1
|
Sadeghi MA, Hemmati S, Yousefi-Manesh H, Foroutani L, Nassireslami E, Yousefi Zoshk M, Hosseini Y, Abbasian K, Dehpour AR, Chamanara M. Cilostazol pretreatment prevents PTSD-related anxiety behavior through reduction of hippocampal neuroinflammation. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2024; 397:133-144. [PMID: 37382600 DOI: 10.1007/s00210-023-02578-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Accepted: 06/13/2023] [Indexed: 06/30/2023]
Abstract
Current pharmacological treatments against post-traumatic stress disorder (PTSD) lack adequate efficacy. As a result, intense research has focused on identifying other molecular pathways mediating the pathogenesis of this condition. One such pathway is neuroinflammation, which has demonstrated a role in PTSD pathogenesis by causing synaptic dysfunction, neuronal death, and functional impairment in the hippocampus. Phosphodiesterase (PDE) inhibitors (PDEIs) have emerged as promising therapeutic agents against neuroinflammation in other neurological conditions. Furthermore, PDEIs have shown some promise in animal models of PTSD. However, the current model of PTSD pathogenesis, which is based on dysregulated fear learning, implies that PDE inhibition in neurons should enhance the acquisition of fear memory from the traumatic event. As a result, we hypothesized that PDEIs may improve PTSD symptoms through inhibiting neuroinflammation rather than long-term potentiation-related mechanisms. To this end, we tested the therapeutic efficacy of cilostazol, a selective inhibitor of PDE3, on PTSD-related anxiety symptoms in the underwater trauma model of PTSD. PDE3 is expressed much more richly in microglia and astrocytes compared to neurons in the murine brain. Furthermore, we used hippocampal indolamine 2,3-dioxygenase 1 (IDO) expression and interleukin 1 beta (IL-1β) concentration as indicators of neuroinflammation. We observed that cilostazol pretreatment prevented the development of anxiety symptoms and the increase in hippocampal IDO and IL-1β following PTSD induction. As a result, PDE3 inhibition ameliorated the neuroinflammatory processes involved in the development of PTSD symptoms. Therefore, cilostazol and other PDEIs may be promising candidates for further investigation as pharmacological therapies against PTSD.
Collapse
Affiliation(s)
- Mohammad Amin Sadeghi
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Sara Hemmati
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hasan Yousefi-Manesh
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Laleh Foroutani
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Nassireslami
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Mojtaba Yousefi Zoshk
- Trauma Research Center, AJA University of Medical Sciences, Tehran, Iran
- Department of Pediatrics, AJA University of Medical Sciences, Tehran, Iran
| | - Yasaman Hosseini
- Cognitive Neuroscience Center, School of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Kourosh Abbasian
- Management and Health Economics Department, AJA University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Dehpour
- Brain and Spinal Cord Injury Research Center, Neuroscience Institute, Tehran University of Medical Sciences, Tehran, Iran
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Toxicology Research Center, AJA University of Medical Sciences, Tehran, Iran.
- Department of Pharmacology, School of Medicine, AJA University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
2
|
R-spondin 3 Inhibits High Glucose-Induced Endothelial Activation Through Leucine-Rich G Protein-Coupled Receptor 4/Wnt/β-catenin Pathway. J Cardiovasc Pharmacol 2022; 80:70-81. [PMID: 35767713 DOI: 10.1097/fjc.0000000000001295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/22/2022] [Indexed: 11/26/2022]
Abstract
ABSTRACT High glucose-induced endothelial activation plays critical roles in the development of diabetic vascular complications. R-spondin 3 could inhibit inflammatory damage, and diabetic vascular inflammation is secondary to endothelial activation. In this article, we identify R-spondin 3 as a novel regulator of high glucose-induced endothelial activation. We found that the serum levels of R-spondin 3 were significantly reduced in type 2 diabetic patients and db/db mice. We observed that the increased expressions of vascular cell adhesion molecule-1, intercellular cell adhesion molecule-1, and monocyte chemoattractant protein-1 (endothelial activation makers) in high glucose-stimulated human umbilical vein endothelial cell lines (HUVECs) could be inhibited by overexpressing R-spondin 3 or human R-spondin 3 recombinant protein. Subsequently, high glucose-induced adhesion and migration of human myeloid leukemia mononuclear cells (THP-1 cells) to HUVECs were markedly suppressed by the overexpression of R-spondin 3 in HUVECs. Moreover, the inhibitory effect of R-spondin 3 on the expressions of vascular cell adhesion molecule-1, intercellular cell adhesion molecule-1, and monocyte chemoattractant protein-1 in high glucose-treated HUVECs could be blocked by knockdown of leucine-rich G protein-coupled receptor 4 (R-spondin 3 receptor) or the specific inhibitor of Wnt/β-catenin pathway. Taken together, R-spondin 3 could suppress high glucose-induced endothelial activation through leucine-rich G protein-coupled receptor 4/Wnt/β-catenin pathway.
Collapse
|
3
|
Bushra S, Al-Sadeq DW, Bari R, Sahara A, Fadel A, Rizk N. Adiponectin Ameliorates Hyperglycemia-Induced Retinal Endothelial Dysfunction, Highlighting Pathways, Regulators, and Networks. J Inflamm Res 2022; 15:3135-3166. [PMID: 35662872 PMCID: PMC9156523 DOI: 10.2147/jir.s358594] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 05/04/2022] [Indexed: 12/25/2022] Open
Abstract
Background The pathophysiology of diabetic retinopathy (DR) is multifaced. A low level of circulating adiponectin (APN) in type 2 diabetes is associated with microvasculature complications, and its role in the evolution of DR is complex. Aim This study is designed to explore the potential impact of APN in the pathogenesis of DR, linking the changes in cellular and biological processes with the pathways, networks, and regulators involved in its actions. Methods Human microvascular retinal endothelial cells (HMRECs) were exposed to 30mM glucose (HG) and treated with globular adiponectin (30μg/mL) for 24 hours. The cells were evaluated for reactive oxidative stress (ROS) and apoptosis. RT-PCR profile arrays were utilized to evaluate the profile of genes involved in endothelial functions, angiogenesis, extracellular matrix, and adhesion molecules for hyperglycemic HMRECs treated with adiponectin. In addition, the barrier function, leukocyte migration, and angiogenesis were evaluated. The differential expressed genes (DEGs) were outlined, and bioinformatic analysis was applied. Results Adiponectin suppresses ROS production and apoptosis in HMRECs under HG conditions. Adiponectin improved migration and barrier functions in hyperglycemic cells. The bioinformatic analysis highlighted that the signaling pathways of integrin, HMGB1, and p38 AMPK, are mainly involved in the actions of APN on HMRECs. APN significantly affects molecular functions, including the adhesion of cells, chemotaxis, migration of WBCs, and angiogenesis. STAT3, NFKB, IKBKB, and mir-8 are the top upstream regulators, which affect the expressions of the genes of the data set, while TNF and TGFB1 are the top regulators. Conclusion Adiponectin significantly counteracts hyperglycemia at various cellular and molecular levels, reducing its impact on the pathophysiological progression towards DR in vitro using HMRECs. Adiponectin ameliorates inflammatory response, oxidative stress, and endothelial barrier dysfunction using a causal network of NFBk complex, TNF, and HMGB1 and integrin pathways.
Collapse
Affiliation(s)
- Sumbul Bushra
- Department of Biomedical Sciences, College of Health Sciences, QU-Health, Qatar University, Doha, Qatar
| | - Duaa W Al-Sadeq
- Department of Biomedical Sciences, College of Health Sciences, QU-Health, Qatar University, Doha, Qatar
| | - Redwana Bari
- Department of Biomedical Sciences, College of Health Sciences, QU-Health, Qatar University, Doha, Qatar
| | - Afifah Sahara
- Department of Biomedical Sciences, College of Health Sciences, QU-Health, Qatar University, Doha, Qatar
| | - Amina Fadel
- Department of Biomedical Sciences, College of Health Sciences, QU-Health, Qatar University, Doha, Qatar
| | - Nasser Rizk
- Department of Biomedical Sciences, College of Health Sciences, QU-Health, Qatar University, Doha, Qatar
- Biomedical Research Center (BRC), Qatar University, Doha, Qatar
- Correspondence: Nasser Rizk, Department of Biomedical Sciences, College of Health Sciences, QU-Health, Qatar University, P.O. Box 2713, Doha, Qatar, Tel +974-4403-4786, Email
| |
Collapse
|
4
|
Xu R, Zhu C, Li Y, Andrade M, Yin DP. Gastric Bypass Regulates Early Inflammatory Responses in High-Fat Diet-Induced Obese Mice. J Surg Res 2022; 273:161-171. [PMID: 35085943 PMCID: PMC8960359 DOI: 10.1016/j.jss.2021.12.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 12/17/2021] [Accepted: 12/27/2021] [Indexed: 11/15/2022]
Abstract
INTRODUCTION Obesity and diabetes are characterized by chronic inflammatory responses. Roux-en-Y gastric bypass (RYGB) is increasingly regarded as an effective approach for the improvement of glucose homeostasis. In this study, we examined the effects of RYGB on the regulation of early inflammatory responses in the liver and adipose tissue in high-fat diet (HFD)-induced obese (DIO) mice. MATERIALS AND METHODS RYGB was performed in DIO mice followed by analyses of adiposity, insulin sensitivity, plasma and tissue cytokines and adipokines, tissue NF-κB and JNK/c-Jun activation, and tissue macrophage and T-cell subsets. RESULTS We found that RYGB resulted in sustained improvement of adiposity and insulin sensitivity. Plasma insulin and leptin levels were increased in untreated DIO mice and reduced in RYGB mice. RYGB maintained plasma adiponectin levels and inhibited monocyte chemoattractant protein-1 and interleukin 6 in white adipose tissue (WAT) and liver. RYGB inhibited NF-κB activation in WAT and muscle, but not in the liver. However, RYGB attenuated the JNK/c-Jun signaling pathway in the liver and WAT at 1 wk after surgery, suggesting that RYGB regulates the tissue-specific inflammatory pathway. RYGB reduced M1-like (F4/80+/CD11c+) differentiation and enhanced M2-like population (F4/80+/CD206c+). RYGB also regulated CD4+ and CD8+ T-cell infiltration and increased Treg cells in the liver and WAT at the same time point. CONCLUSIONS Our findings demonstrate that RYGB improves obesity and insulin resistance, which are associated with the regulation of early inflammatory reactions in the liver and WAT.
Collapse
Affiliation(s)
- Rui Xu
- Department of Surgery, The First College of Clinical Medical Science, Yichang Central People's Hospital, CTGU, Yichang, Hubei, China
| | - Chenyu Zhu
- Department of Surgery, The First College of Clinical Medical Science, Yichang Central People's Hospital, CTGU, Yichang, Hubei, China
| | - Yuxin Li
- Department of Surgery, The University of Chicago, Chicago, Illinois
| | - Michael Andrade
- Department of Surgery, The University of Chicago, Chicago, Illinois
| | - Deng Ping Yin
- Department of Surgery, The University of Chicago, Chicago, Illinois.
| |
Collapse
|
5
|
Freundt GV, von Samson-Himmelstjerna FA, Nitz JT, Luedde M, Waltenberger J, Wieland T, Frey N, Preusch M, Hippe HJ. The orphan receptor GPRC5B activates pro-inflammatory signaling in the vascular wall via Fyn and NFκB. Biochem Biophys Res Commun 2022; 592:60-66. [PMID: 35033869 DOI: 10.1016/j.bbrc.2022.01.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Accepted: 01/04/2022] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND AIMS Atherosclerosis is driven by an inflammatory process of the vascular wall. The novel orphan G-protein coupled receptor 5B of family C (GPRC5B) is involved in drosophila sugar and lipid metabolism as well as mice adipose tissue inflammation. Here, we investigated the role of GPRC5B in the pro-atherogenic mechanisms of hyperglycemia and vascular inflammation. METHODS Immortalized and primary endothelial cells (ECs) and vascular smooth muscle cells (VSMCs) were used for stimulation with high glucose or different cytokines. Adenoviral- or plasmid-driven GPRC5B overexpression and siRNA-mediated knockdown were performed in these cells to analyze functional and mechanistic pathways of GPRC5B. RESULTS In ECs and VSMCs, stimulation with high glucose, TNFα or LPS induced a significant upregulation of endogenous GPRC5B mRNA and protein levels. GPRC5B overexpression and knockdown increased and attenuated, respectively, the expression of the pro-inflammatory cytokines TNFα, IL-1β, IL-6 as well as the pro-atherogenic vascular adhesion molecules ICAM-1 and VCAM-1. Furthermore, the expression and activity of the metalloproteinase MMP-9, a component of atherosclerotic plaque stabilization, were significantly enhanced by GPRC5B overexpression. Mechanistically, GPRC5B increased the phosphorylation of ERK1/2 and activated NFκB through a direct interaction with the tyrosine kinase Fyn. CONCLUSIONS Our findings demonstrate that GPRC5B is upregulated in response to high glucose and pro-inflammatory signaling. GPRC5B functionally modulates the inflammatory activity in cells of the vascular wall, suggesting a pro-atherogenic GPRC5B-dependent positive feedback loop via Fyn and NFκB. Thus, GPRC5B warrants further attention as a novel pharmacological target for the treatment of vascular inflammation and possibly atherogenesis.
