1
|
Dutta P, Annoor A, Dey P, Sultana J, Mokbul MI, Naurin SA, Roy R, Simona SY, Dutta J, Mazumder T, Masud F. The Link Between Metabolic Dysfunction-Associated Steatotic Liver Disease and Gastroesophageal Reflux Disease. Cureus 2024; 16:e71095. [PMID: 39512967 PMCID: PMC11542734 DOI: 10.7759/cureus.71095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2024] [Indexed: 11/15/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD), formerly nonalcoholic fatty liver disease (NAFLD), and gastroesophageal reflux disease (GERD) are prevalent chronic conditions with escalating global incidence. This study delves into the intricate interplay between MASLD and GERD. The primary objective is to comprehensively explore the association between MASLD and GERD, investigating how various factors contribute to the coexistence and potential exacerbation of these conditions. We conducted a literature search in PubMed and Google Scholar of only human studies over the past 10 years. The search included systematic review, meta-analysis, editorial, and cross-sectional studies of patients with MASLD and GERD. The prevalence of GERD in patients with MASLD was higher, with various risk factors coming into play. Obesity was identified as an independent risk factor for both GERD and MASLD. However, obese patients predominantly had higher disease progression. Lifestyle factors like physical activity and dietary modifications emerge as promising strategies to mitigate risk.
Collapse
Affiliation(s)
| | | | - Proma Dey
- Internal Medicine, Chittagong Medical College, Chattagram, BGD
| | - Jakia Sultana
- Internal Medicine, Comilla Medical College, Cumilla, BGD
| | | | | | - Ritwik Roy
- Internal Medicine, Ragib-Rabeya Medical College and Hospital, Chittagong, BGD
| | | | - Jui Dutta
- Internal Medicine, Comilla Medical College, Cumilla, BGD
| | - Tanusree Mazumder
- Family Medicine, Zainul Haque Sikder Women's Medical College and Hospital, Dhaka, BGD
| | - Farjana Masud
- Internal Medicine, Shaheed Ziaur Rahman Medical College, Dhaka, BGD
| |
Collapse
|
2
|
Feng J, Fu S, Luan J. Harnessing fine fibers in decellularized adipose-derived matrix for enhanced adipose regeneration. Mater Today Bio 2024; 25:100974. [PMID: 38322660 PMCID: PMC10844111 DOI: 10.1016/j.mtbio.2024.100974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 01/19/2024] [Accepted: 01/22/2024] [Indexed: 02/08/2024] Open
Abstract
Decellularized Adipose-Derived Matrix (DAM) has the function of inducing adipogenesis, but the distribution of adipogenesis is uneven. We found for the first time that DAM contains two structural components: The tough fibers DAM (T-DAM) and the fine fibers DAM (F-DAM). T-DAM was a dense vortex structure composed of a large number of coarse fibers, characterized by myoblast-related proteins, which cannot achieve fat regeneration and forms a typical "adipose-free zone". While the F-DAM was a loose structure consisting of uniform fine fibers, has more matrix-related proteins and adipose-related proteins. It can not only better promote the adhesion and proliferation of adipose stem cells in vitro, but also achieve the regeneration of adipose tissue in vivo earlier and better, with a uniform range of adipogenesis. The F-DAM is the main and effective kind of DAM to initiate adipose tissue regeneration, which can be picked out by ultrasound fragmentation.
Collapse
Affiliation(s)
- Jiayi Feng
- Department of Aesthetic and Reconstructive Breast Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100144, China
| | | | | |
Collapse
|
3
|
Pinson MR, Bake S, Hurst DA, Samiya NT, Sohrabji F, Miranda RC. Prenatal alcohol alters inflammatory signatures in enteric portal tissues following adult-onset cerebrovascular ischemic stroke. iScience 2023; 26:107920. [PMID: 37810225 PMCID: PMC10550726 DOI: 10.1016/j.isci.2023.107920] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 07/24/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Prenatal alcohol exposure (PAE) impairs recovery from cerebrovascular ischemic stroke in adult rodents. Since the gut becomes dysbiotic following stroke, we assessed links between PAE and enteric portal inflammation. Adult control and PAE rat offspring received a unilateral endothelin-1-induced occlusion of the middle cerebral artery. Post-stroke behavioral disabilities and brain cytokines were assessed. Mesenteric adipose and liver transcriptomes were assessed from stroke-exposed and stroke-naive offspring. We identified, in the liver of stroke-naive animals, a moderate correlation between PAE and a gene network for inflammatory necroptosis. PAE inhibited the acute-phase brain inflammatory cytokine response to stroke. Post-stroke neurological function was correlated with an adipose gene network associated with B-lymphocyte differentiation and nuclear factor κB (NF-κB) signaling and with a liver pro-inflammatory gene network. Collectively, PAE inhibits brain inflammation but results in an inflammatory signature in enteric portal tissues after stroke, suggesting that PAE persistently and adversely impacts the gut-brain axis following adult-onset disease.
Collapse
Affiliation(s)
- Marisa R Pinson
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
| | - Shameena Bake
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
- Women's Health in Neuroscience Program, Texas A&M University School of Medicine, Bryan, TX, USA
| | - David A Hurst
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
| | - Nadia T Samiya
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
| | - Farida Sohrabji
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
- Women's Health in Neuroscience Program, Texas A&M University School of Medicine, Bryan, TX, USA
| | - Rajesh C Miranda
- Department of Neuroscience and Experimental Therapeutics, Texas A&M School of Medicine, Bryan, TX, USA
- Women's Health in Neuroscience Program, Texas A&M University School of Medicine, Bryan, TX, USA
| |
Collapse
|
4
|
Souza-Tavares H, Miranda CS, Vasques-Monteiro IML, Sandoval C, Santana-Oliveira DA, Silva-Veiga FM, Fernandes-da-Silva A, Souza-Mello V. Peroxisome proliferator-activated receptors as targets to treat metabolic diseases: Focus on the adipose tissue, liver, and pancreas. World J Gastroenterol 2023; 29:4136-4155. [PMID: 37475842 PMCID: PMC10354577 DOI: 10.3748/wjg.v29.i26.4136] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 05/26/2023] [Accepted: 06/13/2023] [Indexed: 07/10/2023] Open
Abstract
The world is experiencing reflections of the intersection of two pandemics: Obesity and coronavirus disease 2019. The prevalence of obesity has tripled since 1975 worldwide, representing substantial public health costs due to its comorbidities. The adipose tissue is the initial site of obesity impairments. During excessive energy intake, it undergoes hyperplasia and hypertrophy until overt inflammation and insulin resistance turn adipocytes into dysfunctional cells that send lipotoxic signals to other organs. The pancreas is one of the organs most affected by obesity. Once lipotoxicity becomes chronic, there is an increase in insulin secretion by pancreatic beta cells, a surrogate for type 2 diabetes mellitus (T2DM). These alterations threaten the survival of the pancreatic islets, which tend to become dysfunctional, reaching exhaustion in the long term. As for the liver, lipotoxicity favors lipogenesis and impairs beta-oxidation, resulting in hepatic steatosis. This silent disease affects around 30% of the worldwide population and can evolve into end-stage liver disease. Although therapy for hepatic steatosis remains to be defined, peroxisome proliferator-activated receptors (PPARs) activation copes with T2DM management. Peroxisome PPARs are transcription factors found at the intersection of several metabolic pathways, leading to insulin resistance relief, improved thermogenesis, and expressive hepatic steatosis mitigation by increasing mitochondrial beta-oxidation. This review aimed to update the potential of PPAR agonists as targets to treat metabolic diseases, focusing on adipose tissue plasticity and hepatic and pancreatic remodeling.
Collapse
Affiliation(s)
| | | | | | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Osorno 5310431, Chile
- Departamento de Ciencias Preclínicas, Universidad de la Frontera, Temuco 4780000, Chile
| | | | | | | | - Vanessa Souza-Mello
- Department of Anatomy, Rio de Janeiro State University, Rio de Janeiro 20551030, Brazil
| |
Collapse
|
5
|
Kahraman C, Rendi TA, Sağlam N, Akça İİ, Abidin İ, Alver A. Reciprocal Effects of Adipose Tissue Denervation and High Fat Diet on Serum Metabolic Parameters and Adipokine Levels in Rats: A Long Term Study. J EVOL BIOCHEM PHYS+ 2022. [DOI: 10.1134/s0022093022020090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
6
|
Qian Y, Xia F, Zuo Y, Zhong M, Yang L, Jiang Y, Zou C. Do patients with Prader-Willi syndrome have favorable glucose metabolism? Orphanet J Rare Dis 2022; 17:187. [PMID: 35525976 PMCID: PMC9077846 DOI: 10.1186/s13023-022-02344-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Accepted: 04/26/2022] [Indexed: 11/26/2022] Open
Abstract
Background In recent years, more studies have observed that patients with Prader–Willi syndrome have lower insulin levels and lower insulin resistance than body mass index-matched controls, which may suggest protected glucose metabolism. Method The PubMed and Web of Science online databases were searched to identify relevant studies published in the English language using the terms “Prader–Willi syndrome” with “glucose”, “insulin”, “diabetes mellitus”, “fat”, “adipo*”, “ghrelin”, “oxytocin”, “irisin” or “autonomic nervous system”. Results The prevalence of impaired glucose intolerance, type 2 diabetes mellitus and some other obesity-associated complications in patients with Prader–Willi syndrome tends to be lower when compared to that in general obesity, which is consistent with the hypothetically protected glucose metabolism. Factors including adipose tissue, adiponectin, ghrelin, oxytocin, irisin, growth hormone and the autonomic nervous system possibly modulate insulin sensitivity in patients with Prader–Willi syndrome. Conclusion Although lower insulin levels, lower IR and protected glucose metabolism are widely reported in PWS patients, the causes are still mysterious. Based on existing knowledge, we cannot determine which factor is of utmost importance and what are the underlying mechanisms, and further research is in urgent need.
Collapse
Affiliation(s)
- Yanjie Qian
- Department of Endocrinology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No 3333 Binsheng Road, Hangzhou, 310051, China
| | - Fangling Xia
- Department of Endocrinology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No 3333 Binsheng Road, Hangzhou, 310051, China
| | - Yiming Zuo
- Department of Endocrinology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No 3333 Binsheng Road, Hangzhou, 310051, China
| | - Mianling Zhong
- Department of Endocrinology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No 3333 Binsheng Road, Hangzhou, 310051, China
| | - Lili Yang
- Department of Endocrinology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No 3333 Binsheng Road, Hangzhou, 310051, China
| | - Yonghui Jiang
- Department of Genetics, Yale University School of Medicine, New Haven, USA
| | - Chaochun Zou
- Department of Endocrinology, The Children's Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, No 3333 Binsheng Road, Hangzhou, 310051, China.
| |
Collapse
|
7
|
Kuchler JC, Siqueira BS, Ceglarek VM, Chasko FV, Moura IC, Sczepanhak BF, Vettorazzi JF, Balbo SL, Grassiolli S. The Vagus Nerve and Spleen: Influence on White Adipose Mass and Histology of Obese and Non-obese Rats. Front Physiol 2021; 12:672027. [PMID: 34248663 PMCID: PMC8269450 DOI: 10.3389/fphys.2021.672027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 04/30/2021] [Indexed: 11/30/2022] Open
Abstract
The vagus nerve (VN) and spleen represent a complex interface between neural and immunological functions, affecting both energy metabolism and white adipose tissue (WAT) content. Here, we evaluated whether vagal and splenic axis participates in WAT mass regulation in obese and non-obese male Wistar rats. High doses of monosodium glutamate (M; 4 g/Kg) were administered during the neonatal period to induce hypothalamic lesion and obesity (M-Obese rats). Non-obese or Control (CTL) rats received equimolar saline. At 60 days of life, M-Obese and CTL rats were randomly distributed into experimental subgroups according to the following surgical procedures: sham, subdiaphragmatic vagotomy (SV), splenectomy (SPL), and SV + SPL (n = 11 rats/group). At 150 days of life and after 12 h of fasting, rats were euthanized, blood was collected, and the plasma levels of glucose, triglycerides, cholesterol, insulin, and interleukin 10 (IL10) were analyzed. The visceral and subcutaneous WAT depots were excised, weighed, and histologically evaluated for number and size of adipocytes as well as IL10 protein expression. M-Obese rats showed higher adiposity, hyperinsulinemia, hypertriglyceridemia, and insulin resistance when compared with CTL groups (p < 0.05). In CTL and M-Obese rats, SV reduced body weight gain and triglycerides levels, diminishing adipocyte size without changes in IL10 expression in WAT (p< 0.05). The SV procedure resulted in high IL10 plasma levels in CTL rats, but not in the M-Obese group. The splenectomy prevented the SV anti-adiposity effects, as well as blocked the elevation of IL10 levels in plasma of CTL rats. In contrast, neither SV nor SPL surgeries modified the plasma levels of IL10 and IL10 protein expression in WAT from M-Obese rats. In conclusion, vagotomy promotes body weight and adiposity reduction, elevating IL10 plasma levels in non-obese animals, in a spleen-dependent manner. Under hypothalamic obesity conditions, VN ablation also reduces body weight gain and adiposity, improving insulin sensitivity without changes in IL10 protein expression in WAT or IL10 plasma levels, in a spleen-independent manner. Our findings indicate that the vagal-spleen axis influence the WAT mass in a health state, while this mechanism seems to be disturbed in hypothalamic obese animals.
