151
|
Pedro MN, Magro DO, da Silva EUPP, Guadagnini D, Santos A, de Jesus Pedro R, Saad MJA. Plasma levels of lipopolysaccharide correlate with insulin resistance in HIV patients. Diabetol Metab Syndr 2018; 10:5. [PMID: 29434676 PMCID: PMC5793397 DOI: 10.1186/s13098-018-0308-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Accepted: 01/22/2018] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND In HIV patients using HAART insulin resistance is a central pathophysiological condition that can contribute to the development of diabetes and cardiovascular complications. To examine the role of adipocyte hormones and LPS in insulin resistance in HIV patients, we investigated the role of adiponectin, leptin, visfatin and LPS levels in the insulin resistance of HIV-infected patients treated with HAART. METHODS This study included 67 HIV positive individuals on HAART and ten healthy controls. All participants performed plasma or serum levels of glucose; insulin; lipids, visfatin, leptin, adiponectin, and LPS. The homeostasis model assessment (HOMA-IR), was used to estimate insulin resistance. RESULTS The levels of visfatin, leptin and adiponectin were similar between controls and HIV patients. However, circulating levels of LPS were higher in HIV patients on HAART than in controls. There was a positive correlation between LPS and TG (r = 0.49, p = 0.0001), between LPS and TG/HDL (r = 0.50, p = 0.0001), between LPS and insulin (r = 0.52, p = 0.0003), and between LPS and HOMA-IR (r = 0.52, p = 0.0005), in HIV patients. CONCLUSIONS Our results showed a clear correlation between plasma LPS and markers of insulin resistance, suggesting a relationship between LPS levels and metabolic alterations, particularly affecting lipids and insulin resistance in HIV patients.
Collapse
Affiliation(s)
- Marcelo Nardi Pedro
- Department of Internal Medicine-FCM, University of Campinas-UNICAMP, Campinas, SP Brazil
| | - Daniela Oliveira Magro
- Department of Internal Medicine-FCM, University of Campinas-UNICAMP, Campinas, SP Brazil
| | | | - Dioze Guadagnini
- Department of Internal Medicine-FCM, University of Campinas-UNICAMP, Campinas, SP Brazil
| | - Andrey Santos
- Department of Internal Medicine-FCM, University of Campinas-UNICAMP, Campinas, SP Brazil
| | - Rogerio de Jesus Pedro
- Department of Internal Medicine-FCM, University of Campinas-UNICAMP, Campinas, SP Brazil
| | | |
Collapse
|
152
|
Toll-like receptor-4 signaling mediates inflammation and tissue injury in diabetic nephropathy. J Nephrol 2017; 30:719-727. [DOI: 10.1007/s40620-017-0432-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 08/14/2017] [Indexed: 12/15/2022]
|
153
|
The Contribution of Singlet Oxygen to Insulin Resistance. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:8765972. [PMID: 29081894 PMCID: PMC5610878 DOI: 10.1155/2017/8765972] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 08/07/2017] [Indexed: 12/21/2022]
Abstract
Insulin resistance contributes to the development of diabetes and cardiovascular dysfunctions. Recent studies showed that elevated singlet oxygen-mediated lipid peroxidation precedes and predicts diet-induced insulin resistance (IR), and neutrophils were suggested to be responsible for such singlet oxygen production. This review highlights literature suggesting that insulin-responsive cells such as endothelial cells, hepatocytes, adipocytes, and myocytes also produce singlet oxygen, which contributes to insulin resistance, for example, by generating bioactive aldehydes, inducing endoplasmic reticulum (ER) stress, and modifying mitochondrial DNA. In these cells, nutrient overload leads to the activation of Toll-like receptor 4 and other receptors, leading to the production of both peroxynitrite and hydrogen peroxide, which react to produce singlet oxygen. Cytochrome P450 2E1 and cytochrome c also contribute to singlet oxygen formation in the ER and mitochondria, respectively. Endothelial cell-derived singlet oxygen is suggested to mediate the formation of oxidized low-density lipoprotein which perpetuates IR, partly through neutrophil recruitment to adipose tissue. New singlet oxygen-involving pathways for the formation of IR-inducing bioactive aldehydes such as 4-hydroperoxy-(or hydroxy or oxo)-2-nonenal, malondialdehyde, and cholesterol secosterol A are proposed. Strategies against IR should target the singlet oxygen-producing pathways, singlet oxygen quenching, and singlet oxygen-induced cellular responses.
Collapse
|
154
|
Bagarolli RA, Tobar N, Oliveira AG, Araújo TG, Carvalho BM, Rocha GZ, Vecina JF, Calisto K, Guadagnini D, Prada PO, Santos A, Saad STO, Saad MJA. Probiotics modulate gut microbiota and improve insulin sensitivity in DIO mice. J Nutr Biochem 2017; 50:16-25. [PMID: 28968517 DOI: 10.1016/j.jnutbio.2017.08.006] [Citation(s) in RCA: 164] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 07/05/2017] [Accepted: 08/17/2017] [Indexed: 02/06/2023]
Abstract
Obesity and type 2 diabetes are characterized by subclinical inflammatory process. Changes in composition or modulation of the gut microbiota may play an important role in the obesity-associated inflammatory process. In the current study, we evaluated the effects of probiotics (Lactobacillus rhamnosus, L. acidophilus and Bifidobacterium bifidumi) on gut microbiota, changes in permeability, and insulin sensitivity and signaling in high-fat diet and control animals. More importantly, we investigated the effects of these gut modulations on hypothalamic control of food intake, and insulin and leptin signaling. Swiss mice were submitted to a high-fat diet (HFD) with probiotics or pair-feeding for 5 weeks. Metagenome analyses were performed on DNA samples from mouse feces. Blood was drawn to determine levels of glucose, insulin, LPS, cytokines and GLP-1. Liver, muscle, ileum and hypothalamus tissue proteins were analyzed by Western blotting and real-time polymerase chain reaction. In addition, liver and adipose tissues were analyzed using histology and immunohistochemistry. The HFD induced huge alterations in gut microbiota accompanied by increased intestinal permeability, LPS translocation and systemic low-grade inflammation, resulting in decreased glucose tolerance and hyperphagic behavior. All these obesity-related features were reversed by changes in the gut microbiota profile induced by probiotics. Probiotics also induced an improvement in hypothalamic insulin and leptin resistance. Our data demonstrate that the intestinal microbiome is a key modulator of inflammatory and metabolic pathways in both peripheral and central tissues. These findings shed light on probiotics as an important tool to prevent and treat patients with obesity and insulin resistance.
Collapse
Affiliation(s)
- Renata A Bagarolli
- Department of Internal Medicine, State University of Campinas, 13081-970, Campinas, SP, Brazil
| | - Natália Tobar
- Department of Internal Medicine, State University of Campinas, 13081-970, Campinas, SP, Brazil
| | - Alexandre G Oliveira
- Department of Physical Education, São Paulo State University (UNESP), Bioscience Institute, Rio Claro, SP, Brazil
| | - Tiago G Araújo
- Department of Internal Medicine, State University of Campinas, 13081-970, Campinas, SP, Brazil
| | - Bruno M Carvalho
- Department of Biology Science, Federal University of Pernambuco, Recife, PE, Brazil
| | - Guilherme Z Rocha
- Department of Internal Medicine, State University of Campinas, 13081-970, Campinas, SP, Brazil
| | - Juliana F Vecina
- Department of Internal Medicine, State University of Campinas, 13081-970, Campinas, SP, Brazil
| | - Kelly Calisto
- Department of Internal Medicine, State University of Campinas, 13081-970, Campinas, SP, Brazil
| | - Dioze Guadagnini
- Department of Internal Medicine, State University of Campinas, 13081-970, Campinas, SP, Brazil
| | - Patrícia O Prada
- Department of Internal Medicine, State University of Campinas, 13081-970, Campinas, SP, Brazil
| | - Andrey Santos
- Department of Internal Medicine, State University of Campinas, 13081-970, Campinas, SP, Brazil
| | - Sara T O Saad
- Department of Internal Medicine, State University of Campinas, 13081-970, Campinas, SP, Brazil
| | - Mario J A Saad
- Department of Internal Medicine, State University of Campinas, 13081-970, Campinas, SP, Brazil.
| |
Collapse
|
155
|
Knockdown of Tlr4 in the Arcuate Nucleus Improves Obesity Related Metabolic Disorders. Sci Rep 2017; 7:7441. [PMID: 28785099 PMCID: PMC5547063 DOI: 10.1038/s41598-017-07858-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 07/03/2017] [Indexed: 02/06/2023] Open
Abstract
High-fat diet-induced hypothalamic metabolic inflammation is emerging as a cause for the development of obesity. It is acknowledged that Toll-like receptor4 (TLR4) signaling plays a crucial role in triggering of the hypothalamic metabolic inflammation during the course of diet-induced obesity. Whether hypothalamic arcuate nucleus (ARC)-restricted TLR4 knockdown improves obesity-related metabolic disorders remains unexplored. In this study, we used TLR4 shRNA lentiviral particles to suppress the TLR4 expression in the hypothalamic ARC of diet-induced obese rat model by stereotaxic injection. Our results demonstrate that ARC-restricted TLR4 knockdown protects obese rats from diet-induced weight gain and energy intake, from diet-induced impaired glucose homeostasis and peripheral insulin resistance, and from high-fat diet-induced hepatic steatosis and adipocyte hypertrophy. Thus, we define ARC-restricted TLR4 knockdown as a potential strategy to combat metabolic disorders associated with obesity.
Collapse
|
156
|
Zhong P, Quan D, Peng J, Xiong X, Liu Y, Kong B, Huang H. Role of CaMKII in free fatty acid/hyperlipidemia-induced cardiac remodeling both in vitro and in vivo. J Mol Cell Cardiol 2017; 109:1-16. [DOI: 10.1016/j.yjmcc.2017.06.010] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 06/19/2017] [Accepted: 06/27/2017] [Indexed: 01/24/2023]
|
157
|
Liu L, Jiang Y, Curtiss E, Fukuchi KI, Steinle JJ. TLR4 regulates insulin-resistant proteins to increase apoptosis in the mouse retina. Inflamm Res 2017; 66:993-997. [PMID: 28681194 DOI: 10.1007/s00011-017-1080-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/14/2017] [Accepted: 06/28/2017] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE AND DESIGN Work in multiple organs has suggested that toll-like receptor 4 (TLR4) may play a role in insulin resistance. Additional studies have shown a negative role for TLR4 on retinal health. We have previously reported that β-adrenergic receptors can regulate both TLR4 signal transduction, as well as insulin signaling in the retina and in retinal endothelial cells. Thus, we hypothesized that TLR4 would regulate retinal insulin signaling. MATERIALS AND METHODS We used endothelial cell-specific TLR4 knockout mice, as well as TLR4-overexpressing mice for these studies. METHODS Western blotting and ELISA analyses were done for investigations of insulin receptor, insulin receptor substrate 1 (IRS-1) serine 307, and Akt phosphorylation, as well as cleaved caspase 3 levels in the mouse retina. RESULTS We found that loss of TLR4 led to increased insulin receptor and Akt phosphorylation, as well as decreased IRS-1Ser307 levels. In support of these results, TLR4 overexpression decreased insulin signaling and the cleavage of caspase 3. CONCLUSIONS Therefore, these results suggest that TLR4 plays a key role in insulin signaling in the retina. Reduction of TLR4 levels may be protective to the retina.
