buzadzic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Ana Stancic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Vesna Otasevic
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| | - Péter Ferdinandy
- Department of Pharmacology and PharmacotherapySemmelweis UniversityBudapestHungary
- Pharmahungary GroupSzegedHungary
| | - Andreas Daiber
- Center for Cardiology ‐ Cardiology 1, Molecular CardiologyUniversity Medical CenterMainzGermany
| | - Bato Korac
- Department of Physiology, Institute for Biological Research “Sinisa Stankovic”University of BelgradeBelgradeSerbia
| |
Collapse
|
106
|
Rodriguez-Rodriguez R, Jiménez-Altayó F, Alsina L, Onetti Y, Rinaldi de Alvarenga JF, Claro C, Ogalla E, Casals N, Lamuela-Raventos RM. Mediterranean tomato-based sofrito
protects against vascular alterations in obese Zucker rats by preserving NO bioavailability. Mol Nutr Food Res 2017; 61. [DOI: 10.1002/mnfr.201601010] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Revised: 02/17/2017] [Accepted: 03/03/2017] [Indexed: 11/12/2022]
Affiliation(s)
- Rosalia Rodriguez-Rodriguez
- Basic Sciences Department; Faculty of Medicine and Health Sciences; Universitat Internacional de Catalunya; Sant Cugat del Vallès Barcelona Spain
| | - Francesc Jiménez-Altayó
- Departament de Farmacologia, de Terapèutica i de Toxicologia; Institut de Neurociències; Facultat de Medicina; Universitat Autònoma de Barcelona; Bellaterra Spain
| | - Laia Alsina
- Departament de Farmacologia, de Terapèutica i de Toxicologia; Institut de Neurociències; Facultat de Medicina; Universitat Autònoma de Barcelona; Bellaterra Spain
| | - Yara Onetti
- Departament de Farmacologia, de Terapèutica i de Toxicologia; Institut de Neurociències; Facultat de Medicina; Universitat Autònoma de Barcelona; Bellaterra Spain
| | | | - Carmen Claro
- Departamento de Farmacología; Facultad de Farmacia; Universidad de Sevilla; Sevilla Spain
| | - Elena Ogalla
- Departamento de Farmacología; Facultad de Farmacia; Universidad de Sevilla; Sevilla Spain
| | - Núria Casals
- Basic Sciences Department; Faculty of Medicine and Health Sciences; Universitat Internacional de Catalunya; Sant Cugat del Vallès Barcelona Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
| | - Rosa M. Lamuela-Raventos
- Nutrition, Food Science Department and Gastronomy; XaRTA, INSA-UB Pharmacy; University of Barcelona; Barcelona Spain
- CIBER Fisiopatología de la Obesidad y la Nutrición (CIBEROBN); Instituto de Salud Carlos III; Madrid Spain
| |
Collapse
|
107
|
Di Meo S, Iossa S, Venditti P. Skeletal muscle insulin resistance: role of mitochondria and other ROS sources. J Endocrinol 2017; 233:R15-R42. [PMID: 28232636 DOI: 10.1530/joe-16-0598] [Citation(s) in RCA: 204] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 01/31/2017] [Indexed: 12/12/2022]
Abstract
At present, obesity is one of the most important public health problems in the world because it causes several diseases and reduces life expectancy. Although it is well known that insulin resistance plays a pivotal role in the development of type 2 diabetes mellitus (the more frequent disease in obese people) the link between obesity and insulin resistance is yet a matter of debate. One of the most deleterious effects of obesity is the deposition of lipids in non-adipose tissues when the capacity of adipose tissue is overwhelmed. During the last decade, reduced mitochondrial function has been considered as an important contributor to 'toxic' lipid metabolite accumulation and consequent insulin resistance. More recent reports suggest that mitochondrial dysfunction is not an early event in the development of insulin resistance, but rather a complication of the hyperlipidemia-induced reactive oxygen species (ROS) production in skeletal muscle, which might promote mitochondrial alterations, lipid accumulation and inhibition of insulin action. Here, we review the literature dealing with the mitochondria-centered mechanisms proposed to explain the onset of obesity-linked IR in skeletal muscle. We conclude that the different pathways leading to insulin resistance may act synergistically because ROS production by mitochondria and other sources can result in mitochondrial dysfunction, which in turn can further increase ROS production leading to the establishment of a harmful positive feedback loop.
Collapse
Affiliation(s)
- Sergio Di Meo
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Susanna Iossa
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| | - Paola Venditti
- Department of BiologyUniversity of Naples 'Federico II', Naples, Italy
| |
Collapse
|
108
|
Chan KL, Boroumand P, Milanski M, Pillon NJ, Bilan PJ, Klip A. Deconstructing metabolic inflammation using cellular systems. Am J Physiol Endocrinol Metab 2017; 312:E339-E347. [PMID: 28196858 DOI: 10.1152/ajpendo.00039.2017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 02/09/2017] [Accepted: 02/10/2017] [Indexed: 02/06/2023]
Abstract
Over the past years, we have embarked in a systematic analysis of the effect of obesity or fatty acids on circulating monocytes, microvascular endothelial cells, macrophages, and skeletal muscle cells. With the use of cell culture strategies, we have deconstructed complex physiological systems and then reconstructed "partial equations" to better understand cell-to-cell communication. Through these approaches, we identified that in high saturated fat environments, cell-autonomous proinflammatory pathways are activated in monocytes and endothelial cells, promoting monocyte adhesion and transmigration. We think of this as a paradigm of the conditions promoting immune cell infiltration into tissues during obesity. In concert, it is possible that muscle and adipose tissue secrete immune cell chemoattractants, and indeed, our tissue culture reconstructions reveal that myotubes treated with the saturated fatty acid palmitate, but not the unsaturated fatty acid palmitoleate, release nucleotides that attract monocytes and other compounds that promote proinflammatory classically activated "(M1)-like" polarization in macrophages. In addition, palmitate directly triggers an M1-like macrophage phenotype, and secretions from these activated macrophages confer insulin resistance to target muscle cells. Together, these studies suggest that in pathophysiological conditions of excess fat, the muscle, endothelial and immune cells engage in a synergistic crosstalk that exacerbates tissue inflammation, leukocyte infiltration, polarization, and consequent insulin resistance.
Collapse
Affiliation(s)
- Kenny L Chan
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Physiology, University of Toronto, Ontario, Canada; and
| | - Parastoo Boroumand
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Ontario Canada
| | - Marciane Milanski
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Nicolas J Pillon
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Philip J Bilan
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amira Klip
- Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada;
- Department of Physiology, University of Toronto, Ontario, Canada; and
- Department of Biochemistry, University of Toronto, Ontario Canada
| |
Collapse
|
109
|
Shin HS, Kang SI, Ko HC, Park DB, Kim SJ. Tangeretin Improves Glucose Uptake in a Coculture of Hypertrophic Adipocytes and Macrophages by Attenuating Inflammatory Changes. Dev Reprod 2017; 21:93-100. [PMID: 28484748 PMCID: PMC5409214 DOI: 10.12717/dr.2017.21.1.093] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 03/10/2017] [Accepted: 03/12/2017] [Indexed: 12/12/2022]
Abstract
Obesity is characterized by a state of chronic low-grade inflammation and insulin
resistance, which are aggravated by the interaction between hypertrophic
adipocytes and macrophages. In this study, we investigated the effects of
tangeretin on inflammatory changes and glucose uptake in a coculture of
hypertrophic adipocytes and macrophages. Tangeretin decreased nitric oxide
production and the expression of interleukin (IL)-6, IL-1β, tumor necrosis
factor-α, inducible nitric oxide synthase, and cyclooxygenase-2 in a coculture
of 3T3-L1 adipocytes and RAW 264.7 cells. Tangeretin also increased glucose
uptake in the coculture system, but did not affect the phosphorylation of
insulin receptor substrate (IRS) and Akt. These results suggest that tangeretin
improves insulin resistance by attenuating obesity-induced inflammation in
adipose tissue.
Collapse
Affiliation(s)
- Hye-Sun Shin
- Department of Biology, Jeju National University, Jeju Special Self-Governing Province, Korea
| | - Seong-Il Kang
- Department of Biology, Jeju National University, Jeju Special Self-Governing Province, Korea
| | - Hee-Chul Ko
- Jeju Sasa Industry Development Agency, Jeju National University, Jeju Special Self-Governing Province, Korea
| | - Deok-Bae Park
- Department of Medicine, Jeju National University, Jeju Special Self-Governing Province, Korea
| | - Se-Jae Kim
- Department of Biology, Jeju National University, Jeju Special Self-Governing Province, Korea
| |
Collapse
|
110
|
Filippi BM, Abraham MA, Silva PN, Rasti M, LaPierre MP, Bauer PV, Rocheleau JV, Lam TK. Dynamin-Related Protein 1-Dependent Mitochondrial Fission Changes in the Dorsal Vagal Complex Regulate Insulin Action. Cell Rep 2017; 18:2301-2309. [DOI: 10.1016/j.celrep.2017.02.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 01/26/2017] [Accepted: 02/11/2017] [Indexed: 11/25/2022] Open
|
111
|
van den Berg SM, van Dam AD, Rensen PCN, de Winther MPJ, Lutgens E. Immune Modulation of Brown(ing) Adipose Tissue in Obesity. Endocr Rev 2017; 38:46-68. [PMID: 27849358 DOI: 10.1210/er.2016-1066] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 11/14/2016] [Indexed: 12/13/2022]
Abstract
Obesity is associated with a variety of medical conditions such as type 2 diabetes and cardiovascular diseases and is therefore responsible for high morbidity and mortality rates. Increasing energy expenditure by brown adipose tissue (BAT) is a current novel strategy to reduce the excessive energy stores in obesity. Brown adipocytes burn energy to generate heat and are mainly activated upon cold exposure. As prolonged cold exposure is not a realistic therapy, researchers worldwide are searching for novel ways to activate BAT and/or induce beiging of white adipose tissue. Recently, the contribution of immune cells in the regulation of brown adipocyte activity and beiging of white adipose tissue has gained increased attention, with a prominent role for eosinophils and alternatively activated macrophages. This review discusses the rediscovery of BAT, presents an overview of modes of activation and differentiation of beige and brown adipocytes, and describes the recently discovered immunological pathways that are key in mediating brown/beige adipocyte development and function. Interventions in immunological pathways harbor the potential to provide novel strategies to increase beige and brown adipose tissue activity as a therapeutic target for obesity.