Collapse
Affiliation(s)
- Joice Cristina Kuchler
- Postgraduate Program in Applied Health Sciences, Western Paraná State University, Francisco Beltrão, Brazil
- Laboratory of Endocrine and Metabolic Physiology, Postgraduate Program in Biosciences and Health, Western Paraná State University, Cascavel, Brazil
| | - Bruna Schumaker Siqueira
- Laboratory of Endocrine and Metabolic Physiology, Postgraduate Program in Biosciences and Health, Western Paraná State University, Cascavel, Brazil
| | - Vanessa Marieli Ceglarek
- Department of Physiology, Institute of Basic Health Sciences, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
- Postgraduate Program in Biological Sciences, Physiology, Federal University of Rio Grande do Sul, Porto Alegre, Brazil
| | - Fernanda Vigilato Chasko
- Laboratory of Endocrine and Metabolic Physiology, Postgraduate Program in Biosciences and Health, Western Paraná State University, Cascavel, Brazil
| | - Isllany Carvalho Moura
- Laboratory of Endocrine and Metabolic Physiology, Postgraduate Program in Biosciences and Health, Western Paraná State University, Cascavel, Brazil
| | - Bruna Fatima Sczepanhak
- Laboratory of Endocrine and Metabolic Physiology, Postgraduate Program in Biosciences and Health, Western Paraná State University, Cascavel, Brazil
| | | | - Sandra Lucinei Balbo
- Laboratory of Endocrine and Metabolic Physiology, Postgraduate Program in Biosciences and Health, Western Paraná State University, Cascavel, Brazil
| | - Sabrina Grassiolli
- Postgraduate Program in Applied Health Sciences, Western Paraná State University, Francisco Beltrão, Brazil
- Laboratory of Endocrine and Metabolic Physiology, Postgraduate Program in Biosciences and Health, Western Paraná State University, Cascavel, Brazil
| |
Collapse
|
8
|
Tessaris D, Matarazzo P, Tuli G, Tuscano A, Rabbone I, Spinardi A, Lezo A, Fenocchio G, Buganza R, de Sanctis L. Multidisciplinary Approach for Hypothalamic Obesity in Children and Adolescents: A Preliminary Study. CHILDREN (BASEL, SWITZERLAND) 2021; 8:531. [PMID: 34206290 PMCID: PMC8304472 DOI: 10.3390/children8070531] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 06/15/2021] [Accepted: 06/19/2021] [Indexed: 01/07/2023]
Abstract
Hypothalamic obesity (HO) is delineated by an inexorable weight gain in subjects with hypothalamic disorder (congenital or acquired). The aim of the present study was to evaluate the effect of a multidisciplinary approach on weight trend and metabolic outcome in children and adolescents with hypothalamic disease who were overweight or obese. Thirteen patients (aged 8.1-16.1 years) received a personalized diet, accelerometer-based activity monitoring, and psychological assessment. Height, weight, body mass index (BMI), and serum metabolic parameters were assessed at baseline (T0) and after six months (T1). Metformin was introduced at T1 in four subjects who were then re-evaluated after six months (T2). At T1, weight gain was significantly reduced compared with T0 (0.29 ± 0.79 kg/month vs. 0.84 ± 0.55 kg/month, p = 0.03), and weight standard deviation score (SDS) and BMI SDS did not change significantly, as serum metabolic parameters. The four subjects treated with metformin showed a reduction of weight SDS and BMI SDS at T2. In conclusion, patients treated with our multidisciplinary approach showed, after 6 months, favorable results characterized by decreased weight gain and stabilization of weight SDS and BMI SDS in a condition usually characterized by inexorable weight gain. However, further analysis, larger cohorts, and longer follow-up are needed to confirm these preliminary data.
Collapse
Affiliation(s)
- Daniele Tessaris
- Pediatric Endocrinology Unit, Regina Margherita Children’s Hospital, University of Turin, 10126 Turin, Italy; (P.M.); (G.T.); (I.R.); (R.B.); (L.d.S.)
| | - Patrizia Matarazzo
- Pediatric Endocrinology Unit, Regina Margherita Children’s Hospital, University of Turin, 10126 Turin, Italy; (P.M.); (G.T.); (I.R.); (R.B.); (L.d.S.)
| | - Gerdi Tuli
- Pediatric Endocrinology Unit, Regina Margherita Children’s Hospital, University of Turin, 10126 Turin, Italy; (P.M.); (G.T.); (I.R.); (R.B.); (L.d.S.)
| | - Antonella Tuscano
- Postgraduation School of Pediatrics, University of Turin, 10126 Turin, Italy;
| | - Ivana Rabbone
- Pediatric Endocrinology Unit, Regina Margherita Children’s Hospital, University of Turin, 10126 Turin, Italy; (P.M.); (G.T.); (I.R.); (R.B.); (L.d.S.)
| | | | - Antonella Lezo
- Dietetic and Clinical Nutrition Unit, Regina Margherita Children’s Hospital, 10126 Turin, Italy;
| | - Giorgia Fenocchio
- Clinical Psychology, Regina Margherita Children’s Hospital, 10126 Turin, Italy;
| | - Raffaele Buganza
- Pediatric Endocrinology Unit, Regina Margherita Children’s Hospital, University of Turin, 10126 Turin, Italy; (P.M.); (G.T.); (I.R.); (R.B.); (L.d.S.)
| | - Luisa de Sanctis
- Pediatric Endocrinology Unit, Regina Margherita Children’s Hospital, University of Turin, 10126 Turin, Italy; (P.M.); (G.T.); (I.R.); (R.B.); (L.d.S.)
| |
Collapse
|
9
|
Wegeberg AM, Meldgaard T, Baek A, Drewes AM, Vyberg M, Jessen N, Brock B, Brock C. Subcutaneous adipose tissue composition and function are unaffected by liraglutide-induced weight loss in adults with type 1 diabetes. Basic Clin Pharmacol Toxicol 2021; 128:773-782. [PMID: 33624417 PMCID: PMC8251841 DOI: 10.1111/bcpt.13575] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/18/2022]
Abstract
Adipose tissue is the primary energy reservoir of the human body, which also possesses endocrine functions. The glucagon‐like peptide agonist liraglutide produces weight loss, although the specific effects on adipose tissue are unknown. We aimed to characterize the white adipose tissue composition and pericellular fibrosis of subcutaneous adipose tissue in response to liraglutide treatment. Furthermore, we explored the level of circulating free fatty acids, cluster of differentiation 163 (CD163) macrophage marker, leptin and adiponectin. Thirty‐nine adults with type 1 diabetes and polyneuropathy were randomly assigned to 26 weeks of liraglutide or placebo treatment. Biopsies of subcutaneous tissue were formalin‐fixed stained with picrosirius red to visualize collagen or immunohistochemically stained for CD163. Serum concentrations of free fatty acids, CD163, leptin and adiponectin were assessed with immunoassays or multiplex panels. In comparison with placebo, liraglutide induced weight loss (3.38 kg, 95% CI −5.29; −1.48, P < 0.001), but did not cause any differences in cell size, distribution of CD163‐positive cells, pericellular fibrosis and serum levels of free fatty acids, CD163, leptin or adiponectin (all P < 0.1). Additionally, no associations between weight loss, cell size and serum markers were found (all P > 0.08). In conclusion, despite liraglutide's effect on weight loss, sustained alterations in subcutaneous adipose tissue did not seem to appear.
Collapse
Affiliation(s)
- Anne-Marie Wegeberg
- Mech-Sense, Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark.,Clinical Institute, Aalborg University, Aalborg, Denmark
| | - Theresa Meldgaard
- Mech-Sense, Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark
| | - Amanda Baek
- The Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Asbjørn Mohr Drewes
- Mech-Sense, Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark.,Clinical Institute, Aalborg University, Aalborg, Denmark.,Steno Diabetes Center North Denmark, Aalborg University Hospital and Clinical Institute, Aalborg University, Aalborg, Denmark
| | - Mogens Vyberg
- Clinical Institute, Aalborg University, Aalborg, Denmark
| | - Niels Jessen
- The Research Laboratory for Biochemical Pathology, Department of Clinical Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | | - Christina Brock
- Mech-Sense, Department of Gastroenterology & Hepatology, Aalborg University Hospital, Aalborg, Denmark.,Clinical Institute, Aalborg University, Aalborg, Denmark.,Steno Diabetes Center North Denmark, Aalborg University Hospital and Clinical Institute, Aalborg University, Aalborg, Denmark
| |
Collapse
|
10
|
Fleury G, Masís‐Vargas A, Kalsbeek A. Metabolic Implications of Exposure to Light at Night: Lessons from Animal and Human Studies. Obesity (Silver Spring) 2020; 28 Suppl 1:S18-S28. [PMID: 32700826 PMCID: PMC7497102 DOI: 10.1002/oby.22807] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Revised: 03/13/2020] [Accepted: 03/14/2020] [Indexed: 02/06/2023]
Abstract
Lately, the incidence of overweight, obesity, and type 2 diabetes has shown a staggering increase. To prevent and treat these conditions, one must look at their etiology. As life on earth has evolved under the conditions of nature's 24-hour light/dark cycle, it seems likely that exposure to artificial light at night (LAN) would affect physiology. Indeed, ample evidence has shown that LAN impacts many metabolic parameters, at least partly via the biological clock in the suprachiasmatic nucleus of the hypothalamus. This review focuses on the impact of chronic and acute effects of LAN of different wavelengths on locomotor activity, food intake, the sleep/wake cycle, body temperature, melatonin, glucocorticoids, and glucose and lipid metabolism. While chronic LAN disturbs daily rhythms in these parameters, experiments using short-term LAN exposure also have shown acute negative effects in metabolically active peripheral tissues. Experiments using LAN of different wavelengths not only have indicated an important role for melanopsin, the photopigment found in intrinsically photosensitive retinal ganglion cells, but also provided evidence that each wavelength may have a specific impact on energy metabolism. Importantly, exposure to LAN has been shown to impact glucose homeostasis also in humans and to be associated with an increased incidence of overweight, obesity, and atherosclerosis.
Collapse
Affiliation(s)
- Giulia Fleury
- Department of Endocrinology and MetabolismAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Anayanci Masís‐Vargas
- Department of Endocrinology and MetabolismAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
- Hypothalamic Integration MechanismsNetherlands Institute for Neuroscience (NIN)Amsterdamthe Netherlands
- Institute of Cellular and Integrative Neurosciences (INCI)UPR‐3212 CNRSUniversity of StrasbourgStrasbourgFrance
| | - Andries Kalsbeek
- Department of Endocrinology and MetabolismAmsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
- Hypothalamic Integration MechanismsNetherlands Institute for Neuroscience (NIN)Amsterdamthe Netherlands
| |
Collapse
|
11
|
Qian SW, Wu MY, Wang YN, Zhao YX, Zou Y, Pan JB, Tang Y, Liu Y, Guo L, Tang QQ. BMP4 facilitates beige fat biogenesis via regulating adipose tissue macrophages. J Mol Cell Biol 2020; 11:14-25. [PMID: 29462349 DOI: 10.1093/jmcb/mjy011] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/15/2018] [Indexed: 12/21/2022] Open
Abstract
Thermogenic beige fat improves metabolism and prevents obesity. Emerging evidence shows that the activation of M2 macrophages stimulates beige adipogenesis, whereas the activation of M1 macrophages, which play a major role in inflammation, impedes beige adipogenesis. Thus, the identification of factors that regulate adipose tissue macrophages (ATMs) will help clarify the mechanism involved in beiging. Here, we found that one of the secreted proteins in adipose tissue, namely, BMP4, alters the ATM profile in subcutaneous adipose tissue by activating M2 and inhibiting M1 macrophages. Mechanistically, the BMP4-stimulated p38/MAPK/STAT6/PI3K-AKT signalling pathway is involved. Meanwhile, BMP4 improved the potency of M2 macrophages to induce beige fat biogenesis. Considering that the overexpression of BMP4 in adipose tissue promotes the beiging of subcutaneous adipose tissue and improves insulin sensitivity, these findings provide evidence that BMP4 acts as an activator of beige fat by targeting immuno-metabolic pathways.
Collapse
Affiliation(s)
- Shu-Wen Qian
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education; Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Meng-Yuan Wu
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education; Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yi-Na Wang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education; Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Ya-Xin Zhao
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education; Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Ying Zou
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education; Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Jia-Bao Pan
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education; Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yan Tang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education; Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Yang Liu
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education; Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Liang Guo
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education; Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| | - Qi-Qun Tang
- Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education; Department of Biochemistry and Molecular Biology, Fudan University Shanghai Medical College, Shanghai, China
| |
Collapse
|
12
|
Eichwald T, Talbot S. Neuro-Immunity Controls Obesity-Induced Pain. Front Hum Neurosci 2020; 14:181. [PMID: 32581740 PMCID: PMC7295985 DOI: 10.3389/fnhum.2020.00181] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2020] [Accepted: 04/23/2020] [Indexed: 12/16/2022] Open
Abstract
The prevalence of obesity skyrocketed over the past decades to become a significant public health problem. Obesity is recognized as a low-grade inflammatory disease and is linked with several comorbidities such as diabetes, circulatory disease, common neurodegenerative diseases, as well as chronic pain. Adipocytes are a major neuroendocrine organ that continually, and systemically, releases pro-inflammatory factors. While the exact mechanisms driving obesity-induced pain remain poorly defined, nociceptor hypersensitivity may result from the systemic state of inflammation characteristic of obesity as well as weight surplus-induced mechanical stress. Obesity and pain also share various genetic mutations, lifestyle risk factors, and metabolic pathways. For instance, fat pads are often found hyper-innervated and rich in immune cell types of multiple origins. These immunocytes release cytokines, amplifying nociceptor function, which, in turn, via locally released neuropeptides, sustain immunocytes' function. Here, we posit that along with mechanical stress stemming from extra weight, the local neuro-immune interplay occurring within the fat pads maintains the state of chronic low-grade inflammation and heightens sensory hypersensitivity. Overall, stopping such harmful neuro-immune crosstalk may constitute a novel pathway to prevent obesity-associated comorbidities, including neuronal hypersensitivity.