Collapse
Affiliation(s)
- Li Liu
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, 9314 Scott Hall, Detroit, MI, 48202, USA
| | - Youde Jiang
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, 9314 Scott Hall, Detroit, MI, 48202, USA
| | - Elizabeth Curtiss
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, 9314 Scott Hall, Detroit, MI, 48202, USA
| | - Ken-Ichiro Fukuchi
- Department of Cancer Biology and Pharmacology, University of Illinois College of Medicine at Peoria, Peoria, IL, USA
| | - Jena J Steinle
- Department of Anatomy and Cell Biology, Wayne State University School of Medicine, 9314 Scott Hall, Detroit, MI, 48202, USA. .,Department of Ophthalmology, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
158
|
Camandola S, Mattson MP. Toll-like receptor 4 mediates fat, sugar, and umami taste preference and food intake and body weight regulation. Obesity (Silver Spring) 2017; 25:1237-1245. [PMID: 28500692 PMCID: PMC5487280 DOI: 10.1002/oby.21871] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/08/2017] [Accepted: 04/06/2017] [Indexed: 12/17/2022]
Abstract
OBJECTIVE Immune and inflammatory pathways play important roles in the pathogenesis of metabolic disorders. This study investigated the role of toll-like receptor 4 (TLR4) in orosensory detection of dietary lipids and sugars. METHODS Taste preferences of TLR4 knockout (KO) and wild-type (WT) male mice under a standard and a high-fat, high-sugar diet were assessed with two-bottle tests. Gene expression of taste signaling molecules was analyzed in the tongue epithelium. The role of TLR4 in food intake and weight gain was investigated in TLR4 KO and WT mice fed a high-fat and high-sugar diet for 12 weeks. RESULTS Compared to WT mice, TLR4 KO mice showed reduced preference for lipids, sugars, and umami in a two-bottle preference test. The altered taste perception was associated with decreased levels of key taste regulatory molecules in the tongue epithelium. TLR4 KO mice on a high-fat and high-sugar diet consumed less food and drink, resulting in diminished weight gain. CONCLUSIONS TLR4 signaling promotes ingestion of sugar and fat by a mechanism involving increased preference for such obesogenic foods.
Collapse
Affiliation(s)
- Simonetta Camandola
- Laboratory of Neurosciences, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
| | - Mark P. Mattson
- Laboratory of Neurosciences, National Institute on Aging, Intramural Research Program, Baltimore, MD, USA
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
159
|
Balistreri CR, Ruvolo G, Lio D, Madonna R. Toll-like receptor-4 signaling pathway in aorta aging and diseases: "its double nature". J Mol Cell Cardiol 2017; 110:38-53. [PMID: 28668304 DOI: 10.1016/j.yjmcc.2017.06.011] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 06/20/2017] [Accepted: 06/27/2017] [Indexed: 12/20/2022]
Abstract
Recent advances in the field of innate immunity have revealed a complex role of innate immune signaling pathways in both tissue homeostasis and disease. Among them, the Toll-like receptor 4 (TLR-4) pathways has been linked to various pathophysiological conditions, such as cardiovascular diseases (CVDs). This has been interrogated by developing multiple laboratory tools that have shown in animal models and clinical conditions, the involvement of the TLR-4 signaling pathway in the pathophysiology of different CVDs, such as atherosclerosis, ischemic heart disease, heart failure, ischemia-reperfusion injury and aorta aneurysm. Among these, aorta aneurysm, a very complex pathological condition with uncertain etiology and fatal complications (i.e. dissection and rupture), has been associated with the occurrence of high risk cardiovascular conditions, including thrombosis and embolism. In this review, we discuss the possible role of TLR-4 signaling pathway in the development of aorta aneurysm, considering the emerging evidence from ongoing investigations. Our message is that emphasizing the role of TLR-4 signaling pathway in aorta aneurysm may serve as a starting point for future studies, leading to a better understanding of the pathophysiological basis and perhaps the effective treatment of this difficult human disease.
Collapse
Affiliation(s)
- Carmela Rita Balistreri
- Department of Pathobiology and Medical Biotechnologies, University of Palermo, Corso Tukory 211, 90134 Palermo, Italy.
| | - Giovanni Ruvolo
- Department of Cardiac Surgery, University of Rome 'Tor Vergata', Rome, Italy
| | - Domenico Lio
- Department of Pathobiology and Medical Biotechnologies, University of Palermo, Corso Tukory 211, 90134 Palermo, Italy
| | - Rosalinda Madonna
- Heart Failure Research, Texas Heart Institute, St. Luke's Episcopal Hospital, Houston, TX, United States; Department of Internal Medicine, Cardiology, The University of Texas Health Science Center at Houston, Houston, TX, United States; Center of Aging Sciences and Translational Medicine - CESI-Met and Institute of Cardiology, Department of Neurosciences, Imaging and Clinical Sciences "G. D'Annunzio" University, 66100 Chieti, Italy
| |
Collapse
|
160
|
Garbossa SG, Folli F. Vitamin D, sub-inflammation and insulin resistance. A window on a potential role for the interaction between bone and glucose metabolism. Rev Endocr Metab Disord 2017; 18:243-258. [PMID: 28409320 DOI: 10.1007/s11154-017-9423-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Vitamin D is a key hormone involved in the regulation of calcium/phosphorous balance and recently it has been implicated in the pathogenesis of sub-inflammation, insulin resistance and obesity. The two main forms of vitamin D are cholecalciferol (Vitamin D3) and ergocalciferol (Vitamin D2): the active form (1,25-dihydroxyvitamin D) is the result of two hydroxylations that take place in liver, kidney, pancreas and immune cells. Vitamin D increases the production of some anti-inflammatory cytokines and reduces the release of some pro-inflammatory cytokines. Low levels of Vitamin D are also associated with an up-regulation of TLRs expression and a pro-inflammatory state. Regardless of the effect on inflammation, Vitamin D seems to directly increase insulin sensitivity and secretion, through different mechanisms. Considering the importance of low grade chronic inflammation in metabolic syndrome, obesity and diabetes, many authors hypothesized the involvement of this nutrient/hormone in the pathogenesis of these diseases. Vitamin D status could alter the balance between pro and anti-inflammatory cytokines and thus affect insulin action, lipid metabolism and adipose tissue function and structure. Numerous studies have shown that Vitamin D concentrations are inversely associated with pro-inflammatory markers, insulin resistance, glucose intolerance and obesity. Interestingly, some longitudinal trials suggested also an inverse association between vitamin D status and incident type 2 diabetes mellitus. However, vitamin D supplementation in humans showed controversial effects: with some studies demonstrating improvements in insulin sensitivity, glucose and lipid metabolism while others showing no beneficial effect on glycemic control and on inflammation. In conclusion, although the evidences of a significant role of Vitamin D on inflammation, insulin resistance and insulin secretion in the pathogenesis of obesity, metabolic syndrome and type 2 diabetes, its potential function in treatment and prevention of type 2 diabetes mellitus is unclear. Encouraging results have emerged from Vitamin D supplementation trials on patients at risk of developing diabetes and further studies are needed to fully explore and understand its clinical applications.
Collapse
Affiliation(s)
- Stefania Giuliana Garbossa
- Department of Internal Medicine, Azienda Socio Sanitaria Territoriale Santi Paolo e Carlo, Ospedale San Paolo, Via A. Di Rudini 8, Milan, 20142, Italy
- Program in Nutritional Science, Universitá degli Studi di Milano, Milan, Italy
| | - Franco Folli
- Department of Internal Medicine, Azienda Socio Sanitaria Territoriale Santi Paolo e Carlo, Ospedale San Paolo, Via A. Di Rudini 8, Milan, 20142, Italy.
- Endocrinology and Metabolism, Department of Health Science, Universitá degli Studi di Milano, San Paolo Hospital, Room #737, Via A. Di Rudiní 8, 20142, Milan, Italy.
- Department of Medicine, Diabetes Division, University of Texas Health System, 7703 Floyd Curl Drive, San Antonio, 78229, TX, USA.
| |
Collapse
|
161
|
Ramos-Lobo AM, Donato J. The role of leptin in health and disease. Temperature (Austin) 2017; 4:258-291. [PMID: 28944270 DOI: 10.1080/23328940.2017.1327003] [Citation(s) in RCA: 88] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/27/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Leptin is a master regulator of energy balance and body adiposity. Additionally, leptin exerts important control on glucose homeostasis, thermogenesis, autonomic nervous system and neuroendocrine axes. In metabolic diseases, such as obesity and diabetes mellitus, leptin signaling may be compromised, indicating the important role of this hormone in the etiology and pathophysiological manifestations of these conditions. In the present manuscript, we reviewed important concepts of leptin signaling, as well as about the effects of leptin on several biologic functions. We also discussed the possible therapeutic use of leptin administration and how our current obesogenic environment contributes to the development of leptin resistance. Our objective was to provide a comprehensive and state-of-the-art review about the importance of leptin to maintain the homeostasis and during pathological conditions.
Collapse
Affiliation(s)
- Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
162
|
Bando M, Iwakura H, Ueda Y, Ariyasu H, Inaba H, Furukawa Y, Furuta H, Nishi M, Akamizu T. IL-1β directly suppress ghrelin mRNA expression in ghrelin-producing cells. Mol Cell Endocrinol 2017; 447:45-51. [PMID: 28237719 DOI: 10.1016/j.mce.2017.02.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Revised: 02/20/2017] [Accepted: 02/20/2017] [Indexed: 02/05/2023]
Abstract
In animal models, ghrelin production is suppressed by LPS administration. To elucidate the detailed molecular mechanisms involved in the phenomenon, we investigated the effects of LPS and LPS-inducible cytokines, including TNF-α, IL-1β, and IL-6, on the expression of ghrelin in the ghrelin-producing cell line MGN3-1. These cells expressed IL-1R, and IL-1β significantly suppressed ghrelin mRNA levels. The suppressive effects of IL-1β were attenuated by knockdown of IKKβ, suggesting the involvement of the NF-κB pathway. These results suggested that IL-1β is a major regulator of ghrelin expression during inflammatory processes.
Collapse
Affiliation(s)
- Mika Bando
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Hiroshi Iwakura
- The First Department of Medicine, Wakayama Medical University, Japan.
| | - Yoko Ueda
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Hiroyuki Ariyasu
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Hidefumi Inaba
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Yasushi Furukawa
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Hiroto Furuta
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Masahiro Nishi
- The First Department of Medicine, Wakayama Medical University, Japan
| | - Takashi Akamizu
- The First Department of Medicine, Wakayama Medical University, Japan
| |
Collapse
|
163
|
Zhao GN, Zhang P, Gong J, Zhang XJ, Wang PX, Yin M, Jiang Z, Shen LJ, Ji YX, Tong J, Wang Y, Wei QF, Wang Y, Zhu XY, Zhang X, Fang J, Xie Q, She ZG, Wang Z, Huang Z, Li H. Tmbim1 is a multivesicular body regulator that protects against non-alcoholic fatty liver disease in mice and monkeys by targeting the lysosomal degradation of Tlr4. Nat Med 2017; 23:742-752. [PMID: 28481357 DOI: 10.1038/nm.4334] [Citation(s) in RCA: 104] [Impact Index Per Article: 14.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Accepted: 04/07/2017] [Indexed: 02/08/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is an increasingly prevalent liver pathology that can progress from non-alcoholic fatty liver disease (NAFLD), and it is a leading cause of cirrhosis and hepatocellular carcinoma. There is currently no pharmacological therapy for NASH. Defective lysosome-mediated protein degradation is a key process that underlies steatohepatitis and a well-recognized drug target in a variety of diseases; however, whether it can serve as a therapeutic target for NAFLD and NASH remains unknown. Here we report that transmembrane BAX inhibitor motif-containing 1 (TMBIM1) is an effective suppressor of steatohepatitis and a previously unknown regulator of the multivesicular body (MVB)-lysosomal pathway. Tmbim1 expression in hepatocytes substantially inhibited high-fat diet-induced insulin resistance, hepatic steatosis and inflammation in mice. Mechanistically, Tmbim1 promoted the lysosomal degradation of toll-like receptor 4 by cooperating with the ESCRT endosomal sorting complex to facilitate MVB formation, and the ubiquitination of Tmbim1 by the E3 ubiquitin ligase Nedd4l was required for this process. We also found that overexpression of Tmbim1 in the liver effectively inhibited a severe form of NAFLD in mice and NASH progression in monkeys. Taken together, these findings could lead to the development of promising strategies to treat NASH by targeting MVB regulators to properly orchestrate the lysosome-mediated protein degradation of key mediators of the disease.