Collapse
Affiliation(s)
- Susan M van den Berg
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, 1105AZ The Netherlands
| | - Andrea D van Dam
- Department of Medicine, Division of Endocrinology, and.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; and
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, and.,Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, 2333ZA Leiden, The Netherlands; and
| | - Menno P J de Winther
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, 1105AZ The Netherlands.,Institute for Cardiovascular Prevention, Ludwig Maximilians University of Munich, 80539 Munich, Germany
| | - Esther Lutgens
- Department of Medical Biochemistry, Subdivision of Experimental Vascular Biology, Academic Medical Centre, University of Amsterdam, 1105AZ The Netherlands.,Institute for Cardiovascular Prevention, Ludwig Maximilians University of Munich, 80539 Munich, Germany
| |
Collapse
|
112
|
Guo Q, Xu L, Li H, Sun H, Liu J, Wu S, Zhou B. Progranulin causes adipose insulin resistance via increased autophagy resulting from activated oxidative stress and endoplasmic reticulum stress. Lipids Health Dis 2017; 16:25. [PMID: 28143512 PMCID: PMC5282713 DOI: 10.1186/s12944-017-0425-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 01/25/2017] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Progranulin (PGRN) has recently emerged as an important regulator for insulin resistance. However, the direct effect of progranulin in adipose insulin resistance associated with the autophagy mechanism is not fully understood. METHODS In the present study, progranulin was administered to 3T3-L1 adipocytes and C57BL/6 J mice with/without specific inhibitors of oxidative stress and endoplasmic reticulum stress, and metabolic parameters, oxidative stress, endoplasmic reticulum stress and autophagy markers were assessed. RESULTS Progranulin treatment increased iNOS expression, NO synthesis and ROS generation, and elevated protein expressions of CHOP, GRP78 and the phosphorylation of PERK, and caused a significant increase in Atg7 and LC3-II protein expression and a decreased p62 expression, and decreased insulin-stimulated tyrosine phosphorylation of IRS-1 and glucose uptake, demonstrating that progranulin activated oxidative stress and ER stress, elevated autophagy and induced insulin insensitivity in adipocytes and adipose tissue of mice. Interestingly, inhibition of iNOS and ER stress both reversed progranulin-induced stress response and increased autophagy, protecting against insulin resistance in adipocytes. Furthermore, the administration of the ER stress inhibitor 4-phenyl butyric acid reversed the negative effect of progranulin in vivo. CONCLUSION Our findings showed the clinical potential of the novel adipokine progranulin in the regulation of insulin resistance, suggesting that progranulin might mediate adipose insulin resistance, at least in part, by inducing autophagy via activated oxidative stress and ER stress.
Collapse
Affiliation(s)
- Qinyue Guo
- grid.452438.cDepartment of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi 710061 China
| | - Lin Xu
- 0000 0001 0599 1243grid.43169.39Department of Endocrinology, the Affiliated Guangren Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710004 China
| | - Huixia Li
- 0000 0001 0599 1243grid.43169.39Key Laboratory of Environment and Genes Related to Diseases, Medical School of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 China
| | - Hongzhi Sun
- 0000 0001 0599 1243grid.43169.39Key Laboratory of Environment and Genes Related to Diseases, Medical School of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 China
| | - Jiali Liu
- grid.452672.0Clinical Laboratory, the Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 China
| | - Shufang Wu
- grid.452438.cCenter for Translational Medicine, the First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, Shaanxi 710061 China
| | - Bo Zhou
- grid.452438.cDepartment of Respiratory and Critical Care Medicine, the First Affiliated Hospital of Xi’an Jiaotong University, 277 Yanta West Road, Xi’an, Shaanxi 710061 China
| |
Collapse
|
113
|
Kanuri BN, Kanshana JS, Rebello SC, Pathak P, Gupta AP, Gayen JR, Jagavelu K, Dikshit M. Altered glucose and lipid homeostasis in liver and adipose tissue pre-dispose inducible NOS knockout mice to insulin resistance. Sci Rep 2017; 7:41009. [PMID: 28106120 PMCID: PMC5247703 DOI: 10.1038/srep41009] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 12/14/2016] [Indexed: 12/15/2022] Open
Abstract
On the basis of diet induced obesity and KO mice models, nitric oxide is implied to play an important role in the initiation of dyslipidemia induced insulin resistance. However, outcomes using iNOS KO mice have so far remained inconclusive. The present study aimed to assess IR in iNOS KO mice after 5 weeks of LFD feeding by monitoring body composition, energy homeostasis, insulin sensitivity/signaling, nitrite content and gene expressions changes in the tissues. We found that body weight and fat content in KO mice were significantly higher while the respiratory exchange ratio (RER), volume of carbon dioxide (VCO2), and heat production were lower as compared to WT mice. Furthermore, altered systemic glucose tolerance, tissue insulin signaling, hepatic gluconeogenesis, augmented hepatic lipids, adiposity, as well as gene expression regulating lipid synthesis, catabolism and efflux were evident in iNOS KO mice. Significant reduction in eNOS and nNOS gene expression, hepatic and adipose tissue nitrite content, circulatory nitrite was also observed. Oxygen consumption rate of mitochondrial respiration has remained unaltered in KO mice as measured using extracellular flux analyzer. Our findings establish a link between the NO status with systemic and tissue specific IR in iNOS KO mice at 5 weeks.
Collapse
Affiliation(s)
- Babu Nageswararao Kanuri
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow - 226031, India.,Academy of Scientific and Innovative Research, New Delhi - 110001, India
| | - Jitendra S Kanshana
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow - 226031, India
| | - Sanjay C Rebello
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow - 226031, India
| | - Priya Pathak
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow - 226031, India
| | - Anand P Gupta
- Pharmacokinetics and Metabolism Division, CSIR-Central Drug Research Institute, Lucknow - 226031, India
| | - Jiaur R Gayen
- Pharmacokinetics and Metabolism Division, CSIR-Central Drug Research Institute, Lucknow - 226031, India
| | - Kumaravelu Jagavelu
- Pharmacology Division, CSIR-Central Drug Research Institute, Lucknow - 226031, India
| | - Madhu Dikshit
- Academy of Scientific and Innovative Research, New Delhi - 110001, India
| |
Collapse
|
114
|
Nakazawa H, Chang K, Shinozaki S, Yasukawa T, Ishimaru K, Yasuhara S, Yu YM, Martyn JAJ, Tompkins RG, Shimokado K, Kaneki M. iNOS as a Driver of Inflammation and Apoptosis in Mouse Skeletal Muscle after Burn Injury: Possible Involvement of Sirt1 S-Nitrosylation-Mediated Acetylation of p65 NF-κB and p53. PLoS One 2017; 12:e0170391. [PMID: 28099528 PMCID: PMC5242494 DOI: 10.1371/journal.pone.0170391] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 01/04/2017] [Indexed: 01/28/2023] Open
Abstract
Inflammation and apoptosis develop in skeletal muscle after major trauma, including burn injury, and play a pivotal role in insulin resistance and muscle wasting. We and others have shown that inducible nitric oxide synthase (iNOS), a major mediator of inflammation, plays an important role in stress (e.g., burn)-induced insulin resistance. However, it remains to be determined how iNOS induces insulin resistance. Moreover, the interrelation between inflammatory response and apoptosis is poorly understood, although they often develop simultaneously. Nuclear factor (NF)-κB and p53 are key regulators of inflammation and apoptosis, respectively. Sirt1 inhibits p65 NF-κB and p53 by deacetylating these transcription factors. Recently, we have shown that iNOS induces S-nitrosylation of Sirt1, which inactivates Sirt1 and thereby increases acetylation and activity of p65 NF-κB and p53 in various cell types, including skeletal muscle cells. Here, we show that iNOS enhances burn-induced inflammatory response and apoptotic change in mouse skeletal muscle along with S-nitrosylation of Sirt1. Burn injury induced robust expression of iNOS in skeletal muscle and gene disruption of iNOS significantly inhibited burn-induced increases in inflammatory gene expression and apoptotic change. In parallel, burn increased Sirt1 S-nitrosylation and acetylation and DNA-binding capacity of p65 NF-κB and p53, all of which were reversed or ameliorated by iNOS deficiency. These results indicate that iNOS functions not only as a downstream effector but also as an upstream enhancer of burn-induced inflammatory response, at least in part, by Sirt1 S-nitrosylation-dependent activation (acetylation) of p65 NF-κB. Our data suggest that Sirt1 S-nitrosylation may play a role in iNOS-mediated enhanced inflammatory response and apoptotic change, which, in turn, contribute to muscle wasting and supposedly to insulin resistance after burn injury.
Collapse
Affiliation(s)
- Harumasa Nakazawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
- Shriners Hospitals for Children, Boston, Massachusetts, United States of America
| | - Kyungho Chang
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
| | - Shohei Shinozaki
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
- Department of Geriatrics and Vascular Medicine, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Takashi Yasukawa
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
- Shriners Hospitals for Children, Boston, Massachusetts, United States of America
| | - Kazuhiro Ishimaru
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
- Shriners Hospitals for Children, Boston, Massachusetts, United States of America
| | - Shingo Yasuhara
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
- Shriners Hospitals for Children, Boston, Massachusetts, United States of America
| | - Yong-Ming Yu
- Shriners Hospitals for Children, Boston, Massachusetts, United States of America
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - J. A. Jeevendra Martyn
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
- Shriners Hospitals for Children, Boston, Massachusetts, United States of America
| | - Ronald. G. Tompkins
- Shriners Hospitals for Children, Boston, Massachusetts, United States of America
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kentaro Shimokado
- Department of Geriatrics and Vascular Medicine, Tokyo Medical and Dental University Graduate School, Tokyo, Japan
| | - Masao Kaneki
- Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Charlestown, Massachusetts, United States of America
- Shriners Hospitals for Children, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
115
|
da Cunha NV, Lopes FNC, Panis C, Cecchini R, Pinge-Filho P, Martins-Pinge MC. iNOS inhibition improves autonomic dysfunction and oxidative status in hypertensive obese rats. Clin Exp Hypertens 2017; 39:50-57. [PMID: 28055264 DOI: 10.1080/10641963.2016.1210628] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
| | | | - Carolina Panis
- Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Rubens Cecchini
- Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | - Phileno Pinge-Filho
- Department of Pathological Sciences, State University of Londrina, Londrina, Brazil
| | | |
Collapse
|
116
|
Pereira RM, Moura LPD, Muñoz VR, Silva ASRD, Gaspar RS, Ropelle ER, Pauli JR. Molecular mechanisms of glucose uptake in skeletal muscle at rest and in response to exercise. MOTRIZ: REVISTA DE EDUCACAO FISICA 2017. [DOI: 10.1590/s1980-6574201700si0004] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Affiliation(s)
| | | | | | | | | | | | - José Rodrigo Pauli
- Universidade Estadual de Campinas, Brazil; Universidade Estadual de Campinas, Brazil
| |
Collapse
|
117
|
Zanotto TM, Quaresma PGF, Guadagnini D, Weissmann L, Santos AC, Vecina JF, Calisto K, Santos A, Prada PO, Saad MJA. Blocking iNOS and endoplasmic reticulum stress synergistically improves insulin resistance in mice. Mol Metab 2016; 6:206-218. [PMID: 28180062 PMCID: PMC5279911 DOI: 10.1016/j.molmet.2016.12.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 12/02/2016] [Accepted: 12/12/2016] [Indexed: 12/12/2022] Open
Abstract
OBJECTIVE Recent data show that iNOS has an essential role in ER stress in obesity. However, whether iNOS is sufficient to account for obesity-induced ER stress and Unfolded Protein Response (UPR) has not yet been investigated. In the present study, we used iNOS knockout mice to investigate whether high-fat diet (HFD) can still induce residual ER stress-associated insulin resistance. METHODS For this purpose, we used the intraperitoneal glucose tolerance test (GTT), euglycemic-hyperinsulinemic clamp, western blotting and qPCR in liver, muscle, and adipose tissue of iNOS KO and control mice on HFD. RESULTS The results of the present study demonstrated that, in HFD fed mice, iNOS-induced alteration in insulin signaling is an essential mechanism of insulin resistance in muscle, suggesting that iNOS may represent an important target that could be blocked in order to improve insulin sensitivity in this tissue. However, in liver and adipose tissue, the insulin resistance induced by HFD was only partially dependent on iNOS, and, even in the presence of genetic or pharmacological blockade of iNOS, a clear ER stress associated with altered insulin signaling remained evident in these tissues. When this ER stress was blocked pharmacologically, insulin signaling was improved, and a complete recovery of glucose tolerance was achieved. CONCLUSIONS Taken together, these results reinforce the tissue-specific regulation of insulin signaling in obesity, with iNOS being sufficient to account for insulin resistance in muscle, but in liver and adipose tissue ER stress and insulin resistance can be induced by both iNOS-dependent and iNOS-independent mechanisms.