Collapse
Affiliation(s)
- Tuany Eichwald
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
- Departamento de Bioquímica, Centro de Ciências Biológicas, Universidade Federal de Santa Catarina, Florianópolis, Brazil
| | - Sebastien Talbot
- Département de Pharmacologie et Physiologie, Faculté de Médecine, Université de Montréal, Montreal, QC, Canada
| |
Collapse
|
13
|
Bizino MB, Jazet IM, de Heer P, van Eyk HJ, Dekkers IA, Rensen PCN, Paiman EHM, Lamb HJ, Smit JW. Placebo-controlled randomised trial with liraglutide on magnetic resonance endpoints in individuals with type 2 diabetes: a pre-specified secondary study on ectopic fat accumulation. Diabetologia 2020; 63:65-74. [PMID: 31690988 PMCID: PMC6890592 DOI: 10.1007/s00125-019-05021-6] [Citation(s) in RCA: 82] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Accepted: 08/28/2019] [Indexed: 01/18/2023]
Abstract
AIMS/HYPOTHESIS The aim of this work was to assess the effect of liraglutide on ectopic fat accumulation in individuals with type 2 diabetes mellitus. METHODS This study is a pre-specified subanalysis of the MAGNetic resonance Assessment of VICTOza efficacy in the Regression of cardiovascular dysfunction In type 2 diAbetes mellitus (MAGNA VICTORIA) study, with primary endpoints being the effects of liraglutide on left ventricular diastolic and systolic function. The MAGNA VICTORIA study was a single-centre, parallel-group trial in 50 individuals with type 2 diabetes mellitus (BMI >25 kg/m2) who were randomly assigned (1:1, stratified for sex and insulin use) to receive liraglutide 1.8 mg once daily or placebo for 26 weeks, added to standard care. Participants, study personnel and outcome assessors were blinded to treatment allocation. The secondary endpoints of visceral adipose tissue (VAT), abdominal subcutaneous adipose tissue (SAT) and epicardial fat were measured with MRI. Hepatic triacylglycerol content (HTGC) and myocardial triacylglycerol content (MTGC) were quantified with proton MR spectroscopy. Between-group differences (change from baseline) were tested for significance using ANCOVA. Mean differences with 95% CIs were reported. RESULTS The trial was completed in 2016. Twenty-four participants were randomised to receive liraglutide and 26 to receive placebo. One patient in the liraglutide group withdrew consent before having received the study drug and was not included in the intention-to-treat analysis. Liraglutide (n = 23) vs placebo (n = 26) significantly reduced body weight (liraglutide 98.4 ± 13.8 kg to 94.3 ± 14.9 kg; placebo 94.5 ± 13.1 kg to 93.9 ± 13.2 kg; estimated treatment effect -4.5 [95% CI -6.4, -2.6] kg). HbA1c declined in both groups without a significant treatment effect of liraglutide vs placebo (liraglutide 66.7 ± 11.5 mmol/mol to 55.0 ± 13.2 mmol/mol [8.4 ± 1.1% to 7.3 ± 1.2%]; placebo 64.7 ± 10.2 mmol/mol to 56.9 ± 6.9 mmol/mol [8.2 ± 1.0% to 7.5 ± 0.7%]; estimated treatment effect -2.9 [95% CI -8.1, 2.3] mmol/mol or -0.3 [95% CI -0.8, 0.2]%). VAT did not change significantly between groups (liraglutide 207 ± 87 cm2 to 203 ± 88 cm2; placebo 204 ± 63 cm2 to 200 ± 55 cm2; estimated treatment effect -7 [95% CI -24, 10] cm2), while SAT was reduced by a significantly greater extent with liraglutide than with placebo (liraglutide 361 ± 142 cm2 to 339 ± 131 cm2; placebo 329 ± 107 cm2 to 333 ± 125 cm2; estimated treatment effect -29 [95% CI -51, -8] cm2). Epicardial fat did not change significantly between groups (liraglutide 8.9 ± 4.3 cm2 to 9.1 ± 4.7 cm2; placebo 9.6 ± 4.1 cm2 to 9.6 ± 4.6 cm2; estimated treatment effect 0.2 [95% CI -1.5, 1.8] cm2). Change in HTGC was not different between groups (liraglutide 18.1 ± 11.2% to 12.0 ± 7.7%; placebo 18.4 ± 9.4% to 14.7 ± 10.0%; estimated treatment effect -2.1 [95% CI -5.3, 1.0]%). MTGC was not different after treatment with liraglutide (1.5 ± 0.6% to 1.2 ± 0.6%) vs placebo (1.3 ± 0.5% to 1.2 ± 0.6%), with an estimated treatment effect of -0.1 (95% CI -0.4, 0.2)%. There were no adjudicated serious adverse events. CONCLUSIONS/INTERPRETATION Compared with placebo, liraglutide-treated participants lost significantly more body weight. Liraglutide primarily reduced subcutaneous fat but not visceral, hepatic, myocardial or epicardial fat. Future larger studies are needed to confirm the results of this secondary endpoint study. TRIAL REGISTRATION ClinicalTrials.gov NCT01761318. FUNDING This study was funded by Novo Nordisk A/S (Bagsvaerd, Denmark).
Collapse
Affiliation(s)
- Maurice B Bizino
- Department of Radiology, Leiden University Medical Center, LUMC postzone C2S, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands.
| | - Ingrid M Jazet
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, LUMC post zone C7Q, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Paul de Heer
- Department of Radiology, Leiden University Medical Center, LUMC postzone C2S, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Huub J van Eyk
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, LUMC post zone C7Q, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Ilona A Dekkers
- Department of Radiology, Leiden University Medical Center, LUMC postzone C2S, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Patrick C N Rensen
- Department of Medicine, Division of Endocrinology, Leiden University Medical Center, LUMC post zone C7Q, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
- Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, Leiden, the Netherlands
| | - Elisabeth H M Paiman
- Department of Radiology, Leiden University Medical Center, LUMC postzone C2S, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Hildebrandus J Lamb
- Department of Radiology, Leiden University Medical Center, LUMC postzone C2S, Albinusdreef 2, 2333 ZA, Leiden, the Netherlands
| | - Johannes W Smit
- Department of Medicine, Radboud University Medical Center, Nijmegen, the Netherlands
| |
Collapse
|
14
|
Combination of Olanzapine Pamoate with Melatonin and Metformin: Quantitative Changes in Rat Adipose Tissue. CURRENT HEALTH SCIENCES JOURNAL 2019; 45:372-382. [PMID: 32110439 PMCID: PMC7014984 DOI: 10.12865/chsj.45.04.05] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 12/20/2019] [Indexed: 12/12/2022]
Abstract
Olanzapine is one of the atypical antipsychotics widely used in the treatment of schizophrenia and has been associated with metabolic changes as adverse effects, including hyperglycemia, dyslipidemia, and weight gain. In a batch of adult female Wistar rats, we studied the prolonged-release intramuscular olanzapine pamoate induced quantitative changes of visceral and subcutaneous adipose tissue. We also assessed the effects of the combinations of olanzapine pamoate with melatonin, metformin, and melatonin plus metformin, administered by gastric gavage. A higher mean weight of the visceral and subcutaneous adipose tissue per animal was noted in the olanzapine pamoate exposed group compared to controls. The association with melatonin, metformin, or the combination of melatonin with metformin attenuated the olanzapine-induced adipose deposit tissue growth. The effect was more pronounced for the combination of olanzapine with melatonin and metformin. Because most of the results were not statistically significant we can deduce that in the chronic experiment, adaptive type modifications of the receptors on which both olanzapine and melatonin act can occur.
Collapse
|
15
|
Iozzo P, Guzzardi MA. Imaging of brain glucose uptake by PET in obesity and cognitive dysfunction: life-course perspective. Endocr Connect 2019; 8:R169-R183. [PMID: 31590145 PMCID: PMC6865363 DOI: 10.1530/ec-19-0348] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Accepted: 10/07/2019] [Indexed: 12/17/2022]
Abstract
The prevalence of obesity has reached epidemic proportions and keeps growing. Obesity seems implicated in the pathogenesis of cognitive dysfunction, Alzheimer's disease and dementia, and vice versa. Growing scientific efforts are being devoted to the identification of central mechanisms underlying the frequent association between obesity and cognitive dysfunction. Glucose brain handling undergoes dynamic changes during the life-course, suggesting that its alterations might precede and contribute to degenerative changes or signaling abnormalities. Imaging of the glucose analog 18F-labeled fluorodeoxyglucose (18FDG) by positron emission tomography (PET) is the gold-standard for the assessment of cerebral glucose metabolism in vivo. This review summarizes the current literature addressing brain glucose uptake measured by PET imaging, and the effect of insulin on brain metabolism, trying to embrace a life-course vision in the identification of patterns that may explain (and contribute to) the frequent association between obesity and cognitive dysfunction. The current evidence supports that brain hypermetabolism and brain insulin resistance occur in selected high-risk conditions as a transient phenomenon, eventually evolving toward normal or low values during life or disease progression. Associative studies suggest that brain hypermetabolism predicts low BDNF levels, hepatic and whole body insulin resistance, food desire and an unfavorable balance between anticipated reward from food and cognitive inhibitory control. Emerging mechanistic links involve the microbiota and the metabolome, which correlate with brain metabolism and cognition, deserving attention as potential future prevention targets.
Collapse
Affiliation(s)
- Patricia Iozzo
- Institute of Clinical Physiology, National Research Council (CNR), Pisa, Italy
- Correspondence should be addressed to P Iozzo:
| | | |
Collapse
|
16
|
Chang RY, Tsai HL, Hsiao PG, Tan CW, Lee CP, Chu IT, Chen YP, Koo M. Association between heart rate recovery after exercise and renal function in patients referred for treadmill exercise test. PLoS One 2019; 14:e0222236. [PMID: 31491037 PMCID: PMC6730871 DOI: 10.1371/journal.pone.0222236] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/23/2019] [Indexed: 11/18/2022] Open
Abstract
Introduction Heart rate recovery (HRR) is a marker of parasympathetic activity recovery after exercise, and it is associated with cardiovascular mortality and total mortality. Impaired renal function is also associated with cardiac mortality. The aim of this study was to investigate the association between HRR after exercise and renal function in patients referred for a treadmill exercise test. Patients and methods This cross-sectional study was conducted at a regional hospital in southern Taiwan. Patients who completed a symptom-limited treadmill exercise test from January 2015 to February 2018 were recruited. Before the treadmill exercise test, patients were asked to complete a questionnaire on the past disease history and lifestyle factors. Serum creatinine measurement within two years prior to or after the date of the treadmill exercise test of the patients was also obtained from the medical records for these patients. Estimated glomerular filtration rate (eGFR) was calculated. Simple and multiple linear regression analyses were performed to investigate the association between one-minute HRR and eGFR. Results A total of 2,825 patients completed the treadmill exercise test, and serum creatinine measurement was identified from medical records for 2,153 patients (76.2%). Multiple linear regression analysis revealed that a lower eGFR was significantly associated with lower one-minute HRR (P< 0.001), adjusting for other significant independent factors, including age, waist circumference, type 2 diabetes mellitus, and smoking. Conclusions In this cross-sectional observational study, a lower eGFR was significantly and independently associated with decreased one-minute HRR, suggesting that parasympathetic activity recovery after exercise could be impaired by a decrease in renal function.
Collapse
Affiliation(s)
- Rei-Yeuh Chang
- Division of Cardiology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan
- Chung Jen Junior College of Nursing, Health Sciences and Management, Chiayi, Taiwan
- Min-Hwei Junior College of Health Care Management, Tainan City, Taiwan
| | - Han-Lin Tsai
- Division of Cardiology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan
| | - Ping-Gune Hsiao
- Division of Cardiology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan
| | - Chao-Wen Tan
- Division of Cardiology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan
| | - Chi-Pin Lee
- Division of Cardiology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan
| | - I-Tseng Chu
- Division of Cardiology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan
| | - Yung-Ping Chen
- Division of Cardiology, Department of Internal Medicine, Ditmanson Medical Foundation Chia-Yi Christian Hospital, Chiayi City, Taiwan
| | - Malcolm Koo
- Graduate Institute of Long-term Care, Tzu Chi University of Science and Technology, Hualien City, Hualien, Taiwan
- Dalla Lana School of Public Health, University of Toronto, Toronto, Ontario, Canada
- * E-mail:
| |
Collapse
|
17
|
Rastović M, Srdić-Galić B, Barak O, Stokić E, Polovina S. AGING, HEART RATE VARIABILITY AND METABOLIC IMPACT OF OBESITY. Acta Clin Croat 2019; 58:430-438. [PMID: 31969754 PMCID: PMC6971797 DOI: 10.20471/acc.2019.58.03.05] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The relationship between aging and changes in heart rate variability (HRV) could depend on the metabolic profile of obese people, i.e. metabolically healthy obese (MHO) and metabolically unhealthy obese (MUO). We aimed to determine the age at which obesity related autonomic dysfunction becomes significant and whether it decreases differently according to metabolic profile. We analyzed HRV in 99 adults using Wildman's criteria for metabolic profile and 5-minute HRV for autonomic nervous system. In MHO, high frequency (HF) decreased in the 4th decade of life. In MUO, standard deviation of R-R intervals (SDNN), root mean square of successive differences of all R-R intervals (RMSSD), number of adjacent intervals differing by more than 50 ms expressed as percentage of all intervals in the collecting period (pNN50), HF, low frequency (LF), LF/HF (LF divided by HF) and total power (TP) decreased in the 4th decade of life (partial shared variance 28%-36%). In conclusion, an age dependent decrease of HRV occurs in MUO between the third and fifth decade of life. In MHO, HF significantly decreases around the age of 40 years. Cardiometabolic profile influences metabolic aging, altering the autonomic nervous system.
Collapse
Affiliation(s)
| | - Biljana Srdić-Galić
- 1Subotica General Hospital, Department of Internal Medicine, Division of Endocrinology, Subotica, Serbia; 2University of Novi Sad, Faculty of Medicine, Department of Anatomy, Novi Sad, Serbia; 3University of Novi Sad, Faculty of Medicine, Department of Physiology, Novi Sad, Serbia; 4University of Novi Sad, Faculty of Medicine, Institute of Internal Disease, Department of Endocrinology, Diabetes and Metabolic Disorders, Novi Sad, Serbia; 5Clinical Center of Serbia, Department of Endocrinology, Diabetes and Metabolic Diseases, Belgrade, Serbia; 6University of Novi Sad, Faculty of Pharmacy, Department of Internal Medicine, Novi Sad, Serbia
| | - Otto Barak
- 1Subotica General Hospital, Department of Internal Medicine, Division of Endocrinology, Subotica, Serbia; 2University of Novi Sad, Faculty of Medicine, Department of Anatomy, Novi Sad, Serbia; 3University of Novi Sad, Faculty of Medicine, Department of Physiology, Novi Sad, Serbia; 4University of Novi Sad, Faculty of Medicine, Institute of Internal Disease, Department of Endocrinology, Diabetes and Metabolic Disorders, Novi Sad, Serbia; 5Clinical Center of Serbia, Department of Endocrinology, Diabetes and Metabolic Diseases, Belgrade, Serbia; 6University of Novi Sad, Faculty of Pharmacy, Department of Internal Medicine, Novi Sad, Serbia
| | - Edita Stokić
- 1Subotica General Hospital, Department of Internal Medicine, Division of Endocrinology, Subotica, Serbia; 2University of Novi Sad, Faculty of Medicine, Department of Anatomy, Novi Sad, Serbia; 3University of Novi Sad, Faculty of Medicine, Department of Physiology, Novi Sad, Serbia; 4University of Novi Sad, Faculty of Medicine, Institute of Internal Disease, Department of Endocrinology, Diabetes and Metabolic Disorders, Novi Sad, Serbia; 5Clinical Center of Serbia, Department of Endocrinology, Diabetes and Metabolic Diseases, Belgrade, Serbia; 6University of Novi Sad, Faculty of Pharmacy, Department of Internal Medicine, Novi Sad, Serbia
| | - Snežana Polovina
- 1Subotica General Hospital, Department of Internal Medicine, Division of Endocrinology, Subotica, Serbia; 2University of Novi Sad, Faculty of Medicine, Department of Anatomy, Novi Sad, Serbia; 3University of Novi Sad, Faculty of Medicine, Department of Physiology, Novi Sad, Serbia; 4University of Novi Sad, Faculty of Medicine, Institute of Internal Disease, Department of Endocrinology, Diabetes and Metabolic Disorders, Novi Sad, Serbia; 5Clinical Center of Serbia, Department of Endocrinology, Diabetes and Metabolic Diseases, Belgrade, Serbia; 6University of Novi Sad, Faculty of Pharmacy, Department of Internal Medicine, Novi Sad, Serbia
| |
Collapse
|
18
|
França da Silva AK, Destro Christofaro DG, Manata Vanzella L, Marques Vanderlei F, Lopez Laurino MJ, Marques Vanderlei LC. Relationship of the Aggregation of Cardiovascular Risk Factors in the Parasympathetic Modulation of Young People with Type 1 Diabetes. MEDICINA (KAUNAS, LITHUANIA) 2019; 55:E534. [PMID: 31454959 PMCID: PMC6780872 DOI: 10.3390/medicina55090534] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Revised: 08/20/2019] [Accepted: 08/21/2019] [Indexed: 11/20/2022]
Abstract
Background and objectives: In healthy individuals, autonomic alterations are associated with the aggregation of cardiovascular risk factors. However, in individuals with type 1 diabetes, who are known to present autonomic alterations, mainly characterized by a reduction in parasympathetic modulation, these associations have not yet been investigated. We assess whether the aggregation of cardiovascular risk factors influences parasympathetic indices of heart rate variability in young people with type 1 diabetes. Materials and methods: This cross-sectional study included 39 individuals with type 1 diabetes (22.54 ± 4.31), evaluated in relation to the risk factors: blood pressure, fat percentage, and resting heart rate. For heart rate variability analysis, heart rate was recorded beat-to-beat using a cardio frequency meter (PolarS810i) for 30 min with the volunteers in dorsal decubitus. The parasympathetic heart rate variability indices were calculated: rMSSD, pNN50, high frequency (HF) n.u (normalized units), SD1, 2LV, and 2ULV. Data collection was carried out in 2014 and analyzed in 2017. Results: Individuals with two aggregate risk factors present a reduction in the values of the indices that reflect parasympathetic autonomic modulation compared to individuals without the risk factors analyzed, regardless of sex and age. Conclusion: In young people with type 1 diabetes, the aggregation of cardiovascular risk factors is associated with parasympathetic autonomic impairment.