Collapse
Affiliation(s)
- Guang-Nian Zhao
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,College of Life Sciences, Wuhan University, Wuhan, China
| | - Peng Zhang
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Jun Gong
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,College of Life Sciences, Wuhan University, Wuhan, China
| | - Xiao-Jing Zhang
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pi-Xiao Wang
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Miao Yin
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Zhou Jiang
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,College of Life Sciences, Wuhan University, Wuhan, China
| | - Li-Jun Shen
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan-Xiao Ji
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Jingjing Tong
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,College of Life Sciences, Wuhan University, Wuhan, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yutao Wang
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qiao-Fang Wei
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yong Wang
- Institute of Model Animal of Wuhan University, Wuhan, China
| | - Xue-Yong Zhu
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China
| | - Xin Zhang
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,College of Life Sciences, Wuhan University, Wuhan, China
| | - Jing Fang
- Division of Cardiothoracic and Vascular Surgery, Heart-Lung Transplantation Center, Sino-Swiss Heart-Lung Transplantation Institute, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qingguo Xie
- Biomedical Engineering Department, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi-Gang She
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhihua Wang
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zan Huang
- Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,College of Life Sciences, Wuhan University, Wuhan, China
| | - Hongliang Li
- Medical Science Research Center, Zhongnan Hospital of Wuhan University, Wuhan, China.,Basic Medical School, Wuhan University, Wuhan, China.,Institute of Model Animal of Wuhan University, Wuhan, China.,Medical Research Institute, School of Medicine, Wuhan University, Wuhan, China.,Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
164
|
Contreras GA, Strieder-Barboza C, Raphael W. Adipose tissue lipolysis and remodeling during the transition period of dairy cows. J Anim Sci Biotechnol 2017; 8:41. [PMID: 28484594 PMCID: PMC5420123 DOI: 10.1186/s40104-017-0174-4] [Citation(s) in RCA: 97] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2016] [Accepted: 04/11/2017] [Indexed: 12/12/2022] Open
Abstract
Elevated concentrations of plasma fatty acids in transition dairy cows are significantly associated with increased disease susceptibility and poor lactation performance. The main source of plasma fatty acids throughout the transition period is lipolysis from adipose tissue depots. During this time, plasma fatty acids serve as a source of calories mitigating the negative energy balance prompted by copious milk synthesis and limited dry matter intake. Past research has demonstrated that lipolysis in the adipose organ is a complex process that includes not only the activation of lipolytic pathways in response to neural, hormonal, or paracrine stimuli, but also important changes in the structure and cellular distribution of the tissue in a process known as adipose tissue remodeling. This process involves an inflammatory response with immune cell migration, proliferation of the cellular components of the stromal vascular fraction, and changes in the extracellular matrix. This review summarizes current knowledge on lipolysis in dairy cattle, expands on the new field of adipose tissue remodeling, and discusses how these biological processes affect transition cow health and productivity.
Collapse
Affiliation(s)
- G Andres Contreras
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824 USA
| | - Clarissa Strieder-Barboza
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824 USA
| | - William Raphael
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, MI 48824 USA
| |
Collapse
|
165
|
Nutrients and Inflammatory Diseases. Mediators Inflamm 2017; 2017:6134909. [PMID: 28539705 PMCID: PMC5429948 DOI: 10.1155/2017/6134909] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 03/09/2017] [Indexed: 11/18/2022] Open
|
166
|
Saad MJA, Santos A, Prada PO. Linking Gut Microbiota and Inflammation to Obesity and Insulin Resistance. Physiology (Bethesda) 2017; 31:283-93. [PMID: 27252163 DOI: 10.1152/physiol.00041.2015] [Citation(s) in RCA: 421] [Impact Index Per Article: 60.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Obesity and insulin resistance are the major predisposing factors to comorbidities, such as Type 2 diabetes, nonalcoholic fatty liver disease, cardiovascular and neurodegenerative diseases, and several types of cancer. The prevalence of obesity is still increasing worldwide and now affects a large number of individuals. Here, we review the role of the gut microbiota in the pathophysiology of insulin resistance/obesity. The human intestine is colonized by ∼100 trillion bacteria, which constitute the gut microbiota. Studies have shown that lean and overweight rodents and humans may present differences in the composition of their intestinal flora. Over the past 10 years, data from different sources have established a causal link between the intestinal microbiota and obesity/insulin resistance. It is important to emphasize that diet-induced obesity promotes insulin resistance by mechanisms independent and dependent on gut microbiota. In this review, we present several mechanisms that contribute to explaining the link between intestinal flora and insulin resistance/obesity. The LPS from intestinal flora bacteria can induce a chronic subclinical inflammatory process and obesity, leading to insulin resistance through activation of TLR4. The reduction in circulating SCFA may also have an essential role in the installation of reduced insulin sensitivity and obesity. Other mechanisms include effects of bile acids, branched-chain amino acids (BCAA), and some other lesser-known factors. In the near future, this area should open new therapeutic avenues for obesity/insulin resistance and its comorbidities.
Collapse
Affiliation(s)
- M J A Saad
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, Brazil; and
| | - A Santos
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, Brazil; and
| | - P O Prada
- Department of Internal Medicine, State University of Campinas (UNICAMP), Campinas, Brazil; and School of Applied Sciences, State University of Campinas (UNICAMP), Limeira, Brazil
| |
Collapse
|
167
|
Norde MM, Oki E, Carioca AAF, Castro IA, Souza JMP, Marchioni DML, Fisberg RM, Rogero MM. Influence of toll-like receptor 4 gene variants and plasma fatty acid profile on systemic inflammation: A population-based cross-sectional study. Nutrition 2017; 35:106-111. [PMID: 28241976 DOI: 10.1016/j.nut.2016.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2016] [Revised: 09/30/2016] [Accepted: 11/10/2016] [Indexed: 02/06/2023]
Abstract
OBJECTIVE The aim of this study was to investigate the interaction of toll-like receptor 4 (TLR4) gene single nucleotide polymorphism (SNP) and plasma fatty acid (FA) profile in modulating risk for systemic inflammation. METHODS In all, 262 adult (19-59 y) participants of the Health Survey of São Paulo met the inclusion criteria. Anthropometric parameters, blood pressure, plasma inflammatory biomarker concentration, and fatty acid profile were measured and four SNPs of the TLR4 gene (rs4986790, rs4986791, rs11536889, and rs5030728) were genotyped. Multivariate cluster analysis was performed to stratify individuals based on levels of 11 plasma inflammatory biomarkers into two groups: inflammatory (INF) and noninflammatory (NINF). RESULTS No association was found between any of the SNPs studied and systemic inflammation. The INF cluster had higher palmitic acid levels (P = 0.039) and estimated stearoyl coenzyme A desaturase activity (P = 0.045) and lower polyunsaturated fatty acid (P = 0.011), ω-6 fatty acid (P = 0.018), arachidonic acid (P = 0.002) levels, and estimated δ-5 desaturase activity (P = 0.025) compared with the NINF cluster. Statistically significant interaction between rs11536889 and arachidonic acid/eicosapentaenoic acid (AA/EPA) ratio (P = 0.034) was found to increase the odds of belonging to the INF cluster when individuals had the variant allele C and were at the higher percentile of AA/EPA plasma ratio. CONCLUSION Plasma fatty acid profile modulated the odds of belonging to the INF cluster depending on genotypes of TRL4 gene polymorphisms.
Collapse
Affiliation(s)
| | - Erica Oki
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Antonio A F Carioca
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Inar A Castro
- Department of Food and Experimental Nutrition, Faculty of Pharmaceutical Sciences, University of São Paulo, Brazil
| | - José M P Souza
- Department of Epidemiology, School of Public Health, University of São Paulo, Brazil
| | - Dirce M L Marchioni
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Regina M Fisberg
- Nutrition Department, School of Public Health, University of São Paulo, Brazil
| | - Marcelo M Rogero
- Nutrition Department, School of Public Health, University of São Paulo, Brazil.
| |
Collapse
|
168
|
de Moraes ACF, Fernandes GR, da Silva IT, Almeida-Pititto B, Gomes EP, Pereira ADC, Ferreira SRG. Enterotype May Drive the Dietary-Associated Cardiometabolic Risk Factors. Front Cell Infect Microbiol 2017; 7:47. [PMID: 28280715 PMCID: PMC5322172 DOI: 10.3389/fcimb.2017.00047] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Accepted: 02/07/2017] [Indexed: 01/21/2023] Open
Abstract
Analyses of typical bacterial clusters in humans named enterotypes may facilitate understanding the host differences in the cardiometabolic profile. It stills unknown whether the three previously described enterotypes were present in populations living below the equator. We examined how the identification of enterotypes could be useful to explain the dietary associations with cardiometabolic risk factors in Brazilian subjects. In this cross-sectional study, a convenience sample of 268 adults (54.2% women) reported their dietary habits and had clinical and biological samples collected. In this study, we analyzed biochemical data and metagenomics of fecal microbiota (16SrRNA sequencing, V4 region). Continuous variables were compared using ANOVA, and categorical variables using chi-square test. Vsearch clustered the operational taxonomic units, and Silva Database provided the taxonomic signatures. Spearman coefficient was used to verify the correlation between bacteria abundances within each enterotype. One hundred subjects were classified as omnivore, 102 lacto-ovo-vegetarians, and 66 strict vegetarians. We found the same structure as the three previously described enterotypes: 111 participants were assigned to Bacteroides, 55 to Prevotella, and 102 to Ruminococcaceae enterotype. The Prevotella cluster contained higher amount of strict vegetarians individuals than the other enterotypes (40.0 vs. 20.7 and 20.6, p = 0.04). Subjects in this enterotype had a similar anthropometric profile but a lower mean LDL-c concentration than the Bacteroides enterotype (96 ± 23 vs. 109 ± 32 mg/dL, p = 0.04). We observed significant correlations between bacterial abundances and cardiometabolic risk factors, but coefficients differed depending on the enterotype. In Prevotella enterotype, Eubacterium ventriosum (r BMI = -0.33, p = 0.03, and r HDL-c = 0.33, p = 0.04), Akkermansia (r 2h glucose = -0.35, p = 0.02), Roseburia (r BMI = -0.36, p = 0.02 and r waist = -0.36, p = 0.02), and Faecalibacterium (r insulin = -0.35, p = 0.02) abundances were associated to better cardiometabolic profile. The three enterotypes previously described are present in Brazilians, supporting that those bacterial clusters are not population-specific. Diet-independent lower LDL-c levels in subjects from Prevotella than in other enterotypes suggest that a protective bacterial cluster in the former should be driving this association. Enterotypes seem to be useful to understand the impact of daily diet exposure on cardiometabolic risk factors. Prospective studies are needed to confirm their utility for predicting phenotypes in humans.