Collapse
Affiliation(s)
- Greta Verena Freundt
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3, D-24105, Kiel, Germany
| | | | - Jan-Thorge Nitz
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3, D-24105, Kiel, Germany
| | - Mark Luedde
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3, D-24105, Kiel, Germany
| | - Johannes Waltenberger
- Department of Cardiovascular Medicine, Medical Faculty, University of Münster, D- 48149, Münster, Germany
| | - Thomas Wieland
- Institute of Experimental and Clinical Pharmacology and Toxicology, Medical Faculty Mannheim, University of Heidelberg, Maybachstr. 14, D-68169, Mannheim, Germany
| | - Norbert Frey
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3, D-24105, Kiel, Germany
| | - Michael Preusch
- Department of Cardiology, Angiology and Pneumology, University of Heidelberg, Im Neuenheimer Feld 410, D-69120, Heidelberg, Germany
| | - Hans-Jörg Hippe
- Department of Cardiology and Angiology, University Hospital Schleswig-Holstein, Campus Kiel, Arnold-Heller-Str. 3, D-24105, Kiel, Germany.
| |
Collapse
|
6
|
Stranahan AM. Visceral adiposity, inflammation, and hippocampal function in obesity. Neuropharmacology 2021; 205:108920. [PMID: 34902347 DOI: 10.1016/j.neuropharm.2021.108920] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 11/09/2021] [Accepted: 12/08/2021] [Indexed: 02/06/2023]
Abstract
The 'apple-shaped' anatomical pattern that accompanies visceral adiposity increases risk for multiple chronic diseases, including conditions that impact the brain, such as diabetes and hypertension. However, distinguishing between the consequences of visceral obesity, as opposed to visceral adiposity-associated metabolic and cardiovascular pathologies, presents certain challenges. This review summarizes current literature on relationships between adipose tissue distribution and cognition in preclinical models and highlights unanswered questions surrounding the potential role of tissue- and cell type-specific insulin resistance in these effects. While gaps in knowledge persist related to insulin insensitivity and cognitive impairment in obesity, several recent studies suggest that cells of the neurovascular unit contribute to hippocampal synaptic dysfunction, and this review interprets those findings in the context of progressive metabolic dysfunction in the CNS. Signalling between cerebrovascular endothelial cells, astrocytes, microglia, and neurons has been linked with memory deficits in visceral obesity, and this article describes the cellular changes in each of these populations with respect to their role in amplification or diminution of peripheral signals. The picture emerging from these studies, while incomplete, implicates pro-inflammatory cytokines, insulin resistance, and hyperglycemia in various stages of obesity-induced hippocampal dysfunction. As in the parable of the five blind wanderers holding different parts of an elephant, considerable work remains in order to assemble a model for the underlying mechanisms linking visceral adiposity with age-related cognitive decline.
Collapse
Affiliation(s)
- Alexis M Stranahan
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia, Augusta University, 1462 Laney Walker Blvd, Augusta, GA, 30912, USA.
| |
Collapse
|
7
|
Krinock MJ, Singhal NS. Diabetes, stroke, and neuroresilience: looking beyond hyperglycemia. Ann N Y Acad Sci 2021; 1495:78-98. [PMID: 33638222 DOI: 10.1111/nyas.14583] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Revised: 02/02/2021] [Accepted: 02/05/2021] [Indexed: 12/14/2022]
Abstract
Ischemic stroke is a leading cause of morbidity and mortality among type 2 diabetic patients. Preclinical and translational studies have identified critical pathophysiological mediators of stroke risk, recurrence, and poor outcome in diabetic patients, including endothelial dysfunction and inflammation. Most clinical trials of diabetes and stroke have focused on treating hyperglycemia alone. Pioglitazone has shown promise in secondary stroke prevention for insulin-resistant patients; however, its use is not yet widespread. Additional research into clinical therapies directed at diabetic pathophysiological processes to prevent stroke and improve outcome for diabetic stroke survivors is necessary. Resilience is the process of active adaptation to a stressor. In patients with diabetes, stroke recovery is impaired by insulin resistance, endothelial dysfunction, and inflammation, which impair key neuroresilience pathways maintaining cerebrovascular integrity, resolving poststroke inflammation, stimulating neural plasticity, and preventing neurodegeneration. Our review summarizes the underpinnings of stroke risk in diabetes, the clinical consequences of stroke in diabetic patients, and proposes hypotheses and new avenues of research for therapeutics to stimulate neuroresilience pathways and improve stroke outcome in diabetic patients.
Collapse
Affiliation(s)
- Matthew J Krinock
- Department of Neurology, University of California - San Francisco, San Francisco, California
| | - Neel S Singhal
- Department of Neurology, University of California - San Francisco, San Francisco, California
| |
Collapse
|
8
|
Adipokines and Inflammation: Focus on Cardiovascular Diseases. Int J Mol Sci 2020; 21:ijms21207711. [PMID: 33081064 PMCID: PMC7589803 DOI: 10.3390/ijms21207711] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/14/2020] [Accepted: 10/16/2020] [Indexed: 02/08/2023] Open
Abstract
It is well established that adipose tissue, apart from its energy storage function, acts as an endocrine organ that produces and secretes a number of bioactive substances, including hormones commonly known as adipokines. Obesity is a major risk factor for the development of cardiovascular diseases, mainly due to a low grade of inflammation and the excessive fat accumulation produced in this state. The adipose tissue dysfunction in obesity leads to an aberrant release of adipokines, some of them with direct cardiovascular and inflammatory regulatory functions. Inflammation is a common link between obesity and cardiovascular diseases, so this review will summarise the role of the main adipokines implicated in the regulation of the inflammatory processes occurring under the scenario of cardiovascular diseases.
Collapse
|
9
|
Oduro PK, Fang J, Niu L, Li Y, Li L, Zhao X, Wang Q. Pharmacological management of vascular endothelial dysfunction in diabetes: TCM and western medicine compared based on biomarkers and biochemical parameters. Pharmacol Res 2020; 158:104893. [PMID: 32434053 DOI: 10.1016/j.phrs.2020.104893] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/18/2020] [Accepted: 05/03/2020] [Indexed: 12/20/2022]
Abstract
Diabetes, a worldwide health concern while burdening significant populace of countries with time due to a hefty increase in both incidence and prevalence rates. Hyperglycemia has been buttressed both in clinical and experimental studies to modulate widespread molecular actions that effect macro and microvascular dysfunctions. Endothelial dysfunction, activation, inflammation, and endothelial barrier leakage are key factors contributing to vascular complications in diabetes, plus the development of diabetes-induced cardiovascular diseases. The recent increase in molecular, transcriptional, and clinical studies has brought a new scope to the understanding of molecular mechanisms and the therapeutic targets for endothelial dysfunction in diabetes. In this review, an attempt made to discuss up to date critical and emerging molecular signaling pathways involved in the pathophysiology of endothelial dysfunction and viable pharmacological management targets. Importantly, we exploit some Traditional Chinese Medicines (TCM)/TCM isolated bioactive compounds modulating effects on endothelial dysfunction in diabetes. Finally, clinical studies data on biomarkers and biochemical parameters involved in the assessment of the efficacy of treatment in vascular endothelial dysfunction in diabetes was compared between clinically used western hypoglycemic drugs and TCM formulas.
Collapse
Affiliation(s)
- Patrick Kwabena Oduro
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China
| | - Jingmei Fang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China
| | - Lu Niu
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China
| | - Yuhong Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Lin Li
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Xin Zhao
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China
| | - Qilong Wang
- Institute of Traditional Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China; Key Laboratory of Pharmacology of Traditional Chinese Medical Formulae, Ministry of Education, Tianjin 301617, PR China; Tianjin Key Laboratory of Chinese medicine Pharmacology, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, PR China.
| |
Collapse
|
10
|
Cyclic nucleotide phosphodiesterases: New targets in the metabolic syndrome? Pharmacol Ther 2020; 208:107475. [PMID: 31926200 DOI: 10.1016/j.pharmthera.2020.107475] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Metabolic diseases have a tremendous impact on human morbidity and mortality. Numerous targets regulating adenosine monophosphate kinase (AMPK) have been identified for treating the metabolic syndrome (MetS), and many compounds are being used or developed to increase AMPK activity. In parallel, the cyclic nucleotide phosphodiesterase families (PDEs) have emerged as new therapeutic targets in cardiovascular diseases, as well as in non-resolved pathologies. Since some PDE subfamilies inactivate cAMP into 5'-AMP, while the beneficial effects in MetS are related to 5'-AMP-dependent activation of AMPK, an analysis of the various controversial relationships between PDEs and AMPK in MetS appears interesting. The present review will describe the various PDE families, AMPK and molecular mechanisms in the MetS and discuss the PDEs/PDE modulators related to the tissues involved, thus supporting the discovery of original molecules and the design of new therapeutic approaches in MetS.
Collapse
|
11
|
Ivovic A, Oprescu AI, Koulajian K, Mori Y, Eversley JA, Zhang L, Nino-Fong R, Lewis GF, Donath MY, Karin M, Wheeler MB, Ehses J, Volchuk A, Chan CB, Giacca A. IKKβ inhibition prevents fat-induced beta cell dysfunction in vitro and in vivo in rodents. Diabetologia 2017; 60:2021-2032. [PMID: 28725915 DOI: 10.1007/s00125-017-4345-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Accepted: 05/16/2017] [Indexed: 02/07/2023]
Abstract
AIMS/HYPOTHESIS We have previously shown that oxidative stress plays a causal role in beta cell dysfunction induced by fat. Here, we address whether the proinflammatory kinase inhibitor of (nuclear factor) κB kinase β (IKKβ), which is activated by oxidative stress, is also implicated. METHODS Fat (oleate or olive oil) was infused intravenously in Wistar rats for 48 h with or without the IKKβ inhibitor salicylate. Thereafter, beta cell function was evaluated in vivo using hyperglycaemic clamps or ex vivo in islets isolated from fat-treated rats. We also exposed rat islets to oleate in culture, with or without salicylate and 4(2'-aminoethyl)amino-1,8-dimethylimidazo(1,2-a)quinoxaline; BMS-345541 (BMS, another inhibitor of IKKβ) and evaluated beta cell function in vitro. Furthermore, oleate was infused in mice treated with BMS and in beta cell-specific Ikkb-null mice. RESULTS 48 h infusion of fat impaired beta-cell function in vivo, assessed using the disposition index (DI), in rats (saline: 1.41 ± 0.13; oleate: 0.95 ± 0.11; olive oil [OLO]: 0.87 ± 0.15; p < 0.01 for both fats vs saline) and in mice (saline: 2.51 ± 0.39; oleate: 1.20 ± 0.19; p < 0.01 vs saline) and ex vivo (i.e., insulin secretion, units are pmol insulin islet-1 h-1) in rat islets (saline: 1.51 ± 0.13; oleate: 1.03 ± 0.10; OLO: 0.91 ± 0.13; p < 0.001 for both fats vs saline) and the dysfunction was prevented by co-infusion of salicylate in rats (oleate + salicylate: 1.30 ± 0.09; OLO + salicylate: 1.33 ± 0.23) or BMS in mice (oleate + BMS: 2.25 ± 0.42) in vivo and by salicylate in rat islets ex vivo (oleate + salicylate: 1.74 ± 0.31; OLO + salicylate: 1.54 ± 0.29). In cultured islets, 48 h exposure to oleate impaired beta-cell function ([in pmol insulin islet-1 h-1] control: 0.66 ± 0.12; oleate: 0.23 ± 0.03; p < 0.01 vs saline), an effect prevented by both inhibitors (oleate + salicylate: 0.98 ± 0.08; oleate + BMS: 0.50 ± 0.02). Genetic inhibition of IKKβ also prevented fat-induced beta-cell dysfunction ex vivo ([in pmol insulin islet-1 h-1] control saline: 0.16 ± 0.02; control oleate: 0.10 ± 0.02; knockout oleate: 0.17 ± 0.04; p < 0.05 control saline vs. control oleate) and in vivo (DI: control saline: 3.86 ± 0.40; control oleate: 1.95 ± 0.29; knockout oleate: 2.96 ± 0.24; p < 0.01 control saline vs control oleate). CONCLUSIONS/INTERPRETATION Our results demonstrate a causal role for IKKβ in fat-induced beta cell dysfunction in vitro, ex vivo and in vivo.