Collapse
Affiliation(s)
- Anne Kastelianne França da Silva
- Department of Physical Therapy, Faculty of Science and Technology, São Paulo State University (UNESP), Roberto Simonsen Street, 305, Presidente Prudente, São Paulo 19060-900, Brazil.
| | - Diego Giulliano Destro Christofaro
- Department of Physical Education, Faculty of Science and Technology, São Paulo State University (UNESP), Roberto Simonsen Street, 305, Presidente Prudente, São Paulo 19060-900, Brazil
| | - Laís Manata Vanzella
- Department of Physical Therapy, Faculty of Science and Technology, São Paulo State University (UNESP), Roberto Simonsen Street, 305, Presidente Prudente, São Paulo 19060-900, Brazil
| | - Franciele Marques Vanderlei
- Department of Physical Therapy, Faculty of Science and Technology, São Paulo State University (UNESP), Roberto Simonsen Street, 305, Presidente Prudente, São Paulo 19060-900, Brazil
| | - Maria Júlia Lopez Laurino
- Department of Physical Therapy, Faculty of Science and Technology, São Paulo State University (UNESP), Roberto Simonsen Street, 305, Presidente Prudente, São Paulo 19060-900, Brazil
| | - Luiz Carlos Marques Vanderlei
- Department of Physical Therapy, Faculty of Science and Technology, São Paulo State University (UNESP), Roberto Simonsen Street, 305, Presidente Prudente, São Paulo 19060-900, Brazil
| |
Collapse
|
19
|
Berthoud HR, Neuhuber WL. Vagal mechanisms as neuromodulatory targets for the treatment of metabolic disease. Ann N Y Acad Sci 2019; 1454:42-55. [PMID: 31268181 PMCID: PMC6810744 DOI: 10.1111/nyas.14182] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 05/23/2019] [Accepted: 06/05/2019] [Indexed: 12/30/2022]
Abstract
With few effective treatments available, the global rise of metabolic diseases, including obesity, type 2 diabetes mellitus, and cardiovascular disease, seems unstoppable. Likely caused by an obesogenic environment interacting with genetic susceptibility, the pathophysiology of obesity and metabolic diseases is highly complex and involves crosstalk between many organs and systems, including the brain. The vagus nerve is in a key position to bidirectionally link several peripheral metabolic organs with the brain and is increasingly targeted for neuromodulation therapy to treat metabolic disease. Here, we review the basics of vagal functional anatomy and its implications for vagal neuromodulation therapies. We find that most existing vagal neuromodulation techniques either ignore or misinterpret the rich functional specificity of both vagal efferents and afferents as demonstrated by a large body of literature. This lack of specificity of manipulating vagal fibers is likely the reason for the relatively poor beneficial long‐term effects of such therapies. For these therapies to become more effective, rigorous validation of all physiological endpoints and optimization of stimulation parameters as well as electrode placements will be necessary. However, given the large number of function‐specific fibers in any vagal branch, genetically guided neuromodulation techniques are more likely to succeed.
Collapse
Affiliation(s)
- Hans-Rudolf Berthoud
- Neurobiology of Nutrition and Metabolism Department, Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, Louisiana
| | - Winfried L Neuhuber
- Institut fur Anatomie und Zellbiologie, Friedrich-Alexander Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
20
|
Opperhuizen AL, Foppen E, Jonker M, Wackers P, van Faassen M, van Weeghel M, van Kerkhof L, Fliers E, Kalsbeek A. Effects of Light-at-Night on the Rat Liver - A Role for the Autonomic Nervous System. Front Neurosci 2019; 13:647. [PMID: 31281239 PMCID: PMC6596368 DOI: 10.3389/fnins.2019.00647] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Accepted: 06/05/2019] [Indexed: 12/31/2022] Open
Abstract
Exposure to light at night (LAN) has been associated with serious pathologies, including obesity, diabetes and cancer. Recently we showed that 2 h of LAN impaired glucose tolerance in rats. Several studies have suggested that the autonomic nervous system (ANS) plays an important role in communicating these acute effects of LAN to the periphery. Here, we investigated the acute effects of LAN on the liver transcriptome of male Wistar rats. Expression levels of individual genes were not markedly affected by LAN, nevertheless pathway analysis revealed clustered changes in a number of endocrine pathways. Subsequently, we used selective hepatic denervations [sympathetic (Sx), parasympathetic (Px), total (Tx, i.e., Sx plus Px), sham] to investigate the involvement of the ANS in the effects observed. Surgical removal of the sympathetic or parasympathetic hepatic branches of the ANS resulted in many, but small changes in the liver transcriptome, including a pathway involved with circadian clock regulation, but it clearly separated the four denervation groups. On the other hand, analysis of the liver metabolome was not able to separate the denervation groups, and only 6 out of 78 metabolites were significantly up- or downregulated after denervations. Finally, removal of the sympathetic and parasympathetic hepatic nerves combined with LAN exposure clearly modulated the effects of LAN on the liver transcriptome, but left most endocrine pathways unaffected. Conclusion: One-hour light-at-night acutely affects the liver transcriptome. Part of this effect is mediated via the nervous innervation, as a hepatectomy modulated and reduced the effect of LAN on liver transcripts.
Collapse
Affiliation(s)
- Anne-Loes Opperhuizen
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, Netherlands.,Laboratory of Endocrinology, Amsterdam University Medical Center, Department of Clinical Chemistry, University of Amsterdam, Amsterdam, Netherlands
| | - Ewout Foppen
- Laboratory of Endocrinology, Amsterdam University Medical Center, Department of Clinical Chemistry, University of Amsterdam, Amsterdam, Netherlands
| | - Martijs Jonker
- MAD - Dutch Genomics Service and Support Provider, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, Netherlands
| | - Paul Wackers
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University Medical Centre Groningen, University of Groningen, Groningen, Netherlands
| | - Michel van Weeghel
- Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Linda van Kerkhof
- Centre for Health Protection, National Institute for Public Health and the Environment, Bilthoven, Netherlands
| | - Eric Fliers
- Amsterdam University Medical Center, Department of Endocrinology and Metabolism, University of Amsterdam, Amsterdam, Netherlands
| | - Andries Kalsbeek
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience, Amsterdam, Netherlands.,Laboratory of Endocrinology, Amsterdam University Medical Center, Department of Clinical Chemistry, University of Amsterdam, Amsterdam, Netherlands.,Amsterdam University Medical Center, Department of Endocrinology and Metabolism, University of Amsterdam, Amsterdam, Netherlands
| |
Collapse
|
21
|
Zhu Q, Glazier BJ, Hinkel BC, Cao J, Liu L, Liang C, Shi H. Neuroendocrine Regulation of Energy Metabolism Involving Different Types of Adipose Tissues. Int J Mol Sci 2019; 20:E2707. [PMID: 31159462 PMCID: PMC6600468 DOI: 10.3390/ijms20112707] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 05/24/2019] [Accepted: 05/29/2019] [Indexed: 12/17/2022] Open
Abstract
Despite tremendous research efforts to identify regulatory factors that control energy metabolism, the prevalence of obesity has been continuously rising, with nearly 40% of US adults being obese. Interactions between secretory factors from adipose tissues and the nervous system innervating adipose tissues play key roles in maintaining energy metabolism and promoting survival in response to metabolic challenges. It is currently accepted that there are three types of adipose tissues, white (WAT), brown (BAT), and beige (BeAT), all of which play essential roles in maintaining energy homeostasis. WAT mainly stores energy under positive energy balance, while it releases fuels under negative energy balance. Thermogenic BAT and BeAT dissipate energy as heat under cold exposure to maintain body temperature. Adipose tissues require neural and endocrine communication with the brain. A number of WAT adipokines and BAT batokines interact with the neural circuits extending from the brain to cooperatively regulate whole-body lipid metabolism and energy homeostasis. We review neuroanatomical, histological, genetic, and pharmacological studies in neuroendocrine regulation of adipose function, including lipid storage and mobilization of WAT, non-shivering thermogenesis of BAT, and browning of BeAT. Recent whole-tissue imaging and transcriptome analysis of differential gene expression in WAT and BAT yield promising findings to better understand the interaction between secretory factors and neural circuits, which represents a novel opportunity to tackle obesity.
Collapse
Affiliation(s)
- Qi Zhu
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Bradley J Glazier
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Benjamin C Hinkel
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Jingyi Cao
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Lin Liu
- Program of Bioinformatics, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Chun Liang
- Program of Bioinformatics, Department of Biology, Miami University, Oxford, OH 45056, USA.
| | - Haifei Shi
- Program of Physiology and Neuroscience, Department of Biology, Miami University, Oxford, OH 45056, USA.
| |
Collapse
|
22
|
Blaszkiewicz M, Willows JW, Johnson CP, Townsend KL. The Importance of Peripheral Nerves in Adipose Tissue for the Regulation of Energy Balance. BIOLOGY 2019; 8:E10. [PMID: 30759876 PMCID: PMC6466238 DOI: 10.3390/biology8010010] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/05/2019] [Accepted: 02/06/2019] [Indexed: 12/29/2022]
Abstract
Brown and white adipose tissues are essential for maintenance of proper energy balance and metabolic health. In order to function efficiently, these tissues require both endocrine and neural communication with the brain. Brown adipose tissue (BAT), as well as the inducible brown adipocytes that appear in white adipose tissue (WAT) after simulation, are thermogenic and energy expending. This uncoupling protein 1 (UCP1)-mediated process requires input from sympathetic nerves releasing norepinephrine. In addition to sympathetic noradrenergic signaling, adipose tissue contains sensory nerves that may be important for relaying fuel status to the brain. Chemical and surgical denervation studies of both WAT and BAT have clearly demonstrated the role of peripheral nerves in browning, thermogenesis, lipolysis, and adipogenesis. However, much is still unknown about which subtypes of nerves are present in BAT versus WAT, what nerve products are released from adipose nerves and how they act to mediate metabolic homeostasis, as well as which cell types in adipose are receiving synaptic input. Recent advances in whole-depot imaging and quantification of adipose nerve fibers, as well as other new research findings, have reinvigorated this field of research. This review summarizes the history of research into adipose innervation and brain⁻adipose communication, and also covers landmark and recent research on this topic to outline what we currently know and do not know about adipose tissue nerve supply and communication with the brain.
Collapse
Affiliation(s)
- Magdalena Blaszkiewicz
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA.
| | - Jake W Willows
- School of Biology and Ecology, University of Maine, Orono, ME 04469, USA.
| | - Cory P Johnson
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA.
| | - Kristy L Townsend
- Graduate School of Biomedical Science and Engineering, University of Maine, Orono, ME 04469, USA.
- School of Biology and Ecology, University of Maine, Orono, ME 04469, USA.
| |
Collapse
|
23
|
Chen SH, Chen HC, Hsieh CL, Chao PM. Electric stimulation of ears accelerates body weight loss mediated by high-fat to low-fat diet switch accompanied by increased white adipose tissue browning in C57BL/6 J mice. BMC COMPLEMENTARY AND ALTERNATIVE MEDICINE 2018; 18:323. [PMID: 30518367 PMCID: PMC6282328 DOI: 10.1186/s12906-018-2388-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 11/27/2018] [Indexed: 01/09/2023]
Abstract
Background Weight reduction frequently occurs in patients receiving vagus nerve stimulation (VNS) therapy. Therefore, we hypothesized that during dietary intervention for weight loss, auricular electric stimulation (AES), an alternative of VNS, accelerates weight loss by increasing white adipose tissue (WAT) browning and increases energy expenditure. Methods C57BL/6J male mice were fed a high-fat diet for 5 wk. to induce obesity, then switched to a low-fat diet for 5 wk. and allocated into 3 groups to receive 2 Hz electric stimulation on ears, electrode clamps only, or nothing (AES, Sham and Ctrl, respectively). Results Switching to a low-fat diet reduced body weight progressively in all 3 groups, with the greatest reduction in the AES group. In accordance with a mild decrease in feed intake, hypothalamus mRNA levels of Npy, AgRP tended to be reduced, while Pomc tended to be increased by AES. Mice in the AES group had the highest concentrations of norepinephrine in serum and inguinal WAT, and expression levels of uncoupling protein-1 (UCP-1) and tyrosine hydroxylase in inguinal WAT. Furthermore, their subcutaneous adipocytes had multilocular and UCP-1+ characteristics, along with a smaller cell size. Conclusion AES, by increasing WAT browning, could be used in conjunction with a low-fat diet to augment weight loss in addition to suppressing appetite.