Collapse
Key Words
- AKT, Protein kinase B
- ATF6, activating transcription factor 6
- Blocking
- ER, endoplasmic reticulum
- Endoplasmic reticulum stress
- GAPDH, glyceraldehyde 3-phosphate dehydrogenase
- GTT, glucose tolerance test
- HFD, high-fat diet
- IKK, kappa α/β kinase
- IRE1, inositol requiring enzyme 1
- ITT, insulin tolerance test
- Improving
- Insulin resistance
- JNK, c-JunN-terminal kinase
- NO, nitric oxide
- PERK, protein kinase RNA-like ER kinase
- UPR, unfolded protein response
- iNOS
- iNOS, inducible nitric oxide synthase
- qPCR, real time PCR
Collapse
Affiliation(s)
- Tamires M Zanotto
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Paula G F Quaresma
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Dioze Guadagnini
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Lais Weissmann
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Andressa C Santos
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Juliana F Vecina
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Kelly Calisto
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Andrey Santos
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Patrícia O Prada
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; School of Applied Sciences, State University of Campinas (UNICAMP), Limeira, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil
| | - Mario J A Saad
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, SP, Brazil; Department of Medical Clinics, Obesity and Comorbidities Research Center (O.C.R.C.), State University of Campinas (UNICAMP), Campinas, SP, Brazil.
| |
Collapse
|
118
|
Diabetic Microvascular Disease and Pulmonary Fibrosis: The Contribution of Platelets and Systemic Inflammation. Int J Mol Sci 2016; 17:ijms17111853. [PMID: 27834824 PMCID: PMC5133853 DOI: 10.3390/ijms17111853] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 10/24/2016] [Accepted: 11/01/2016] [Indexed: 12/29/2022] Open
Abstract
Diabetes is strongly associated with systemic inflammation and oxidative stress, but its effect on pulmonary vascular disease and lung function has often been disregarded. Several studies identified restrictive lung disease and fibrotic changes in diabetic patients and in animal models of diabetes. While microvascular dysfunction is a well-known complication of diabetes, the mechanisms leading to diabetes-induced lung injury have largely been disregarded. We described the potential involvement of diabetes-induced platelet-endothelial interactions in perpetuating vascular inflammation and oxidative injury leading to fibrotic changes in the lung. Changes in nitric oxide synthase (NOS) activation and decreased NO bioavailability in the diabetic lung increase platelet activation and vascular injury and may account for platelet hyperreactivity reported in diabetic patients. Additionally, the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway has been reported to mediate pancreatic islet damage, and is implicated in the onset of diabetes, inflammation and vascular injury. Many growth factors and diabetes-induced agonists act via the JAK/STAT pathway. Other studies reported the contribution of the JAK/STAT pathway to the regulation of the pulmonary fibrotic process but the role of this pathway in the development of diabetic lung fibrosis has not been considered. These observations may open new therapeutic perspectives for modulating multiple pathways to mitigate diabetes onset or its pulmonary consequences.
Collapse
|
119
|
Brinkmann C, Schulte-Körne B, Grau M, Obels S, Kemmerling R, Schiffer T, Bloch W, Brixius K. Effects of Endurance Training on the Skeletal Muscle Nitric Oxide Metabolism in Insulin-Independent Type 2 Diabetic Men-A Pilot Study. Metab Syndr Relat Disord 2016; 15:52-58. [PMID: 27782779 DOI: 10.1089/met.2016.0092] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Increases in the amount of inducible nitric oxide synthase (iNOS) protein and abnormal production of nitric oxide (NO) in skeletal muscle have been suggested to be associated with peripheral insulin resistance in patients with type 2 diabetes mellitus (T2DM). This pilot study analyzed whether a 3-month endurance training can affect iNOS protein and NO metabolite levels in the vastus lateralis muscle of insulin-independent T2DM men, thereby affecting the patients` glycemic control. Furthermore, serum molecules, which have been shown to activate iNOS protein expression in in vitro experiments, were quantified. METHODS Eight overweight/obese T2DM men (years = 61 ± 10) participated in the study. Muscle biopsies and venous blood collections were performed at T1 (6 weeks before training), T2 (1 week before training), and T3 (3 to 4 days after training). Protein contents (iNOS) were determined by Western blotting, nitrite concentrations by chemiluminescence, and serum molecule levels by enzyme-linked immunosorbent assay kits. RESULTS The training reduced iNOS protein contents significantly (T2-T3: approximately -31%, P = 0.018). Nitrite concentrations as well as fasting glucose and HbA1c decreased, but not significantly. Serum tumor necrosis factor-α, thiobarbituric acid-reactive substances (lipid peroxidation as an indirect measure of reactive oxygen species), lipopolysaccharide binding protein, interferon-γ, and interleukin-1β showed no significant changes. CONCLUSIONS The data indicate that the endurance training performed in the present study can reduce iNOS protein contents in insulin-independent T2DM men. Future studies should identify key molecules in iNOS regulation in vivo and fully clarify whether iNOS downregulation can help improve insulin sensitivity in T2DM patients in the long term.
Collapse
Affiliation(s)
- Christian Brinkmann
- 1 Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne , Cologne, Germany
| | - Benedikt Schulte-Körne
- 1 Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne , Cologne, Germany
| | - Marijke Grau
- 1 Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne , Cologne, Germany
| | - Sinja Obels
- 1 Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne , Cologne, Germany
| | - Roman Kemmerling
- 1 Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne , Cologne, Germany
| | - Thorsten Schiffer
- 2 Outpatient Clinic for Sports Traumatology and Public Health Consultation, German Sport University Cologne , Cologne, Germany
| | - Wilhelm Bloch
- 1 Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne , Cologne, Germany
| | - Klara Brixius
- 1 Department of Molecular and Cellular Sport Medicine, Institute of Cardiovascular Research and Sport Medicine, German Sport University Cologne , Cologne, Germany
| |
Collapse
|
120
|
Roy B, Curtis ME, Fears LS, Nahashon SN, Fentress HM. Molecular Mechanisms of Obesity-Induced Osteoporosis and Muscle Atrophy. Front Physiol 2016; 7:439. [PMID: 27746742 PMCID: PMC5040721 DOI: 10.3389/fphys.2016.00439] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2016] [Accepted: 09/15/2016] [Indexed: 12/19/2022] Open
Abstract
Obesity and osteoporosis are two alarming health disorders prominent among middle and old age populations, and the numbers of those affected by these two disorders are increasing. It is estimated that more than 600 million adults are obese and over 200 million people have osteoporosis worldwide. Interestingly, both of these abnormalities share some common features including a genetic predisposition, and a common origin: bone marrow mesenchymal stromal cells. Obesity is characterized by the expression of leptin, adiponectin, interleukin 6 (IL-6), interleukin 10 (IL-10), monocyte chemotactic protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), macrophage colony stimulating factor (M-CSF), growth hormone (GH), parathyroid hormone (PTH), angiotensin II (Ang II), 5-hydroxy-tryptamine (5-HT), Advance glycation end products (AGE), and myostatin, which exert their effects by modulating the signaling pathways within bone and muscle. Chemical messengers (e.g., TNF-α, IL-6, AGE, leptins) that are upregulated or downregulated as a result of obesity have been shown to act as negative regulators of osteoblasts, osteocytes and muscles, as well as positive regulators of osteoclasts. These additive effects of obesity ultimately increase the risk for osteoporosis and muscle atrophy. The aim of this review is to identify the potential cellular mechanisms through which obesity may facilitate osteoporosis, muscle atrophy and bone fractures.
Collapse
Affiliation(s)
- Bipradas Roy
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| | - Mary E Curtis
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| | - Letimicia S Fears
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| | - Samuel N Nahashon
- Department of Agricultural and Environmental Sciences, Tennessee State University Nashville, TN, USA
| | - Hugh M Fentress
- Department of Biological Sciences, Tennessee State University Nashville, TN, USA
| |
Collapse
|
121
|
Assmann TS, Brondani LA, Bouças AP, Rheinheimer J, de Souza BM, Canani LH, Bauer AC, Crispim D. Nitric oxide levels in patients with diabetes mellitus: A systematic review and meta-analysis. Nitric Oxide 2016; 61:1-9. [PMID: 27677584 DOI: 10.1016/j.niox.2016.09.009] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Revised: 08/30/2016] [Accepted: 09/23/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Altered serum nitric oxide (NO) levels in patients with diabetes mellitus (DM) have been reported by different studies; however, results are still controversial. Until this date, no meta-analysis evaluated the association of NO levels with DM. Thus, this paper describes a meta-analysis conducted to evaluate if there is a relationship between NO levels and type 1 DM (T1DM) or type 2 DM (T2DM). METHODS A literature search was done to identify all studies that investigated NO levels between T1DM or T2DM patients (cases) and non-diabetic subjects (controls). Measurement of nitrate and nitrite (NOx - the stable NO products) were used to estimate NO concentrations because they closely reflect NO bioavailability. Weighted mean differences (WMD) of NOx levels between case and control samples were calculated for T1DM and T2DM groups. RESULTS Thirty studies were eligible for inclusion in the meta-analysis (8 in T1DM samples and 22 in T2DM samples). NOx levels were increased in European T1DM patients compared with controls [random effect model (REM) WMD = 8.55, 95% CI 2.88 - 14.21]. No other ethnicity was evaluated in T1DM studies. NOx levels were also increased in both European (REM WMD = 18.76, 95% CI 1.67 - 35.85) and Asian (REM WMD = 18.41, 95% CI 8.01 - 28.81) T2DM patients, but not in Latin American patients compared with controls. CONCLUSIONS This meta-analysis detected a significant increase in NOx levels in European T1DM patients as well as European and Asian T2DM patients. Further studies in other ethnicities are necessary to confirm these data.
Collapse
Affiliation(s)
- Taís S Assmann
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Letícia A Brondani
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Ana P Bouças
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Jakeline Rheinheimer
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Bianca M de Souza
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Luís H Canani
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Andrea C Bauer
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil
| | - Daisy Crispim
- Endocrine Division, Hospital de Clínicas de Porto Alegre, Porto Alegre, RS, Brazil; Post-graduation Program in Medical Sciences: Endocrinology, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil.
| |
Collapse
|
122
|
Jang JE, Ko MS, Yun JY, Kim MO, Kim JH, Park HS, Kim AR, Kim HJ, Kim BJ, Ahn YE, Oh JS, Lee WJ, Harris RA, Koh EH, Lee KU. Nitric Oxide Produced by Macrophages Inhibits Adipocyte Differentiation and Promotes Profibrogenic Responses in Preadipocytes to Induce Adipose Tissue Fibrosis. Diabetes 2016; 65:2516-28. [PMID: 27246913 DOI: 10.2337/db15-1624] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 05/18/2016] [Indexed: 11/13/2022]
Abstract
Fibrosis of adipose tissue induces ectopic fat accumulation and insulin resistance by inhibiting adipose tissue expandability. Mechanisms responsible for the induction of adipose tissue fibrosis may provide therapeutic targets but are poorly understood. In this study, high-fat diet (HFD)-fed wild-type (WT) and iNOS(-/-) mice were used to examine the relationship between nitric oxide (NO) produced by macrophages and adipose tissue fibrosis. In contrast to WT mice, iNOS(-/-) mice fed an HFD were protected from infiltration of proinflammatory macrophages and adipose tissue fibrosis. Hypoxia-inducible factor 1α (HIF-1α) protein level was increased in adipose tissue of HFD-fed WT mice, but not iNOS(-/-) mice. In contrast, the expression of mitochondrial biogenesis factors was decreased in HFD-fed WT mice, but not iNOS(-/-) mice. In studies with cultured cells, macrophage-derived NO decreased the expression of mitochondrial biogenesis factors, and increased HIF-1α protein level, DNA damage, and phosphorylated p53 in preadipocytes. By activating p53 signaling, NO suppressed peroxisome proliferator-activated receptor γ coactivator 1α expression, which induced mitochondrial dysfunction and inhibited preadipocyte differentiation in adipocytes. The effects of NO were blocked by rosiglitazone. The findings suggest that NO produced by macrophages induces mitochondrial dysfunction in preadipocytes by activating p53 signaling, which in turn increases HIF-1α protein level and promotes a profibrogenic response in preadipocytes that results in adipose tissue fibrosis.