Collapse
Affiliation(s)
- Ana C F de Moraes
- Department of Epidemiology, School of Public Health, University of São Paulo São Paulo, Brazil
| | | | - Isis T da Silva
- Department of Epidemiology, School of Public Health, University of São Paulo São Paulo, Brazil
| | | | - Everton P Gomes
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), University of São Paulo Medical School São Paulo, Brazil
| | - Alexandre da Costa Pereira
- Laboratory of Genetics and Molecular Cardiology, Heart Institute (Incor), University of São Paulo Medical School São Paulo, Brazil
| | - Sandra R G Ferreira
- Department of Epidemiology, School of Public Health, University of São Paulo São Paulo, Brazil
| |
Collapse
|
169
|
Lei Y, Li X, Yuan F, Liu L, Zhang J, Yang Y, Zhao J, Han Y, Ren J, Fu X. Toll-like receptor 4 ablation rescues against paraquat-triggered myocardial dysfunction: Role of ER stress and apoptosis. ENVIRONMENTAL TOXICOLOGY 2017; 32:656-668. [PMID: 27442881 DOI: 10.1002/tox.22267] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 03/18/2016] [Accepted: 03/20/2016] [Indexed: 06/06/2023]
Abstract
Paraquat is a nitrogen herbicide imposing severe organ toxicity in human leading to acute lung injury and heart failure. The present study was designed to examine the impact of ablation of the innate proinflammatory mediator toll-like receptor 4 (TLR4) in paraquat-induced cardiac contractile dysfunction and the underlying mechanisms involved with a focus on endoplasmic reticulum (ER) stress and apoptosis. Adult male wild-type (WT) and TLR4 knockout (TLR4-/- ) mice were challenged with paraquat (45 mg/kg, i.p.) for 48 h prior to the assessment of myocardial and cardiomyocyte sarcomere function, ER stress, apoptosis and inflammation. Acute paraquat challenge exerted myocardial functional and geometric alterations including enlarged left ventricular end systolic diameter (LVESD), reduced fractional shortening, decreased sarcomere shortening, maximal velocities of sarcomere shortening and relengthening associated with unchanged LV posterior wall thickness, septal thickness, LV end diastolic diameter (LVEDD), heart rate, sarcomere length, time-to-peak shortening and time-to-90% relengthening. Although TLR4 ablation did not affect mechanical properties in the heart, it significantly attenuated or ablated paraquat-induced cardiac contractile anomalies. Moreover, paraquat imposed overt ER stress, apoptosis and inflammation as evidenced by upregulation of Bip, CHOP, Caspase-3, -9, Bax, Bad, and IL-1β, phosphorylation of PERK, eIF2α and IΚB, as well as activation of the stress molecules ERK and p38, with unchanged Caspase-8, Bcl2, TNF-α, p53, HMGB1, MyD88 and phosphorylation of Akt, GSK3β and JNK, the effects of which were attenuated or negated by TLR4 knockout. Taken together, our results suggested that TLR4 ablation alleviated paraquat-induced myocardial contractile dysfunction possibly through attenuation of ER stress, apoptosis and inflammation. © 2016 Wiley Periodicals, Inc. Environ Toxicol 32: 656-668, 2017.
Collapse
Affiliation(s)
- Yonghong Lei
- Institute of Wound Healing and Cell Biology Laboratory, the First Affiliated Hospital, Beijing, 100048, China
| | - Xue Li
- Cardiovascular Department, Tangdu Hospital, Xi'an, 710038, China
| | - Fang Yuan
- Department of Orthopedics, General Hospital of Chinese PLA, Beijing, 100853, China
| | - Lu Liu
- Department of Clinical Nutrition, General Hospital of Chinese PLA, Beijing, 100853, China
| | - Juan Zhang
- Cardiovascular Department, Tangdu Hospital, Xi'an, 710038, China
| | - Yanping Yang
- Cardiovascular Department, Tangdu Hospital, Xi'an, 710038, China
| | - Jieqiong Zhao
- Cardiovascular Department, Tangdu Hospital, Xi'an, 710038, China
| | - Yan Han
- Department of Plastic Surgery, General Hospital of Chinese PLA, Beijing, 100853, China
| | - Jun Ren
- Department of Cardiology, Fudan University, Zhongshan Hospital, Shanghai, 210032, China
| | - Xiaobing Fu
- Institute of Wound Healing and Cell Biology Laboratory, the First Affiliated Hospital, Beijing, 100048, China
| |
Collapse
|
170
|
TLR4 knockout attenuated high fat diet-induced cardiac dysfunction via NF-κB/JNK-dependent activation of autophagy. Biochim Biophys Acta Mol Basis Dis 2017; 1863:2001-2011. [PMID: 28108421 DOI: 10.1016/j.bbadis.2017.01.010] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Revised: 01/09/2017] [Accepted: 01/12/2017] [Indexed: 12/19/2022]
Abstract
Obesity is commonly associated with a low grade systemic inflammation, which may contribute to the onset and development of myocardial remodeling and contractile dysfunction. Toll-like receptor 4 (TLR4) plays an important role in innate immunity and inflammation although its role in high fat diet-induced obesity cardiac dysfunction remains elusive. This study was designed to examine the effect of TLR4 ablation on high fat diet intake-induced cardiac anomalies, if any, and underlying mechanism(s) involved. Wild-type (WT) and TLR4 knockout mice were fed normal or high fat (60% calorie from fat) diet for 12weeks prior to assessment of mechanical and intracellular Ca2+ properties. The inflammatory signaling proteins (TLR4, NF-κB, and JNK) and autophagic markers (Atg5, Atg12, LC3B and p62) were evaluated. Our results revealed that high fat diet intake promoted obesity, marked decrease in fractional shortening, and cardiomyocyte contractile capacity with dampened intracellular Ca2+ release and clearance, elevated ROS generation and oxidative stress as measured by aconitase activity, the effects of which were significantly attenuated by TLR4 knockout. In addition, high fat intake downregulated levels of Atg5, Atg12 and LC3B, while increasing p62 accumulation. TLR4 knockout itself did not affect Atg5, Atg12, LC3B and p62 levels while it reconciled high fat diet intake-induced changes in autophagy. In addition, TLR4 knockout alleviated high fat diet-induced phosphorylation of IKKβ, JNK and mTOR. In vitro study revealed that palmitic acid suppressed cardiomyocyte contractile function, the effect of which was inhibited the TLR4 inhibitor CLI-095, the JNK inhibitor AS601245 or the NF-κB inhibitor Celastrol. Taken together, these data showed that TLR4 knockout ameliorated high fat diet-induced cardiac contractile and intracellular Ca2+ anomalies through inhibition of inflammation and ROS, possibly through a NF-κB/JNK-dependent activation of autophagy. This article is part of a Special Issue entitled: Genetic and epigenetic control of heart failure - edited by Jun Ren & Megan Yingmei Zhang.
Collapse
|
171
|
Pascoal LB, Bombassaro B, Ramalho AF, Coope A, Moura RF, Correa-da-Silva F, Ignacio-Souza L, Razolli D, de Oliveira D, Catharino R, Velloso LA. Resolvin RvD2 reduces hypothalamic inflammation and rescues mice from diet-induced obesity. J Neuroinflammation 2017; 14:5. [PMID: 28086928 PMCID: PMC5234140 DOI: 10.1186/s12974-016-0777-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/09/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Diet-induced hypothalamic inflammation is an important mechanism leading to dysfunction of neurons involved in controlling body mass. Studies have shown that polyunsaturated fats can reduce hypothalamic inflammation. Here, we evaluated the presence and function of RvD2, a resolvin produced from docosahexaenoic acid, in the hypothalamus of mice. METHODS Male Swiss mice were fed either chow or a high-fat diet. RvD2 receptor and synthetic enzymes were evaluated by real-time PCR and immunofluorescence. RvD2 was determined by mass spectrometry. Dietary and pharmacological approaches were used to modulate the RvD2 system in the hypothalamus, and metabolic phenotype consequences were determined. RESULTS All enzymes involved in the synthesis of RvD2 were detected in the hypothalamus and were modulated in response to the consumption of dietary saturated fats, leading to a reduction of hypothalamic RvD2. GPR18, the receptor for RvD2, which was detected in POMC and NPY neurons, was also modulated by dietary fats. The substitution of saturated by polyunsaturated fats in the diet resulted in increased hypothalamic RvD2, which was accompanied by reduced body mass and improved glucose tolerance. The intracerebroventricular treatment with docosahexaenoic acid resulted in increased expression of the RvD2 synthetic enzymes, increased expression of anti-inflammatory cytokines and improved metabolic phenotype. Finally, intracerebroventricular treatment with RvD2 resulted in reduced adiposity, improved glucose tolerance and increased hypothalamic expression of anti-inflammatory cytokines. CONCLUSIONS Thus, RvD2 is produced in the hypothalamus, and its receptor and synthetic enzymes are modulated by dietary fats. The improved metabolic outcomes of RvD2 make this substance an attractive approach to treat obesity.
Collapse
Affiliation(s)
- Livia B Pascoal
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Albina F Ramalho
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Andressa Coope
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Rodrigo F Moura
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Felipe Correa-da-Silva
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Leticia Ignacio-Souza
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Daniela Razolli
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Diogo de Oliveira
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Rodrigo Catharino
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil.
| |
Collapse
|
172
|
Kuga GK, Botezelli JD, Gaspar RC, Gomes RJ, Pauli JR, Leme JACDA. Hippocampal insulin signaling and neuroprotection mediated by physical exercise in Alzheimer´s Disease. MOTRIZ: REVISTA DE EDUCACAO FISICA 2017. [DOI: 10.1590/s1980-6574201700si0008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|
173
|
Abstract
PURPOSE OF REVIEW Physical and cognitive frailty are interrelated and synergistic syndromes more frequently seen in old age, which represent intermediate stages between aging successfully and disability. Poor nutrition is a fundamental determinant for both conditions, while various dietary components are proposed to prevent and/or improve them. This updated review discusses the possible influence of nutritional factors on cognitive frailty and its potential mediators. RECENT FINDINGS Oxidative stress, low-grade systemic inflammation, neuroinflammation, and altered autophagy, all associated with obesity, metabolic syndrome and insulin resistance, are proposed mechanisms to explain the influence of nutrition on cognitive health. Even if no single food or supplement has definitively demonstrated to affect physical frailty and cognitive impairment, combining various dietary and lifestyle components in the Mediterranean dietary pattern has shown benefit. SUMMARY Cognitive frailty is a potential useful construct for the early detection of cognitive impairment and physical frailty, in order to implement timely interventions. Validation of this construct is eagerly needed. Nutritional status is a fundamental part of physical frailty, and potentially important in the prevention of cognitive decline. Identifying and treating protein/calorie and individual nutrients insufficiency is mandatory in all older adults. Conversely, overeating in middle age has been associated with cognitive decline in older age. A lifelong balance diet, such as the Mediterranean diet, combined with regular physical and mental exercise, is perhaps the best preventive strategy against cognitive decline in old age.
Collapse
Affiliation(s)
- Ligia J Dominguez
- Geriatric Unit, Department of Internal Medicine and Geriatrics, University of Palermo, Palermo, Italy
| | | |
Collapse
|
174
|
Cavadas C, Aveleira CA, Souza GFP, Velloso LA. The pathophysiology of defective proteostasis in the hypothalamus - from obesity to ageing. Nat Rev Endocrinol 2016; 12:723-733. [PMID: 27388987 DOI: 10.1038/nrendo.2016.107] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Hypothalamic dysfunction has emerged as an important mechanism involved in the development of obesity and its comorbidities, as well as in the process of ageing and age-related diseases, such as type 2 diabetes mellitus, hypertension and Alzheimer disease. In both obesity and ageing, inflammatory signalling is thought to coordinate many of the cellular events that lead to hypothalamic neuronal dysfunction. This process is triggered by the activation of signalling via the toll-like receptor 4 pathway and endoplasmic reticulum stress, which in turn results in intracellular inflammatory signalling. However, the process that connects inflammation with neuronal dysfunction is complex and includes several regulatory mechanisms that ultimately control the homeostasis of intracellular proteins and organelles (also known as 'proteostasis'). This Review discusses the evidence for the key role of proteostasis in the control of hypothalamic neurons and the involvement of this process in regulating whole-body energy homeostasis and lifespan.