Collapse
Affiliation(s)
- Aleksandar Ivovic
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada, M5S 1A8
| | - Andrei I Oprescu
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Khajag Koulajian
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada, M5S 1A8
| | - Yusaku Mori
- Division of Diabetes, Metabolism, and Endocrinology, Showa University School of Medicine, Shinagawa, Tokyo, Japan
| | - Judith A Eversley
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada, M5S 1A8
| | - Liling Zhang
- Division of Cellular and Molecular Biology, Toronto General Research Institute, University Health Network, Toronto, ON, Canada
| | - Rodolfo Nino-Fong
- Department of Biomedical Sciences, Ross University School of Veterinary Medicine, Basseterre, St Kitts and Nevis
| | - Gary F Lewis
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada, M5S 1A8
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada
| | - Marc Y Donath
- Department of Endocrinology, Diabetes, and Metabolism, University Hospital Basel, Basel, Switzerland
| | - Michael Karin
- Department of Pharmacology, University of California, San Diego, School of Medicine, La Jolla, CA, USA
| | - Michael B Wheeler
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada, M5S 1A8
| | - Jan Ehses
- Department of Surgery, Faculty of Medicine, University of British Columbia, Vancouver, BC, Canada
- Child and Family Research Institute, Vancouver, BC, Canada
| | - Allen Volchuk
- Keenan Research Centre for Biomedical Science, St Michael's Hospital, Toronto, ON, Canada
| | - Catherine B Chan
- Department of Agriculture, Food and Nutritional Sciences, Faculty of Agricultural, Life and Environmental Sciences, University of Alberta, Edmonton, AB, Canada
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Adria Giacca
- Department of Physiology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada, M5S 1A8.
- Institute of Medical Science, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Department of Medicine, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
- Banting and Best Diabetes Centre, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
12
|
Jeon J, Kim YJ, Shin H, Ha UH. T3SS effector ExoY reduces inflammasome-related responses by suppressing bacterial motility and delaying activation of NF-κB and caspase-1. FEBS J 2017; 284:3392-3403. [PMID: 28815941 DOI: 10.1111/febs.14199] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Revised: 07/05/2017] [Accepted: 08/11/2017] [Indexed: 11/28/2022]
Abstract
Type III-secreted effectors are essential for modulating host immune responses during the pathogenesis of Pseudomonas aeruginosa infections. Little is known about the impact of one of the effectors, ExoY, on inflammasome activation, which results in IL-1β production and pyroptotic cell death. In this study, we found that transcriptional expression of Il-1β was induced to a lesser extent in response to an exoY-harboring strain than to a deleted mutant. This suppressive effect of ExoY was verified by complementation assay as well as by direct translocation of exoY into host cells. In addition to the production of IL-1β, pyroptotic cell death was also diminished in response to an exoY-harboring strain. These inflammasome responses were mediated by the adenylate cyclase activity of ExoY, which plays a role in delaying the activation of NF-κB and caspase-1, a key component of inflammasome-mediated responses. Moreover, the negative effects of ExoY on these responses were in part conferred by the suppression of bacterial motility, which could reduce the degree of bacterial contact with cells. Together, these results demonstrate that the adenylate cyclase activity of P. aeruginosa ExoY can reduce inflammasome-related responses by influencing both the host and the bacterium itself by delaying the activation of inflammatory pathways and suppressing bacterial motility.
Collapse
Affiliation(s)
- Jisu Jeon
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| | - Yong-Jae Kim
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| | - Heesung Shin
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| | - Un-Hwan Ha
- Department of Biotechnology and Bioinformatics, Korea University, Sejong, Korea
| |
Collapse
|
13
|
Tong W, Wang Q, Sun D, Suo J. Curcumin suppresses colon cancer cell invasion via AMPK-induced inhibition of NF-κB, uPA activator and MMP9. Oncol Lett 2016; 12:4139-4146. [PMID: 27895783 DOI: 10.3892/ol.2016.5148] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 07/01/2016] [Indexed: 01/31/2023] Open
Abstract
Curcumin, an active nontoxic ingredient of turmeric, possesses potent anti-inflammatory, antioxidant and anti-cancer properties; however, the molecular mechanisms of curcumin are not fully understood. The transcription factor nuclear factor-κB (NF-κB) is key in cellular processes, and the expression/activation of urokinase-type plasminogen activator (uPA) and matrix metalloproteinase-9 (MMP9) are crucial for cell invasion. The present study investigated the hypothesis that curcumin inhibits colon cancer cell invasion by modulating NF-κB-mediated expression and activation of uPA and MMP9. Human colon cancer SW480 and LoVo cells were treated with various concentrations of curcumin. Curcumin was demonstrated to dose-dependently inhibit the adhesion and proliferation ability of LoVo and SW480 cells using Transwell and MTT assays, respectively. In addition, curcumin activated 5' AMP-activated protein kinase (AMPK) and suppressed p65 NF-κB phosphorylation, as shown by western blot analysis. Compound C, a potent AMPK inhibitor, abolished curcumin-induced inhibition of NF-κB, uPA and MMP9, suggesting that AMPK activation is responsible for curcumin-mediated NF-κB, uPA and MMP9 inhibition. The binding activity of NF-κB to DNA was examined and western blotting and quantitative polymerase reaction was performed to detect the effect of curcumin on the expression of uPA and MMP9. The present results revealed that curcumin significantly decreased the expression of uPA and MMP9 and NF-κB DNA binding activity. Furthermore, curcumin decreased the level of the p65 subunit of NF-κB binding to the promoter of the gene encoding uPA and MMP9, which suppressed transcriptional activation of uPA and MMP9. Overall, the present data suggest that curcumin inhibits colon cancer cell invasion via AMPK activation and subsequent inhibition of p65 NF-κB, uPA and MMP9. The therapeutic potential of curcumin for colon cancer metastasis required additional study.
Collapse
Affiliation(s)
- Weihua Tong
- Department of Gastrointestinal-Colorectal Surgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Quan Wang
- Department of Gastrointestinal-Colorectal Surgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Donghui Sun
- Department of Gastrointestinal-Colorectal Surgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| | - Jian Suo
- Department of Gastrointestinal-Colorectal Surgery, First Hospital of Jilin University, Changchun, Jilin 130021, P.R. China
| |
Collapse
|
14
|
Kumase F, Takeuchi K, Morizane Y, Suzuki J, Matsumoto H, Kataoka K, Al-Moujahed A, Maidana DE, Miller JW, Vavvas DG. AMPK-Activated Protein Kinase Suppresses Ccr2 Expression by Inhibiting the NF-κB Pathway in RAW264.7 Macrophages. PLoS One 2016; 11:e0147279. [PMID: 26799633 PMCID: PMC4723067 DOI: 10.1371/journal.pone.0147279] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2015] [Accepted: 01/02/2016] [Indexed: 01/08/2023] Open
Abstract
C-C chemokine receptor 2 (Ccr2) is a key pro-inflammatory marker of classic (M1) macrophage activation. Although Ccr2 is known to be expressed both constitutively and inductively, the full regulatory mechanism of its expression remains unclear. AMP-activated protein kinase (AMPK) is not only a master regulator of energy homeostasis but also a central regulator of inflammation. In this study, we sought to assess AMPK's role in regulating RAW264.7 macrophage Ccr2 protein levels in resting (M0) or LPS-induced M1 states. In both M0 and M1 RAW264.7 macrophages, knockdown of the AMPKα1 subunit by siRNA led to increased Ccr2 levels whereas pharmacologic (A769662) activation of AMPK, attenuated LPS-induced increases in Ccr2 expression in an AMPK dependent fashion. The increases in Ccr2 levels by AMPK downregulation were partially reversed by NF-κB inhibition whereas TNF-a inhibition had minimal effects. Our results indicate that AMPK is a negative regulator of Ccr2 expression in RAW264.7 macrophages, and that the mechanism of action of AMPK inhibition of Ccr2 is mediated, in part, through the NF-κB pathway.
Collapse
Affiliation(s)
- Fumiaki Kumase
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Kimio Takeuchi
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Yuki Morizane
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Ophthalmology, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Jun Suzuki
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Ophthalmology, Tokyo Medical University, Tokyo, Japan
| | - Hidetaka Matsumoto
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Keiko Kataoka
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Ahmad Al-Moujahed
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Daniel E. Maidana
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Joan W. Miller
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Demetrios G. Vavvas
- Retina Service, Angiogenesis Laboratory, Massachusetts Eye and Ear Infirmary, Department of Ophthalmology, Harvard Medical School, Boston, Massachusetts, United States of America
| |
Collapse
|
15
|
Kanno Y, Ishisaki A, Kawashita E, Kuretake H, Ikeda K, Matsuo O. uPA Attenuated LPS-induced Inflammatory Osteoclastogenesis through the Plasmin/PAR-1/Ca(2+)/CaMKK/AMPK Axis. Int J Biol Sci 2016; 12:63-71. [PMID: 26722218 PMCID: PMC4679399 DOI: 10.7150/ijbs.12690] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 08/25/2015] [Indexed: 02/03/2023] Open
Abstract
Chronic inflammatory diseases, such as rheumatoid arthritis and periodontitis-caused bone destruction, results from an increase of bone-resorbing osteoclasts (OCs) induced by inflammation. However, the detailed mechanisms underlying this disorder remain unclear. We herein investigated that the effect of urokinase-type plasminogen activator (uPA) on inflammatory osteoclastogenesis induced by lipopolysaccharide (LPS), which is a potent stimulator of bone resorption in inflammatory diseases. We found that the uPA deficiency promoted inflammatory osteoclastogenesis and bone loss induced by LPS. We also showed that LPS induced the expression of uPA, and the uPA treatment attenuated the LPS-induced inflammatory osteoclastogenesis of RAW264.7 mouse monocyte/macrophage lineage cells. Additionally, we showed that the uPA-attenuated inflammatory osteoclastgenesis is associated with the activation of plasmin/protease-activated receptor (PAR)-1 axis by uPA. Moreover, we examined the mechanism underlying the effect of uPA on inflammatory osteoclastogenesis, and found that uPA/plasmin/PAR-1 activated the adenosine monophosphate-activated protein kinase (AMPK) pathway through Ca2+/calmodulin dependent protein kinase kinase (CaMKK) activation, and attenuated inflammatory osteoclastogenesis by inactivation of NF-κB in RAW264.7 cells. These data suggest that uPA attenuated inflammatory osteoclastogenesis through the plasmin/PAR-1/Ca2+/CaMKK/AMPK axis. Our findings may provide a novel therapeutic approach to bone loss caused by inflammatory diseases.
Collapse
Affiliation(s)
- Yosuke Kanno
- 1. Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's College of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto 610-0395, Japan
| | - Akira Ishisaki
- 2. Division of Cellular Biosignal Sciences, Department of Biochemistry, Iwate Medical University, 2-1-1 Nishitokuta, Yahaba-cho, Shiwa-gun, Iwate 028-3694, Japan
| | - Eri Kawashita
- 1. Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's College of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto 610-0395, Japan
| | - Hiromi Kuretake
- 1. Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's College of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto 610-0395, Japan
| | - Kanako Ikeda
- 1. Department of Clinical Pathological Biochemistry, Faculty of Pharmaceutical Science, Doshisha Women's College of Liberal Arts, 97-1 Kodo Kyo-tanabe, Kyoto 610-0395, Japan
| | - Osamu Matsuo
- 3. Kinki University Faculty of Medicine 377-2 Ohnohigashi, Osaka-sayama 589-8511, Japan
| |
Collapse
|
16
|
Ebrahimi-Mamaeghani M, Mohammadi S, Arefhosseini SR, Fallah P, Bazi Z. Adiponectin as a potential biomarker of vascular disease. Vasc Health Risk Manag 2015; 11:55-70. [PMID: 25653535 PMCID: PMC4303398 DOI: 10.2147/vhrm.s48753] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The increasing prevalence of diabetes and its complications heralds an alarming situation worldwide. Obesity-associated changes in circulating adiponectin concentrations have the capacity to predict insulin sensitivity and are a link between obesity and a number of vascular diseases. One obvious consequence of obesity is a decrease in circulating levels of adiponectin, which are associated with cardiovascular disorders and associated vascular comorbidities. Human and animal studies have demonstrated decreased adiponectin to be an independent risk factor for cardiovascular disease. However, in animal studies, increased circulating adiponectin alleviates obesity-induced endothelial dysfunction and hypertension, and also prevents atherosclerosis, myocardial infarction, and diabetic cardiac tissue disorders. Further, metabolism of a number of foods and medications are affected by induction of adiponectin. Adiponectin has beneficial effects on cardiovascular cells via its antidiabetic, anti-inflammatory, antioxidant, antiapoptotic, antiatherogenic, vasodilatory, and antithrombotic activity, and consequently has a favorable effect on cardiac and vascular health. Understanding the molecular mechanisms underlying the regulation of adiponectin secretion and signaling is critical for designing new therapeutic strategies. This review summarizes the recent evidence for the physiological role and clinical significance of adiponectin in vascular health, identification of the receptor and post-receptor signaling events related to the protective effects of the adiponectin system on vascular compartments, and its potential use as a target for therapeutic intervention in vascular disease.