Collapse
Affiliation(s)
- Jung Eun Jang
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Myoung Seok Ko
- Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Ji-Young Yun
- Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Mi-Ok Kim
- Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Jin Hee Kim
- Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Hye Sun Park
- Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Ah-Ram Kim
- Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Hyuk-Joong Kim
- Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Bum Joong Kim
- Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Young Eun Ahn
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Jin Sun Oh
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Woo Je Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Robert A Harris
- Richard L. Roudebush VA Medical Center and the Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN
| | - Eun Hee Koh
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| | - Ki-Up Lee
- Department of Internal Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea Metabolism Research Unit, Asan Institute for Life Sciences, Seoul, Republic of Korea
| |
Collapse
|
123
|
Affourtit C. Mitochondrial involvement in skeletal muscle insulin resistance: A case of imbalanced bioenergetics. BIOCHIMICA ET BIOPHYSICA ACTA-BIOENERGETICS 2016; 1857:1678-93. [PMID: 27473535 DOI: 10.1016/j.bbabio.2016.07.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/19/2016] [Accepted: 07/23/2016] [Indexed: 12/16/2022]
Abstract
Skeletal muscle insulin resistance in obesity associates with mitochondrial dysfunction, but the causality of this association is controversial. This review evaluates mitochondrial models of nutrient-induced muscle insulin resistance. It transpires that all models predict that insulin resistance arises as a result of imbalanced cellular bioenergetics. The nature and precise origin of the proposed insulin-numbing molecules differ between models but all species only accumulate when metabolic fuel supply outweighs energy demand. This observation suggests that mitochondrial deficiency in muscle insulin resistance is not merely owing to intrinsic functional defects, but could instead be an adaptation to nutrient-induced changes in energy expenditure. Such adaptive effects are likely because muscle ATP supply is fully driven by energy demand. This market-economic control of myocellular bioenergetics offers a mechanism by which insulin-signalling deficiency can cause apparent mitochondrial dysfunction, as insulin resistance lowers skeletal muscle anabolism and thus dampens ATP demand and, consequently, oxidative ATP synthesis.
Collapse
Affiliation(s)
- Charles Affourtit
- School of Biomedical and Healthcare Sciences, Plymouth University Peninsula Schools of Medicine and Dentistry, Plymouth University, Drake Circus, PL4 8AA Plymouth, UK.
| |
Collapse
|
124
|
Cinar R, Iyer MR, Liu Z, Cao Z, Jourdan T, Erdelyi K, Godlewski G, Szanda G, Liu J, Park JK, Mukhopadhyay B, Rosenberg AZ, Liow JS, Lorenz RG, Pacher P, Innis RB, Kunos G. Hybrid inhibitor of peripheral cannabinoid-1 receptors and inducible nitric oxide synthase mitigates liver fibrosis. JCI Insight 2016; 1:87336. [PMID: 27525312 DOI: 10.1172/jci.insight.87336] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Liver fibrosis, a consequence of chronic liver injury and a way station to cirrhosis and hepatocellular carcinoma, lacks effective treatment. Endocannabinoids acting via cannabinoid-1 receptors (CB1R) induce profibrotic gene expression and promote pathologies that predispose to liver fibrosis. CB1R antagonists produce opposite effects, but their therapeutic development was halted due to neuropsychiatric side effects. Inducible nitric oxide synthase (iNOS) also promotes liver fibrosis and its underlying pathologies, but iNOS inhibitors tested to date showed limited therapeutic efficacy in inflammatory diseases. Here, we introduce a peripherally restricted, orally bioavailable CB1R antagonist, which accumulates in liver to release an iNOS inhibitory leaving group. In mouse models of fibrosis induced by CCl4 or bile duct ligation, the hybrid CB1R/iNOS antagonist surpassed the antifibrotic efficacy of the CB1R antagonist rimonabant or the iNOS inhibitor 1400W, without inducing anxiety-like behaviors or CB1R occupancy in the CNS. The hybrid inhibitor also targeted CB1R-independent, iNOS-mediated profibrotic pathways, including increased PDGF, Nlrp3/Asc3, and integrin αvβ6 signaling, as judged by its ability to inhibit these pathways in cnr1-/- but not in nos2-/- mice. Additionally, it was able to slow fibrosis progression and to attenuate established fibrosis. Thus, dual-target peripheral CB1R/iNOS antagonists have therapeutic potential in liver fibrosis.
Collapse
Affiliation(s)
| | | | - Ziyi Liu
- Laboratory of Physiologic Studies and
| | - Zongxian Cao
- Laboratory of Oxidative Stress and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, Washington, DC, USA
| | | | - Katalin Erdelyi
- Laboratory of Oxidative Stress and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, Washington, DC, USA
| | | | | | - Jie Liu
- Laboratory of Physiologic Studies and
| | | | | | - Avi Z Rosenberg
- Kidney Diseases Section, National Institute on Diabetes, Digestive, and Kidney Diseases, Washington, DC, USA.,Children's National Medical Center, Washington, DC, USA
| | - Jeih-San Liow
- Molecular Imaging Branch, National Institute on Mental Health, NIH, Bethesda, Maryland, USA
| | - Robin G Lorenz
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Pal Pacher
- Laboratory of Oxidative Stress and Tissue Injury, National Institute on Alcohol Abuse and Alcoholism, Washington, DC, USA
| | - Robert B Innis
- Molecular Imaging Branch, National Institute on Mental Health, NIH, Bethesda, Maryland, USA
| | | |
Collapse
|
125
|
Stegen S, Stegen B, Aldini G, Altomare A, Cannizzaro L, Orioli M, Gerlo S, Deldicque L, Ramaekers M, Hespel P, Derave W. Plasma carnosine, but not muscle carnosine, attenuates high-fat diet-induced metabolic stress. Appl Physiol Nutr Metab 2016; 40:868-76. [PMID: 26307517 DOI: 10.1139/apnm-2015-0042] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
There is growing in vivo evidence that the dipeptide carnosine has protective effects in metabolic diseases. A critical unanswered question is whether its site of action is tissues or plasma. This was investigated using oral carnosine versus β-alanine supplementation in a high-fat diet rat model. Thirty-six male Sprague-Dawley rats received a control diet (CON), a high-fat diet (HF; 60% of energy from fat), the HF diet with 1.8% carnosine (HFcar), or the HF diet with 1% β-alanine (HFba), as β-alanine can increase muscle carnosine without increasing plasma carnosine. Insulin sensitivity, inflammatory signaling, and lipoxidative stress were determined in skeletal muscle and blood. In a pilot study, urine was collected. The 3 HF groups were significantly heavier than the CON group. Muscle carnosine concentrations increased equally in the HFcar and HFba groups, while elevated plasma carnosine levels and carnosine-4-hydroxy-2-nonenal adducts were detected only in the HFcar group. Elevated plasma and urine N(ε)-(carboxymethyl)lysine in HF rats was reduced by ∼50% in the HFcar group but not in the HFba group. Likewise, inducible nitric oxide synthase mRNA was decreased by 47% (p < 0.05) in the HFcar group, but not in the HFba group, compared with HF rats. We conclude that plasma carnosine, but not muscle carnosine, is involved in preventing early-stage lipoxidation in the circulation and inflammatory signaling in the muscle of rats.
Collapse
Affiliation(s)
- Sanne Stegen
- a Department of Movement and Sport Sciences, Ghent University, Watersportlaan 2, 9000 Ghent, Belgium
| | - Bram Stegen
- a Department of Movement and Sport Sciences, Ghent University, Watersportlaan 2, 9000 Ghent, Belgium
| | - Giancarlo Aldini
- b Department of Pharmaceutical Sciences, Università degli Studi di Milano, via Mangiagalli 25, 20133 Milan, Italy
| | - Alessandra Altomare
- b Department of Pharmaceutical Sciences, Università degli Studi di Milano, via Mangiagalli 25, 20133 Milan, Italy
| | - Luca Cannizzaro
- b Department of Pharmaceutical Sciences, Università degli Studi di Milano, via Mangiagalli 25, 20133 Milan, Italy
| | - Marica Orioli
- b Department of Pharmaceutical Sciences, Università degli Studi di Milano, via Mangiagalli 25, 20133 Milan, Italy
| | - Sarah Gerlo
- c VIB Department of Medical Protein Research, Ghent University, Albert Baertsoenkaai 3, 9000 Ghent, Belgium
| | - Louise Deldicque
- d Department of Kinesiology, Research Group in Exercise Physiology, KU Leuven, Tervuursevest 101, Box 1500, 3001 Leuven, Belgium
| | - Monique Ramaekers
- d Department of Kinesiology, Research Group in Exercise Physiology, KU Leuven, Tervuursevest 101, Box 1500, 3001 Leuven, Belgium
| | - Peter Hespel
- d Department of Kinesiology, Research Group in Exercise Physiology, KU Leuven, Tervuursevest 101, Box 1500, 3001 Leuven, Belgium
| | - Wim Derave
- a Department of Movement and Sport Sciences, Ghent University, Watersportlaan 2, 9000 Ghent, Belgium
| |
Collapse
|
126
|
Control of leukemia relapse after allogeneic hematopoietic stem cell transplantation: integrating transplantation with genetically modified T cell therapies. Curr Opin Hematol 2016; 22:489-96. [PMID: 26335421 DOI: 10.1097/moh.0000000000000177] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Leukemia relapse remains a significant cause of failure after allogeneic hematopoietic stem cell transplantation (HSCT). Although it is widely accepted that immunological components of the stem cell graft play a critical role in promoting leukemia eradication (graft versus leukemia effect), it is also evident that their efficacy is frequently inadequate and leukemia relapse still occurs. This article reviews recent insights into T cell-based posttransplant immunotherapy approaches aimed at preventing or controlling leukemia relapse. RECENT FINDINGS Donor lymphocyte infusion with T cells genetically modified with safety switches improves the patient's immune reconstitution while offering appropriate control of graft versus host disease. T lymphocytes engineered with artificial tumor-specific receptors such as αβT-cell receptor chains or chimeric antigen receptors are major players in promoting antileukemia effects after allogeneic HSCT. SUMMARY The landscape of adoptive T cell therapies after allogeneic HSCT has seen significant achievements with the introduction of T cell engineering. Gene transfer grants the generation of T cell products characterized by standardizable specificity and functionality. This aspect is critical for scalable and reproducible approaches for application in large clinical studies. The clinical results so far reported are encouraging and multicenter studies conducted by pharmaceutical companies will provide definitive conclusions on the clinical impact of these new methodologies.