Collapse
Affiliation(s)
- Cláudia Cavadas
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
- Faculty of Pharmacy, University of Coimbra, Coimbra, 3004-504, Portugal
| | - Célia A Aveleira
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, 3004-504, Portugal
| | - Gabriela F P Souza
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, 1308-970, Brazil
| | - Lício A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, University of Campinas, Campinas, 1308-970, Brazil
| |
Collapse
|
175
|
Sokolova M, Vinge LE, Alfsnes K, Olsen MB, Eide L, Kaasbøll OJ, Attramadal H, Torp MK, Fosshaug LE, Rashidi A, Lien E, Finsen AV, Sandanger Ø, Aukrust P, Ranheim T, Yndestad A. Palmitate promotes inflammatory responses and cellular senescence in cardiac fibroblasts. Biochim Biophys Acta Mol Cell Biol Lipids 2016; 1862:234-245. [PMID: 27845246 DOI: 10.1016/j.bbalip.2016.11.003] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 10/30/2016] [Accepted: 11/04/2016] [Indexed: 12/29/2022]
Abstract
Palmitate triggers inflammatory responses in several cell types, but its effects on cardiac fibroblasts are at present unknown. The aims of the study were to (1) assess the potential of palmitate to promote inflammatory signaling in cardiac fibroblasts through TLR4 and the NLRP3 inflammasome and (2) characterize the cellular phenotype of cardiac fibroblasts exposed to palmitate. We examined whether palmitate induces inflammatory responses in cardiac fibroblasts from WT, NLRP3-/- and ASC-/-mice (C57BL/6 background). Exposure to palmitate caused production of TNF, IL-6 and CXCL2 via TLR4 activation. NLRP3 inflammasomes are activated in a two-step manner. Whereas palmitate did not prime the NLRP3 inflammasome, it induced activation in LPS-primed cardiac fibroblasts as indicated by IL-1β, IL-18 production and NLRP3-ASC co-localization. Palmitate-induced NLRP3 inflammasome activation in LPS-primed cardiac fibroblasts was associated with reduced AMPK activity, mitochondrial reactive oxygen species production and mitochondrial dysfunction. The cardiac fibroblast phenotype caused by palmitate, in an LPS and NLRP3 independent manner, was characterized by decreased cellular proliferation, contractility, collagen and MMP-2 expression, as well as increased senescence-associated β-galactosidase activity, and consistent with a state of cellular senescence. This study establishes that in vitro palmitate exposure of cardiac fibroblasts provides inflammatory responses via TLR4 and NLRP3 inflammasome activation. Palmitate also modulates cardiac fibroblast functionality, in a NLRP3 independent manner, resulting in a phenotype related to cellular senescence. These effects of palmitate could be of importance for myocardial dysfunction in obese and diabetic patients.
Collapse
Affiliation(s)
- Marina Sokolova
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, University of Oslo, Norway; KG Jebsen Center for Inflammation Research, University of Oslo, Norway; Center for Heart Failure Research, University of Oslo, Norway
| | - Leif Erik Vinge
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Center for Heart Failure Research, University of Oslo, Norway; Department of Cardiology, Oslo University Hospital Rikshospitalet, Norway; Department of Medicine, Diakonhjemmet Hospital, Oslo, Norway
| | - Katrine Alfsnes
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; KG Jebsen Center for Inflammation Research, University of Oslo, Norway
| | - Maria Belland Olsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, University of Oslo, Norway; KG Jebsen Center for Inflammation Research, University of Oslo, Norway; Center for Heart Failure Research, University of Oslo, Norway
| | - Lars Eide
- Institute of Clinical Medicine, University of Oslo, Norway; Department of Medical Biochemistry, Oslo University Hospital Rikshospitalet, Norway
| | - Ole Jørgen Kaasbøll
- Center for Heart Failure Research, University of Oslo, Norway; Institute of Surgical Research, Oslo University Hospital Rikshospitalet, Norway
| | - Håvard Attramadal
- Center for Heart Failure Research, University of Oslo, Norway; Institute of Surgical Research, Oslo University Hospital Rikshospitalet, Norway
| | - May-Kristin Torp
- Center for Heart Failure Research, University of Oslo, Norway; Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Linn E Fosshaug
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; KG Jebsen Center for Inflammation Research, University of Oslo, Norway; Center for Heart Failure Research, University of Oslo, Norway; Department of Medicine, Diakonhjemmet Hospital, Oslo, Norway
| | - Azita Rashidi
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; KG Jebsen Center for Inflammation Research, University of Oslo, Norway
| | - Egil Lien
- Division of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA, USA; Centre of Molecular Inflammation Research, NTNU, Trondheim, Norway
| | - Alexandra Vanessa Finsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; KG Jebsen Center for Inflammation Research, University of Oslo, Norway; Center for Heart Failure Research, University of Oslo, Norway
| | - Øystein Sandanger
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; KG Jebsen Center for Inflammation Research, University of Oslo, Norway; Center for Heart Failure Research, University of Oslo, Norway
| | - Pål Aukrust
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, University of Oslo, Norway; KG Jebsen Center for Inflammation Research, University of Oslo, Norway; Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Norway
| | - Trine Ranheim
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, University of Oslo, Norway; KG Jebsen Center for Inflammation Research, University of Oslo, Norway; Center for Heart Failure Research, University of Oslo, Norway
| | - Arne Yndestad
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, Norway; Institute of Clinical Medicine, University of Oslo, Norway; KG Jebsen Center for Inflammation Research, University of Oslo, Norway; Center for Heart Failure Research, University of Oslo, Norway.
| |
Collapse
|
176
|
Chen YH, Hu XG, Zhou Y, Yu Z, Fu L, Zhang GB, Bo QL, Wang H, Zhang C, Xu DX. Obeticholic Acid Protects against Lipopolysaccharide-Induced Fetal Death and Intrauterine Growth Restriction through Its Anti-Inflammatory Activity. THE JOURNAL OF IMMUNOLOGY 2016; 197:4762-4770. [PMID: 27821667 DOI: 10.4049/jimmunol.1601331] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 10/08/2016] [Indexed: 12/16/2022]
Abstract
Farnesoid X receptor (FXR) is expressed in human and rodent placentas. Nevertheless, its function remains obscure. This study investigated the effects of obeticholic acid (OCA), a novel synthetic FXR agonist, on LPS-induced fetal death and intrauterine growth restriction. All pregnant mice except controls were i.p. injected with LPS (100 μg/kg) daily from gestational day (GD) 15 to GD17. Some pregnant mice were orally administered with OCA (5 mg/kg) daily from GD13 to GD17. As expected, placental FXR signaling was activated by OCA. OCA pretreatment protected against LPS-induced fetal death. In addition, OCA pretreatment alleviated LPS-induced reduction of fetal weight and crown-rump length. Additional experiments showed that OCA inhibited LPS-evoked TNF-α in maternal serum and amniotic fluid. Moreover, OCA significantly attenuated LPS-induced upregulation of placental proinflammatory genes including Tnf-α, Il-1β, IL-6, Il-12, Mip-2, Kc, and Mcp-1 By contrast, OCA elevated anti-inflammatory cytokine IL-10 in maternal serum, amniotic fluid, and placenta. Further analysis showed that OCA blocked nuclear translocation of NF-κB p65 and p50 subunits in trophoblast giant cells of the labyrinth zone. These results provide a mechanistic explanation for placental FXR-mediated anti-inflammatory activity. Overall, this study provides evidence for roles of FXR as an important regulator of placental inflammation.
Collapse
Affiliation(s)
- Yuan-Hua Chen
- Department of Toxicology, Anhui Medical University, Hefei 230032, China.,Laboratory of Environmental Toxicology, Hefei 230032, China; and.,Department of Histology and Embryology, Anhui Medical University, Hefei 230032, China
| | - Xiao-Guang Hu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China.,Laboratory of Environmental Toxicology, Hefei 230032, China; and
| | - Yan Zhou
- Department of Toxicology, Anhui Medical University, Hefei 230032, China.,Laboratory of Environmental Toxicology, Hefei 230032, China; and
| | - Zhen Yu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China.,Laboratory of Environmental Toxicology, Hefei 230032, China; and
| | - Lin Fu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Gui-Bin Zhang
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - Qing-Li Bo
- Department of Toxicology, Anhui Medical University, Hefei 230032, China.,Laboratory of Environmental Toxicology, Hefei 230032, China; and
| | - Hua Wang
- Department of Toxicology, Anhui Medical University, Hefei 230032, China.,Laboratory of Environmental Toxicology, Hefei 230032, China; and
| | - Cheng Zhang
- Department of Toxicology, Anhui Medical University, Hefei 230032, China
| | - De-Xiang Xu
- Department of Toxicology, Anhui Medical University, Hefei 230032, China; .,Laboratory of Environmental Toxicology, Hefei 230032, China; and
| |
Collapse
|
177
|
Vinuesa A, Pomilio C, Menafra M, Bonaventura MM, Garay L, Mercogliano MF, Schillaci R, Lux Lantos V, Brites F, Beauquis J, Saravia F. Juvenile exposure to a high fat diet promotes behavioral and limbic alterations in the absence of obesity. Psychoneuroendocrinology 2016; 72:22-33. [PMID: 27337091 DOI: 10.1016/j.psyneuen.2016.06.004] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 06/03/2016] [Accepted: 06/04/2016] [Indexed: 01/09/2023]
Abstract
The incidence of metabolic disorders including obesity, type 2 diabetes and metabolic syndrome have seriously increased in the last decades. These diseases - with growing impact in modern societies - constitute major risk factors for neurodegenerative disorders such as Alzheimer's disease (AD), sharing insulin resistance, inflammation and associated cognitive impairment. However, cerebral cellular and molecular pathways involved are not yet clearly understood. Thus, our aim was to study the impact of a non-severe high fat diet (HFD) that resembles western-like alimentary habits, particularly involving juvenile stages where the brain physiology and connectivity are in plain maturation. To this end, one-month-old C57BL/6J male mice were given either a control diet or HFD during 4 months. Exposure to HFD produced metabolic alterations along with changes in behavioral and central parameters, in the absence of obesity. Two-month-old HFD mice showed increased glycemia and plasmatic IL1β but these values normalized at the end of the HFD protocol at 5 months of age, probably representing an acute response that is compensated at later stages. After four months of HFD exposure, mice presented dyslipidemia, increased Lipoprotein-associated phospholipase A2 (Lp-PLA2) activity, hepatic insulin resistance and inflammation. Alterations in the behavioral profile of the HFD group were shown by the impediment in nest building behavior, deficiencies in short and mid-term spatial memories, anxious and depressive- like behavior. Regarding the latter disruptions in emotional processing, we found an increased neural activity in the amygdala, shown by a greater number of c-Fos+ nuclei. We found that hippocampal adult neurogenesis was decreased in HFD mice, showing diminished cell proliferation measured as Ki67+ cells and neuronal differentiation in SGZ by doublecortin labeling. These phenomena were accompanied by a neuroinflammatory and insulin-resistant state in the hippocampus, depicted by a reactive phenotype in Iba1+ microglia cells (increased in number and soma size) and an impaired response to insulin given by decreased phosphorylated Akt levels and increased levels of inhibitory phosphorylation of IRS1. Our data portray a set of alterations in behavioral and neural parameters as a consequence of an early-life exposure to a quite moderate high fat diet, many of which can resemble AD-related features. These results highly emphasize the need to study how metabolic and neurodegenerative disorders are interrelated in deep, thus allowing the finding of successful preventive and therapeutic approaches.