Collapse
|
24
|
Dos Santos F, Moraes-Silva IC, Moreira ED, Irigoyen MC. The role of the baroreflex and parasympathetic nervous system in fructose-induced cardiac and metabolic alterations. Sci Rep 2018; 8:10970. [PMID: 30030527 PMCID: PMC6054615 DOI: 10.1038/s41598-018-29336-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/18/2018] [Indexed: 12/19/2022] Open
Abstract
It is well-established that baroreflex sensitivity is essential for blood pressure control, and also plays a key role in the modulation of disease-induced metabolic alterations. In order to investigate the role of the baroreflex in the cardiometabolic and inflammatory derangements promoted by fructose overload, Wistar rats underwent sinoaortic denervation (SAD) or sham surgery and were studied 90 days after receiving tap water (Den and Ctrl) or a 10% fructose solution (Fruc and Den-Fruc). All experimental groups showed marked and similar degree of baroreflex impairment compared to Ctrl. As expected, fructose overload effectively induced metabolic syndrome; however, when it was associated with SAD, several alterations were attenuated. While Fruc rats displayed increased sympathetic modulation and tone and reduced vagal modulation compared to Ctrl animals, Den-Fruc rats showed greater vagal tone and modulation when compared to the Fruc group. Moreover, the Den-Fruc group showed augmented expression of β1 adrenergic receptors and TNF/IL-10 ratio and reduction of β2 in the left ventricle. The increase in vagal function was correlated with improved insulin sensitivity (r2 = 0.76), and decreased abdominal fat (r2 = -0.78) and β2 receptors (r2 = -0.85). Our results showed that: (1) chronic fructose overload induced severe baroreflex impairment, i.e. in a similar magnitude to that observed in SAD rats, which is accompanied by cardiometabolic dysfunctions; (2) the compensatory enhancement in parasympathetic function in SAD rats submitted to fructose intake may point out the possibility of use of approaches that improve vagal function as therapeutic target to attenuate fructose-induced cardiometabolic dysfunctions.
Collapse
Affiliation(s)
- Fernando Dos Santos
- Heart Institute (InCor), School of Medicine, University of Sao Paulo (FMUSP), Sao Paulo, Brazil.
| | - Ivana C Moraes-Silva
- Heart Institute (InCor), School of Medicine, University of Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Edson D Moreira
- Heart Institute (InCor), School of Medicine, University of Sao Paulo (FMUSP), Sao Paulo, Brazil
| | - Maria-Claudia Irigoyen
- Heart Institute (InCor), School of Medicine, University of Sao Paulo (FMUSP), Sao Paulo, Brazil
| |
Collapse
|
25
|
Wang X, Villar VA, Tiu A, Upadhyay KK, Cuevas S. Dopamine D2 receptor upregulates leptin and IL-6 in adipocytes. J Lipid Res 2018; 59:607-614. [PMID: 29472382 PMCID: PMC5880505 DOI: 10.1194/jlr.m081000] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2017] [Revised: 02/12/2018] [Indexed: 12/12/2022] Open
Abstract
Leptin is a pro-inflammatory cytokine secreted by the adipose tissue. Dopamine D2 receptors (D2Rs) have anti-inflammatory effects in the brain and kidney tissues. Mouse and human adipocytes express D2R; D2R protein was 10-fold greater in adipocytes from human visceral tissue than subcutaneous tissue. However, the function of D2R in adipocytes is not well understood. 3T3-L1 cells were treated with D2-like receptor agonist quinpirole, and immunoblot and quantitative PCR were performed. Quinpirole increased the protein and mRNA expression of leptin and IL-6, but not adiponectin and visfatin (24 h). It also increased the mRNA expression of TNF-α , MCP1, and NFkB-p50. An acute increase in the protein expression of leptin and TNF-α was also found in the cells treated with quinpirole. The leptin concentration in the culture media was increased by quinpirole-bathing the 3T3-L1 adipocytes. These quinpirole effects on leptin and IL-6 expression were prevented by the D2R antagonist L741,626. Similarly, siRNA-mediated silencing of Drd2 decreased the leptin, IL-6, mRNA, and protein expressions. The D2R-mediated increase in leptin expression was prevented by the phosphoinositide 3-kinase inhibitor LY294002. Acute quinpirole treatment in C57Bl/6J mice increased serum leptin concentration and leptin mRNA in visceral adipocyte tissue but not in subcutaneous adipocytes, confirming the stimulatory effect of D2R on leptin in vivo. Our results suggest that the stimulation of D2R increases leptin production and may have a tissue-specific pro-inflammatory effect in adipocytes.
Collapse
Affiliation(s)
- Xiaoyan Wang
- Department of Medicine, Division of Renal Diseases & Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Van Anthony Villar
- Department of Medicine, Division of Renal Diseases & Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Andrew Tiu
- Department of Medicine, Division of Renal Diseases & Hypertension, The George Washington University School of Medicine and Health Sciences, Washington, DC 20037
| | - Kiran K Upadhyay
- Department of Pediatrics, Division of Pediatric Nephrology, University of Florida, Gainesville, FL 32607
| | - Santiago Cuevas
- Research Center for Genetic Medicine, Children's National Health System, Washington DC 20010.
| |
Collapse
|
26
|
Lee KA, Lee NY, Park TS, Jin HY. Morphologic Comparison of Peripheral Nerves in Adipocyte Tissue from db/db Diabetic versus Normal Mice. Diabetes Metab J 2018; 42:169-172. [PMID: 29676547 PMCID: PMC5911521 DOI: 10.4093/dmj.2018.42.2.169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Accepted: 01/15/2018] [Indexed: 11/18/2022] Open
Abstract
Present study investigated the morphologic changes of autonomic nerves in the adipose tissue in diabetic animal model. Male obese type 2 diabetic db/db mice and age matched non-diabetic db/m control mice were used. Epididymal adipose tissue from diabetic db/db mice with that from control heterozygous db/m mice was compared using confocal microscopy-based method to visualize intact whole adipose tissue. Immunohistochemistry with tyrosine hydroxylase for sympathetic (SP), choline acetyltransferase for parasympathetic (PSP), and protein gene product 9.5 (PGP 9.5) for whole autonomic nerves was performed. The quantity of immunostained portion of SP, PSP, and PGP 9.5 stained nerve fibers showed decreased trend in diabetic group; however, the ratio of SP/PSP of adipose tissue was higher in diabetic group compared with control group as follows (0.70±0.30 vs. 0.95±0.25, P<0.05; normal vs. diabetic, respectively). Both SP and PSP nerve fibers were observed in white adipose tissue and PSP nerve fibers were suggested as more decreased in diabetes based on our observation.
Collapse
Affiliation(s)
- Kyung Ae Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Na Young Lee
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Tae Sun Park
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea
| | - Heung Yong Jin
- Division of Endocrinology and Metabolism, Department of Internal Medicine, Chonbuk National University Medical School, Research Institute of Clinical Medicine of Chonbuk National University-Biomedical Research Institute of Chonbuk National University Hospital, Jeonju, Korea.
| |
Collapse
|
27
|
Dezfuli G, Gillis RA, Tatge JE, Duncan KR, Dretchen KL, Jackson PG, Verbalis JG, Sahibzada N. Subdiaphragmatic Vagotomy With Pyloroplasty Ameliorates the Obesity Caused by Genetic Deletion of the Melanocortin 4 Receptor in the Mouse. Front Neurosci 2018; 12:104. [PMID: 29545738 PMCID: PMC5838008 DOI: 10.3389/fnins.2018.00104] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 02/12/2018] [Indexed: 12/22/2022] Open
Abstract
Background/Objectives: We tested the hypothesis that abolishing vagal nerve activity will reverse the obesity phenotype of melanocortin 4 receptor knockout mice (Mc4r−/−). Subjects/Methods: In two separate studies, we examined the efficacy of bilateral subdiaphragmatic vagotomy (SDV) with pyloroplasty in the prevention and treatment of obesity in Mc4r−/− mice. Results: In the first study, SDV prevented >20% increase in body weight (BW) associated with this genotype. This was correlated with a transient reduction in overall food intake (FI) in the preventative arm of the study. Initially, SDV mice had reduced weekly FI; however, FI normalized to that of controls and baseline FI within the 8-week study period. In the second study, the severe obesity that is characteristic of the adult Mc4r−/− genotype was significantly improved by SDV with a magnitude of 30% loss in excess BW over a 4-week period. Consistent with the first preventative study, within the treatment arm, SDV mice also demonstrated a transient reduction in FI relative to control and baseline levels that normalized over subsequent weeks. In addition to the accompanying loss in weight, mice subjected to SDV showed a decrease in respiratory exchange ratio (RER), and an increase in locomotor activity (LA). Analysis of the white fat-pad deposits of these mice showed that they were significantly less than the control groups. Conclusions: Altogether, our data demonstrates that SDV both prevents gain in BW and causes weight loss in severely obese Mc4r−/− mice. Moreover, it suggests that an important aspect of weight reduction for this type of monogenic obesity involves loss of signaling in vagal motor neurons.
Collapse
Affiliation(s)
- Ghazaul Dezfuli
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Richard A Gillis
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Jaclyn E Tatge
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Kimbell R Duncan
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Kenneth L Dretchen
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| | - Patrick G Jackson
- Department of Surgery, Georgetown University Medical Center, Washington, DC, United States
| | - Joseph G Verbalis
- Department of Medicine, Georgetown University Medical Center, Washington, DC, United States
| | - Niaz Sahibzada
- Department of Pharmacology and Physiology, Georgetown University Medical Center, Washington, DC, United States
| |
Collapse
|
28
|
Abstract
Interactions between the brain and distinct adipose depots have a key role in maintaining energy balance, thereby promoting survival in response to metabolic challenges such as cold exposure and starvation. Recently, there has been renewed interest in the specific central neuronal circuits that regulate adipose depots. Here, we review anatomical, genetic and pharmacological studies on the neural regulation of adipose function, including lipolysis, non-shivering thermogenesis, browning and leptin secretion. In particular, we emphasize the role of leptin-sensitive neurons and the sympathetic nervous system in modulating the activity of brown, white and beige adipose tissues. We provide an overview of advances in the understanding of the heterogeneity of the brain regulation of adipose tissues and offer a perspective on the challenges and paradoxes that the community is facing regarding the actions of leptin on this system.
Collapse
Affiliation(s)
- Alexandre Caron
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Syann Lee
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joel K. Elmquist
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Laurent Gautron
- Division of Hypothalamic Research and Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
29
|
van der Spek R, Fliers E, la Fleur SE, Kalsbeek A. Daily Gene Expression Rhythms in Rat White Adipose Tissue Do Not Differ Between Subcutaneous and Intra-Abdominal Depots. Front Endocrinol (Lausanne) 2018; 9:206. [PMID: 29760682 PMCID: PMC5936761 DOI: 10.3389/fendo.2018.00206] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 04/12/2018] [Indexed: 01/05/2023] Open
Abstract
White adipose tissue (WAT) is present in different depots throughout the body. Although all depots are exposed to systemic humoral signals, they are not functionally identical. Studies in clock gene knockout animals and in shift workers suggest that daily rhythmicity may play an important role in lipid metabolism. Differences in rhythmicity between fat depots might explain differences in depot function; therefore, we measured mRNA expression of clock genes and metabolic genes on a 3-h interval over a 24-h period in the subcutaneous inguinal depot and in the intra-abdominal perirenal, epididymal, and mesenteric depots of male Wistar rats. We analyzed rhythmicity using CircWave software. Additionally, we measured plasma concentrations of glucose, insulin, corticosterone, and leptin. The clock genes (Bmal1/Per2/Cry1/Cry2/RevErbα/DBP) showed robust daily gene expression rhythms, which did not vary between WAT depots. Metabolic gene expression rhythms (SREBP1c/PPARα/PPARγ/FAS/LPL/Glut4/HSL/CPT1b/leptin/visfatin/resistin) were more variable between depots. However, no distinct differences between intra-abdominal and subcutaneous rhythms were found. Concluding, specific fat depots are not associated with differences in clock gene expression rhythms and, therefore, do not provide a likely explanation for the differences in metabolic function between different fat depots.
Collapse
Affiliation(s)
- Rianne van der Spek
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
- *Correspondence: Rianne van der Spek,
| | - Eric Fliers
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
| | - Susanne E. la Fleur
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
| | - Andries Kalsbeek
- Department of Endocrinology and Metabolism, Academic Medical Center (AMC), University of Amsterdam, Amsterdam, Netherlands
- Hypothalamic Integration Mechanisms, Netherlands Institute for Neuroscience (NIN), Amsterdam, Netherlands
| |
Collapse
|
30
|
Guarino D, Nannipieri M, Iervasi G, Taddei S, Bruno RM. The Role of the Autonomic Nervous System in the Pathophysiology of Obesity. Front Physiol 2017; 8:665. [PMID: 28966594 PMCID: PMC5606212 DOI: 10.3389/fphys.2017.00665] [Citation(s) in RCA: 164] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Accepted: 08/22/2017] [Indexed: 12/18/2022] Open
Abstract
Obesity is reaching epidemic proportions globally and represents a major cause of comorbidities, mostly related to cardiovascular disease. The autonomic nervous system (ANS) dysfunction has a two-way relationship with obesity. Indeed, alterations of the ANS might be involved in the pathogenesis of obesity, acting on different pathways. On the other hand, the excess weight induces ANS dysfunction, which may be involved in the haemodynamic and metabolic alterations that increase the cardiovascular risk of obese individuals, i.e., hypertension, insulin resistance and dyslipidemia. This article will review current evidence about the role of the ANS in short-term and long-term regulation of energy homeostasis. Furthermore, an increased sympathetic activity has been demonstrated in obese patients, particularly in the muscle vasculature and in the kidneys, possibily contributing to increased cardiovascular risk. Selective leptin resistance, obstructive sleep apnea syndrome, hyperinsulinemia and low ghrelin levels are possible mechanisms underlying sympathetic activation in obesity. Weight loss is able to reverse metabolic and autonomic alterations associated with obesity. Given the crucial role of autonomic dysfunction in the pathophysiology of obesity and its cardiovascular complications, vagal nerve modulation and sympathetic inhibition may serve as therapeutic targets in this condition.