Collapse
MESH Headings
- Adiponectin/immunology
- Adiponectin/metabolism
- Adipose Tissue/immunology
- Adipose Tissue/metabolism
- Adipose Tissue/physiopathology
- Animals
- Biomarkers/metabolism
- Endothelium, Vascular/immunology
- Endothelium, Vascular/metabolism
- Endothelium, Vascular/physiopathology
- Humans
- Muscle, Smooth, Vascular/immunology
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/physiopathology
- Prognosis
- Protective Factors
- Receptors, Adiponectin/metabolism
- Risk Factors
- Signal Transduction
- Vascular Diseases/immunology
- Vascular Diseases/metabolism
- Vascular Diseases/physiopathology
- Vascular Diseases/prevention & control
Collapse
Affiliation(s)
| | - Somayeh Mohammadi
- Department of Nutrition, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyed Rafie Arefhosseini
- Department of Food Technology, Faculty of Nutrition Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parviz Fallah
- Department of Molecular Biology and Genetic Engineering, Stem Cell Technology Research Center, Tehran, Iran
| | - Zahra Bazi
- Department of Biotechnology, Faculty of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
17
|
Peixoto CA, Nunes AKS, Garcia-Osta A. Phosphodiesterase-5 Inhibitors: Action on the Signaling Pathways of Neuroinflammation, Neurodegeneration, and Cognition. Mediators Inflamm 2015; 2015:940207. [PMID: 26770022 PMCID: PMC4681825 DOI: 10.1155/2015/940207] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Accepted: 11/08/2015] [Indexed: 12/16/2022] Open
Abstract
Phosphodiesterase type 5 inhibitors (PDE5-Is) have recently emerged as a potential therapeutic strategy for neuroinflammatory, neurodegenerative, and memory loss diseases. Mechanistically, PDE5-Is produce an anti-inflammatory and neuroprotection effect by increasing expression of nitric oxide synthases and accumulation of cGMP and activating protein kinase G (PKG), the signaling pathway of which is thought to play an important role in the development of several neurodiseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). The aim of this paper was to review present knowledge of the signaling pathways that underlie the use of PDE5-Is in neuroinflammation, neurogenesis, learning, and memory.
Collapse
Affiliation(s)
- Christina Alves Peixoto
- 1Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), 50.740-465 Recife, PE, Brazil
- *Christina Alves Peixoto:
| | - Ana Karolina Santana Nunes
- 1Laboratório de Ultraestrutura, Centro de Pesquisa Aggeu Magalhães (FIOCRUZ), 50.740-465 Recife, PE, Brazil
- 2Universidade Federal de Pernambuco, 50.670-901 Recife, PE, Brazil
| | - Ana Garcia-Osta
- 3Neurobiology of Alzheimer's disease, Neurosciences Division, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona, Spain
| |
Collapse
|
18
|
Abstract
The increased prevalence of obesity has mandated extensive research focused on mechanisms responsible for associated clinical complications. Emerging from the focus on adipose tissue biology as a vitally important adipokine is adiponectin which is now believed to mediate anti-diabetic, anti-atherosclerotic, anti-inflammatory, cardioprotective and cancer modifying actions. Adiponectin mediates these primarily beneficial effects via direct signaling effects and via enhancing insulin sensitivity via crosstalk with insulin signaling pathways. Reduced adiponectin action is detrimental and occurs in obesity via decreased circulating levels of adiponectin action or development of adiponectin resistance. This review will focus on cellular mechanisms of adiponectin action, their crosstalk with insulin signaling and the resultant role of adiponectin in cardiovascular disease, diabetes and cancer and reviews data from in vitro cell based studies through animal models to clinical observations.
Collapse
Affiliation(s)
- Michael P Scheid
- Department of Biology, York University, Toronto, ON, M3J 1P3, Canada
| | | |
Collapse
|
19
|
Caselli C, D'Amico A, Cabiati M, Prescimone T, Del Ry S, Giannessi D. Back to the heart: the protective role of adiponectin. Pharmacol Res 2014; 82:9-20. [PMID: 24657240 DOI: 10.1016/j.phrs.2014.03.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Revised: 02/25/2014] [Accepted: 03/06/2014] [Indexed: 12/15/2022]
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide and the prevalence of obesity and diabetes are increasing. In obesity, adipose tissue increases the secretion of bioactive mediators (adipokines) that may represent a key mechanism linking obesity to CVD. Adiponectin, extensively studied in metabolic diseases, exerts anti-diabetic, anti-atherogenic and anti-inflammatory activities. Due to these positive actions, the role of adiponectin in cardiovascular protection has been evaluated in recent years. In particular, for its potential therapeutic benefits in humans, adiponectin has become the subject of intense preclinical research. In the cardiovascular context, understanding of the cellular and molecular mechanisms underlying the adiponectin system, throughout its secretion, regulation and signaling, is critical for designing new drugs that target adiponectin system molecules. This review focused on recent advances regarding molecular mechanisms related to protective effects of the adiponectin system on both cardiac and vascular compartments and its potential use as a target for therapeutic intervention of CVD.
Collapse
Affiliation(s)
- C Caselli
- Consiglio Nazionale delle Ricerche (CNR), Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa 56100, Italy.
| | - A D'Amico
- Scuola Superiore S. Anna, Pisa, Italy
| | - M Cabiati
- Consiglio Nazionale delle Ricerche (CNR), Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa 56100, Italy
| | - T Prescimone
- Consiglio Nazionale delle Ricerche (CNR), Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa 56100, Italy
| | - S Del Ry
- Consiglio Nazionale delle Ricerche (CNR), Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa 56100, Italy
| | - D Giannessi
- Consiglio Nazionale delle Ricerche (CNR), Institute of Clinical Physiology, Laboratory of Cardiovascular Biochemistry, Pisa 56100, Italy
| |
Collapse
|
20
|
Harmel E, Grenier E, Bendjoudi Ouadda A, El Chebly M, Ziv E, Beaulieu JF, Sané A, Spahis S, Laville M, Levy E. AMPK in the small intestine in normal and pathophysiological conditions. Endocrinology 2014; 155:873-88. [PMID: 24424053 DOI: 10.1210/en.2013-1750] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The role of AMPK in regulating energy storage and depletion remains unexplored in the intestine. This study will to define its status, composition, regulation and lipid function, as well as to examine the impact of insulin resistance and type 2 diabetes on intestinal AMPK activation, insulin sensitivity, and lipid metabolism. Caco-2/15 cells and Psammomys obesus (P. obesus) animal models were experimented. We showed the predominance of AMPKα1 and the prevalence of α1/β2/γ1 heterotrimer in Caco-2/15 cells. The activation of AMPK by 5-aminoimidazole-4-carboxamide ribonucleoside and metformin resulted in increased phospho(p)-ACC. However, the down-regulation of p-AMPK by compound C and high glucose lowered p-ACC without affecting 3-hydroxy-3-methylglutaryl-coenzyme A reductase. Administration of metformin to P. obesus with insulin resistance and type 2 diabetes led to 1) an up-regulation of intestinal AMPK signaling pathway typified by ascending p-AMPKα(-Thr172); 2) a reduction in ACC activity; 3) an elevation of carnitine palmitoyltransferase 1; 4) a trend of increase in insulin sensitivity portrayed by augmentation of p-Akt and phospho-glycogen synthetase kinase 3β; 5) a reduced phosphorylation of p38-MAPK and ERK1/2; and 6) a decrease in diabetic dyslipidemia following lowering of intracellular events that govern lipoprotein assembly. These data suggest that AMPK fulfills key functions in metabolic processes in the small intestine.
Collapse
Affiliation(s)
- Elodie Harmel
- Research Center (E.H., E.G., A.B.O., M.E.C., A.S., S.S., E.L.), Sainte-Justine MUHC, Montreal, Quebec, Canada, H3T 1C5; Department of Nutrition (E.H., E.G., S.S., E.L.) and Department of Biochemistry (M.E.C.), Université de Montréal, Montreal, Quebec, Canada, H3T 1C5; Diabetes Unit (E.Z.), Division of Internal Medicine, Hadassah Ein Kerem Hospital, 120 Jerusalem, Israel-91; Canadian Institutes for Health Research Team on the Digestive Epithelium (J.F.B., E.L.), Department of Anatomy and Cellular Biology, Faculty of Medicine and Health Sciences, Université de Sherbrooke, Sherbrooke, Quebec, Canada, J1H 5N4; and CRNH Rhône-Alpes (E.H., M.L.), Université Lyon 1, Institut National de la Santé et de la Recherche Médicale Unité Mixte de Recherche 1060, CENS, Centre Hospitalier Lyon-Sud, F-69310 Pierre Bénite, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
The effect of resveratrol on the expression of AdipoR1 in kidneys of diabetic nephropathy. Mol Biol Rep 2014; 41:2151-9. [PMID: 24413998 DOI: 10.1007/s11033-014-3064-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 01/04/2014] [Indexed: 12/17/2022]
Abstract
Adiponectin is an adipocyte derived protein that plays pivotal roles in anti-oxidation, anti-inflammatory and insulin-sensitizing properties by activating two receptors, AdipoR1 and AdipoR2. Recent studies have shown that the down-regulation of AdipoR1 is a known cause of diabetic nephropathy (DN). Resveratrol (Resv), a natural polyphenol, has been identified as a potent activator of forkhead transcription factor O1 (FoxO1) which can up-regulate the expression of AdipoR1. In the present study, we have investigated whether Resv can up-regulate the expression of AdipoR1 by activating FoxO1 that is in kidney of DN rats and mesangial cells (MCs) cultured in high glucose (HG, 30 mmol/L) medium. In vivo, we show that, in the renal cortex of diabetic rats, the expression of AdipoR1 was significantly reduced and correlated with an increase in the generation of malondialdehyde (MDA), Collagen IV and fibronectin proteins. However, administration with Resv significantly increased the expression of AdipoR1. This correlated with not only a decrease in generation of MDA, Collagen IV and fibronectin proteins levels but also more improved kidney pathological and biochemical indicators changes. In vitro, we show that HG-induced depression of FoxO1 activity was associated with the expression of Adipor1 in MCs. Treatment with Resv (20 μmol/L) caused an elevation in the activity of FoxO1 and a significantly increase in the expression of AdipoR1. Furthermore, inhibition of FoxO1 through short hairpin RNA markedly reduced the expression of Adipor1 in MCs cultured by Resv. In conclusion, Resv can significantly increase the expression of AdipoR1 by activating FoxO1 in diabetic kidney. These data also suggest that Resv may serve as a promising agent for preventing or treating DN.
Collapse
|
22
|
Zhang Y, Wang XL, Zhao J, Wang YJ, Lau WB, Yuan YX, Gao EH, Koch WJ, Ma XL. Adiponectin inhibits oxidative/nitrative stress during myocardial ischemia and reperfusion via PKA signaling. Am J Physiol Endocrinol Metab 2013; 305:E1436-43. [PMID: 24129398 PMCID: PMC3882378 DOI: 10.1152/ajpendo.00445.2013] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The cardioprotective effects of adiponectin (APN) against myocardial ischemia/reperfusion (MI/R) injury are well known. However, comprehension of the mechanisms mediating intracellular APN signaling remains incomplete. We recently demonstrate the antioxidant/antinitrative effects of APN are not dependent on AMPK. Protein kinase A (PKA) has been previously shown to be activated by APN, with uncertain relevance to APN cardiac protection. The current study determined whether the antioxidative/antinitrative effect of APN is mediated by PKA. Administration of APN (2 μg/g) 10 min before reperfusion significantly enhanced cardiac PKA activity, reduced oxidative stress, and decreased infarct size. Knockdown of cardiac PKA expression (PKA-KD) by intramyocardial injection of PKA-siRNAs (>70% suppression) significantly inhibited APN cardioprotection determined by cardiac apoptosis, infarct size, and cardiac function. Moreover, PKA-KD virtually abolished the suppressive effect of APN on MI/R-induced NADPH oxidase overexpression and superoxide overproduction and partially inhibited the effect of APN on nitrative protein modification in MI/R heart. Mechanistically, APN significantly inhibited MI/R-induced IKK/IκB phosphorylation and NF-κB activation, which were blocked in PKA-KD heart. Finally, the PKA-mediated antioxidant/antinitrative and cardioprotective effects of APN are intact in AMPK-deficient mice, suggesting that there is no cross talk between AMPK and PKA signaling in APN cardioprotection. Collectively, we demonstrate for the first time that APN inhibits oxidative/nitrative stress during MI/R via PKA-dependent NF-κB inhibition.