Collapse
Affiliation(s)
- Angeles Vinuesa
- Neurobiology of Aging, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina; Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Carlos Pomilio
- Neurobiology of Aging, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina; Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Martin Menafra
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | | | - Laura Garay
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | | | - Roxana Schillaci
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Victoria Lux Lantos
- Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Fernando Brites
- Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, Buenos Aires, Argentina
| | - Juan Beauquis
- Neurobiology of Aging, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina; Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina
| | - Flavia Saravia
- Neurobiology of Aging, Departamento de Química Biológica, Facultad de Ciencias Exactas y Naturales, Universidad de Buenos Aires, Argentina; Instituto de Biología y Medicina Experimental, CONICET, Buenos Aires, Argentina.
| |
Collapse
|
178
|
Miele L, Giorgio V, Alberelli MA, De Candia E, Gasbarrini A, Grieco A. Impact of Gut Microbiota on Obesity, Diabetes, and Cardiovascular Disease Risk. Curr Cardiol Rep 2016; 17:120. [PMID: 26497040 DOI: 10.1007/s11886-015-0671-z] [Citation(s) in RCA: 104] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Gut microbiota has been recently established to have a contributory role in the development of cardiometabolic disorders, such as atherosclerosis, obesity, and type 2 diabetes. Growing interest has focused on the modulation of gut microbiota as a therapeutic strategy in cardiovascular diseases and metabolic disorders. In this paper, we have reviewed the impact of gut microbiota on metabolic disorders and cardiovascular disease risk, focusing on the newest findings in this field.
Collapse
Affiliation(s)
- Luca Miele
- Institute of Internal Medicine, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy. .,Clinical Division of Internal Medicine, Gastroenterology and Liver Unit, Complesso Integrato Columbus Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| | - Valentina Giorgio
- Pediatric Department, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| | - Maria Adele Alberelli
- Institute of Internal Medicine, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| | - Erica De Candia
- Institute of Internal Medicine, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| | - Antonio Gasbarrini
- Institute of Internal Medicine, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| | - Antonio Grieco
- Institute of Internal Medicine, Policlinico Gemelli Hospital, Catholic University of Sacred Heart of Rome, Rome, Italy.
| |
Collapse
|
179
|
Araujo EP, Moraes JC, Cintra DE, Velloso LA. MECHANISMS IN ENDOCRINOLOGY: Hypothalamic inflammation and nutrition. Eur J Endocrinol 2016; 175:R97-R105. [PMID: 27006108 DOI: 10.1530/eje-15-1207] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/22/2016] [Indexed: 12/29/2022]
Abstract
Selected subpopulations of hypothalamic neurons play important roles in the regulation of whole body energy homeostasis. Studies have shown that the saturated fats present in large amounts in western diets can activate an inflammatory response in the hypothalamus, affecting the capacity of such neurons to respond appropriately to satiety and adipostatic signals. In the first part of this review, we will explore the mechanisms behind saturated fatty acid-induced hypothalamic dysfunction. Next, we will present and discuss recent studies that have identified the mechanisms that mediate some of the anti-inflammatory actions of unsaturated fatty acids in the hypothalamus and the potential for exploring these mechanisms to prevent or treat obesity.
Collapse
Affiliation(s)
- Eliana P Araujo
- Laboratory of Cell SignalingUniversity of Campinas, Campinas, São Paulo, Brazil Faculty of NursingUniversity of Campinas, Campinas, São Paulo, Brazil
| | - Juliana C Moraes
- Laboratory of Cell SignalingUniversity of Campinas, Campinas, São Paulo, Brazil
| | - Dennys E Cintra
- Laboratory of Cell SignalingUniversity of Campinas, Campinas, São Paulo, Brazil Faculty of Applied SciencesUniversity of Campinas, Campinas, São Paulo, Brazil
| | - Licio A Velloso
- Laboratory of Cell SignalingUniversity of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
180
|
Aburasayn H, Al Batran R, Ussher JR. Targeting ceramide metabolism in obesity. Am J Physiol Endocrinol Metab 2016; 311:E423-35. [PMID: 27382035 DOI: 10.1152/ajpendo.00133.2016] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 07/04/2016] [Indexed: 12/12/2022]
Abstract
Obesity is a major health concern that increases the risk for insulin resistance, type 2 diabetes (T2D), and cardiovascular disease. Thus, an enormous research effort has been invested into understanding how obesity-associated dyslipidemia and obesity-induced alterations in lipid metabolism increase the risk for these diseases. Accordingly, it has been proposed that the accumulation of lipid metabolites in organs such as the liver, skeletal muscle, and heart is critical to these obesity-induced pathologies. Ceramide is one such lipid metabolite that accumulates in tissues in response to obesity, and both pharmacological and genetic strategies that reduce tissue ceramide levels yield salutary actions on overall metabolic health. We will review herein why ceramide accumulates in tissues during obesity and how an increase in intracellular ceramide impacts cellular signaling and function as well as potential mechanisms by which reducing intracellular ceramide levels improves insulin resistance, T2D, atherosclerosis, and heart failure. Because a reduction in skeletal muscle ceramide levels is frequently associated with improvements in insulin sensitivity in humans, the beneficial findings reported for reducing ceramides in preclinical studies may have clinical application in humans. Therefore, modulating ceramide metabolism may be a novel, exciting target for preventing and/or treating obesity-related diseases.
Collapse
Affiliation(s)
- Hanin Aburasayn
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - Rami Al Batran
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| | - John R Ussher
- Faculty of Pharmacy and Pharmaceutical Sciences, University of Alberta, Edmonton, Alberta, Canada; and Alberta Diabetes Institute, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
181
|
de Toledo Baldi E, Dias Bóbbo VC, Melo Lima MH, Velloso LA, Pereira de Araujo E. Tumor necrosis factor-alpha levels in blood cord is directly correlated with the body weight of mothers. Obes Sci Pract 2016; 2:210-214. [PMID: 27840691 PMCID: PMC5089573 DOI: 10.1002/osp4.47] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2015] [Revised: 04/14/2016] [Accepted: 05/02/2016] [Indexed: 11/06/2022] Open
Abstract
Background Obesity has emerged as major public health problem leading to increased morbidity and mortality. Epidemiological studies indicate that in many regions of the world, children and teenagers are increasingly affected by obesity, which contributes for a pessimistic projection for the near future. Maternal obesity has been implicated in metabolic disorders of the offspring, but there are no biological markers that can be detected early on life that predict the development of obesity in the offspring. Objective To evaluate the expression of inflammatory markers in the umbilical cord blood of babies of mothers with obesity/overweight, and correlate these markers with the body weight at age 9 months. Methods Anthropometric data of mothers and babies were obtained during prenatal evaluation, at birth and 9 months after birth. Cord blood was collected during delivery of 54 babies from mothers with obesity/overweight and of 50 babies from lean mothers. Tumour necrosis factor‐alpha (TNF‐α), transforming growth factor 1 beta, monocyte chemoattractant protein‐1 and 2 (MCP‐1/MCP‐2) were determined in serum samples using enzyme‐linked immunosorbent assay methods. Correlations were evaluated using the Spearman correlation coefficient, and comparisons were evaluated using the non‐parametric Mann–Whitney U‐test. Results Cord blood TNF‐α was positively correlated with maternal body mass index. There was an inverse correlation between cord blood transforming growth factor 1 beta and baby body weight at birth. There was no biological marker that predicted body weight at age 9 months. Conclusion Although we have not found a biological marker to predict increased body weight at 9 months of age, the study shows that maternal obesity exposes the baby to higher TNF‐α level in the early stages of life, and this can affect metabolic and inflammatory parameters during adulthood.
Collapse
Affiliation(s)
- E de Toledo Baldi
- Obesity and Comorbidities Research Center (OCRC) University of Campinas Campinas São Paulo Brazil; Faculty of Nursing University of Campinas Campinas São Paulo Brazil
| | - V C Dias Bóbbo
- Faculty of Nursing University of Campinas Campinas São Paulo Brazil
| | - M H Melo Lima
- Obesity and Comorbidities Research Center (OCRC) University of Campinas Campinas São Paulo Brazil; Faculty of Nursing University of Campinas Campinas São Paulo Brazil
| | - L A Velloso
- Obesity and Comorbidities Research Center (OCRC) University of Campinas Campinas São Paulo Brazil; Faculty of Medical Science University of Campinas Campinas São Paulo Brazil
| | - E Pereira de Araujo
- Obesity and Comorbidities Research Center (OCRC) University of Campinas Campinas São Paulo Brazil; Faculty of Nursing University of Campinas Campinas São Paulo Brazil
| |
Collapse
|
182
|
Si YC, Miao WN, He JY, Wang XF, Wang YL, Ding WJ. Intricate interactions of obesity, intestinal flora and Toll-like receptors. Shijie Huaren Xiaohua Zazhi 2016; 24:2361-2367. [DOI: 10.11569/wcjd.v24.i15.2361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The rapidly increasing incidence of obesity has resulted in a severe public problem globally. Obesity is associated with subclinical inflammation, causing elevated levels of inflammatory cytokines, as well as disorders of the immune function, which are involved in the dysfunction of intestinal flora. Intestinal flora maintains a dynamic equilibrium with intestinal mucosal immunity. Obesity-related inflammation is mainly trigged by endoplasmic reticulum stress, Toll-like receptor 4 (TLR4) activation and changes of gut flora. Among them, TLR4 plays a central role in sensing intestinal pathogens and inducing mucosal immunity. On the other hand, metabolism, genetics, gut flora and immune state are integrally regulating the TLR function. In the present paper we explore the intricate interactions of obesity, intestinal flora and TLRs, in order to find novel targets for the treatment of obesity.
Collapse
|
183
|
Coope A, Torsoni AS, Velloso LA. MECHANISMS IN ENDOCRINOLOGY: Metabolic and inflammatory pathways on the pathogenesis of type 2 diabetes. Eur J Endocrinol 2016; 174:R175-87. [PMID: 26646937 DOI: 10.1530/eje-15-1065] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 12/07/2015] [Indexed: 12/17/2022]
Abstract
Obesity is the main risk factor for type 2 diabetes (T2D). Studies performed over the last 20 years have identified inflammation as the most important link between these two diseases. During the development of obesity, there is activation of subclinical inflammatory activity in tissues involved in metabolism and energy homeostasis. Intracellular serine/threonine kinases activated in response to inflammatory factors can catalyse the inhibitory phosphorylation of key proteins of the insulin-signalling pathway, leading to insulin resistance. Moreover, during the progression of obesity and insulin resistance, the pancreatic islets are also affected by inflammation, contributing to β-cell failure and leading to the onset of T2D. In this review, we will present the main mechanisms involved in the activation of obesity-associated metabolic inflammation and discuss potential therapeutic opportunities that can be developed to treat obesity-associated metabolic diseases.