Collapse
|
127
|
Haddad Y, Couture R. Interplay between the kinin B1 receptor and inducible nitric oxide synthase in insulin resistance. Br J Pharmacol 2016; 173:1988-2000. [PMID: 27059924 DOI: 10.1111/bph.13491] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Revised: 02/23/2016] [Accepted: 03/26/2016] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND AND PURPOSE Kinins are vasoactive and pro-inflammatory peptides whose biological effects are mediated by two GPCRs, named B1 and B2 receptors. While the B2 receptor plays a protective role in the cardiovascular system via the activation of endothelial NOS, the B1 receptor is associated with vascular inflammation, insulin resistance and diabetic complications. Because the B1 receptor is a potent activator of the inducible form of NOS (iNOS), this study has addressed the role of iNOS in the deleterious effects of B1 receptors in insulin resistance. EXPERIMENTAL APPROACH Male Sprague-Dawley rats (50-75 g) had free access to a drinking solution containing 10% d-glucose or tap water (control) for 9 weeks. During the last week, a selective iNOS inhibitor (1400W, 1 mg·kg(-1) twice daily) or its vehicle was administered s.c. KEY RESULTS Prolonged glucose treatment caused insulin resistance and several hallmarks of type 2 diabetes. Whereas the treatment with 1400W had no impact on the elevated systolic blood pressure and leptin levels in glucose-fed rats, it significantly reversed or attenuated hyperglycaemia, hyperinsulinaemia, insulin resistance (HOMA index), body weight gain, peroxynitrite formation (nitrotyrosine expression) and the up-regulation of biomarkers of inflammation (B1 receptor, carboxypeptidase M, iNOS and IL-1β) in renal cortex and aorta and to some extent in the liver. CONCLUSIONS AND IMPLICATIONS Pharmacological blockade of iNOS prevents the formation of peroxynitrite, which amplifies the pro-inflammatory effects of B1 receptors through a positive feedback mechanism. Hence, targeting iNOS can prevent the deleterious effects of B1 receptors in insulin resistance and peripheral inflammation.
Collapse
Affiliation(s)
- Youssef Haddad
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| | - Réjean Couture
- Department of Molecular and Integrative Physiology, Faculty of Medicine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
128
|
Hespe GE, Kataru RP, Savetsky IL, García Nores GD, Torrisi JS, Nitti MD, Gardenier JC, Zhou J, Yu JZ, Jones LW, Mehrara BJ. Exercise training improves obesity-related lymphatic dysfunction. J Physiol 2016; 594:4267-82. [PMID: 26931178 PMCID: PMC4967732 DOI: 10.1113/jp271757] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 02/24/2016] [Indexed: 01/04/2023] Open
Abstract
Key points Obesity results in perilymphatic inflammation and lymphatic dysfunction. Lymphatic dysfunction in obesity is characterized by decreased lymphatic vessel density, decreased collecting lymphatic vessel pumping frequency, decreased lymphatic trafficking of immune cells, increased lymphatic vessel leakiness and changes in the gene expression patterns of lymphatic endothelial cells. Aerobic exercise, independent of weight loss, decreases perilymphatic inflammatory cell accumulation, improves lymphatic function and reverses pathological changes in gene expression in lymphatic endothelial cells.
Abstract Although previous studies have shown that obesity markedly decreases lymphatic function, the cellular mechanisms that regulate this response remain unknown. In addition, it is unclear whether the pathological effects of obesity on the lymphatic system are reversible with behavioural modifications. The purpose of this study, therefore, was to analyse lymphatic vascular changes in obese mice and to determine whether these pathological effects are reversible with aerobic exercise. We randomized obese mice to either aerobic exercise (treadmill running for 30 min per day, 5 days a week, for 6 weeks) or a sedentary group that was not exercised and analysed lymphatic function using a variety of outcomes. We found that sedentary obese mice had markedly decreased collecting lymphatic vessel pumping capacity, decreased lymphatic vessel density, decreased lymphatic migration of immune cells, increased lymphatic vessel leakiness and decreased expression of lymphatic specific markers compared with lean mice (all P < 0.01). Aerobic exercise did not cause weight loss but markedly improved lymphatic function compared with sedentary obese mice. Exercise had a significant anti‐inflammatory effect, resulting in decreased perilymphatic accumulation of inflammatory cells and inducible nitric oxide synthase expression. In addition, exercise normalized isolated lymphatic endothelial cell gene expression of lymphatic specific genes, including VEGFR‐3 and Prox1. Taken together, our findings suggest that obesity impairs lymphatic function via multiple mechanisms and that these pathological changes can be reversed, in part, with aerobic exercise, independent of weight loss. In addition, our study shows that obesity‐induced lymphatic endothelial cell gene expression changes are reversible with behavioural modifications. Obesity results in perilymphatic inflammation and lymphatic dysfunction. Lymphatic dysfunction in obesity is characterized by decreased lymphatic vessel density, decreased collecting lymphatic vessel pumping frequency, decreased lymphatic trafficking of immune cells, increased lymphatic vessel leakiness and changes in the gene expression patterns of lymphatic endothelial cells. Aerobic exercise, independent of weight loss, decreases perilymphatic inflammatory cell accumulation, improves lymphatic function and reverses pathological changes in gene expression in lymphatic endothelial cells.
Collapse
Affiliation(s)
- Geoffrey E Hespe
- The Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Raghu P Kataru
- The Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Ira L Savetsky
- The Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gabriela D García Nores
- The Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jeremy S Torrisi
- The Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew D Nitti
- The Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jason C Gardenier
- The Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jie Zhou
- The Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Jessie Z Yu
- The Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lee W Jones
- The Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Babak J Mehrara
- The Department of Surgery, Division of Plastic and Reconstructive Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
129
|
A high fat diet induces sex-specific differences in hepatic lipid metabolism and nitrite/nitrate in rats. Nitric Oxide 2016; 54:51-9. [DOI: 10.1016/j.niox.2016.02.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Revised: 02/17/2016] [Accepted: 02/23/2016] [Indexed: 02/08/2023]
|
130
|
Wellhauser L, Chalmers JA, Belsham DD. Nitric Oxide Exerts Basal and Insulin-Dependent Anorexigenic Actions in POMC Hypothalamic Neurons. Mol Endocrinol 2016; 30:402-16. [PMID: 26930171 DOI: 10.1210/me.2015-1275] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
The arcuate nucleus of the hypothalamus represents a key center for the control of appetite and feeding through the regulation of 2 key neuronal populations, notably agouti-related peptide/neuropeptide Y and proopimelanocortin (POMC)/cocaine- and amphetamine-regulated transcript neurons. Altered regulation of these neuronal networks, in particular the dysfunction of POMC neurons upon high-fat consumption, is a major pathogenic mechanism involved in the development of obesity and type 2 diabetes mellitus. Efforts are underway to preserve the integrity or enhance the functionality of POMC neurons in order to prevent or treat these metabolic diseases. Here, we report for the first time that the nitric oxide (NO(-)) donor, sodium nitroprusside (SNP) mediates anorexigenic actions in both hypothalamic tissue and hypothalamic-derived cell models by mediating the up-regulation of POMC levels. SNP increased POMC mRNA in a dose-dependent manner and enhanced α-melanocortin-secreting hormone production and secretion in mHypoA-POMC/GFP-2 cells. SNP also enhanced insulin-driven POMC expression likely by inhibiting the deacetylase activity of sirtuin 1. Furthermore, SNP enhanced insulin-dependent POMC expression, likely by reducing the transcriptional repression of Foxo1 on the POMC gene. Prolonged SNP exposure prevented the development of insulin resistance. Taken together, the NO(-) donor SNP enhances the anorexigenic potential of POMC neurons by promoting its transcriptional expression independent and in cooperation with insulin. Thus, increasing cellular NO(-) levels represents a hormone-independent method of promoting anorexigenic output from the existing POMC neuronal populations and may be advantageous in the fight against these prevalent disorders.
Collapse
Affiliation(s)
- Leigh Wellhauser
- Department of Physiology (L.W., J.A.C., D.D.B.), University of Toronto, Toronto, Ontario, Canada M5G 1A8; and Departments of Obstetrics, Gynaecology, and Medicine (D.D.B.), University of Toronto and Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Jennifer A Chalmers
- Department of Physiology (L.W., J.A.C., D.D.B.), University of Toronto, Toronto, Ontario, Canada M5G 1A8; and Departments of Obstetrics, Gynaecology, and Medicine (D.D.B.), University of Toronto and Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| | - Denise D Belsham
- Department of Physiology (L.W., J.A.C., D.D.B.), University of Toronto, Toronto, Ontario, Canada M5G 1A8; and Departments of Obstetrics, Gynaecology, and Medicine (D.D.B.), University of Toronto and Division of Cellular and Molecular Biology, Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada M5S 1A8
| |
Collapse
|
131
|
Khodabandehloo H, Gorgani-Firuzjaee S, Panahi G, Meshkani R. Molecular and cellular mechanisms linking inflammation to insulin resistance and β-cell dysfunction. Transl Res 2016; 167:228-56. [PMID: 26408801 DOI: 10.1016/j.trsl.2015.08.011] [Citation(s) in RCA: 203] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 08/29/2015] [Accepted: 08/31/2015] [Indexed: 12/13/2022]
Abstract
Obesity is a major public health problem worldwide, and it is associated with an increased risk of developing type 2 diabetes. It is now commonly accepted that chronic inflammation associated with obesity induces insulin resistance and β-cell dysfunction in diabetic patients. Obesity-associated inflammation is characterized by increased abundance of macrophages and enhanced production of inflammatory cytokines in adipose tissue. Adipose tissue macrophages are suggested to be the major source of local and systemic inflammatory mediators such as tumor necrosis factor α, interleukin (IL)-1β, and IL-6. These cytokines induce insulin resistance in insulin target tissues by activating the suppressors of cytokine signaling proteins, several kinases such as c-Jun N-terminal kinase, IκB kinase β, and protein kinase C, inducible nitric oxide synthase, extracellular signal-regulated kinase, and protein tyrosine phosphatases such as protein tyrosine phosphatase 1B. These activated factors impair the insulin signaling at the insulin receptor and the insulin receptor substrates levels. The same process most likely occurs in the pancreas as it contains a pool of tissue-resident macrophages. High concentrations of glucose or palmitate via the chemokine production promote further immune cell migration and infiltration into the islets. These events ultimately induce inflammatory responses leading to the apoptosis of the pancreatic β cells. In this review, the cellular and molecular players that participate in the regulation of obesity-induced inflammation are discussed, with particular attention being placed on the roles of the molecular players linking inflammation to insulin resistance and β-cell dysfunction.