Collapse
Affiliation(s)
- Yanqing Zhang
- Department of Anesthesiology, Shanxi Medical University, Taiyuan, China
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Wedellova Z, Kovacova Z, Tencerova M, Vedral T, Rossmeislova L, Siklova-Vitkova M, Stich V, Polak J. The Impact of Full-Length, Trimeric and Globular Adiponectin on Lipolysis in Subcutaneous and Visceral Adipocytes of Obese and Non-Obese Women. PLoS One 2013; 8:e66783. [PMID: 23805277 PMCID: PMC3689658 DOI: 10.1371/journal.pone.0066783] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Accepted: 05/13/2013] [Indexed: 01/14/2023] Open
Abstract
Contribution of individual adiponectin isoforms to lipolysis regulation remains unknown. We investigated the impact of full-length, trimeric and globular adiponectin isoforms on spontaneous lipolysis in subcutaneous abdominal (SCAAT) and visceral adipose tissues (VAT) of obese and non-obese subjects. Furthermore, we explored the role of AMPK (5'-AMP-activated protein kinase) in adiponectin-dependent lipolysis regulation and expression of adiponectin receptors type 1 and 2 (AdipoR1 and AdipoR2) in SCAAT and VAT. Primary adipocytes isolated from SCAAT and VAT of obese and non-obese women were incubated with 20 µg/ml of: A) full-length adiponectin (physiological mixture of all adiponectin isoforms), B) trimeric adiponectin isoform or C) globular adiponectin isoform. Glycerol released into media was used as a marker of lipolysis. While full-length adiponectin inhibited lipolysis by 22% in non-obese SCAAT, globular isoform inhibited lipolysis by 27% in obese SCAAT. No effect of either isoform was detected in non-obese VAT, however trimeric isoform inhibited lipolysis by 21% in obese VAT (all p<0.05). Trimeric isoform induced Thr172 p-AMPK in differentiated preadipocytes from a non-obese donor, while globular isoform induced Ser79 p-ACC by 32% (p<0.05) and Ser565 p-HSL by 52% (p = 0.08) in differentiated preadipocytes from an obese donor. AdipoR2 expression was 17% and 37% higher than AdipoR1 in SCAAT of obese and non-obese groups and by 23% higher in VAT of obese subjects (all p<0.05). In conclusion, the anti-lipolytic effect of adiponectin isoforms is modified with obesity: while full-length adiponectin exerts anti-lipolytic action in non-obese SCAAT, globular and trimeric isoforms show anti-lipolytic activity in obese SCAAT and VAT, respectively.
Collapse
Affiliation(s)
- Zuzana Wedellova
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Czech Republic
- 2 Internal Medicine Department, University Hospital of Kralovske Vinohrady, Prague, Czech Republic
| | - Zuzana Kovacova
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Michaela Tencerova
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Tomas Vedral
- General Surgery Department, University Hospital of Kralovske Vinohrady, Prague, Czech Republic
| | - Lenka Rossmeislova
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine and INSERM Unite 586, Charles University, Prague, Czech Republic
| | - Michaela Siklova-Vitkova
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Czech Republic
| | - Vladimir Stich
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Czech Republic
- Franco-Czech Laboratory for Clinical Research on Obesity, Third Faculty of Medicine and INSERM Unite 586, Charles University, Prague, Czech Republic
| | - Jan Polak
- Department of Sport Medicine, Third Faculty of Medicine, Charles University in Prague, Czech Republic
- 2 Internal Medicine Department, University Hospital of Kralovske Vinohrady, Prague, Czech Republic
- Division of Pulmonary and Critical Care Medicine, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
24
|
Nobe K, Fujii A, Saito K, Negoro T, Ogawa Y, Nakano Y, Hashimoto T, Honda K. Adiponectin enhances calcium dependency of mouse bladder contraction mediated by protein kinase Cα expression. J Pharmacol Exp Ther 2013; 345:62-8. [PMID: 23365260 DOI: 10.1124/jpet.112.202028] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
Adiponectin is an adipose tissue-secreted protein and is a multifunctional adipocytokine. However, the association of adiponectin with bladder contraction has not been investigated. In this study, the adiponectin-sense transgenic mouse (Adip-Sen mouse; age, 16-24 weeks; male) and age-matched controls (C57Bl mouse) were studied. The Adip-Sen mouse showed a significant increase in plasma adiponectin levels (56.2%; P < 0.01), compared with those in the C57Bl mouse, without affecting other lipid parameters. Isometric force development in bladder smooth muscle tissues were detected using an organ-bath system. Although carbachol (CCh)-induced (0.1-100 µM) time- and dose-dependent contractions in Adip-Sen mouse bladder were slightly enhanced, compared with those in the C57Bl mouse during a low range (0.3-1.0 µM) of CCh, differences could not be detected with other CCh concentrations. However, the reduction in contraction under Ca(2+)-replaced conditions was significantly different between Adip-Sen and C57Bl mice (94.1 and 66.3% of normal contraction, respectively; n = 5). A parameter of Ca(2+) sensitivity, the relation between intracellular Ca(2+) concentration and contraction, was increased in the Adip-Sen mouse, compared with that in the C57B1 mouse. This Ca(2+) dependency in the Adip-Sen mouse was reduced by a protein kinase C (PKC) inhibitor, but not by a Rho kinase inhibitor. Expression of the calcium-dependent isoform of PKC, PKCα, was increased in the Adip-Sen mouse bladder, and CCh-induced phosphorylation of PKCα was also enhanced, compared with those in the C57Bl mouse. In conclusion, adiponectin is associated with bladder smooth muscle contraction, which involves an increase in Ca(2+) dependency of contraction mediated by PKCα expression.
Collapse
Affiliation(s)
- Koji Nobe
- Department of Pharmacology, School of Pharmacy, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Northcott JM, Yeganeh A, Taylor CG, Zahradka P, Wigle JT. Adipokines and the cardiovascular system: mechanisms mediating health and disease. Can J Physiol Pharmacol 2012; 90:1029-59. [DOI: 10.1139/y2012-053] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
This review focuses on the role of adipokines in the maintenance of a healthy cardiovascular system, and the mechanisms by which these factors mediate the development of cardiovascular disease in obesity. Adipocytes are the major cell type comprising the adipose tissue. These cells secrete numerous factors, termed adipokines, into the blood, including adiponectin, leptin, resistin, chemerin, omentin, vaspin, and visfatin. Adipose tissue is a highly vascularised endocrine organ, and different adipose depots have distinct adipokine secretion profiles, which are altered with obesity. The ability of many adipokines to stimulate angiogenesis is crucial for adipose tissue expansion; however, excessive blood vessel growth is deleterious. As well, some adipokines induce inflammation, which promotes cardiovascular disease progression. We discuss how these 7 aforementioned adipokines act upon the various cardiovascular cell types (endothelial progenitor cells, endothelial cells, vascular smooth muscle cells, pericytes, cardiomyocytes, and cardiac fibroblasts), the direct effects of these actions, and their overall impact on the cardiovascular system. These were chosen, as these adipokines are secreted predominantly from adipocytes and have known effects on cardiovascular cells.
Collapse
Affiliation(s)
- Josette M. Northcott
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E OJ9, Canada
- Institute of Cardiovascular Sciences, and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
| | - Azadeh Yeganeh
- Department of Physiology, University of Manitoba, Winnipeg, MB R3E OJ9, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R3T 2N2, Canada
| | - Carla G. Taylor
- Department of Physiology, University of Manitoba, Winnipeg, MB R3E OJ9, Canada
- Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3E OJ9, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R3T 2N2, Canada
| | - Peter Zahradka
- Department of Physiology, University of Manitoba, Winnipeg, MB R3E OJ9, Canada
- Department of Human Nutritional Sciences, University of Manitoba, Winnipeg, MB R3E OJ9, Canada
- Canadian Centre for Agri-Food Research in Health and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R3T 2N2, Canada
| | - Jeffrey T. Wigle
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, MB R3E OJ9, Canada
- Institute of Cardiovascular Sciences, and Medicine, St. Boniface Hospital Research Centre, Winnipeg, MB R2H 2A6, Canada
| |
Collapse
|
26
|
Regulation of autophagy by glucose in Mammalian cells. Cells 2012; 1:372-95. [PMID: 24710481 PMCID: PMC3901114 DOI: 10.3390/cells1030372] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2012] [Revised: 06/22/2012] [Accepted: 07/13/2012] [Indexed: 02/07/2023] Open
Abstract
Autophagy is an evolutionarily conserved process that contributes to maintain cell homeostasis. Although it is strongly regulated by many extracellular factors, induction of autophagy is mainly produced by starvation of nutrients. In mammalian cells, the regulation of autophagy by amino acids, and also by the hormone insulin, has been extensively investigated, but knowledge about the effects of other autophagy regulators, including another nutrient, glucose, is more limited. Here we will focus on the signalling pathways by which environmental glucose directly, i.e., independently of insulin and glucagon, regulates autophagy in mammalian cells, but we will also briefly mention some data in yeast. Although glucose deprivation mainly induces autophagy via AMPK activation and the subsequent inhibition of mTORC1, we will also comment other signalling pathways, as well as evidences indicating that, under certain conditions, autophagy can be activated by glucose. A better understanding on how glucose regulates autophagy not only will expand our basic knowledge of this important cell process, but it will be also relevant to understand common human disorders, such as cancer and diabetes, in which glucose levels play an important role.
Collapse
|
27
|
Wang B, Yu Y, Han L. Adiponectin improves endothelial dysfunction caused by elevated FFAs levels, partially through cAMP-dependent pathway. Diabetes Res Clin Pract 2012; 97:119-24. [PMID: 22502813 DOI: 10.1016/j.diabres.2012.02.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 02/11/2012] [Accepted: 02/14/2012] [Indexed: 02/05/2023]
Abstract
AIMS To determine whether adiponectin can attenuate endothelial dysfunction caused by elevated free fatty acids (FFAs) concentration, and if so, to explore the underlying mechanism. METHODS Male Sprague-Dawley rat thoracic aortas were isolated then cut into four vascular rings, incubated in organ bath containing Krebs-Henseleit buffer with different agents separately: 800 μmol/L Palmic acid (FFA, n = 14), 800 μmol/L Palmic acid + 2 μg/mL adiponectin (FFA + gAd, n = 14), 800 μmol/L Palmic acid + 2 μg/mL adiponectin + 200 μmol/L adenylate cyclase inhibitor dideoxyadenosine (FFA + gAd + ddAdo, n = 7), blank control (NC, n=10). The endothelial dependent vasodilatation (EDV) and endothelial independent vasodilatation (EIV) were assessed by acetylcholine (Ach) induced contraction of the aortas. Nuclear transcription factor kappa B (NF-κB) expression in rat aortic section was evaluated immunohistochemically. RESULTS Ach caused a concentration dependent vascular relaxation in all pre-constricted aortic rings. PA treatment impaired the Ach induced EDV which was significantly attenuated by pretreatment with adiponectin. Dideoxyadenosine partly abolished the vascular protective effect of adiponectin. Sodium nitroprusside (SNP) had no significant effect on the vasodilatation among four groups. Increased NF-κB expression was noted in FFA group. Pretreatment with adiponectin partly decreased NF-κB expression when compared with FFA group. CONCLUSION Adiponectin may independently mitigate endothelial dysfunction caused by elevated FFAs concentration through the cross talk between cAMP and NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ben Wang
- Department of Endocrinology and Metabolism, West China Hospital, Sichuan University, Chengdu City, Sichuan Province, China
| | | | | |
Collapse
|
28
|
Kalariya NM, Shoeb M, Ansari NH, Srivastava SK, Ramana KV. Antidiabetic drug metformin suppresses endotoxin-induced uveitis in rats. Invest Ophthalmol Vis Sci 2012; 53:3431-40. [PMID: 22562515 DOI: 10.1167/iovs.12-9432] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
PURPOSE To investigate the therapeutic effects of metformin, a commonly used antidiabetic drug, in preventing endotoxin-induced uveitis (EIU) in rats. METHODS EIU in Lewis rats was developed by subcutaneous injection of lipopolysaccharide (LPS; 150 μg). Metformin (300 mg/kg body weight, intraperitoneally) or its carrier was injected either 12 hours before or 2 hours after LPS induction. Three and 24 hours after EIU, eyes were enucleated and aqueous humor (AqH) was collected. The MILLIPLEX-MAG Rat cytokine-chemokine magnetic bead array was used to determine inflammatory cytokines. The expression of Cox-2, phosphorylation of AMPK, and NF-κB (p65) were determined immunohistochemically. Primary human nonpigmented ciliary epithelial cells (HNPECs) were used to determine the in vitro efficacy of metformin. RESULTS Compared with controls, the EIU rat AqH had significantly increased number of infiltrating cells and increased levels of various cytokines and chemokines (TNF-α, MCP-1, IL-1β, MIP-1α, IL-6, Leptin, and IL-18) and metformin significantly prevented the increase. Metformin also prevented the expression of Cox-2 and phosphorylation of p65, and increased the activation of AMPK in the ciliary bodies and retinal tissues. Moreover, metformin prevented the expression of Cox-2, iNOS, and activation of NF-kB in the HNPECs and decreased the levels of NO and PGE2 in cell culture media. CONCLUSIONS Our results for the first time demonstrate a novel role of the antidiabetic drug, metformin, in suppressing uveitis in rats and suggest that this drug could be developed to prevent uveitis complications.