Collapse
Affiliation(s)
- Andressa Coope
- Laboratory of Cell SignalingApplied Sciences FacultyUniversity of Campinas, 13084-970 Campinas, São Paulo, Brazil
| | - Adriana S Torsoni
- Laboratory of Cell SignalingApplied Sciences FacultyUniversity of Campinas, 13084-970 Campinas, São Paulo, Brazil
| | - Licio A Velloso
- Laboratory of Cell SignalingApplied Sciences FacultyUniversity of Campinas, 13084-970 Campinas, São Paulo, Brazil
| |
Collapse
|
184
|
Fowler SPG. Low-calorie sweetener use and energy balance: Results from experimental studies in animals, and large-scale prospective studies in humans. Physiol Behav 2016; 164:517-523. [PMID: 27129676 DOI: 10.1016/j.physbeh.2016.04.047] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 04/21/2016] [Accepted: 04/22/2016] [Indexed: 12/21/2022]
Abstract
For more than a decade, pioneering animal studies conducted by investigators at Purdue University have provided evidence to support a central thesis: that the uncoupling of sweet taste and caloric intake by low-calorie sweeteners (LCS) can disrupt an animal's ability to predict the metabolic consequences of sweet taste, and thereby impair the animal's ability to respond appropriately to sweet-tasting foods. These investigators' work has been replicated and extended internationally. There now exists a body of evidence, from a number of investigators, that animals chronically exposed to any of a range of LCSs - including saccharin, sucralose, acesulfame potassium, aspartame, or the combination of erythritol+aspartame - have exhibited one or more of the following conditions: increased food consumption, lower post-prandial thermogenesis, increased weight gain, greater percent body fat, decreased GLP-1 release during glucose tolerance testing, and significantly greater fasting glucose, glucose area under the curve during glucose tolerance testing, and hyperinsulinemia, compared with animals exposed to plain water or - in many cases - even to calorically-sweetened foods or liquids. Adverse impacts of LCS have appeared diminished in animals on dietary restriction, but were pronounced among males, animals genetically predisposed to obesity, and animals with diet-induced obesity. Impacts have been especially striking in animals on high-energy diets: diets high in fats and sugars, and diets which resemble a highly-processed 'Western' diet, including trans-fatty acids and monosodium glutamate. These studies have offered both support for, and biologically plausible mechanisms to explain, the results from a series of large-scale, long-term prospective observational studies conducted in humans, in which longitudinal increases in weight, abdominal adiposity, and incidence of overweight and obesity have been observed among study participants who reported using diet sodas and other LCS-sweetened beverages daily or more often at baseline. Furthermore, frequent use of diet beverages has been associated prospectively with increased long-term risk and/or hazard of a number of cardiometabolic conditions usually considered to be among the sequelae of obesity: hypertension, metabolic syndrome, diabetes, depression, kidney dysfunction, heart attack, stroke, and even cardiovascular and total mortality. Reverse causality does not appear to explain fully the increased risk observed across all of these studies, the majority of which have included key potential confounders as covariates. These have included body mass index or waist circumference at baseline; total caloric intake and specific macronutrient intake; physical activity; smoking; demographic and other relevant risk factors; and/or family history of disease. Whether non-LCS ingredients in diet beverages might have independently increased the weight gain and/or cardiometabolic risk observed among frequent consumers of LCS-sweetened beverages deserves further exploration. In the meantime, however, there is a striking congruence between results from animal research and a number of large-scale, long-term observational studies in humans, in finding significantly increased weight gain, adiposity, incidence of obesity, cardiometabolic risk, and even total mortality among individuals with chronic, daily exposure to low-calorie sweeteners - and these results are troubling.
Collapse
Affiliation(s)
- Sharon P G Fowler
- Department of Medicine, The University of Texas Health Science Center at San Antonio, Texas, United States.
| |
Collapse
|
185
|
Amouzou C, Breuker C, Fabre O, Bourret A, Lambert K, Birot O, Fédou C, Dupuy AM, Cristol JP, Sutra T, Molinari N, Maimoun L, Mariano-Goulart D, Galtier F, Avignon A, Stanke-Labesque F, Mercier J, Sultan A, Bisbal C. Skeletal Muscle Insulin Resistance and Absence of Inflammation Characterize Insulin-Resistant Grade I Obese Women. PLoS One 2016; 11:e0154119. [PMID: 27111539 PMCID: PMC4844150 DOI: 10.1371/journal.pone.0154119] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2015] [Accepted: 04/08/2016] [Indexed: 01/14/2023] Open
Abstract
CONTEXT Obesity is associated with insulin-resistance (IR), the key feature of type 2 diabetes. Although chronic low-grade inflammation has been identified as a central effector of IR development, it has never been investigated simultaneously at systemic level and locally in skeletal muscle and adipose tissue in obese humans characterized for their insulin sensitivity. OBJECTIVES We compared metabolic parameters and inflammation at systemic and tissue levels in normal-weight and obese subjects with different insulin sensitivity to better understand the mechanisms involved in IR development. METHODS 30 post-menopausal women were classified as normal-weight insulin-sensitive (controls, CT) and obese (grade I) insulin-sensitive (OIS) or insulin-resistant (OIR) according to their body mass index and homeostasis model assessment of IR index. They underwent a hyperinsulinemic-euglycemic clamp, blood sampling, skeletal muscle and subcutaneous adipose tissue biopsies, an activity questionnaire and a self-administrated dietary recall. We analyzed insulin sensitivity, inflammation and IR-related parameters at the systemic level. In tissues, insulin response was assessed by P-Akt/Akt expression and inflammation by macrophage infiltration as well as cytokines and IκBα expression. RESULTS Systemic levels of lipids, adipokines, inflammatory cytokines, and lipopolysaccharides were equivalent between OIS and OIR subjects. In subcutaneous adipose tissue, the number of anti-inflammatory macrophages was higher in OIR than in CT and OIS and was associated with higher IL-6 level. Insulin induced Akt phosphorylation to the same extent in CT, OIS and OIR. In skeletal muscle, we could not detect any inflammation even though IκBα expression was lower in OIR compared to CT. However, while P-Akt/Akt level increased following insulin stimulation in CT and OIS, it remained unchanged in OIR. CONCLUSION Our results show that systemic IR occurs without any change in systemic and tissues inflammation. We identified a muscle defect in insulin response as an early mechanism of IR development in grade I obese post-menopausal women.
Collapse
Affiliation(s)
- Cacylde Amouzou
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Cyril Breuker
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | - Odile Fabre
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Annick Bourret
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Karen Lambert
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| | - Olivier Birot
- Faculty of Health, York University, York, Ontario, Canada
| | - Christine Fédou
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | - Anne-Marie Dupuy
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | - Jean-Paul Cristol
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | - Thibault Sutra
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | - Nicolas Molinari
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | - Laurent Maimoun
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | - Denis Mariano-Goulart
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | - Florence Galtier
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | - Antoine Avignon
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | | | - Jacques Mercier
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | - Ariane Sultan
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
- Centre Hospitalier Régional Universitaire (CHRU) Montpellier, Montpellier, France
| | - Catherine Bisbal
- PhyMedExp, University of Montpellier, INSERM U1046, CNRS UMR 9214, Montpellier, France
| |
Collapse
|
186
|
Song J, Kim J. Degeneration of Dopaminergic Neurons Due to Metabolic Alterations and Parkinson's Disease. Front Aging Neurosci 2016; 8:65. [PMID: 27065205 PMCID: PMC4811934 DOI: 10.3389/fnagi.2016.00065] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 03/17/2016] [Indexed: 12/25/2022] Open
Abstract
The rates of metabolic diseases, such as type 2 diabetes mellitus (T2DM), obesity, and cardiovascular disease (CVD), markedly increase with age. In recent years, studies have reported an association between metabolic changes and various pathophysiological mechanisms in the central nervous system (CNS) in patients with metabolic diseases. Oxidative stress and hyperglycemia in metabolic diseases lead to adverse neurophysiological phenomena, including neuronal loss, synaptic dysfunction, and improper insulin signaling, resulting in Parkinson’s disease (PD). In addition, several lines of evidence suggest that alterations of CNS environments by metabolic changes influence the dopamine neuronal loss, eventually affecting the pathogenesis of PD. Thus, we reviewed recent findings relating to degeneration of dopaminergic neurons during metabolic diseases. We highlight the fact that using a metabolic approach to manipulate degeneration of dopaminergic neurons can serve as a therapeutic strategy to attenuate pathology of PD.
Collapse
Affiliation(s)
- Juhyun Song
- Department of Biomedical Engineering, Dongguk University Seoul, South Korea
| | - Jongpil Kim
- Department of Biomedical Engineering, Dongguk University Seoul, South Korea
| |
Collapse
|
187
|
Wang S, Xiang N, Yang L, Zhu C, Zhu X, Wang L, Gao P, Xi Q, Zhang Y, Shu G, Jiang Q. Linoleic acid and stearic acid elicit opposite effects on AgRP expression and secretion via TLR4-dependent signaling pathways in immortalized hypothalamic N38 cells. Biochem Biophys Res Commun 2016; 471:566-71. [DOI: 10.1016/j.bbrc.2016.02.031] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Accepted: 02/09/2016] [Indexed: 12/28/2022]
|
188
|
Iwakura H, Dote K, Bando M, Koyama H, Hosoda K, Kangawa K, Nakao K. Establishment of Leptin-Responsive Cell Lines from Adult Mouse Hypothalamus. PLoS One 2016; 11:e0148639. [PMID: 26849804 PMCID: PMC4744015 DOI: 10.1371/journal.pone.0148639] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2015] [Accepted: 01/21/2016] [Indexed: 12/03/2022] Open
Abstract
Leptin resistance is considered to be the primary cause of obesity. However, the cause of leptin resistance remains incompletely understood, and there is currently no cure for the leptin-resistant state. In order to identify novel drug-target molecules that could overcome leptin resistance, it would be useful to develop in vitro assay systems for evaluating leptin resistance. In this study, we established immortalized adult mouse hypothalamus—derived cell lines, termed adult mouse hypothalamus (AMH) cells, by developing transgenic mice in which SV40 Tag was overexpressed in chromogranin A—positive cells in a tamoxifen-dependent manner. In order to obtain leptin-responsive clones, we selected clones based on the phosphorylation levels of STAT3 induced by leptin. The selected clones were fairly responsive to leptin in terms of STAT3, ERK, and Akt phosphorylation and induction of c-Fos mRNA induction. Pretreatment with leptin, insulin, and palmitate attenuated the c-Fos mRNA response to leptin, suggesting that certain aspects of leptin resistance might be reconstituted in this cellular model. These cell lines are useful tools for understanding the molecular nature of the signal disturbance in the leptin-resistant state and for identifying potential target molecules for drugs that relieve leptin resistance, although they have drawbacks including de-differentiated nature and lack of long-time stability.
Collapse
Affiliation(s)
- Hiroshi Iwakura
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- * E-mail:
| | - Katsuko Dote
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Mika Bando
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Hiroyuki Koyama
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Diabetes and Clinical Nutrition, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kiminori Hosoda
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- Department of Human Health Sciences, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Kenji Kangawa
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
- National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan
| | - Kazuwa Nakao
- Medical Innovation Center, Kyoto University Graduate School of Medicine, Kyoto, Japan
| |
Collapse
|
189
|
Arantes EL, Dragano N, Ramalho A, Vitorino D, de-Souza GF, Lima MHM, Velloso LA, Araújo EP. Topical Docosahexaenoic Acid (DHA) Accelerates Skin Wound Healing in Rats and Activates GPR120. Biol Res Nurs 2016; 18:411-9. [DOI: 10.1177/1099800415621617] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background: The development of methods for improving skin wound healing may have an impact on the outcomes of a number of medical conditions. The topical use of polyunsaturated fatty acids (PUFAs) can accelerate skin wound healing through mechanisms that involve, at least in part, the modulation of inflammatory activity. Purpose: We evaluated whether G-protein-coupled receptor 120 (GPR120), a recently identified receptor for docosahexaenoic acid (DHA) with anti-inflammatory activity, is expressed in the skin and responds to topical DHA. Method: Male Wistar rats were submitted to an 8.0-mm wound on the back and were immediately administered a topical treatment of a solution containing 30 μM of DHA once a day. The healing process was photodocumented, and tissues were collected on Days 5, 9, and 15 for protein and RNA analyses and histological evaluation. Results: GPR120 was expressed in the intact skin and in the wound. Keratinocytes expressed the most skin GPR120, while virtually no expression was detected in fibroblasts. Upon DHA topical treatment, wound healing was significantly accelerated and was accompanied by the molecular activation of GPR120, as determined by its association with β-arrestin-2. In addition, DHA promoted a reduction in the expression of interleukin (IL) 1β and an increase in the expression of IL-6. Furthermore, there was a significant increase in expression of transforming growth factor β (TGF-β) and the keratinocyte marker involucrin. Discussion: Topical DHA improved skin wound healing. The activation of GPR120 is potentially involved in this process.