Collapse
Affiliation(s)
- Hadi Khodabandehloo
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Sattar Gorgani-Firuzjaee
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Ghodratollah Panahi
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran
| | - Reza Meshkani
- Department of Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Islamic Republic of Iran.
| |
Collapse
|
132
|
Yin R, Fang L, Li Y, Xue P, Li Y, Guan Y, Chang Y, Chen C, Wang N. Pro-inflammatory Macrophages suppress PPARγ activity in Adipocytes via S-nitrosylation. Free Radic Biol Med 2015; 89:895-905. [PMID: 26475041 DOI: 10.1016/j.freeradbiomed.2015.10.406] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 09/23/2015] [Accepted: 10/10/2015] [Indexed: 11/29/2022]
Abstract
Peroxisome proliferator-activated receptor-γ (PPARγ) is a ligand-activated nuclear receptor and plays an essential role in insulin signaling. Macrophage infiltration into adipose tissue is a character of metabolic inflammation and closely related to insulin resistance in type 2 diabetes. The mechanism by which pro-inflammatory macrophages cause insulin resistance remains to be elucidated. Here we showed that co-culture with macrophages significantly suppressed the transcriptional activity of PPARγ on its target genes in 3T3-L1 preadipocytes and diabetic primary adipocytes, depending on inducible nitric oxide synthase (iNOS). We further showed that PPARγ underwent S-nitrosylation in response to nitrosative stress. Mass-spectrometry and site-directed mutagenesis revealed that S-nitrosylation at cysteine 168 was responsible for the impairment of PPARγ function. Extended exposure to NO instigated the proteasome-dependent degradation of PPARγ. Consistently, in vivo evidence revealed an association of the decreased PPARγ protein level with increased macrophage infiltration in visceral adipose tissue (VAT) of obese diabetic db/db mice. Together, our results demonstrated that pro-inflammatory macrophages suppressed PPARγ activity in adipocytes via S-nitrosylation, suggesting a novel mechanism linking metabolic inflammation with insulin resistance.
Collapse
Affiliation(s)
- Ruiying Yin
- Institute of Cardiovascular Science and Diabetes Center, Peking University, Beijing 100191, China
| | - Li Fang
- Institute of Cardiovascular Science and Diabetes Center, Peking University, Beijing 100191, China
| | - Yingjia Li
- Institute of Cardiovascular Science and Diabetes Center, Peking University, Beijing 100191, China
| | - Peng Xue
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Yazi Li
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
| | - Youfei Guan
- The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China
| | - Yongsheng Chang
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Science, Chinese Academy of Medical Science and Peking Union Medical College, Beijing 100005, China
| | - Chang Chen
- Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China.
| | - Nanping Wang
- Institute of Cardiovascular Science and Diabetes Center, Peking University, Beijing 100191, China; The Advanced Institute for Medical Sciences, Dalian Medical University, Dalian 116044, China.
| |
Collapse
|
133
|
Porojan MD, Cătană A, Popp RA, Dumitrascu DL, Bala C. The role of NOS2A -954G/C and vascular endothelial growth factor +936C/T polymorphisms in type 2 diabetes mellitus and diabetic nonproliferative retinopathy risk management. Ther Clin Risk Manag 2015; 11:1743-8. [PMID: 26664124 PMCID: PMC4669928 DOI: 10.2147/tcrm.s93172] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Type 2 diabetes mellitus (T2DM) remains one of the major health problems in Europe. Retinopathy is one of the major causes of morbidity in T2DM, strongly influencing the evolution and prognosis of these patients. In the last 2 decades, several studies have been conducted to identify the possible genetic susceptibility factors involved in the pathogenesis of the disease. However, there is little data related to the involvement of vascular endothelial growth factor (VEGF) and nitric oxide synthase (NOS) gene polymorphisms in the T2DM Caucasian population. The objective of this study was to identify a possible connection between NOS2A −954G/C (rs2297518) and VEGF +936C/T (rs3025039) polymorphisms and the risk of developing T2DM and nonproliferative diabetic retinopathy in a Caucasian population group. We investigated 200 patients diagnosed with T2DM and 208 controls. Genotypes were determined by multiplex polymerase chain reaction-restriction fragment length polymorphism. Statistical and comparative analyses (Fisher’s exact test) for dominant and recessive models of NOS2A −954G/C and VEGF +936C/T polymorphisms revealed an increased risk of T2DM (χ2=8.14, phi =0.141, P=0.004, odds ratio [OR] =2.795, 95% confidence interval [CI] =1.347–5.801; χ2=18.814, phi =0.215, P<0.001, OR =2.59, 95% CI =1.675–4.006, respectively). Also, comparative analysis for the recessive model (using Pearson’s chi-square test [χ2] and the phi coefficient [phi]) reveals that the variant CC genotype of NOS2A gene is more frequently associated with T2DM without retinopathy (χ2=3.835, phi =−0.138, P=0.05, OR =0.447, 95% CI =0.197–1.015). In conclusion, the results of the study place VEGF +936C/T polymorphisms among the genetic risk factor for T2DM, whereas NOS2A −954G/C polymorphisms act like a protective individual factor for nonproliferative retinopathy.
Collapse
Affiliation(s)
- Mihai Dumitru Porojan
- Department of Internal Medicine, University of Pharmacy and Medicine, Cluj Napoca, Romania
| | - Andreea Cătană
- Department of Molecular Sciences, University of Pharmacy and Medicine, Cluj Napoca, Romania
| | - Radu A Popp
- Department of Molecular Sciences, University of Pharmacy and Medicine, Cluj Napoca, Romania
| | - Dan L Dumitrascu
- Department of Internal Medicine, University of Pharmacy and Medicine, Cluj Napoca, Romania
| | - Cornelia Bala
- Department of Diabetes, Nutrition and Metabolic Diseases, Iuliu Hatieganu, University of Pharmacy and Medicine, Cluj Napoca, Romania
| |
Collapse
|
134
|
Wojcik M, Zieleniak A, Zurawska-Klis M, Cypryk K, Wozniak LA. Increased expression of immune-related genes in leukocytes of patients with diagnosed gestational diabetes mellitus (GDM). Exp Biol Med (Maywood) 2015; 241:457-65. [PMID: 26568332 DOI: 10.1177/1535370215615699] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 10/14/2015] [Indexed: 12/16/2022] Open
Abstract
Compelling evidence indicates that the immune system is linked to metabolism in gestational diabetes mellitus (GDM), but factors participating in these processes still are awaiting identification. Inducible nitric oxide synthase, encoded by the NOS2 gene, and surfactant protein D, encoded by the SFTPD gene, have been implicated in diabetes. We investigated NOS2 and SFTPD mRNA levels in leukocytes obtained from 125 pregnant women with (n = 87) or without (control group; n = 38) GDM, and, in turn, correlated their expression with clinical parameters of subjects. Leukocytes were isolated from the blood of pregnant women and NOS2 and SFTPD expression in these cells was determined by quantitative real time PCR (qRT-PCR). Univariate correlation analyses were performed to assess an association between leukocyte NOS2 and SFTPD expression and clinical characteristics of patients. qRT-PCR experiments disclosed significantly increased leukocyte NOS2 and SFTPD mRNA levels in hyperglycemic GDM patients (P < 0.05). In the entire study group, there were significant positive associations of leukocyte NOS2 and SFTPD mRNAs with C-reactive protein. Additionally, transcript level of SFTPD also correlated positively with fasting glycemia and insulin resistance. This study demonstrates that an impaired glucose metabolism in GDM may be predominant predictor of leukocyte NOS2 and SFTPD overexpression in diabetic patients. Furthermore, alterations in the expression of these genes are associated with glucose metabolism dysfunction and/or inflammation during pregnancy. In addition, these findings support the utilization of leukocytes as good experimental model to study a relationship between immune-related genes and metabolic changes in women with GDM, as well as to assess the potential mechanisms underlying these alterations.
Collapse
Affiliation(s)
- Marzena Wojcik
- Department of Structural Biology, Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, 90-752 Lodz, Poland
| | - Andrzej Zieleniak
- Department of Structural Biology, Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, 90-752 Lodz, Poland
| | - Monika Zurawska-Klis
- Diabetology and Metabolic Diseases Department, Medical University of Lodz, 92-213 Lodz, Poland Diabetological Medical Center "OmniMed", 93-338 Lodz, Poland
| | - Katarzyna Cypryk
- Diabetology and Metabolic Diseases Department, Medical University of Lodz, 92-213 Lodz, Poland Diabetological Medical Center "OmniMed", 93-338 Lodz, Poland
| | - Lucyna Alicja Wozniak
- Department of Structural Biology, Faculty of Biomedical Sciences and Postgraduate Education, Medical University of Lodz, 90-752 Lodz, Poland
| |
Collapse
|
135
|
Resveratrol suppresses inflammatory responses and improves glucose uptake in adipocytes interacted with macrophages. Genes Genomics 2015. [DOI: 10.1007/s13258-015-0348-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
|
136
|
Macrophage polarization: the link between inflammation and related diseases. Inflamm Res 2015; 65:1-11. [DOI: 10.1007/s00011-015-0874-1] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2015] [Revised: 08/16/2015] [Accepted: 08/25/2015] [Indexed: 01/04/2023] Open
|
137
|
Tian L, Zeng K, Shao W, Yang BB, Fantus IG, Weng J, Jin T. Short-Term Curcumin Gavage Sensitizes Insulin Signaling in Dexamethasone-Treated C57BL/6 Mice. J Nutr 2015; 145:2300-7. [PMID: 26338887 DOI: 10.3945/jn.115.216853] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 07/24/2015] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Long-term dietary curcumin (>12 wk) improves metabolic homeostasis in obese mice by sensitizing insulin signaling and reducing hepatic gluconeogenesis. Whether these occur only secondary to its chronic anti-inflammatory and antioxidative functions is unknown. OBJECTIVE In this study, we assessed the insulin sensitization effect of short-term curcumin gavage in a rapid dexamethasone-induced insulin resistance mouse model, in which the chronic anti-inflammatory function is eliminated. METHODS Six-week-old male C57BL/6 mice received an intraperitoneal injection of dexamethasone (100 mg/kg body weight) or phosphate-buffered saline every day for 5 d, with or without simultaneous curcumin gavage (500 mg/kg body weight). On day 7, insulin tolerance tests were performed. After a booster dexamethasone injection and curcumin gavage on day 8, blood glucose and insulin concentrations were measured. Liver tissues were collected on day 10 for quantitative polymerase chain reaction and Western blotting to assess gluconeogenic gene expression, insulin signaling, and the expression of fibroblast growth factor 21 (FGF21). Primary hepatocytes from separate, untreated C57BL/6 mice were used for testing the in vitro effect of curcumin treatment. RESULTS Dexamethasone injection impaired insulin tolerance (P < 0.05) and elevated ambient plasma insulin concentrations by ~2.7-fold (P < 0.01). Concomitant curcumin administration improved insulin sensitivity and reduced hepatic gluconeogenic gene expression. The insulin sensitization effect of curcumin was demonstrated by increased stimulation of S473 phosphorylation of protein kinase B (P < 0.01) in the dexamethasone-treated mouse liver, as well as the repression of glucose production in primary hepatocytes (P < 0.001). Finally, curcumin gavage increased FGF21 expression by 2.1-fold in the mouse liver (P < 0.05) and curcumin treatment increased FGF21 expression in primary hepatocytes. CONCLUSION These observations suggest that the early beneficial effect of curcumin intervention in dexamethasone-treated mice is the sensitization of insulin signaling, involving the stimulation of FGF21 production, a known insulin sensitizer.