Collapse
Affiliation(s)
- Daniela Guarino
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy.,Institute of Clinical Physiology of CNRPisa, Italy.,Scuola Superiore Sant'AnnaPisa, Italy
| | - Monica Nannipieri
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | | | - Stefano Taddei
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| | - Rosa Maria Bruno
- Department of Clinical and Experimental Medicine, University of PisaPisa, Italy
| |
Collapse
|
31
|
Vestergaard ET, Cichosz SL, Møller N, Jørgensen JOL, Fleischer J. Short-term acipimox treatment is associated with decreased cardiac parasympathetic modulation. Br J Clin Pharmacol 2017; 83:2671-2677. [PMID: 28736944 DOI: 10.1111/bcp.13384] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 07/02/2017] [Accepted: 07/19/2017] [Indexed: 11/28/2022] Open
Abstract
AIMS The nicotinic acid analogue acipimox is an antilipolytic agent, which acutely inhibits lipolysis and suppresses systemic levels of free fatty acids (FFA) and improves insulin sensitivity in obese patients. These effects of acipimox are transient due to a counter-regulatory increase in growth hormone levels that reverse the antilipolytic effect of acipimox. Hypopituitary patients constitute a viable model to study the growth hormone-independent effects of acipimox and the impact of isolated changes in FFA concentrations and insulin sensitivity on parasympathetic nervous activity. The aim of the present study was to investigate if pharmacological antilipolysis with acipimox acutely affects autonomic tone. METHODS We studied heart rate variability as a measure of autonomic tone in eight hypopituitary men with and without acipimox treatment. The standard deviation of normal-to-normal intervals, root mean square of successive differences and high frequency were measured as heart rate variability parameters. The patients were studied in the basal and insulin-stimulated state with clamped plasma glucose on two occasions in a randomized, double-blind and placebo-controlled crossover study. RESULTS Plasma glucose (4.7 vs. 4.9 mmol l-1 , P = 0.02) and serum FFA (0.05 vs. 0.41 mmol l-1 , P < 0.001) were significantly decreased during acipimox treatment. Acipimox had an inhibitory effect on standard deviation of normal-to-normal intervals (41.3 vs. 45.3 ms, P = 0.01), root mean square of successive differences (23.2 vs. 11 ms, P = 0.03) and high frequency (3.79 vs 3.60 ln (ms2 ), P = 0.02) and these effects were reversed during clamping. CONCLUSIONS Short-term inhibition of lipolysis by acipimox treatment lowered circulating FFA levels, improved insulin sensitivity, and was accompanied by reduced parasympathetic tone. The effect of acipimox on the parasympathetic modulation was reversed by hyperinsulinaemia.
Collapse
Affiliation(s)
- Esben Thyssen Vestergaard
- Medical Research Laboratory, Aarhus University, Aarhus, Denmark.,Department of Pediatrics, Randers Regional Hospital, Randers, Denmark
| | - Simon Lebech Cichosz
- Department of Health Science and Technology, Aalborg University, Aalborg East, Denmark
| | - Niels Møller
- Medical Research Laboratory, Aarhus University, Aarhus, Denmark.,Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | - Jens Otto Lunde Jørgensen
- Medical Research Laboratory, Aarhus University, Aarhus, Denmark.,Department of Endocrinology and Internal Medicine, Aarhus University Hospital, Aarhus, Denmark
| | | |
Collapse
|
32
|
Cohen P, Spiegelman BM. Cell biology of fat storage. Mol Biol Cell 2017; 27:2523-7. [PMID: 27528697 PMCID: PMC4985254 DOI: 10.1091/mbc.e15-10-0749] [Citation(s) in RCA: 136] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Accepted: 06/17/2016] [Indexed: 01/09/2023] Open
Abstract
The worldwide epidemic of obesity and type 2 diabetes has greatly increased interest in the biology and physiology of adipose tissues. Adipose (fat) cells are specialized for the storage of energy in the form of triglycerides, but research in the last few decades has shown that fat cells also play a critical role in sensing and responding to changes in systemic energy balance. White fat cells secrete important hormone-like molecules such as leptin, adiponectin, and adipsin to influence processes such as food intake, insulin sensitivity, and insulin secretion. Brown fat, on the other hand, dissipates chemical energy in the form of heat, thereby defending against hypothermia, obesity, and diabetes. It is now appreciated that there are two distinct types of thermogenic fat cells, termed brown and beige adipocytes. In addition to these distinct properties of fat cells, adipocytes exist within adipose tissue, where they are in dynamic communication with immune cells and closely influenced by innervation and blood supply. This review is intended to serve as an introduction to adipose cell biology and to familiarize the reader with how these cell types play a role in metabolic disease and, perhaps, as targets for therapeutic development.
Collapse
Affiliation(s)
- Paul Cohen
- Laboratory of Molecular Metabolism, Rockefeller University, New York, NY 10065
| | - Bruce M Spiegelman
- Dana-Farber Cancer Institute and Department of Cell Biology, Harvard Medical School, Boston, MA 02115
| |
Collapse
|
33
|
Messina G, Valenzano A, Moscatelli F, Salerno M, Lonigro A, Esposito T, Monda V, Corso G, Messina A, Viggiano A, Triggiani AI, Chieffi S, Guglielmi G, Monda M, Cibelli G. Role of Autonomic Nervous System and Orexinergic System on Adipose Tissue. Front Physiol 2017; 8:137. [PMID: 28344558 PMCID: PMC5344930 DOI: 10.3389/fphys.2017.00137] [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: 01/10/2017] [Accepted: 02/23/2017] [Indexed: 01/16/2023] Open
Abstract
Adipose tissue, defined as white adipose tissue (WAT) and brown adipose tissue (BAT), is a biological caloric reservoir; in response to over-nutrition it expands and, in response to energy deficit, it releases lipids. The WAT primarily stores energy as triglycerides, whereas BAT dissipates chemical energy as heat. In mammals, the BAT is a key site for heat production and an attractive target to promote weight loss. The autonomic nervous system (ANS) exerts a direct control at the cellular and molecular levels in adiposity. The sympathetic nervous system (SNS) provides a complex homeostatic control to specifically coordinate function and crosstalk of both fat pads, as indicated by the increase of the sympathetic outflow to BAT, in response to cold and high-fat diet, but also by the increase or decrease of the sympathetic outflow to selected WAT depots, in response to different lipolytic requirements of these two conditions. More recently, a role has been attributed to the parasympathetic nervous system (PNS) in modulating both adipose tissue insulin-mediated glucose uptake and fatty free acid (FFA) metabolism in an anabolic way and its endocrine function. The regulation of adipose tissue is unlikely to be limited to the autonomic control, since a number of signaling cytokines and neuropeptides play an important role, as well. In this review, we report some experimental evidences about the role played by both the ANS and orexins into different fat pads, related to food intake and energy expenditure, with a special emphasis on body weight status and fat mass (FM) content.
Collapse
Affiliation(s)
- Giovanni Messina
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
- Department of Experimental Medicine, Second University of NaplesNaples, Italy
| | - Anna Valenzano
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Fiorenzo Moscatelli
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Monica Salerno
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Antonio Lonigro
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Teresa Esposito
- Department of Experimental Medicine, Second University of NaplesNaples, Italy
| | - Vincenzo Monda
- Department of Experimental Medicine, Second University of NaplesNaples, Italy
| | - Gaetano Corso
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Antonietta Messina
- Department of Experimental Medicine, Second University of NaplesNaples, Italy
| | - Andrea Viggiano
- Department of Medicine and Surgery, University of SalernoSalerno, Italy
| | - Antonio I. Triggiani
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Sergio Chieffi
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Giuseppe Guglielmi
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| | - Marcellino Monda
- Department of Experimental Medicine, Second University of NaplesNaples, Italy
| | - Giuseppe Cibelli
- Department of Clinical and Experimental Medicine, University of FoggiaFoggia, Italy
| |
Collapse
|
34
|
Porfirio MC, Gomes de Almeida JP, Stornelli M, Giovinazzo S, Purper-Ouakil D, Masi G. Can melatonin prevent or improve metabolic side effects during antipsychotic treatments? Neuropsychiatr Dis Treat 2017; 13:2167-2174. [PMID: 28860773 PMCID: PMC5560235 DOI: 10.2147/ndt.s127564] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the last two decades, second-generation antipsychotics (SGAs) were more frequently used than typical antipsychotics for treating both psychotic and nonpsychotic psychiatric disorders in both children and adolescents, because of their lower risk of adverse neurological effects, that is, extrapyramidal symptoms. Recent studies have pointed out their effect on weight gain and increased visceral adiposity as they induce metabolic syndrome. Patients receiving SGAs often need to be treated with other substances to counteract metabolic side effects. In this paper, we point out the possible protective effect of add-on melatonin treatment in preventing, mitigating, or even reversing SGAs metabolic effects, improving quality of life and providing safer long-term treatments in pediatric patients. Melatonin is an endogenous indolamine secreted during darkness by the pineal gland; it plays a key role in regulating the circadian rhythm, generated by the suprachiasmatic nuclei (SCN) of the hypothalamus, and has many other biological functions, including chronobiotic, antioxidant and neuroprotective properties, anti-inflammatory and free radical scavenging effects, and diminishing oxidative injury and fat distribution. It has been hypothesized that SGAs cause adverse metabolic effects that may be restored by nightly administration of melatonin because of its influence on autonomic and hormonal outputs. Interestingly, atypical anti-psychotics (AAPs) can cause several sleep disorders, and circadian misalignment can influence hormones involved in the metabolic regulation, such as insulin, leptin, and ghrelin; furthermore, a relationship between obesity and sleep curtailment has been demonstrated, as well as sleep deprivation in rats has been associated with hyperphagia. Metabolic effects of melatonin, both central and peripheral, direct and indirect, target most metabolic disorders reported during and after SGA treatment in children, adolescents, and adults. Further systematic studies on psychiatric patients are needed to explore the effect of add-on melatonin on metabolic side effects of SGAs, independent of energy intake, diet, and exercise.
Collapse
Affiliation(s)
| | | | - Maddalena Stornelli
- Unit of Child Neurology and Psychiatry, "Tor Vergata" University of Rome, Italy
| | - Silvia Giovinazzo
- Unit of Child Neurology and Psychiatry, "Tor Vergata" University of Rome, Italy
| | - Diane Purper-Ouakil
- Unit of Child and Adolescent Psychiatry, Saint Eloi Hospital, Montpellier, France
| | - Gabriele Masi
- IRCCS Stella Maris, Scientific Institute of Child Neurology and Psychiatry, Calambrone, Pisa, Italy
| |
Collapse
|
35
|
Gómez-Hernández A, Beneit N, Díaz-Castroverde S, Escribano Ó. Differential Role of Adipose Tissues in Obesity and Related Metabolic and Vascular Complications. Int J Endocrinol 2016; 2016:1216783. [PMID: 27766104 PMCID: PMC5059561 DOI: 10.1155/2016/1216783] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Revised: 07/19/2016] [Accepted: 08/04/2016] [Indexed: 12/18/2022] Open
Abstract
This review focuses on the contribution of white, brown, and perivascular adipose tissues to the pathophysiology of obesity and its associated metabolic and vascular complications. Weight gain in obesity generates excess of fat, usually visceral fat, and activates the inflammatory response in the adipocytes and then in other tissues such as liver. Therefore, low systemic inflammation responsible for insulin resistance contributes to atherosclerotic process. Furthermore, an inverse relationship between body mass index and brown adipose tissue activity has been described. For these reasons, in recent years, in order to combat obesity and its related complications, as a complement to conventional treatments, a new insight is focusing on the role of the thermogenic function of brown and perivascular adipose tissues as a promising therapy in humans. These lines of knowledge are focused on the design of new drugs, or other approaches, in order to increase the mass and/or activity of brown adipose tissue or the browning process of beige cells from white adipose tissue. These new treatments may contribute not only to reduce obesity but also to prevent highly prevalent complications such as type 2 diabetes and other vascular alterations, such as hypertension or atherosclerosis.
Collapse
Affiliation(s)
- Almudena Gómez-Hernández
- Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, IdISSC, Instituto de Salud Carlos III, Madrid, Spain
| | - Nuria Beneit
- Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, IdISSC, Instituto de Salud Carlos III, Madrid, Spain
| | - Sabela Díaz-Castroverde
- Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, IdISSC, Instituto de Salud Carlos III, Madrid, Spain
| | - Óscar Escribano
- Biochemistry and Molecular Biology Department, School of Pharmacy, Complutense University of Madrid, Madrid, Spain
- CIBER of Diabetes and Associated Metabolic Diseases, Madrid, Spain
- Instituto de Investigación Sanitaria Hospital Clínico San Carlos, IdISSC, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
36
|
Fidan-Yaylali G, Yaylali YT, Erdogan Ç, Can B, Senol H, Gedik-Topçu B, Topsakal S. The Association between Central Adiposity and Autonomic Dysfunction in Obesity. Med Princ Pract 2016; 25:442-8. [PMID: 27194294 PMCID: PMC5639620 DOI: 10.1159/000446915] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Accepted: 05/18/2016] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE To determine the relationship between central adiposity parameters and autonomic nervous system (ANS) dysfunction. SUBJECTS AND METHODS The study included 114 obese individuals without any cardiovascular risk factors. Weight (in kg), height (in m), and waist circumference (WC; in cm) were measured and body mass index was calculated. Echocardiographic examination was performed to measure left ventricular mass and epicardial fat thickness (EFT). All the participants underwent an exercise test and electrophysiological evaluation using electromyography. Heart rate recovery (HRR) at 1-5 min, R-R interval variation at rest and during hyperventilation, and sympathetic skin response were measured. Pearson's correlation analysis was used. Multiple linear regression analysis was used to identify the factors associated with autonomic dysfunction. RESULTS The HRR at 1-5 min was negatively correlated with WC and age (WC-HRR1: r = -0.32; WC-HRR2: r = -0.31; WC-HRR3: r = -0.26; WC-HRR4: r = -0.23; WC-HRR5: r = -0.21; age-HRR2: r = -0.32; age-HRR3: r = -0.28; age-HRR4: r = -0.41; age-HRR5: r = -0.42). Age was the only independent predictor of reduced HRR at 1-5 min. In addition, WC predicted a reduced HRR at 3 min. There were no significant associations between central obesity and electrophysiological parameters. EFT was not associated with ANS dysfunction. CONCLUSION In this study, central adiposity and aging were associated with ANS dysfunction in obese individuals. The WC could be a marker of ANS dysfunction in obese individuals without any cardiovascular risk factors. The HRR assessment at a later decay phase could be more valuable for evaluating ANS function than during early recovery.