Collapse
Affiliation(s)
- Nilesh M Kalariya
- Department of Ophthalmology & Visual Sciences, University of Texas Medical Branch, Galveston, Texas 77555, USA
| | | | | | | | | |
Collapse
|
29
|
Abstract
UNLABELLED The global epidemic of obesity is accompanied by an increased prevalence of cardiovascular disease (CVD), in particular stroke and heart attack. Dysfunctional adipose tissue links obesity to CVD by secreting a multitude of bioactive lipids and pro-inflammatory factors (adipokines) with detrimental effects on the cardiovascular system. Adiponectin is one of the few adipokines that possesses multiple salutary effects on insulin sensitivity and cardiovascular health. Clinical investigations have identified adiponectin deficiency (hypoadiponectinaemia) as an independent risk factor for CVD. In animals, elevation of plasma adiponectin by either pharmacological or genetic approaches alleviates obesity-induced endothelial dysfunction and hypertension, and also prevents atherosclerosis, myocardial infarction and diabetic cardiomyopathy. Furthermore, many therapeutic benefits of the peroxisome-proliferator activated receptor gamma agonists, the thiazolidinediones, are mediated by induction of adiponectin. Adiponectin protects cardiovascular health through its vasodilator, anti-apoptotic, anti-inflammatory and anti-oxidative activities in both cardiac and vascular cells. This review summarizes recent findings in the understanding of the physiological role and clinical relevance of adiponectin in cardiovascular health, and in the identification of the receptor and postreceptor signalling events that mediate the cardiovascular actions of adiponectin. It also discusses adiponectin-targeted drug discovery strategies for treating obesity, diabetes and CVD. LINKED ARTICLES This article is part of a themed section on Fat and Vascular Responsiveness. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2012.165.issue-3.
Collapse
Affiliation(s)
- Xiaoyan Hui
- Department of Medicine, the University of Hong Kong, Hong Kong
| | | | | | | |
Collapse
|
30
|
Jung MY, Kim HS, Hong HJ, Youn BS, Kim TS. Adiponectin induces dendritic cell activation via PLCγ/JNK/NF-κB pathways, leading to Th1 and Th17 polarization. THE JOURNAL OF IMMUNOLOGY 2012; 188:2592-601. [PMID: 22345647 DOI: 10.4049/jimmunol.1102588] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Adiponectin (APN) is a crucial regulator for many inflammatory processes, but its effect on Th cell-mediated responses has not been fully understood. Thus, we investigated the immune-modulatory effects of APN on dendritic cells (DCs) controlling Th cell polarization. APN induced maturation and activation of DCs, as demonstrated by the increased expression of MHC class II, costimulatory molecules in both mouse and human DCs, and it significantly enhanced production of proinflammatory cytokines. APN triggered degradation of IκB proteins, nuclear translocation of NF-κB p65 subunit, and phosphorylation of MAPKs in DCs. Pretreatment with a phospholipase C (PLC)γ inhibitor and a JNK inhibitor suppressed IL-12 production and NF-κB binding activity. Additionally, PLCγ inhibitor downregulated phosphorylation of JNK, indicating that PLCγ and JNK may be upstream molecules of NF-κB. Importantly, APN-treated DCs significantly induced both Th1 and Th17 responses in allogeneic CD4(+) T cells. The addition of a neutralizing anti-IL-12 mAb to the cocultures abolished the secretion of IFN-γ, whereas the blockage of IL-23 and IL-1β suppressed APN-induced IL-17 production. Immunization of mice with OVA-pulsed, APN-treated DCs efficiently led to Ag-specific Th1 and Th17 cell responses. Taken together, these results demonstrated that APN effectively induced activation of DCs through PLCγ/JNK/NF-κB-signaling pathways, leading to enhanced Th1 and Th17 responses.
Collapse
Affiliation(s)
- Mi Young Jung
- Division of Life Sciences, School of Life Sciences and Biotechnology, Korea University, Seoul 136-701, Republic of Korea
| | | | | | | | | |
Collapse
|
31
|
|
32
|
Xiao X, Dong Y, Zhong J, Cao R, Zhao X, Wen G, Liu J. Adiponectin protects endothelial cells from the damages induced by the intermittent high level of glucose. Endocrine 2011; 40:386-93. [PMID: 21948177 DOI: 10.1007/s12020-011-9531-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2011] [Accepted: 08/30/2011] [Indexed: 11/24/2022]
Abstract
Globular adiponectin (gAd) has anti-atherogenic effects on the vascular wall. Intermittent hyperglycemia induces endothelial cells (ECs) injury but the physiological factors that may protect against ECs damage are largely unknown. In the present study, we investigated the effect of gAd on ECs dysfunction induced by intermittent high glucose. The gAd significantly attenuated intermittent high glucose-induced apoptosis and oxidative stress in human umbilical vein endothelial cells. This was achieved by decreasing caspase-3 and 3-nitrotyrosine protein expression, increasing nitric oxide (NO) secretion and phosphorylation of Akt, AMPK, and endothelial nitric oxide synthase protein expression. Pretreatment with a phosphatidylinositol 3' kinase (PI3K) inhibitor, LY294002, partly reversed adiponectin's anti-apoptotic effect. Taken together, our results indicate that gAd acts as a critical physiological factor which protects against fluctuating high glucose-induced endothelial damage. It may act via attenuating apoptosis and increasing synthesis of NO through both the PI3K/AKT and AMPK signaling pathway to reduce oxidative stress and cell apoptosis.
Collapse
Affiliation(s)
- Xinhua Xiao
- Department of Metabolism & Endocrinology, The First Affiliated Hospital, University of South China, 69 Chuanshan Road, Hengyang, 421001, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
33
|
Gerlo S, Kooijman R, Beck IM, Kolmus K, Spooren A, Haegeman G. Cyclic AMP: a selective modulator of NF-κB action. Cell Mol Life Sci 2011; 68:3823-41. [PMID: 21744067 PMCID: PMC11114830 DOI: 10.1007/s00018-011-0757-8] [Citation(s) in RCA: 149] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Revised: 06/10/2011] [Accepted: 06/16/2011] [Indexed: 10/18/2022]
Abstract
It has been known for several decades that cyclic AMP (cAMP), a prototypical second messenger, transducing the action of a variety of G-protein-coupled receptor ligands, has potent immunosuppressive and anti-inflammatory actions. These actions have been attributed in part to the ability of cAMP-induced signals to interfere with the function of the proinflammatory transcription factor Nuclear Factor-kappaB (NF-κB). NF-κB plays a crucial role in switching on the gene expression of a plethora of inflammatory and immune mediators, and as such is one of the master regulators of the immune response and a key target for anti-inflammatory drug design. A number of fundamental molecular mechanisms, contributing to the overall inhibitory actions of cAMP on NF-κB function, are well established. Paradoxically, recent reports indicate that cAMP, via its main effector, the protein kinase A (PKA), also promotes NF-κB activity. Indeed, cAMP actions appear to be highly cell type- and context-dependent. Importantly, several novel players in the cAMP/NF-κB connection, which selectively direct cAMP action, have been recently identified. These findings not only open up exciting new research avenues but also reveal novel opportunities for the design of more selective, NF-κB-targeting, anti-inflammatory drugs.
Collapse
Affiliation(s)
- Sarah Gerlo
- VIB Department of Medical Protein Research, Ghent University (UGent), Albert Baertsoenkaai, Belgium.
| | | | | | | | | | | |
Collapse
|
34
|
Lugnier C. PDE inhibitors: a new approach to treat metabolic syndrome? Curr Opin Pharmacol 2011; 11:698-706. [PMID: 22018840 DOI: 10.1016/j.coph.2011.09.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Accepted: 09/27/2011] [Indexed: 01/16/2023]
Abstract
About one third of people in the world suffer from metabolic syndrome (MetS), with symptoms such as hypertension and elevated blood cholesterol, and with increased risk of developing additional diseases such as diabetes mellitus and heart disease. The progression of this multifactorial pathology, which targets various tissues and organs, might necessitate a renewal in therapeutic approaches. Since cyclic nucleotide phosphodiesterases (PDEs), enzymes which hydrolyze cyclic AMP and cyclic GMP, play a crucial role in regulating endocrine and cardiovascular functions, inflammation, oxidative stress, and cell proliferation, all of which contribute to MetS, we wonder whether PDE inhibitors might represent new therapeutic approaches for preventing and treating MetS.
Collapse
Affiliation(s)
- Claire Lugnier
- CNRS UMR 7213, Laboratoire de Biophotonique et Pharmacologie, Université de Strasbourg, Faculté de Pharmacie, 74 route du Rhin, 67401 Illkirch, France.
| |
Collapse
|
35
|
Lau WB, Tao L, Wang Y, Li R, Ma XL. Systemic adiponectin malfunction as a risk factor for cardiovascular disease. Antioxid Redox Signal 2011; 15:1863-73. [PMID: 21091079 PMCID: PMC3159112 DOI: 10.1089/ars.2010.3743] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Adiponectin (Ad) is an abundant protein hormone regulatory of numerous metabolic processes. The 30 kDa protein originates from adipose tissue, with full-length and globular domain circulatory forms. A collagenous domain within Ad leads to spontaneous self-assemblage into various oligomeric isoforms, including trimers, hexamers, and high-molecular-weight multimers. Two membrane-spanning receptors for Ad have been identified, with differing concentration distribution in various body tissues. The major intracellular pathway activated by Ad includes phosphorylation of AMP-activated protein kinase, which is responsible for many of Ad's metabolic regulatory, anti-inflammatory, vascular protective, and anti-ischemic properties. Additionally, several AMP-activated protein kinase-independent mechanisms responsible for Ad's anti-inflammatory and anti-ischemic (resulting in cardioprotective) effects have also been discovered. Since its 1995 discovery, Ad has garnered considerable attention for its role in diabetic and cardiovascular pathology. Clinical observations have demonstrated the association of hypoadiponectinemia in patients with obesity, cardiovascular disease, and insulin resistance. In this review, we elaborate currently known information about Ad malfunction and deficiency pertaining to cardiovascular disease risk (including atherosclerosis, endothelial dysfunction, and cardiac injury), as well as review evidence supporting Ad resistance as a novel risk factor for cardiovascular injury, providing insight about the future of Ad research and the protein's potential therapeutic benefits.
Collapse
Affiliation(s)
- Wayne Bond Lau
- Department of Emergency Medicine, Thomas Jefferson University, 1020 Sansom Street, Philadelphia, PA 19107, USA
| | | | | | | | | |
Collapse
|
36
|
Vaiopoulos AG, Marinou K, Christodoulides C, Koutsilieris M. The role of adiponectin in human vascular physiology. Int J Cardiol 2011; 155:188-93. [PMID: 21907426 DOI: 10.1016/j.ijcard.2011.07.047] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2010] [Revised: 06/22/2011] [Accepted: 07/11/2011] [Indexed: 12/20/2022]
Abstract
Adiponectin (ApN) is an adipose tissue-derived hormone which is involved in a wide variety of physiological processes including energy metabolism, inflammation, and vascular physiology via actions on a broad spectrum of target organs including liver, skeletal muscle, and vascular endothelium. Besides possessing insulin sensitizing and anti-inflammatory properties ApN also exerts a pivotal role in vascular protection through activation of multiple intracellular signaling cascades. Enhancement of nitric oxide generation and attenuation of reactive oxygen species production in endothelial cells along with reduced vascular smooth muscle cell proliferation and migration constitute some of ApN's vasoprotective actions. Additionally, recent data indicate that ApN has direct myocardio-protective effects. Decreased plasma ApN levels are implicated in the pathogenesis of the metabolic syndrome and atherosclerosis and may serve as a diagnostic and prognostic biomarker as well as a rational pharmaco-therapeutic target to treat these disorders. This review article summarizes recent work on the cardiovascular actions of ApN.