Collapse
Affiliation(s)
- Lili Tian
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Canada; Department of Medicine and Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Canada
| | - Kejing Zeng
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Canada; Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Weijuan Shao
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Canada
| | | | - I George Fantus
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Canada; Department of Medicine and Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada
| | - Jianping Weng
- Department of Endocrinology and Metabolism, Third Affiliated Hospital of Sun Yat-Sen University, and Guangdong Provincial Key Laboratory of Diabetology, Guangzhou, China
| | - Tianru Jin
- Division of Advanced Diagnostics, Toronto General Research Institute, University Health Network, Toronto, Canada; Department of Physiology, University of Toronto, Toronto, Canada
| |
Collapse
|
138
|
McNally B, Griffin JL, Roberts LD. Dietary inorganic nitrate: From villain to hero in metabolic disease? Mol Nutr Food Res 2015; 60:67-78. [PMID: 26227946 PMCID: PMC4863140 DOI: 10.1002/mnfr.201500153] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Revised: 07/13/2015] [Accepted: 07/23/2015] [Indexed: 12/22/2022]
Abstract
Historically, inorganic nitrate was believed to be an inert by‐product of nitric oxide (NO) metabolism that was readily excreted by the body. Studies utilising doses of nitrate far in excess of dietary and physiological sources reported potentially toxic and carcinogenic effects of the anion. However, nitrate is a significant component of our diets, with the majority of the anion coming from green leafy vegetables, which have been consistently shown to offer protection against obesity, type 2 diabetes and metabolic diseases. The discovery of a metabolic pathway in mammals, in which nitrate is reduced to NO, via nitrite, has warranted a re‐examination of the physiological role of this small molecule. Obesity, type 2 diabetes and the metabolic syndrome are associated with a decrease in NO bioavailability. Recent research suggests that the nitrate‐nitrite‐NO pathway may be harnessed as a therapeutic to supplement circulating NO concentrations, with both anti‐obesity and anti‐diabetic effects, as well as improving vascular function. In this review, we examine the key studies that have led to the re‐evaluation of the physiological function of inorganic nitrate, from toxic and carcinogenic metabolite, to a potentially important and beneficial agent in the treatment of metabolic disease.
Collapse
Affiliation(s)
- Ben McNally
- Elsie Widdowson Laboratory, Medical Research Council - Human Nutrition Research, Cambridge, UK.,Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Julian L Griffin
- Elsie Widdowson Laboratory, Medical Research Council - Human Nutrition Research, Cambridge, UK.,Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| | - Lee D Roberts
- Elsie Widdowson Laboratory, Medical Research Council - Human Nutrition Research, Cambridge, UK.,Department of Biochemistry and Cambridge Systems Biology Centre, University of Cambridge, Cambridge, UK
| |
Collapse
|
139
|
Yang L, Calay ES, Fan J, Arduini A, Kunz RC, Gygi SP, Yalcin A, Fu S, Hotamisligil GS. METABOLISM. S-Nitrosylation links obesity-associated inflammation to endoplasmic reticulum dysfunction. Science 2015; 349:500-6. [PMID: 26228140 DOI: 10.1126/science.aaa0079] [Citation(s) in RCA: 185] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The association between inflammation and endoplasmic reticulum (ER) stress has been observed in many diseases. However, if and how chronic inflammation regulates the unfolded protein response (UPR) and alters ER homeostasis in general, or in the context of chronic disease, remains unknown. Here, we show that, in the setting of obesity, inflammatory input through increased inducible nitric oxide synthase (iNOS) activity causes S-nitrosylation of a key UPR regulator, IRE1α, which leads to a progressive decline in hepatic IRE1α-mediated XBP1 splicing activity in both genetic (ob/ob) and dietary (high-fat diet-induced) models of obesity. Finally, in obese mice with liver-specific IRE1α deficiency, reconstitution of IRE1α expression with a nitrosylation-resistant variant restored IRE1α-mediated XBP1 splicing and improved glucose homeostasis in vivo. Taken together, these data describe a mechanism by which inflammatory pathways compromise UPR function through iNOS-mediated S-nitrosylation of IRE1α, which contributes to defective IRE1α activity, impaired ER function, and prolonged ER stress in obesity.
Collapse
Affiliation(s)
- Ling Yang
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ediz S Calay
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Jason Fan
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Alessandro Arduini
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Ryan C Kunz
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Abdullah Yalcin
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Suneng Fu
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Gökhan S Hotamisligil
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA. Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| |
Collapse
|
140
|
Serum nitric oxide metabolites are associated with the risk of hypertriglyceridemic-waist phenotype in women: Tehran Lipid and Glucose Study. Nitric Oxide 2015; 50:52-57. [PMID: 26284308 DOI: 10.1016/j.niox.2015.08.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 08/12/2015] [Indexed: 11/23/2022]
Abstract
BACKGROUND AND AIM There are some controversial issues regarding the association of nitric oxide and obesity-related states. This study was conducted to investigate whether serum nitric oxide metabolites (NOx) could predict the occurrence of visceral lipid accumulation, defined as hypertriglyceridemic-waist (HTW) phenotype. METHODS We used a prospective approach for this study conducted on participants of the Tehran Lipid and Glucose Study, 2243 adult men and women were followed for a median of 6.3 years. Serum NOx concentrations were measured at baseline (2006-2008), and demographics, anthropometrics and biochemical variables were evaluated at baseline and again after a 3-year (2009-2011) and a 6-year follow-up (2012-2014). The occurrence of HTW phenotype, defined as waist circumference ≥90 cm in men and ≥85 cm in women, along with serum triglyceride levels ≥177 mg/dL, were assessed across serum NOx tertiles. RESULTS Mean age of participants was 41.5 ± 14.5 years at baseline and 39.4% were male. The cumulative incidence of HTW phenotype was 37.6% (33.2% in men, 40.5% in women). There was no significant association between serum NOx and the occurrence of HTW phenotype in men. After adjustment of confounding variables, risk of HTW phenotype in women, in the highest compared to the lowest tertile of serum NOx (≥30.9 vs. <19.9 μmol/L), increased by 39% (OR = 1.39, 95% CI = 1.05-1.93, P for trend = 0.053). CONCLUSION Serum NOx level was an independent predictor of HTW phenotype in women.
Collapse
|
141
|
Nepali S, Son JS, Poudel B, Lee JH, Lee YM, Kim DK. Luteolin is a bioflavonoid that attenuates adipocyte-derived inflammatory responses via suppression of nuclear factor-κB/mitogen-activated protein kinases pathway. Pharmacogn Mag 2015; 11:627-35. [PMID: 26246742 PMCID: PMC4522853 DOI: 10.4103/0973-1296.160470] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2014] [Revised: 12/27/2014] [Accepted: 07/10/2015] [Indexed: 01/13/2023] Open
Abstract
Background: Inflammation of adipocytes has been a therapeutic target for treatment of obesity and metabolic disorders which cause insulin resistance and hence lead to type II diabetes. Luteolin is a bioflavonoid with many beneficial properties such as antioxidant, antiproliferative, and anti-cancer. Objectives: To elucidate the potential anti-inflammatory response and the underlying mechanism of luteolin in 3T3-L1 adipocytes. Materials and Methods: We stimulated 3T3-L1 adipocytes with the mixture of tumor necrosis factor-α, lipopolysaccharide, and interferon-γ (TLI) in the presence or absence of luteolin. We performed Griess’ method for nitric oxide (NO) production and measure mRNA and protein expressions by real-time polymerase chain reaction and western blotting, respectively. Results: Luteolin opposed the stimulation of inducible nitric oxide synthase and NO production by simultaneous treatment of adipocytes with TLI. Furthermore, it reduced the pro-inflammatory genes such as cyclooxygenase-2, interleukin-6, resistin, and monocyte chemoattractant protein-1. Furthermore, luteolin improved the insulin sensitivity by enhancing the expression of insulin receptor substrates (IRS1/2) and glucose transporter-4 via phosphatidylinositol-3K signaling pathway. This inhibition was associated with suppression of Iκ-B-α degradation and subsequent inhibition of nuclear factor-κB (NF-κB) p65 translocation to the nucleus. In addition, luteolin blocked the phosphorylation of ERK1/2, c-Jun N-terminal Kinases and also p38 mitogen-activated protein kinases (MAPKs). Conclusions: These results illustrate that luteolin attenuates inflammatory responses in the adipocytes through suppression of NF-κB and MAPKs activation, and also improves insulin sensitivity in 3T3-L1 cells, suggesting that luteolin may represent a therapeutic agent to prevent obesity-associated inflammation and insulin resistance.
Collapse
Affiliation(s)
- Sarmila Nepali
- Department of Immunology and Institute of Medical Sciences, Medical School, Chonbuk National University, Jeonju, Jeonbuk 561-756, Korea
| | - Ji-Seon Son
- Department of Anesthesiology and Pain Medicine, Medical School, Chonbuk National University, Jeonju, Jeonbuk 561-756, Korea
| | - Barun Poudel
- Department of Immunology and Institute of Medical Sciences, Medical School, Chonbuk National University, Jeonju, Jeonbuk 561-756, Korea
| | - Ji-Hyun Lee
- Department of Immunology and Institute of Medical Sciences, Medical School, Chonbuk National University, Jeonju, Jeonbuk 561-756, Korea
| | - Young-Mi Lee
- Department of Oriental Pharmacy, College of Pharmacy and Wonkwang-Oriental Medicines Research Institute, Wonkwang University, Iksan, Jeonbuk, 570-749, Korea
| | - Dae-Ki Kim
- Department of Immunology and Institute of Medical Sciences, Medical School, Chonbuk National University, Jeonju, Jeonbuk 561-756, Korea
| |
Collapse
|
142
|
Wicks SE, Nguyen TT, Breaux C, Kruger C, Stadler K. Diet-induced obesity and kidney disease - In search of a susceptible mouse model. Biochimie 2015; 124:65-73. [PMID: 26248309 DOI: 10.1016/j.biochi.2015.08.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 08/02/2015] [Indexed: 01/09/2023]
Abstract
Obesity and metabolic syndrome are independent risk factors for chronic kidney disease, even without diabetes or hyperglycemia. Here, we compare two mouse models that are susceptible to diet-induced obesity: the relatively renal injury resistant C57BL/6J strain and the DBA2/J strain which is more sensitive to renal injury. Our studies focused on characterizing the effects of high fat diet feeding on renal oxidative stress, albuminuria, fibrosis and podocyte loss/insulin resistance. While the C57BL/6J strain does not develop significant pathological changes in the kidney, at least on lard based diets within the time frame investigated, it does show increased renal iNOS and nitrotyrosine levels and elevated mitochondrial respiration which may be indicative of mitochondrial lipid overfueling. Restricting the high fat diet to decrease adiposity decreased the levels of cellular oxidative stress markers, indicating that adiposity-related proinflammatory changes such as increased iNOS levels may trigger similar responses in the kidney. Mitochondrial respiration remained higher, suggesting that eating excess lipids, despite normal adiposity may still lead to renal mitochondrial overfueling. In comparison, DBA/2J mice developed albuminuria on similar diets, signs of fibrosis, oxidative stress, early signs of podocyte loss (evaluated by the markers podocin and WT-1) and podocyte insulin resistance (unable to phosphorylate their glomerular Akt when insulin was given). To summarize, while the C57BL/6J strain is not particularly susceptible to renal disease, changes in its mitochondrial lipid handling combined with the easy availability of transgenic technology may be an advantage to design new knockout models related to mitochondrial lipid metabolism. The DBA/2J model could serve as a basis for studying podocyte insulin resistance and identifying early renal markers in obesity before more severe kidney disease develops. Based on our observations, we encourage further critical evaluation of mouse models for obesity related chronic kidney disease.