Collapse
Affiliation(s)
- Güzin Fidan-Yaylali
- Department of Endocrinology and Metabolic Diseases, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Yalin Tolga Yaylali
- Department of Cardiology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Çağdaş Erdogan
- Department of Neurology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Beray Can
- Department of Internal Medicine, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Hande Senol
- Department of Biostatistics, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Bengi Gedik-Topçu
- Department of Neurology, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| | - Senay Topsakal
- Department of Endocrinology and Metabolic Diseases, Faculty of Medicine, Pamukkale University, Denizli, Turkey
| |
Collapse
|
37
|
Lopes HF, Corrêa-Giannella ML, Consolim-Colombo FM, Egan BM. Visceral adiposity syndrome. Diabetol Metab Syndr 2016; 8:40. [PMID: 27437032 PMCID: PMC4950710 DOI: 10.1186/s13098-016-0156-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 07/10/2016] [Indexed: 02/08/2023] Open
Abstract
The association of anthropometric (waist circumference) and hemodynamic (blood pressure) changes with abnormalities in glucose and lipid metabolism has been motivation for a lot of discussions in the last 30 years. Nowadays, blood pressure, body mass index/abdominal circumference, glycemia, triglyceridemia, and HDL-cholesterol concentrations are considered in the definition of Metabolic syndrome, referred as Visceral adiposity syndrome (VAS) in the present review. However, more than 250 years ago an association between visceral and mediastinal obesity with hypertension, gout, and obstructive apnea had already been recognized. Expansion of visceral adipose tissue secondary to chronic over-consumption of calories stimulates the recruitment of macrophages, which assume an inflammatory phenotype and produce cytokines that directly interfere with insulin signaling, resulting in insulin resistance. In turn, insulin resistance (IR) manifests itself in various tissues, contributing to the overall phenotype of VAS. For example, in white adipose tissue, IR results in lipolysis, increased free fatty acids release and worsening of inflammation, since fatty acids can bind to Toll-like receptors. In the liver, IR results in increased hepatic glucose production, contributing to hyperglycemia; in the vascular endothelium and kidney, IR results in vasoconstriction, sodium retention and, consequently, arterial hypertension. Other players have been recognized in the development of VAS, such as genetic predisposition, epigenetic factors associated with exposure to an unfavourable intrauterine environment and the gut microbiota. More recently, experimental and clinical studies have shown the autonomic nervous system participates in modulating visceral adipose tissue. The sympathetic nervous system is related to adipose tissue function and differentiation through beta1, beta2, beta3, alpha1, and alpha2 adrenergic receptors. The relation is bidirectional: sympathetic denervation of adipose tissue blocks lipolysis to a variety of lipolytic stimuli and adipose tissue send inputs to the brain. An imbalance of sympathetic/parasympathetic and alpha2 adrenergic/beta3 receptor is related to visceral adipose tissue storage and insulin sensitivity. Thus, in addition to the well-known factors classically associated with VAS, abnormal autonomic activity also emerges as an important factor regulating white adipose tissue, which highlights complex role of adipose tissue in the VAS.
Collapse
Affiliation(s)
- Heno F. Lopes
- />Universidade Nove de Julho-UNINOVE, Rua Vergueiro 235/249, 2 subsolo, Liberdade, São Paulo, CEP: 01504-001 Brazil
- />Instituto do Coração do Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Maria Lúcia Corrêa-Giannella
- />Laboratório de Investigação Médica (LIM-18) e Centro de Terapia Celular e Molecular (NUCEL/NETCEM) da Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP Brazil
| | - Fernanda M. Consolim-Colombo
- />Universidade Nove de Julho-UNINOVE, Rua Vergueiro 235/249, 2 subsolo, Liberdade, São Paulo, CEP: 01504-001 Brazil
- />Instituto do Coração do Hospital das Clinicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo, Brazil
| | - Brent M. Egan
- />Greenville Health System and Department of Medicine, Care Coordination Institute, University of South Carolina-Greenville, Greenville, SC USA
| |
Collapse
|
38
|
Bischoff SJ, Schmidt M, Lehmann T, Schwab M, Matziolis G, Saemann A, Schiffner R. Renal glucose release during hypoglycemia is partly controlled by sympathetic nerves - a study in pigs with unilateral surgically denervated kidneys. Physiol Rep 2015; 3:3/11/e12603. [PMID: 26564063 PMCID: PMC4673633 DOI: 10.14814/phy2.12603] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
Abstract
Catecholamines are known to increase renal glucose release during hypoglycemia. The specific extent of the contribution of different sources of catecholamines, endocrine delivery via circulation or release from autonomous sympathetic renal nerves, though, is unknown. We tested the hypothesis that sympathetic renal innervation plays a major role in the regulation of renal gluconeogenesis. For this purpose, instrumented adolescent pigs had one kidney surgically denervated while the other kidney served as a control. A hypoglycemic clamp with arterial blood glucose below 2 mmol/L was maintained for 75 min. Arteriovenous blood glucose difference, inulin clearance, p-aminohippurate clearance, and sodium excretion were measured in intervals of 15 min separately for both kidneys. Blood glucose was lowered to 0.84 ± 0.33 mmol/L for 75 min. The side-dependent renal net glucose release (SGN) decreased significantly after the unilateral ablation of renal nerves. In the linear mixed model, renal denervation had a significant inhibitory effect on renal net glucose release (P = 0.036). The SGN of the ablated kidney decreased by 0.02 mmol/min and was equivalent to 43.3 ± 23.2% of the control (nonablated) kidney in the pigs. This allows the conclusion that renal glucose release is partly controlled by sympathetic nerves. This may be relevant in humans as well, and could explain the increased risk of severe hypoglycemia of patients with diabetes mellitus and autonomous neuropathy. The effects of denervation on renal glucose metabolism should be critically taken into account when considering renal denervation as a therapy in diabetic patients.
Collapse
Affiliation(s)
- Sabine J Bischoff
- Institute for Laboratory Animals and Welfare, Jena University Hospital, Jena, Germany
| | - Martin Schmidt
- Institute for Biochemistry II, Jena University Hospital, Jena, Germany
| | - Thomas Lehmann
- Institute of Medical Statistics, Computer Sciences and Documentation Science, Jena University Hospital, Jena, Germany
| | - Matthias Schwab
- Department of Neurology, Jena University Hospital, Jena, Germany
| | - Georg Matziolis
- Orthopaedic Department, Jena University Hospital, Jena, Germany
| | - Alexander Saemann
- Department of Internal Medicine II, Helios Hospital, Erfurt, Germany
| | - René Schiffner
- Department of Neurology, Jena University Hospital, Jena, Germany Orthopaedic Department, Jena University Hospital, Jena, Germany
| |
Collapse
|
39
|
Mateos J, Landeira-Abia A, Fafián-Labora JA, Fernández-Pernas P, Lesende-Rodríguez I, Fernández-Puente P, Fernández-Moreno M, Delmiro A, Martín MA, Blanco FJ, Arufe MC. iTRAQ-based analysis of progerin expression reveals mitochondrial dysfunction, reactive oxygen species accumulation and altered proteostasis. Stem Cell Res Ther 2015; 6:119. [PMID: 26066325 PMCID: PMC4487579 DOI: 10.1186/s13287-015-0110-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 10/14/2014] [Accepted: 06/04/2015] [Indexed: 02/08/2023] Open
Abstract
INTRODUCTION Nuclear accumulation of a mutant form of the nuclear protein Lamin-A, called Progerin (PG) or Lamin AΔ50, occurs in Hutchinson-Gilford Progeria Syndrome (HGPS) or Progeria, an accelerated aging disease. One of the main symptoms of this genetic disorder is a loss of sub-cutaneous fat due to a dramatic lipodystrophy. METHODS We stably induced the expression of human PG and GFP -Green Fluorescent Protein- as control in 3T3L1 cells using a lentiviral system to study the effect of PG expression in the differentiation capacity of this cell line, one of the most used adipogenic models. Quantitative proteomics (iTRAQ) was done to study the effect of the PG accumulation. Several of the modulated proteins were validated by immunoblotting and real-time PCR. Mitochondrial function was analyzed by measurement of a) the mitochondrial basal activity, b) the superoxide anion production and c) the individual efficiency of the different complex of the respiratory chain. RESULTS We found that over-expression PG by lentiviral gene delivery leads to a decrease in the proliferation rate and to defects in adipogenic capacity when compared to the control. Quantitative proteomics analysis showed 181 proteins significantly (p<0.05) modulated in PG-expressing preadipocytes. Mitochondrial function is impaired in PG-expressing cells. Specifically, we have detected an increase in the activity of the complex I and an overproduction of Superoxide anion. Incubation with Reactive Oxygen Species (ROS) scavenger agents drives to a decrease in autophagic proteolysis as revealed by LC3-II/LC3-I ratio. CONCLUSION PG expression in 3T3L1 cells promotes changes in several Biological Processes, including structure of cytoskeleton, lipid metabolism, calcium regulation, translation, protein folding and energy generation by the mitochondria. Our data strengthen the contribution of ROS accumulation to the premature aging phenotype and establish a link between mitochondrial dysfunction and loss of proteostasis in HGPS.
Collapse
Affiliation(s)
- Jesús Mateos
- Grupo de Proteómica-ProteoRed/Plataforma PBR2-ISCIII, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña, As Xubias, 15006, A Coruña, Spain.
| | - Arancha Landeira-Abia
- Grupo de Proteómica-ProteoRed/Plataforma PBR2-ISCIII, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña, As Xubias, 15006, A Coruña, Spain.
| | - Juan Antonio Fafián-Labora
- Cellular Therapy and Medicine Regenerative Group, Department of Medicine, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña, As Xubias, 15006, A Coruña, Spain.
| | - Pablo Fernández-Pernas
- Cellular Therapy and Medicine Regenerative Group, Department of Medicine, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña, As Xubias, 15006, A Coruña, Spain.
- Rheumatology Division, CIBER-BBN/ISCII, Instituto de Investigación Biomédica de A Coruña INIBIC-Hospital Universitario A Coruña, 15006, A Coruña, Spain.
| | - Iván Lesende-Rodríguez
- Cellular Therapy and Medicine Regenerative Group, Department of Medicine, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña, As Xubias, 15006, A Coruña, Spain.
| | - Patricia Fernández-Puente
- Grupo de Proteómica-ProteoRed/Plataforma PBR2-ISCIII, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña, As Xubias, 15006, A Coruña, Spain.
| | - Mercedes Fernández-Moreno
- Rheumatology Division, CIBER-BBN/ISCII, Instituto de Investigación Biomédica de A Coruña INIBIC-Hospital Universitario A Coruña, 15006, A Coruña, Spain.
- Grupo de Genómica, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña, As Xubias, 15006, A Coruña, Spain.
| | - Aitor Delmiro
- Laboratorio de Enfermedades Mitocondriales, Instituto de Investigación Hospital 12 de Octubre (i + 12), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), U723, Madrid, E-28041, Spain.
| | - Miguel A Martín
- Laboratorio de Enfermedades Mitocondriales, Instituto de Investigación Hospital 12 de Octubre (i + 12), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), U723, Madrid, E-28041, Spain.
| | - Francisco J Blanco
- Grupo de Proteómica-ProteoRed/Plataforma PBR2-ISCIII, Servicio de Reumatología, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña, As Xubias, 15006, A Coruña, Spain.
- Rheumatology Division, CIBER-BBN/ISCII, Instituto de Investigación Biomédica de A Coruña INIBIC-Hospital Universitario A Coruña, 15006, A Coruña, Spain.
| | - María C Arufe
- Cellular Therapy and Medicine Regenerative Group, Department of Medicine, Instituto de Investigación Biomédica de A Coruña (INIBIC), Complexo Hospitalario Universitario de A Coruña (CHUAC), Sergas, Universidade da Coruña, As Xubias, 15006, A Coruña, Spain.
- Rheumatology Division, CIBER-BBN/ISCII, Instituto de Investigación Biomédica de A Coruña INIBIC-Hospital Universitario A Coruña, 15006, A Coruña, Spain.
| |
Collapse
|
40
|
Ramkisoensing A, Meijer JH. Synchronization of Biological Clock Neurons by Light and Peripheral Feedback Systems Promotes Circadian Rhythms and Health. Front Neurol 2015; 6:128. [PMID: 26097465 PMCID: PMC4456861 DOI: 10.3389/fneur.2015.00128] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 05/19/2015] [Indexed: 12/16/2022] Open
Abstract
In mammals, the suprachiasmatic nucleus (SCN) functions as a circadian clock that drives 24-h rhythms in both physiology and behavior. The SCN is a multicellular oscillator in which individual neurons function as cell-autonomous oscillators. The production of a coherent output rhythm is dependent upon mutual synchronization among single cells and requires both synaptic communication and gap junctions. Changes in phase-synchronization between individual cells have consequences on the amplitude of the SCN’s electrical activity rhythm, and these changes play a major role in the ability to adapt to seasonal changes. Both aging and sleep deprivation negatively affect the circadian amplitude of the SCN, whereas behavioral activity (i.e., exercise) has a positive effect on amplitude. Given that the amplitude of the SCN’s electrical activity rhythm is essential for achieving robust rhythmicity in physiology and behavior, the mechanisms that underlie neuronal synchronization warrant further study. A growing body of evidence suggests that the functional integrity of the SCN contributes to health, well-being, cognitive performance, and alertness; in contrast, deterioration of the 24-h rhythm is a risk factor for neurodegenerative disease, cancer, depression, and sleep disorders.
Collapse
Affiliation(s)
- Ashna Ramkisoensing
- Laboratory for Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center , Leiden , Netherlands
| | - Johanna H Meijer
- Laboratory for Neurophysiology, Department of Molecular Cell Biology, Leiden University Medical Center , Leiden , Netherlands
| |
Collapse
|
41
|
Daubenbüchel AMM, Müller HL. Neuroendocrine Disorders in Pediatric Craniopharyngioma Patients. J Clin Med 2015; 4:389-413. [PMID: 26239246 PMCID: PMC4470135 DOI: 10.3390/jcm4030389] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2014] [Revised: 01/14/2015] [Accepted: 01/29/2015] [Indexed: 12/31/2022] Open
Abstract
Childhood-onset craniopharyngiomas are partly cystic embryonic malformations of the sellar/parasellar region. The therapy of choice in patients with favorable tumor localization is complete resection with a specific focus on maintaining optical and hypothalamic neuroendocrine functions. In patients with unfavorable tumor localization (i.e., hypothalamic involvement), a limited hypothalamus-sparing surgical strategy followed by local irradiation is recommended. Involvement and/or surgical lesions of posterior hypothalamic areas cause major neuroendocrine sequelae. The overall survival rates are high (92%) but neuroendocrine disorders such as obesity and metabolic syndrome due to involvement and/or treatment-related hypothalamic lesions have major negative impact on survival and quality of life. Recurrences and progressions are frequent post-surgical events. Because irradiation is efficient in preventing tumor progression, appropriate timing of post-surgical irradiation is currently under investigation in a randomized multinational trial (KRANIOPHARYNGEOM 2007). Childhood-onset craniopharyngioma should be recognized as a chronic disease requiring treatment and constant monitoring of the clinical and quality of life consequences, frequently impaired due to neuroendocrine disorders, by experienced multidisciplinary teams in order to provide optimal care of surviving patients.
Collapse
Affiliation(s)
- Anna M M Daubenbüchel
- Department of Pediatrics, Klinikum Oldenburg, Medical Campus University Oldenburg, Rahel-Straus-Strasse 10, Oldenburg 26133, Germany.