Collapse
|
37
|
AMP-activated protein kinase inhibits NF-κB signaling and inflammation: impact on healthspan and lifespan. JOURNAL OF MOLECULAR MEDICINE (BERLIN, GERMANY) 2011. [PMID: 21431325 DOI: 10.1007/s00109-011-0748-0.] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 09/29/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a crucial regulator of energy metabolic homeostasis and thus a major survival factor in a variety of metabolic stresses and also in the aging process. Metabolic syndrome is associated with a low-grade, chronic inflammation, primarily in adipose tissue. A low-level of inflammation is also present in the aging process. There are emerging results indicating that AMPK signaling can inhibit the inflammatory responses induced by the nuclear factor-κB (NF-κB) system. The NF-κB subunits are not direct phosphorylation targets of AMPK, but the inhibition of NF-κB signaling is mediated by several downstream targets of AMPK, e.g., SIRT1, PGC-1α, p53, and Forkhead box O (FoxO) factors. AMPK signaling seems to enhance energy metabolism while it can repress inflammatory responses linked to chronic stress, e.g., in nutritional overload and during the aging process. AMPK can inhibit endoplasmic reticulum and oxidative stresses which are involved in metabolic disorders and the aging process. Interestingly, many target proteins of AMPK are so-called longevity factors, e.g., SIRT1, p53, and FoxOs, which not only can increase the stress resistance and extend the lifespan of many organisms but also inhibit the inflammatory responses. The activation capacity of AMPK declines in metabolic stress and with aging which could augment the metabolic diseases and accelerate the aging process. We will review the AMPK pathways involved in the inhibition of NF-κB signaling and suppression of inflammation. We also emphasize that the capacity of AMPK to repress inflammatory responses can have a significant impact on both healthspan and lifespan.
Collapse
|
38
|
Salminen A, Hyttinen JMT, Kaarniranta K. AMP-activated protein kinase inhibits NF-κB signaling and inflammation: impact on healthspan and lifespan. J Mol Med (Berl) 2011; 89:667-76. [PMID: 21431325 PMCID: PMC3111671 DOI: 10.1007/s00109-011-0748-0] [Citation(s) in RCA: 651] [Impact Index Per Article: 46.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2011] [Revised: 02/24/2011] [Accepted: 02/28/2011] [Indexed: 12/27/2022]
Abstract
Adenosine monophosphate-activated protein kinase (AMPK) is a crucial regulator of energy metabolic homeostasis and thus a major survival factor in a variety of metabolic stresses and also in the aging process. Metabolic syndrome is associated with a low-grade, chronic inflammation, primarily in adipose tissue. A low-level of inflammation is also present in the aging process. There are emerging results indicating that AMPK signaling can inhibit the inflammatory responses induced by the nuclear factor-κB (NF-κB) system. The NF-κB subunits are not direct phosphorylation targets of AMPK, but the inhibition of NF-κB signaling is mediated by several downstream targets of AMPK, e.g., SIRT1, PGC-1α, p53, and Forkhead box O (FoxO) factors. AMPK signaling seems to enhance energy metabolism while it can repress inflammatory responses linked to chronic stress, e.g., in nutritional overload and during the aging process. AMPK can inhibit endoplasmic reticulum and oxidative stresses which are involved in metabolic disorders and the aging process. Interestingly, many target proteins of AMPK are so-called longevity factors, e.g., SIRT1, p53, and FoxOs, which not only can increase the stress resistance and extend the lifespan of many organisms but also inhibit the inflammatory responses. The activation capacity of AMPK declines in metabolic stress and with aging which could augment the metabolic diseases and accelerate the aging process. We will review the AMPK pathways involved in the inhibition of NF-κB signaling and suppression of inflammation. We also emphasize that the capacity of AMPK to repress inflammatory responses can have a significant impact on both healthspan and lifespan.
Collapse
Affiliation(s)
- Antero Salminen
- Department of Neurology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
- Department of Neurology, Kuopio University Hospital, P.O. Box 1777, 70211 Kuopio, Finland
| | - Juha M. T. Hyttinen
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
| | - Kai Kaarniranta
- Department of Ophthalmology, Institute of Clinical Medicine, University of Eastern Finland, P.O. Box 1627, 70211 Kuopio, Finland
- Department of Ophthalmology, Kuopio University Hospital, P.O. Box 1777, 70211 Kuopio, Finland
| |
Collapse
|
39
|
Chen X, Zhang H, McAfee S, Zhang C. The reciprocal relationship between adiponectin and LOX-1 in the regulation of endothelial dysfunction in ApoE knockout mice. Am J Physiol Heart Circ Physiol 2010; 299:H605-12. [PMID: 20581092 DOI: 10.1152/ajpheart.01096.2009] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We hypothesized that the reciprocal association between adiponectin and lectin-like oxidized LDL (ox-LDL) receptor (LOX)-1 contributes to the regulation of aortic endothelial dysfunction in atherosclerosis. To test this hypothesis, endothelium-dependent (ACh) and endothelium-independent (sodium nitroprusside) vasorelaxation of isolated aortic rings from control mice, apolipoprotein E (ApoE) knockout (KO) mice, and ApoE KO mice treated with either adiponectin (15 microg x day(-1) x mouse(-1) sc for 8 days) or neutralizing antibody to LOX-1 (anti-LOX-1, 16 microg/ml, 0.1 ml/mouse ip for 7 days) were examined. Although vasorelaxation to sodium nitroprusside was not different between control and ApoE KO mice, relaxation to ACh was impaired in ApoE KO mice. Adiponectin and anti-LOX-1 restored nitric oxide-mediated endothelium-dependent vasorelaxation in ApoE KO mice. Aortic ROS formation and ox-LDL uptake were increased in ApoE KO mice. Both adiponectin and anti-LOX-1 treatment reduced ROS production and aortic ox-LDL uptake. In mouse coronary artery endothelial cells, TNF-alpha incubation increased endothelial LOX-1 expression. Adiponectin reduced TNF-alpha-induced LOX-1 expression. Consistently, in ApoE KO mice, adiponectin treatment reversed elevated LOX-1 expression in aortas. Immunofluorescence staining showed that adiponectin was mainly colocalized with endothelial cells. Although adiponectin expression was lower in ApoE KO versus control mice, anti-LOX-1 increased aortic adiponectin expression, suggesting a reciprocal regulation between adiponectin and LOX-1. Moreover, both adiponectin and anti-LOX-1 reduced NF-kappaB expression in ApoE KO mice. Thus, adiponectin and LOX-1 may converge on NF-kappaB signaling to regulate their function. In conclusion, our results indicate that the reciprocal regulation between adiponectin and LOX-1 amplifies oxidative stress and ox-LDL uptake, leading to endothelial dysfunction in atherosclerosis.
Collapse
Affiliation(s)
- Xiuping Chen
- Department of Internal Medicine, Dalton CardiovascularResearch Center, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | | |
Collapse
|
40
|
Total adiponectin does not predict cardiovascular events in middle-aged men in a prospective, long-term follow-up study. DIABETES & METABOLISM 2010; 36:137-43. [DOI: 10.1016/j.diabet.2009.10.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2009] [Revised: 10/15/2009] [Accepted: 10/16/2009] [Indexed: 01/31/2023]
|
41
|
Reduced vascular responsiveness to adiponectin in hyperlipidemic rats--mechanisms and significance. J Mol Cell Cardiol 2010; 49:508-15. [PMID: 20303976 DOI: 10.1016/j.yjmcc.2010.03.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2010] [Revised: 03/04/2010] [Accepted: 03/05/2010] [Indexed: 01/14/2023]
Abstract
Deficiency of adiponectin (APN), an adipocyte-derived vascular protective molecule, contributes to diabetic vascular injury. The current study determined whether obesity/hyperlipidemia may alter the vascular response to APN, and investigated the involved mechanisms and pathologic significance. Adult male Sprague-Dawley rats were fed a regular or high-fat diet (HF) for 4-16 weeks. Circulating APN levels, aortic pAMPK/AMPK, peNOS/eNOS, and APN receptor expression levels were determined. Compared to time-matched animals fed control diet, plasma APN levels in HF-diet animals were significantly increased at 8 weeks, and rapidly declined thereafter. Despite unchanged or elevated circulating APN levels, phosphorylated AMPK and eNOS in vascular tissue were significantly reduced at all observed time points. Recombinant full-length APN (rAPN)-induced AMPK/eNOS phosphorylation and vasodilatation were significantly reduced in 16-week obese/hyperlipidemic aortic segments. Vascular APN receptor 1 (AdipoR1) and receptor 2 (AdipoR2) expression were significantly reduced 16 weeks after HF-diet. Pre-incubation of rAPN with obese/hyperlipidemic plasma, but not with normal plasma, significantly reduced its AMPK and eNOS activation effect, and blunted its protective effect against TNFalpha-induced HUVEC apoptosis. This study demonstrated for the first time that obesity/hyperlipidemia reduces vascular responsiveness to APN. Modification/inactivation of APN by unidentified factors present in obese/hyperlipidemic plasma, decreased vascular AdipoR1/R2 expression, and reduced circulating APN levels contribute to reduced vascular responsiveness to APN at different stages of the obese condition. Reduced APN bioactivity allows unmitigated TNFalpha pro-apoptotic and pro-inflammatory actions, contributing to vascular injury in obesity/hyperlipidemia.
Collapse
|
42
|
Sung RJ, Wu YH, Lai NHJ, Teng CH, Luo CH, Tien HC, Lo CP, Wu SN. β-Adrenergic modulation of arrhythmogenesis and identification of targeted sites of antiarrhythmic therapy in Timothy (LQT8) syndrome: a theoretical study. Am J Physiol Heart Circ Physiol 2010; 298:H33-44. [DOI: 10.1152/ajpheart.00232.2009] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Timothy syndrome (TS) is a malignant form of congenital long QT syndrome with a mode of arrhythmia onset often triggered by enhanced sympathetic tone. We sought to explore mechanisms by which β-adrenergic stimulation (BAS) modulates arrhythmogenesis and to identify potential targeted sites of antiarrhythmic therapy in TS. Using a dynamic Luo-Rudy ventricular myocyte model incorporated with detailed intracellular Ca2+ cycling, along with its one-dimensional multicellular strand, we simulated various clinical scenarios of TS, with stepwise increase in the percentage of G406R Cav1.2 channels from 0 to 11.5 and 23%, and to 38.5 and 77%, respectively, for heterozygous and homozygous states of TS1 and TS2. Progressive prolongation of action potential duration (APD) and QT interval, accompanied by amplification of transmural dispersion of repolarization, steepening of APD restitution, induction of delayed afterdepolariztions (DADs), and both DAD and phase 3 early afterdepolariztion-mediated triggered activities, correlated well with the extent of G406R Cav1.2 channel mutation. BAS amplified transmural dispersion of repolarization, steepened APD restitution, and facilitated inducibility of DAD-mediated triggered activity. Systematic analysis of intracellular Ca2+ cycling revealed that sarcoplasmic reticulum Ca2+ ATPase (uptake current) played an essential role in BAS-induced facilitation of DAD-mediated triggered activity and, in addition to L-type calcium current, it could be an effective site of antiarrhythmic therapy under the influence of BAS. Thus G406R Cav1.2 channel mutation confers not only a trigger, but also a substrate for lethal ventricular arrhythmias, which can be exaggerated by BAS. It is suggested that, besides β-adrenergic blockers and L-type calcium current channel blockers, an agent aimed at reduction of sarcoplasmic reticulum Ca2+ ATPase uptake current may provide additional antiarrhythmic effect in patients with TS.
Collapse
Affiliation(s)
- Ruey J. Sung
- Institute of Life Sciences, College of Sciences, National Central University, Taoyuan
- Division of Cardiovascular Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, California
| | - Yung-Han Wu
- Institute of Basic Medical Research, College of Medicine,
| | | | | | - Ching-Hsing Luo
- Institute of Electrical Engineering, College of Engineering and Computer Science, National Cheng Kung University, Tainan; and
| | - Hui-Chun Tien
- Department of Applied Mathematics, Providence University, Taichung, Taiwan; and
| | - Chu-Pin Lo
- Department of Applied Mathematics, Providence University, Taichung, Taiwan; and
| | - Sheng-Nan Wu
- Institute of Basic Medical Research, College of Medicine,
| |
Collapse
|
43
|
Cai XJ, Chen L, Li L, Feng M, Li X, Zhang K, Rong YY, Hu XB, Zhang MX, Zhang Y, Zhang M. Adiponectin inhibits lipopolysaccharide-induced adventitial fibroblast migration and transition to myofibroblasts via AdipoR1-AMPK-iNOS pathway. Mol Endocrinol 2009; 24:218-28. [PMID: 19889816 DOI: 10.1210/me.2009-0128] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Adiponectin is an important antiatherogenic adipocytokine that inhibits inflammation, insulin resistance, and oxide stress. Inflammation in the vascular adventitia is a crucial factor in the pathogenesis of atherosclerosis. Adventitial fibroblasts (AFs) can proliferate, divide into myofibroblasts, and migrate to the intima to become a new component of atherosclerotic plaque under inflammation and atherosclerosis. We investigated whether adiponectin might prevent AFs from proliferating, migrating, and transforming into myofibroblasts. Cultured AFs were stimulated with lipopolysaccharide (LPS) in the presence or absence of adiponectin. Methyl thiazolyl tetrazolium assay and migration and scratch-wound assays demonstrated that adiponectin reduced the AF proliferation and migration induced by LPS, respectively, whereas treatment with AdipoR1 small interfering (si) RNA (siAdipoR1), AMP-activated protein kinase (AMPK) siRNA (siAMPK), and an AMPK inhibitor reversed the effect. Immunocytochemistry and Western blot revealed that adiponectin reduced the transition of AFs to myofibroblasts, and treatment with siAdipoR1, siAMPK, and the AMPK inhibitor increased the transition. RT-PCR, Western blotting, and nitric oxide (NO) assay showed that adiponectin reduces induced NO synthase (iNOS) and nitrotyrosine expression and NO and ONOO(-) production induced by LPS. Treatment with siAdipoR1, siAMPK, and the AMPK inhibitor significantly attenuated adiponectin-induced phosphorylation of AMPK and its downstream target acetyl-coenzyme A carboxylase and up-regulated iNOS mRNA and protein expression, which resulted in a marked increase of NO and ONOO(-) production. In apolipoprotein E-deficient mice, immunohistochemistry of treated vascular adventitia showed that both iNOS expression and ONOO(-) production could be reversed with an adenovirus-adiponectin vector. Taken together, these results suggest that adiponectin reduces LPS-induced NO production and nitrosative stress and prevents AFs from proliferating, transforming to myoflbroblasts, and migrating to the intima, thus worsening atherosclerosis, by inhibiting the AdipoR1-AMPK-iNOS pathway in AFs.