Collapse
Affiliation(s)
- Eva L. Arantes
- Nursing School, University of Campinas, Campinas SP, Brazil
| | - Nathalia Dragano
- Laboratory of Cell Signaling, University of Campinas, Campinas SP, Brazil
| | - Albina Ramalho
- Laboratory of Cell Signaling, University of Campinas, Campinas SP, Brazil
| | - Daniele Vitorino
- Laboratory of Cell Signaling, University of Campinas, Campinas SP, Brazil
| | | | | | - Licio A. Velloso
- Laboratory of Cell Signaling, University of Campinas, Campinas SP, Brazil
| | - Eliana P. Araújo
- Nursing School, University of Campinas, Campinas SP, Brazil
- Laboratory of Cell Signaling, University of Campinas, Campinas SP, Brazil
| |
Collapse
|
190
|
Abstract
PURPOSE OF REVIEW Aging of the world population is a major contributor to the growing prevalence of the metabolic syndrome, as older persons are frequently affected by the constellation of cardiovascular and metabolic risk factors that constitute the syndrome. The metabolic syndrome has been related to the increasing prevalence of obesity, which is escalating even among older age groups. The present review covers data on the novel proposed biological mediators of the metabolic syndrome, which are as well linked to the aging process. RECENT FINDINGS Relevant biological mediators of metabolic syndrome and unhealthy aging include sarcopenic obesity, insulin resistance with ectopic fat accumulation, magnesium metabolism alterations, systemic and hypothalamic inflammation, shortening of telomeres length, epigenetics, and circadian rhythm disturbances. SUMMARY Metabolic syndrome is related to increased accumulation of central adiposity and ectopic fat infiltration in the skeletal muscle and the liver, linked to overeating and sedentarism with deleterious consequences in late life. Obesity may be complicated with sarcopenia, which refers to loss of muscle mass, strength, and quality in older populations. Prevention of obesity and metabolic syndrome is a priority through the promotion of healthier lifestyles and policies for sugar and saturated fats, which might be widely implemented.
Collapse
Affiliation(s)
- Ligia J Dominguez
- Geriatric Unit, Department of Internal Medicine and Geriatrics, University of Palermo, Palermo, Italy
| | | |
Collapse
|
191
|
Wang L, Zhao B, Chen Y, Ma L, Chen EZ, Mao EQ. Biliary tract external drainage protects against intestinal barrier injury in hemorrhagic shock rats. World J Gastroenterol 2015; 21:12800-12813. [PMID: 26668504 PMCID: PMC4671035 DOI: 10.3748/wjg.v21.i45.12800] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 07/20/2015] [Accepted: 09/15/2015] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of biliary tract external drainage (BTED) on intestinal barrier injury in rats with hemorrhagic shock (HS).
METHODS: BTED was performed via cannula insertion into the bile duct of rats. HS was induced by drawing blood from the femoral artery at a rate of 1 mL/min until a mean arterial pressure (MAP) of 40 ± 5 mmHg was achieved. That MAP was maintained for 60 min. A total of 99 Sprague-Dawley rats were randomized into a sham group, an HS group and an HS + BTED group. Nine rats in the sham group were sacrificed 0.5 h after surgery. Nine rats in each of the HS and HS + BTED groups were sacrificed 0.5 h, 1 h, 2 h, 4 h and 6 h after resuscitation. Plasma tumor necrosis factor-α (TNF-α), interleukin-6 (IL-6), and lipopolysaccharide (LPS) levels were analyzed using enzyme-linked immunosorbent assay. Plasma D-lactate levels were analyzed using colorimetry. The expression levels of occludin and claudin-1 in the ileum were analyzed using Western blot and immunohistochemistry. Histology of the ileum was evaluated by hematoxylin and eosin staining.
RESULTS: Plasma TNF-α levels in the HS + BTED group decreased significantly compared with the HS group at 1 h and 6 h after resuscitation (P < 0.05). Plasma IL-6 levels in the HS + BTED group decreased significantly compared with the HS group at 0.5 h, 1 h and 2 h after resuscitation (P < 0.05). Plasma D-lactate and LPS levels in the HS + BTED group decreased significantly compared with the HS group at 6 h after resuscitation (P < 0.05). The expression levels of occludin in the HS + BTED group increased significantly compared with the HS group at 4 h and 6 h after resuscitation (P < 0.05). The expression levels of claudin-1 in the HS + BTED group increased significantly compared with the HS group at 6 h after resuscitation (P < 0.05). Phenomena of putrescence and desquamation of epithelial cells in the ileal mucosa were attenuated in the HS + BTED group. Ileal histopathologic scores in the HS + BTED group decreased significantly compared with the HS group at 2 h, 4 h and 6 h after resuscitation (P < 0.05).
CONCLUSION: BTED protects against intestinal barrier injury in HS rats.
Collapse
|
192
|
Abstract
The innate immune system includes several classes of pattern recognition receptors (PRRs), including membrane-bound Toll-like receptors (TLRs) and nucleotide-binding oligomerization domain (NOD)-like receptors (NLRs). These receptors detect pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) in the extracellular and intracellular space. Intracellular NLRs constitute inflammasomes, which activate and release caspase-1, IL-1β, and IL-18 thereby initiating an inflammatory response. Systemic and local low-grade inflammation and release of proinflammatory cytokines are implicated in the development and progression of diabetes mellitus and diabetic nephropathy. TLR2, TLR4, and the NLRP3 inflammasome can induce the production of various proinflammatory cytokines and are critically involved in inflammatory responses in pancreatic islets, and in adipose, liver and kidney tissues. This Review describes how innate immune system-driven inflammatory processes can lead to apoptosis, tissue fibrosis, and organ dysfunction resulting in insulin resistance, impaired insulin secretion, and renal failure. We propose that careful targeting of TLR2, TLR4, and NLRP3 signalling pathways could be beneficial for the treatment of diabetes mellitus and diabetic nephropathy.
Collapse
|
193
|
Affiliation(s)
- Ele Ferrannini
- Institute of Clinical Physiology, Consiglio Nazionale delle Ricerche, Pisa, Italy
| |
Collapse
|
194
|
Fazolini NPB, Cruz ALS, Werneck MBF, Viola JPB, Maya-Monteiro CM, Bozza PT. Leptin activation of mTOR pathway in intestinal epithelial cell triggers lipid droplet formation, cytokine production and increased cell proliferation. Cell Cycle 2015; 14:2667-76. [PMID: 26017929 PMCID: PMC4614828 DOI: 10.1080/15384101.2015.1041684] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Accumulating evidence suggests that obesity and enhanced inflammatory reactions are predisposing conditions for developing colon cancer. Obesity is associated with high levels of circulating leptin. Leptin is an adipocytokine that is secreted by adipose tissue and modulates immune response and inflammation. Lipid droplets (LD) are organelles involved in lipid metabolism and production of inflammatory mediators, and increased numbers of LD were observed in human colon cancer. Leptin induces the formation of LD in macrophages in a PI3K/mTOR pathway-dependent manner. Moreover, the mTOR is a serine/threonine kinase that plays a key role in cellular growth and is frequently altered in tumors. We therefore investigated the role of leptin in the modulation of mTOR pathway and regulation of lipid metabolism and inflammatory phenotype in intestinal epithelial cells (IEC-6 cells). We show that leptin promotes a dose- and time-dependent enhancement of LD formation. The biogenesis of LD was accompanied by enhanced CXCL1/CINC-1, CCL2/MCP-1 and TGF-β production and increased COX-2 expression in these cells. We demonstrated that leptin-induced increased phosphorylation of STAT3 and AKT and a dose and time-dependent mTORC activation with enhanced phosphorilation of the downstream protein P70S6K protein. Pre-treatment with rapamycin significantly inhibited leptin effects in LD formation, COX-2 and TGF-β production in IEC-6 cells. Moreover, leptin was able to stimulate the proliferation of epithelial cells on a mTOR-dependent manner. We conclude that leptin regulates lipid metabolism, cytokine production and proliferation of intestinal cells through a mechanism largely dependent on activation of the mTOR pathway, thus suggesting that leptin-induced mTOR activation may contribute to the obesity-related enhanced susceptibility to colon carcinoma.
Collapse
Affiliation(s)
- Narayana P B Fazolini
- a Laboratory of Immunopharmacology; Oswaldo Cruz Institute; FIOCRUZ ; Rio de Janeiro , Brazil
| | | | | | | | | | | |
Collapse
|
195
|
Chen J, Tellez G, Richards JD, Escobar J. Identification of Potential Biomarkers for Gut Barrier Failure in Broiler Chickens. Front Vet Sci 2015; 2:14. [PMID: 26664943 PMCID: PMC4672187 DOI: 10.3389/fvets.2015.00014] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Accepted: 05/08/2015] [Indexed: 01/10/2023] Open
Abstract
The objective of the present study was to identify potential biomarkers for gut barrier failure in chickens. A total of 144 day-of-hatch Ross 308 male broiler chickens were housed in 24 battery cages with six chicks per cage. Cages were randomly assigned to either a control group (CON) or gut barrier failure (GBF) group. During the first 13 days, birds in CON or GBF groups were fed a common corn–soy starter diet. On day 14, CON chickens were switched to a corn grower diet, and GBF chickens were switched to rye–wheat–barley grower diet. In addition, on day 21, GBF chickens were orally challenged with a coccidiosis vaccine. At days 21 and 28, birds were weighed by cage and feed intake was recorded to calculate feed conversion ratio. At day 28, one chicken from each cage was euthanized to collect intestinal samples for morphometric analysis, blood for serum, and intestinal mucosa scrapings for gene expression. Overall performance and feed efficiency was severely affected (P < 0.05) by a GBF model when compared with CON group at days 21 and 28. Duodenum of GBF birds had wider villi, longer crypt depth, and higher crypt depth/villi height ratio than CON birds. Similarly, GBF birds had longer crypt depth in jejunum and ileum when compared with CON birds. Protein levels of endotoxin and α1-acid glycoprotein (AGP) in serum, as well as mRNA levels of interleukin (IL)-8, IL-1β, transforming growth factor (TGF)-β4, and fatty acid-binding protein (FABP) 6 were increased (P < 0.05) in GBF birds compared to CON birds; however, mRNA levels of FABP2, occludin, and mucin 2 (MUC2) were reduced by 34% (P < 0.05), 24% (P = 0.107), and 29% (P = 0.088), respectively, in GBF birds compared to CON birds. The results from the present study suggest that serum endotoxin and AGP, as well as, gene expression of FABP2, FABP6, IL-8, IL-1β, TGF-β4, occludin, and MUC2 in mucosa may work as potential biomarkers for gut barrier health in chickens.
Collapse
Affiliation(s)
- Juxing Chen
- Novus International, Inc. , St. Charles, MO , USA
| | - Guillermo Tellez
- Department of Poultry Science, University of Arkansas , Fayetteville, AR , USA
| | | | | |
Collapse
|