- University Medical Center Groningen, University of Groningen, Groningen 9713, The Netherlands.
| | - Hermann L Müller
- Department of Pediatrics, Klinikum Oldenburg, Medical Campus University Oldenburg, Rahel-Straus-Strasse 10, Oldenburg 26133, Germany.
| |
Collapse
|
42
|
Bartness TJ, Liu Y, Shrestha YB, Ryu V. Neural innervation of white adipose tissue and the control of lipolysis. Front Neuroendocrinol 2014; 35:473-93. [PMID: 24736043 PMCID: PMC4175185 DOI: 10.1016/j.yfrne.2014.04.001] [Citation(s) in RCA: 242] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 03/10/2014] [Accepted: 04/04/2014] [Indexed: 01/22/2023]
Abstract
White adipose tissue (WAT) is innervated by the sympathetic nervous system (SNS) and its activation is necessary for lipolysis. WAT parasympathetic innervation is not supported. Fully-executed SNS-norepinephrine (NE)-mediated WAT lipolysis is dependent on β-adrenoceptor stimulation ultimately hinging on hormone sensitive lipase and perilipin A phosphorylation. WAT sympathetic drive is appropriately measured electrophysiologically and neurochemically (NE turnover) in non-human animals and this drive is fat pad-specific preventing generalizations among WAT depots and non-WAT organs. Leptin-triggered SNS-mediated lipolysis is weakly supported, whereas insulin or adenosine inhibition of SNS/NE-mediated lipolysis is strongly supported. In addition to lipolysis control, increases or decreases in WAT SNS drive/NE inhibit and stimulate white adipocyte proliferation, respectively. WAT sensory nerves are of spinal-origin and sensitive to local leptin and increases in sympathetic drive, the latter implicating lipolysis. Transsynaptic viral tract tracers revealed WAT central sympathetic and sensory circuits including SNS-sensory feedback loops that may control lipolysis.
Collapse
Affiliation(s)
- Timothy J Bartness
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302-4010, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA.
| | - Yang Liu
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302-4010, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA; Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Yogendra B Shrestha
- Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| | - Vitaly Ryu
- Department of Biology, Center for Obesity Reversal, Georgia State University, Atlanta, GA 30302-4010, USA; Center for Behavioral Neuroscience, Georgia State University, Atlanta, GA 30302-4010, USA; Metabolic Diseases Branch, NIDDK, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
43
|
Abstract
There has been an upsurge of interest in the adipocyte coincident with the onset of the obesity epidemic and the realization that adipose tissue plays a major role in the regulation of metabolic function. The past few years, in particular, have seen significant changes in the way that we classify adipocytes and how we view adipose development and differentiation. We have new perspective on the roles played by adipocytes in a variety of homeostatic processes and on the mechanisms used by adipocytes to communicate with other tissues. Finally, there has been significant progress in understanding how these relationships are altered during metabolic disease and how they might be manipulated to restore metabolic health.
Collapse
Affiliation(s)
- Evan D Rosen
- Division of Endocrinology, Beth Israel Deaconess Medical Center, Boston, MA 02215, USA; Departments of Genetics and Cell Biology, Harvard Medical School, Boston, MA 02215, USA; Broad Institute of Harvard and MIT, Cambridge, MA 02142, USA.
| | - Bruce M Spiegelman
- Departments of Genetics and Cell Biology, Harvard Medical School, Boston, MA 02215, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Murphy KT, Schwartz GJ, Nguyen NLT, Mendez JM, Ryu V, Bartness TJ. Leptin-sensitive sensory nerves innervate white fat. Am J Physiol Endocrinol Metab 2013; 304:E1338-47. [PMID: 23612999 PMCID: PMC3680695 DOI: 10.1152/ajpendo.00021.2013] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Leptin, the primary white adipose tissue (WAT) adipokine, is thought to convey lipid reserve information to the brain via the circulation. Because WAT responds to environmental/internal signals in a fat pad-specific (FPS) manner, systemic signals such as leptin would fail to communicate such distinctive information. Saturation of brain leptin transport systems also would fail to convey increased lipid levels beyond that point. WAT possesses sensory innervation exemplified by proven sensory-associated peptides in nerves within the tissue and by viral sensory nerve-specific transneuronal tract tracer, H129 strain of herpes simplex virus 1 labeling of dorsal root ganglia (DRG) pseudounipolar neurons, spinal cord and central sensory circuits. Leptin as a paracrine factor activating WAT sensory innervation could supply the brain with FPS information. Therefore, we tested for and found the presence of the long form of the leptin receptor (Ob-Rb) on DRG pseudounipolar neurons immunohistochemically labeled after injections of Fluorogold, a retrograde tract tracer, into inguinal WAT (IWAT). Intra-IWAT leptin injections (300 ng) significantly elevated IWAT nerve spike rate within 5 min and persisted for at least 30 min. Intra-IWAT leptin injections also induced significant c-Fos immunoreactivity (ir), indicating neural activation across DRG pseudounipolar sensory neurons labeled with Fluorogold IWAT injections. Intraperitoneal leptin injection did not increase c-Fos-ir in DRG or the arcuate nucleus, nor did it increase arcuate signal transducer and activator of transcription 3 phosphorylation-ir. Collectively, these results strongly suggest that endogenous leptin secreted from white adipocytes functions as a paracrine factor to activate spinal sensory nerves innervating the tissue.
Collapse
Affiliation(s)
- Keegan T Murphy
- Department of Biology, Obesity Reversal Center, Georgia State University, Atlanta, Georgia; and
| | | | | | | | | | | |
Collapse
|
45
|
Czech MP, Tencerova M, Pedersen DJ, Aouadi M. Insulin signalling mechanisms for triacylglycerol storage. Diabetologia 2013; 56:949-64. [PMID: 23443243 PMCID: PMC3652374 DOI: 10.1007/s00125-013-2869-1] [Citation(s) in RCA: 193] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2012] [Accepted: 01/22/2013] [Indexed: 02/06/2023]
Abstract
Insulin signalling is uniquely required for storing energy as fat in humans. While de novo synthesis of fatty acids and triacylglycerol occurs mostly in liver, adipose tissue is the primary site for triacylglycerol storage. Insulin signalling mechanisms in adipose tissue that stimulate hydrolysis of circulating triacylglycerol, uptake of the released fatty acids and their conversion to triacylglycerol are poorly understood. New findings include (1) activation of DNA-dependent protein kinase to stimulate upstream stimulatory factor (USF)1/USF2 heterodimers, enhancing the lipogenic transcription factor sterol regulatory element binding protein 1c (SREBP1c); (2) stimulation of fatty acid synthase through AMP kinase modulation; (3) mobilisation of lipid droplet proteins to promote retention of triacylglycerol; and (4) upregulation of a novel carbohydrate response element binding protein β isoform that potently stimulates transcription of lipogenic enzymes. Additionally, insulin signalling through mammalian target of rapamycin to activate transcription and processing of SREBP1c described in liver may apply to adipose tissue. Paradoxically, insulin resistance in obesity and type 2 diabetes is associated with increased triacylglycerol synthesis in liver, while it is decreased in adipose tissue. This and other mysteries about insulin signalling and insulin resistance in adipose tissue make this topic especially fertile for future research.
Collapse
Affiliation(s)
- M P Czech
- Program in Molecular Medicine, University of Massachusetts Medical School, 373 Plantation Street, Worcester, MA 01605, USA.
| | | | | | | |
Collapse
|
46
|
Salgado-Delgado RC, Saderi N, Basualdo MDC, Guerrero-Vargas NN, Escobar C, Buijs RM. Shift work or food intake during the rest phase promotes metabolic disruption and desynchrony of liver genes in male rats. PLoS One 2013; 8:e60052. [PMID: 23565183 PMCID: PMC3615006 DOI: 10.1371/journal.pone.0060052] [Citation(s) in RCA: 123] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2012] [Accepted: 02/20/2013] [Indexed: 12/11/2022] Open
Abstract
In the liver, clock genes are proposed to drive metabolic rhythms. These gene rhythms are driven by the suprachiasmatic nucleus (SCN) mainly by food intake and via autonomic and hormonal pathways. Forced activity during the normal rest phase, induces also food intake, thus neglecting the signals of the SCN, leading to conflicting time signals to target tissues of the SCN. The present study explored in a rodent model of night-work the influence of food during the normal sleep period on the synchrony of gene expression between clock genes and metabolic genes in the liver. Male Wistar rats were exposed to forced activity for 8 h either during the rest phase (day) or during the active phase (night) by using a slow rotating wheel. In this shift work model food intake shifts spontaneously to the forced activity period, therefore the influence of food alone without induced activity was tested in other groups of animals that were fed ad libitum, or fed during their rest or active phase. Rats forced to be active and/or eating during their rest phase, inverted their daily peak of Per1, Bmal1 and Clock and lost the rhythm of Per2 in the liver, moreover NAMPT and metabolic genes such as Pparα lost their rhythm and thus their synchrony with clock genes. We conclude that shift work or food intake in the rest phase leads to desynchronization within the liver, characterized by misaligned temporal patterns of clock genes and metabolic genes. This may be the cause of the development of the metabolic syndrome and obesity in individuals engaged in shift work.
Collapse
Affiliation(s)
- Roberto C Salgado-Delgado
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Distrito Federal, México
| | | | | | | | | | | |
Collapse
|
47
|
Abstract
A craniopharyngioma (CP) is an embryonic malformation of the sellar and parasellar region. The annual incidence is 0.5-2.0 cases/million/year and approximately 60 % of CP are seen in adulthood. Craniopharyngiomas have the highest mortality of all pituitary tumors. Typical initial manifestations at diagnosis in adults are visual disturbances, hypopituitarism and symptoms of elevated intracranial pressure. The long-term morbidity is substantial with hypopituitarism, increased cardiovascular risk, hypothalamic damage, visual and neurological deficits, reduced bone health, and reduction in quality of life and cognitive function. Therapy of choice is surgery, followed by cranial radiotherapy in about half of the patients. The standardised overall mortality rate varies 2.88-9.28 in cohort studies. Patients with CP have a 3-19 fold higher cardiovascular mortality in comparison to the general population. Women with CP have an even higher risk.
Collapse
Affiliation(s)
- Eva Marie Erfurth
- Department of Endocrinology, Skånes University Hospital, 221 85, Lund, Sweden.
| | | | | |
Collapse
|
48
|
Abstract
Craniopharyngiomas (CP) are partly cystic embryogenic malformations of the sellar and parasellar region. With an overall incidence of 0.5-2.0 new cases/million population/year, approximately 30-50 % of all cases represent childhood CP. Typical manifestations at diagnosis are headache, visual impairment, polyuria/polydypsia, growth retardation, puberty development disturbances, and significant weight gain. Therapy of choice in children with favorable tumor localization is complete resection with the intention to maintain optic nerve and hypothalamic-pituitary functions. In children with unfavorable tumor localization (hypothalamic involvement), a limited resection followed by local irradiation is recommended. Although overall surgical survival rates are high (92 %), recurrence after complete resection and progression after incomplete resection are typical post-surgical events. Particularly troublesome for the pediatric patient are the disturbances to their pubescent development and overall growth. Accordingly, the appropriate time point of irradiation after incomplete resection is under investigation in a randomized multinational trial (KRANIOPHARYNGEOM 2007). Quality of life is substantially reduced in approximately 50 % of long-term survivors due to sequelae, notably morbid hypothalamic obesity. CP should be recognized as a chronic disease requiring constant monitoring of the early life as well as post-pubescent consequences and appropriate medical resources for treatment in order to provide optimal quality of survival for patients.
Collapse
Affiliation(s)
- Hermann L Müller
- Department of Pediatrics, Klinikum Oldenburg, Rahel-Straus-Strasse 10, 26133, Oldenburg, Germany.
| |
Collapse
|
49
|
Abstract
The vagus nerve has an important role in regulation of metabolic homeostasis, and efferent vagus nerve-mediated cholinergic signalling controls immune function and proinflammatory responses via the inflammatory reflex. Dysregulation of metabolism and immune function in obesity are associated with chronic inflammation, a critical step in the pathogenesis of insulin resistance and type 2 diabetes mellitus. Cholinergic mechanisms within the inflammatory reflex have, in the past 2 years, been implicated in attenuating obesity-related inflammation and metabolic complications. This knowledge has led to the exploration of novel therapeutic approaches in the treatment of obesity-related disorders.
Collapse
Affiliation(s)
- Valentin A Pavlov
- Center for Biomedical Science, The Feinstein Institute for Medical Research, 350 Community Drive, Manhasset, NY 11030, USA.
| | | |
Collapse
|
50
|
Adler ES, Hollis JH, Clarke IJ, Grattan DR, Oldfield BJ. Neurochemical characterization and sexual dimorphism of projections from the brain to abdominal and subcutaneous white adipose tissue in the rat. J Neurosci 2012; 32:15913-21. [PMID: 23136429 PMCID: PMC6621617 DOI: 10.1523/jneurosci.2591-12.2012] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 08/05/2012] [Accepted: 08/20/2012] [Indexed: 11/21/2022] Open
Abstract
Retroperitoneal white adipose tissue (rWAT) and subcutaneous (inguinal) white adipose tissue (iWAT) are both innervated and regulated by sympathetic efferents, but the distribution and identity of the cells in the brain that regulate sympathetic outflow are poorly characterized. Our aim was to use two isogenic strains of a neurotropic virus (pseudorabies, Bartha) tagged with either green or red fluorescent reporters to identify cells in the brain that project to rWAT and/or iWAT. These viruses were injected into separate WAT depots in male and female Sprague Dawley rats. Retrogradely labeled neurons in the CNS were characterized by immunohistochemistry and PCR. For the latter, laser capture of individual virally labeled neurons was used. All virally labeled brain regions contained neurons projecting to either and both WAT depots. Neurons to abdominal fat were the most abundant in males, whereas females contained a greater proportion of neurons to subcutaneous via private lines and collateral branches. Retrogradely labeled neurons directed to WAT expressed estrogen receptor-α (ERα), and fewer neurons to subcutaneous WAT expressed ERα in males. Regardless of sex, projections from the arcuate nucleus were predominantly from pro-opiomelanocortin cells, with a notable lack of projections from agouti-related protein-expressing neurons. Within the lateral hypothalamus, neurons directed to rWAT and iWAT expressed orexin and melanin-concentrating hormone (MCH), but male rats had a predominance of MCH directed to iWAT. In conclusion, the neurochemical substrates that project through polysynaptic pathways to iWAT and rWAT are different in male and female rats, suggesting that metabolic regulation of rWAT and iWAT is sexually dimorphic.
Collapse
Affiliation(s)
- Elaine S. Adler
- Department of Physiology, Monash University, Clayton 3800, Victoria, Australia, and
| | - Jacob H. Hollis
- Department of Physiology, Monash University, Clayton 3800, Victoria, Australia, and
| | - Iain J. Clarke
- Department of Physiology, Monash University, Clayton 3800, Victoria, Australia, and
| | - David R. Grattan
- Centre for Neuroendocrinology and Department of Anatomy, University of Otago, Dunedin 9054, New Zealand
| | - Brian J. Oldfield
- Department of Physiology, Monash University, Clayton 3800, Victoria, Australia, and
| |
Collapse
|