Collapse
Affiliation(s)
- Xiao-jun Cai
- The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Shandong University Qilu Hospital, Jinan, Shandong, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Jeon BT, Shin HJ, Kim JB, Kim YK, Lee DH, Kim KH, Kim HJ, Kang SS, Cho GJ, Choi WS, Roh GS. Adiponectin protects hippocampal neurons against kainic acid-induced excitotoxicity. ACTA ACUST UNITED AC 2009; 61:81-8. [DOI: 10.1016/j.brainresrev.2009.05.002] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2008] [Revised: 04/23/2009] [Accepted: 05/11/2009] [Indexed: 01/05/2023]
|
45
|
Lee DE, Kehlenbrink S, Lee H, Hawkins M, Yudkin JS. Getting the message across: mechanisms of physiological cross talk by adipose tissue. Am J Physiol Endocrinol Metab 2009; 296:E1210-29. [PMID: 19258492 DOI: 10.1152/ajpendo.00015.2009] [Citation(s) in RCA: 88] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Obesity is associated with resistance of skeletal muscle to insulin-mediated glucose uptake, as well as resistance of different organs and tissues to other metabolic and vascular actions of insulin. In addition, the body is exquisitely sensitive to nutrient imbalance, with energy excess or a high-fat diet rapidly increasing insulin resistance, even before noticeable changes occur in fat mass. There is a growing acceptance of the fact that, as well as acting as a storage site for surplus energy, adipose tissue is an important source of signals relevant to, inter alia, energy homeostasis, fertility, and bone turnover. It has also been widely recognized that obesity is a state of low-grade inflammation, with adipose tissue generating substantial quantities of proinflammatory molecules. At a cellular level, the understanding of the signaling pathways responsible for such alterations has been intensively investigated. What is less clear, however, is how alterations of physiology, and of signaling, within one cell or one tissue are communicated to other parts of the body. The concepts of cell signals being disseminated systemically through a circulating "endocrine" signal have been complemented by the view that local signaling may similarly occur through autocrine or paracrine mechanisms. Yet, while much elegant work has focused on the alterations in signaling that are found in obesity or energy excess, there has been less attention paid to ways in which such signals may propagate to remote organs. This review of the integrative physiology of obesity critically appraises the data and outlines a series of hypotheses as to how interorgan cross talk takes place. The hypotheses presented include the "fatty acid hypothesis,", the "portal hypothesis,", the "endocrine hypothesis,", the "inflammatory hypothesis,", the "overflow hypothesis,", a novel "vasocrine hypothesis," and a "neural hypothesis," and the strengths and weaknesses of each hypothesis are discussed.
Collapse
Affiliation(s)
- Do-Eun Lee
- Department of Internal Medicine, Division of Endocrinology, Winthrop University Hospital, London, United Kingdom
| | | | | | | | | |
Collapse
|
46
|
Chang LC, Huang KC, Wu YW, Kao HL, Chen CL, Lai LP, Hwang JJ, Yang WS. The Clinical Implications of Blood Adiponectin in Cardiometabolic Disorders. J Formos Med Assoc 2009; 108:353-66. [DOI: 10.1016/s0929-6646(09)60079-6] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
|
47
|
Loss of p53 enhances catalytic activity of IKKbeta through O-linked beta-N-acetyl glucosamine modification. Proc Natl Acad Sci U S A 2009; 106:3431-6. [PMID: 19202066 DOI: 10.1073/pnas.0813210106] [Citation(s) in RCA: 160] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The IkappaB kinase (IKK)-NF-kappaB pathway plays a critical role in oncogenesis. Recently, we have shown that p53 regulates glucose metabolism through the IKK-NF-kappaB pathway and that, in the absence of p53, the positive feedback loop between IKK-NF-kappaB and glycolysis has an integral role in oncogene-induced cell transformation. Here, we demonstrate that IKKbeta, a component of the IKK complex, was constitutively modified with O-linked beta-N-acetyl glucosamine (O-GlcNAc) in both p53-deficient mouse embryonic fibroblasts (MEFs) and transformed human fibroblasts. In p53-deficient cells, the O-GlcNAcylated IKKbeta and the activating phosphorylation of IKK were decreased by p65/NF-kappaB knockdown or glucose depletion. We also found that high glucose induced the O-GlcNAcylation of IKKbeta and sustained the TNFalpha-dependent IKKbeta activity. Moreover, the O-GlcNAcase inhibitor streptozotocin intensified O-GlcNAcylation and concomitant activating phosphorylation of IKKbeta. Mutational analysis revealed that O-GlcNAcylation of IKKbeta occurred at Ser 733 in the C-terminal domain, which was identified as an inactivating phosphorylation site, suggesting that IKKbeta O-GlcNAcylation regulates its catalytic activity. Taken together, we propose a novel mechanism for the enhancement of NF-kappaB activity by loss of p53, which evokes positive feedback regulation from enhanced glucose metabolism to IKK in oncogenesis.
Collapse
|
48
|
Edinger RS, Lebowitz J, Li H, Alzamora R, Wang H, Johnson JP, Hallows KR. Functional regulation of the epithelial Na+ channel by IkappaB kinase-beta occurs via phosphorylation of the ubiquitin ligase Nedd4-2. J Biol Chem 2009; 284:150-157. [PMID: 18981174 PMCID: PMC2610498 DOI: 10.1074/jbc.m807358200] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Revised: 10/28/2008] [Indexed: 12/31/2022] Open
Abstract
We have previously shown that IkappaB kinase-beta (IKKbeta) interacts with the epithelial Na+ channel (ENaC) beta-subunit and enhances ENaC activity by increasing its surface expression in Xenopus oocytes. Here, we show that the IKKbeta-ENaC interaction is physiologically relevant in mouse polarized kidney cortical collecting duct (mpkCCDc14) cells, as RNA interference-mediated knockdown of endogenous IKKbeta in these cells by approximately 50% resulted in a similar reduction in transepithelial ENaC-dependent equivalent short circuit current. Although IKKbeta binds to ENaC, there was no detectable phosphorylation of ENaC subunits by IKKbeta in vitro. Because IKKbeta stimulation of ENaC activity occurs through enhanced channel surface expression and the ubiquitin-protein ligase Nedd4-2 has emerged as a central locus for ENaC regulation at the plasma membrane, we tested the role of Nedd4-2 in this regulation. IKKbeta-dependent phosphorylation of Xenopus Nedd4-2 expressed in HEK-293 cells occurred both in vitro and in vivo, suggesting a potential mechanism for regulation of Nedd4-2 and thus ENaC activity. 32P labeling studies utilizing wild-type or mutant forms of Xenopus Nedd4-2 demonstrated that Ser-444, a key SGK1 and protein kinase A-phosphorylated residue, is also an important IKKbeta phosphorylation target. ENaC stimulation by IKKbeta was preserved in oocytes expressing wild-type Nedd4-2 but blocked in oocytes expressing either a dominant-negative (C938S) or phospho-deficient (S444A) Nedd4-2 mutant, suggesting that Nedd4-2 function and phosphorylation by IKKbeta are required for IKKbeta regulation of ENaC. In summary, these results suggest a novel mode of ENaC regulation that occurs through IKKbeta-dependent Nedd4-2 phosphorylation at a recognized SGK1 and protein kinase A target site.
Collapse
Affiliation(s)
- Robert S Edinger
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Jonathan Lebowitz
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Hui Li
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Rodrigo Alzamora
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Huamin Wang
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - John P Johnson
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261; Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261
| | - Kenneth R Hallows
- Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261; Renal-Electrolyte Division, Departments of Medicine and Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15261.
| |
Collapse
|
49
|
Bikman BT, Zheng D, Pories WJ, Chapman W, Pender JR, Bowden RC, Reed MA, Cortright RN, Tapscott EB, Houmard JA, Tanner CJ, Lee J, Dohm GL. Mechanism for improved insulin sensitivity after gastric bypass surgery. J Clin Endocrinol Metab 2008; 93:4656-63. [PMID: 18765510 PMCID: PMC2729236 DOI: 10.1210/jc.2008-1030] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
CONTEXT Surgical treatments of obesity have been shown to induce rapid and prolonged improvements in insulin sensitivity. OBJECTIVE The aim of the study was to investigate the effects of gastric bypass surgery and the mechanisms that explain the improvement in insulin sensitivity. DESIGN We performed a cross-sectional, nonrandomized, controlled study. SETTING This study was conducted jointly between the Departments of Exercise Science and Physiology at East Carolina University in Greenville, North Carolina. SUBJECTS Subjects were recruited into four groups: 1) lean [body mass index (BMI) < 25 kg/m(2); n = 93]; 2) weight-matched (BMI = 25 to 35 kg/m(2); n = 310); 3) morbidly obese (BMI > 35 kg/m(2); n = 43); and 4) postsurgery patients (BMI approximately 30 kg/m(2); n = 40). Postsurgery patients were weight stable 1 yr after surgery. MAIN OUTCOME MEASURES Whole-body insulin sensitivity, muscle glucose transport, and muscle insulin signaling were assessed. RESULTS Postsurgery subjects had insulin sensitivity index values that were similar to the lean and higher than morbidly obese and weight-matched control subjects. Glucose transport was higher in the postsurgery vs. morbidly obese and weight-matched groups. IRS1-pSer(312) in the postsurgery group was lower than morbidly obese and weight-matched groups. Inhibitor kappaBalpha was higher in the postsurgery vs. the morbidly obese and weight-matched controls, indicating reduced inhibitor of kappaB kinase beta activity. CONCLUSIONS Insulin sensitivity and glucose transport are greater in the postsurgery patients than predicted from the weight-matched group, suggesting that improved insulin sensitivity after bypass is due to something other than, or in addition to, weight loss. Improved insulin sensitivity is related to reduced inhibitor of kappaB kinase beta activity and enhanced insulin signaling in muscle.
Collapse
Affiliation(s)
- Benjamin T Bikman
- The Metabolic Institute for the Study of Diabetes and Obesity, East Carolina University, 6N98 600 Moye Boulevard, Greenville, North Carolina 27834, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Goldstein BJ, Scalia RG, Ma XL. Protective vascular and myocardial effects of adiponectin. ACTA ACUST UNITED AC 2008; 6:27-35. [PMID: 19029992 DOI: 10.1038/ncpcardio1398] [Citation(s) in RCA: 207] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2008] [Accepted: 09/30/2008] [Indexed: 02/07/2023]
Abstract
Adiponectin is an abundant plasma protein secreted from adipocytes that elicits protective effects in the vasculature and myocardium. In obesity and insulin-resistant states, adiponectin levels are reduced and loss of its protective effects might contribute to the excess cardiovascular risk observed in these conditions. Adiponectin ameliorates the progression of macrovascular disease in rodent models, consistent with its correlation with improved vascular outcomes in epidemiological studies. The mechanisms of adiponectin signaling are multiple and vary among its cellular sites of action. In endothelial cells, adiponectin enhances production of nitric oxide, suppresses production of reactive oxygen species, and protects cells from inflammation that results from exposure to high glucose levels or tumor necrosis factor, through activation of AMP-activated protein kinase and cyclic AMP-dependent protein kinase (also known as protein kinase A) signaling cascades. In the myocardium, adiponectin-mediated protection from ischemia-reperfusion injury is linked to cyclo-oxygenase-2-mediated suppression of tumor necrosis factor signaling, inhibition of apoptosis by AMP-activated protein kinase, and inhibition of excess peroxynitrite-induced oxidative and nitrative stress. In this Review, we provide an update of studies of the signaling effects of adiponectin in endothelial cells and cardiomyocytes.
Collapse
Affiliation(s)
- Barry J Goldstein
- Harvard Medical School and the Joslin Diabetes Center in Boston, USA
| | | | | |
Collapse
|