Collapse
Affiliation(s)
- Shawna E Wicks
- Gene Nutrient Interactions Laboratory, Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA, USA
| | - Trang-Tiffany Nguyen
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA, USA
| | - Chelsea Breaux
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA, USA
| | - Claudia Kruger
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA, USA
| | - Krisztian Stadler
- Oxidative Stress and Disease Laboratory, Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA, USA.
| |
Collapse
|
143
|
Sato H, Ishikawa M, Sugai H, Funaki A, Kimura Y, Sumitomo M, Ueno K. Sex hormones influence expression and function of peroxisome proliferator-activated receptor γ in adipocytes: pathophysiological aspects. Horm Mol Biol Clin Investig 2015; 20:51-61. [PMID: 25415639 DOI: 10.1515/hmbci-2014-0026] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Accepted: 10/01/2014] [Indexed: 01/02/2023]
Abstract
Abstract Adipose tissue plays important roles not only in storing fat but also in maintaining metabolic homeostasis by regulating hundreds of biological signaling events and the secretion of various cytokines. One of the central regulators of adipocyte differentiation is peroxisome proliferator-activated receptor γ (PPARγ), which promotes downstream transcriptional activities, such as adiponectin. Disruption of homeostasis leads to the onset of metabolic diseases such as type 2 diabetes and other triggers for metabolic syndrome. Males and post-menopausal females are more likely to be affected with metabolic diseases than pre-menopausal females, suggesting that sex hormones might be involved in the pathogenesis and development of metabolic diseases. Indeed, 17β-estradiol, testosterone, dihydrotestosterone, and their receptors clearly play a role in adipose regulation: they can alter fat distribution and can modify the expression and activities of PPARγ and its downstream adipocytokines. Furthermore, sex hormones affect inflammatory factors such as nitric oxygen, nitric oxygen synthase, and their surrounding components. Sex hormones are also suggested to be involved with sex differences in the efficacy of the PPARγ agonist thiazolidinediones. Therefore, thorough investigation of how sex hormone-dependent regulation of metabolic homeostasis occurs is necessary in order to develop individualized clinical therapies optimized with regard to each patient's biological condition and drug sensitivities.
Collapse
|
144
|
Shi L, Zhang T, Liang X, Hu Q, Huang J, Zhou Y, Chen M, Zhang Q, Zhu J, Mi M. Dihydromyricetin improves skeletal muscle insulin resistance by inducing autophagy via the AMPK signaling pathway. Mol Cell Endocrinol 2015; 409:92-102. [PMID: 25797177 DOI: 10.1016/j.mce.2015.03.009] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2014] [Revised: 03/12/2015] [Accepted: 03/16/2015] [Indexed: 01/10/2023]
Abstract
Skeletal muscle insulin resistance (SMIR) plays an important role in the pathogenesis of type 2 diabetes. Dihydromyricetin (DHM), a natural flavonoid, exerts various bioactivities including anti-oxidative and hepatoprotective effects. Herein, we intended to determine the effect of DHM on SMIR and the underlying mechanisms. We found that DHM increased the expression of phosphorylated insulin receptor substrate-1, phosphorylated Akt and glucose uptake capacity in palmitate-treated L6 myotubes under insulin-stimulated conditions. The expression of light chain 3, Beclin 1, autophagy-related gene 5 (Atg5), the degradation of sequestosome 1 and the formation of autophagosomes were also upregulated by DHM. Suppression of autophagy by 3-methyladenine and bafilomycin A1 or Atg5 and Beclin1 siRNA abolished the favorable effects of DHM on SMIR. Furthermore, DHM increased the levels of phosphorylated AMP-activated protein kinase (AMPK) and Ulk1, and decreased phosphorylated mTOR levels. AMPK inhibitor compound C (CC) and AMPK siRNA abrogated DHM-induced autophagy, subsequently suppressed DHM-induced SMIR improvement. Additionally, DHM inhibited the activity of F1F0-ATPase thereby activating AMPK. Finally, the results of in vivo study conducted in high fat diet-fed rats were consistent with the findings of in vitro study. In conclusion, DHM improved SMIR by inducing autophagy via the activation of AMPK signaling pathway.
Collapse
Affiliation(s)
- Linying Shi
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Ting Zhang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Xinyu Liang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Qin Hu
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Juan Huang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Yong Zhou
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Mingliang Chen
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Qianyong Zhang
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China
| | - Jundong Zhu
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China.
| | - Mantian Mi
- Research Center for Nutrition and Food Safety, Institute of Military Preventive Medicine, Third Military Medical University, Chongqing Key Laboratory of Nutrition and Food Safety, Chongqing 400038, China.
| |
Collapse
|
145
|
Hill AA, Reid Bolus W, Hasty AH. A decade of progress in adipose tissue macrophage biology. Immunol Rev 2015; 262:134-52. [PMID: 25319332 DOI: 10.1111/imr.12216] [Citation(s) in RCA: 174] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
One decade has passed since seminal publications described macrophage infiltration into adipose tissue (AT) as a key contributor to inflammation and obesity-related insulin resistance. Currently, a PubMed search for 'adipose tissue inflammation' reveals over 3500 entries since these original reports. We now know that resident macrophages in lean AT are alternatively activated, M2-like, and play a role in AT homeostasis. In contrast, the macrophages in obese AT are dramatically increased in number and are predominantly classically activated, M1-like, and promote inflammation and insulin resistance. Mediators of AT macrophage (ATM) phenotype include adipokines and fatty acids secreted from adipocytes as well as cytokines secreted from other immune cells in AT. There are several mechanisms that could explain the large increase in ATMs in obesity. These include recruitment-dependent mechanisms such as adipocyte death, chemokine release, and lipolysis of fatty acids. Newer evidence also points to recruitment-independent mechanisms such as impaired apoptosis, increased proliferation, and decreased egress. Although less is known about the homeostatic function of M2-like resident ATMs, recent evidence suggests roles in AT expansion, thermoregulation, antigen presentation, and iron homeostasis. The field of immunometabolism has come a long way in the past decade, and many exciting new discoveries are bound to be made in the coming years that will expand our understanding of how AT stands at the junction of immune and metabolic co-regulation.
Collapse
Affiliation(s)
- Andrea A Hill
- Department of Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | |
Collapse
|
146
|
Tissue inflammation and nitric oxide-mediated alterations in cardiovascular function are major determinants of endotoxin-induced insulin resistance. Cardiovasc Diabetol 2015; 14:56. [PMID: 25986700 PMCID: PMC4484635 DOI: 10.1186/s12933-015-0223-2] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 05/05/2015] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Endotoxin (i.e. LPS) administration induces a robust inflammatory response with accompanying cardiovascular dysfunction and insulin resistance. Overabundance of nitric oxide (NO) contributes to the vascular dysfunction. However, inflammation itself also induces insulin resistance in skeletal muscle. We sought to investigate whether the cardiovascular dysfunction induced by increased NO availability without inflammatory stress can promote insulin resistance. Additionally, we examined the role of inducible nitric oxide synthase (iNOS or NOS2), the source of the increase in NO availability, in modulating LPS-induced decrease in insulin-stimulated muscle glucose uptake (MGU). METHODS The impact of NO donor infusion on insulin-stimulated whole-body and muscle glucose uptake (hyperinsulinemic-euglycemic clamps), and the cardiovascular system was assessed in chronically catheterized, conscious mice wild-type (WT) mice. The impact of LPS on insulin action and the cardiovascular system were assessed in WT and global iNOS knockout (KO) mice. Tissue blood flow and cardiac function were assessed using microspheres and echocardiography, respectively. Insulin signaling activity, and gene expression of pro-inflammatory markers were also measured. RESULTS NO donor infusion decreased mean arterial blood pressure, whole-body glucose requirements, and MGU in the absence of changes in skeletal muscle blood flow. LPS lowered mean arterial blood pressure and glucose requirements in WT mice, but not in iNOS KO mice. Lastly, despite an intact inflammatory response, iNOS KO mice were protected from LPS-mediated deficits in cardiac output. LPS impaired MGU in vivo, regardless of the presence of iNOS. However, ex vivo, insulin action in muscle obtained from LPS treated iNOS KO animals was protected. CONCLUSION Nitric oxide excess and LPS impairs glycemic control by diminishing MGU. LPS impairs MGU by both the direct effect of inflammation on the myocyte, as well as by the indirect NO-driven cardiovascular dysfunction.
Collapse
|
147
|
Bhattacharya I, Domínguez AP, Drägert K, Humar R, Haas E, Battegay EJ. Hypoxia potentiates tumor necrosis factor-α induced expression of inducible nitric oxide synthase and cyclooxygenase-2 in white and brown adipocytes. Biochem Biophys Res Commun 2015; 461:287-92. [DOI: 10.1016/j.bbrc.2015.04.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Accepted: 04/03/2015] [Indexed: 01/04/2023]
|
148
|
Deficiency of iNOS-derived NO accelerates lipid accumulation-independent liver fibrosis in non-alcoholic steatohepatitis mouse model. BMC Gastroenterol 2015; 15:42. [PMID: 25881230 PMCID: PMC4387704 DOI: 10.1186/s12876-015-0269-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2014] [Accepted: 02/27/2015] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Although many of the factors and molecules closely associated with non-alcoholic steatohepatitis (NASH) have been reported, the role of inducible nitric oxide synthase (iNOS)-derived nitric oxide (NO) on the progression of NASH remains unclear. We therefore investigated the role of iNOS-derived NO in NASH pathogenesis with a long-term follow-up study using systemic iNOS-knockout mice under high-fat diet (HFD) conditions. METHODS iNOS-knockout and wild-type mice were fed a basal or HFD for 10 or 48 weeks. Lipid accumulation, fibrosis, and inflammation were evaluated, and various factors and molecules closely associated with NASH were analyzed. RESULTS Marked fibrosis and inflammation (indicators of NASH) were observed in the livers of iNOS-knockout mice compared to wild-type mice after 48 weeks of a HFD; however, lipid accumulation in iNOS-knockout mice livers was less than in the wild-type. Increased expressions of various cytokines that are transcriptionally controlled by NF-kB in iNOS-deficient mice livers were observed during HFD conditions. CONCLUSIONS iNOS-derived NO may play a protective role against the progression to NASH during an HFD by preventing fibrosis and inflammation, which are mediated by NF-kB activation in Kupffer cells. A lack of iNOS-derived NO accelerates progression to NASH without excessive lipid accumulation.
Collapse
|
149
|
Stanimirovic J, Obradovic M, Zafirovic S, Resanovic I, Bogdanovic N, Gluvic Z, Mousa SA, Isenovic ER. Effects of altered hepatic lipid metabolism on regulation of hepatic iNOS. CLINICAL LIPIDOLOGY 2015; 10:167-175. [DOI: 10.2217/clp.15.8] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Julijana Stanimirovic
- Vinca Institute of Nuclear Sciences, Laboratory of Radiobiology & Molecular Genetics, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| | - Milan Obradovic
- Vinca Institute of Nuclear Sciences, Laboratory of Radiobiology & Molecular Genetics, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| | - Sonja Zafirovic
- Vinca Institute of Nuclear Sciences, Laboratory of Radiobiology & Molecular Genetics, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| | - Ivana Resanovic
- Vinca Institute of Nuclear Sciences, Laboratory of Radiobiology & Molecular Genetics, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| | - Nikola Bogdanovic
- Vinca Institute of Nuclear Sciences, Laboratory of Radiobiology & Molecular Genetics, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| | - Zoran Gluvic
- Department of Endocrinology & ITU, Clinical Hospital Center-Zemun, Vukova Street 9, 11080 Zemun, Serbia
| | - Shaker A Mousa
- The Pharmaceutical Research Institute, Albany College of Pharmacy & Health Sciences, 1 Discovery Drive, Rensselaer, NY 12144, USA
| | - Esma R Isenovic
- Vinca Institute of Nuclear Sciences, Laboratory of Radiobiology & Molecular Genetics, University of Belgrade, P.O. Box 522, 11001 Belgrade, Serbia
| |
Collapse
|
150
|
|