151
|
Griffin C, Eter L, Lanzetta N, Abrishami S, Varghese M, McKernan K, Muir L, Lane J, Lumeng CN, Singer K. TLR4, TRIF, and MyD88 are essential for myelopoiesis and CD11c + adipose tissue macrophage production in obese mice. J Biol Chem 2018; 293:8775-8786. [PMID: 29636416 DOI: 10.1074/jbc.ra117.001526] [Citation(s) in RCA: 61] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Revised: 04/04/2018] [Indexed: 12/12/2022] Open
Abstract
Obesity-induced chronic inflammation is associated with metabolic disease. Results from mouse models utilizing a high-fat diet (HFD) have indicated that an increase in activated macrophages, including CD11c+ adipose tissue macrophages (ATMs), contributes to insulin resistance. Obesity primes myeloid cell production from hematopoietic stem cells (HSCs) and Toll-like receptor 4 (TLR4), and the downstream TIR domain-containing adapter protein-inducing interferon-β (TRIF)- and MyD88-mediated pathways regulate production of similar myeloid cells after lipopolysaccharide stimulation. However, the role of these pathways in HFD-induced myelopoiesis is unknown. We hypothesized that saturated fatty acids and HFD alter myelopoiesis by activating TLR4 pathways in HSCs, differentially producing pro-inflammatory CD11c+ myeloid cells that contribute to obesity-induced metabolic disease. Results from reciprocal bone marrow transplants (BMTs) with Tlr4-/- and WT mice indicated that TLR4 is required for HFD-induced myelopoiesis and production of CD11c+ ATMs. Experiments with homozygous knockouts of Irakm (encoding a suppressor of MyD88 inactivation) and Trif in competitive BMTs revealed that MyD88 is required for HFD expansion of granulocyte macrophage progenitors and that Trif is required for pregranulocyte macrophage progenitor expansion. A comparison of WT, Tlr4-/-, Myd88-/-, and Trif-/- mice on HFD demonstrated that TLR4 plays a role in the production of CD11c+ ATMs, and both Myd88-/- and Trif-/- mice produced fewer ATMs than WT mice. Moreover, HFD-induced TLR4 activation inhibited macrophage proliferation, leading to greater accumulation of recruited CD11c+ ATMs. Our results indicate that HFD potentiates TLR4 and both its MyD88- and TRIF-mediated downstream pathways within progenitors and adipose tissue and leads to macrophage polarization.
Collapse
Affiliation(s)
- Cameron Griffin
- From the Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Leila Eter
- From the Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Nico Lanzetta
- From the Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Simin Abrishami
- From the Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Mita Varghese
- From the Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Kaitlin McKernan
- From the Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Lindsey Muir
- From the Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Jamie Lane
- From the Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Carey N Lumeng
- From the Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan 48109
| | - Kanakadurga Singer
- From the Department of Pediatrics and Communicable Disease, University of Michigan Medical School, Ann Arbor, Michigan 48109
| |
Collapse
|
152
|
Deng Y, Xiao Y, Yuan F, Liu Y, Jiang X, Deng J, Fejes-Toth G, Naray-Fejes-Toth A, Chen S, Chen Y, Ying H, Zhai Q, Shu Y, Guo F. SGK1/FOXO3 Signaling in Hypothalamic POMC Neurons Mediates Glucocorticoid-Increased Adiposity. Diabetes 2018; 67:569-580. [PMID: 29321171 DOI: 10.2337/db17-1069] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Accepted: 01/02/2018] [Indexed: 11/13/2022]
Abstract
Although the central nervous system has been implicated in glucocorticoid-induced gain of fat mass, the underlying mechanisms are poorly understood. The aim of this study was to investigate the possible involvement of hypothalamic serum- and glucocorticoid-regulated kinase 1 (SGK1) in glucocorticoid-increased adiposity. It is well known that SGK1 expression is induced by acute glucocorticoid treatment, but it is interesting that we found its expression to be decreased in the arcuate nucleus of the hypothalamus, including proopiomelanocortin (POMC) neurons, following chronic dexamethasone (Dex) treatment. To study the role of SGK1 in POMC neurons, we produced mice that developed or experienced adult-onset SGK1 deletion in POMC neurons (PSKO). As observed in Dex-treated mice, PSKO mice exhibited increased adiposity and decreased energy expenditure. Mice overexpressing constitutively active SGK1 in POMC neurons consistently had the opposite phenotype and did not experience Dex-increased adiposity. Finally, Dex decreased hypothalamic α-melanocyte-stimulating hormone (α-MSH) content and its precursor Pomc expression via SGK1/FOXO3 signaling, and intracerebroventricular injection of α-MSH or adenovirus-mediated FOXO3 knockdown in the arcuate nucleus largely reversed the metabolic alterations in PSKO mice. These results demonstrate that POMC SGK1/FOXO3 signaling mediates glucocorticoid-increased adiposity, providing new insights into the mechanistic link between glucocorticoids and fat accumulation and important hints for possible treatment targets for obesity.
Collapse
Affiliation(s)
- Yalan Deng
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yuzhong Xiao
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Feixiang Yuan
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yaping Liu
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, Beijing Normal University, Beijing, China
| | - Xiaoxue Jiang
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Jiali Deng
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Geza Fejes-Toth
- Department of Physiology, Geisel School of Medicine, Dartmouth College, Lebanon, NH
| | | | - Shanghai Chen
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yan Chen
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Hao Ying
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Qiwei Zhai
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| | - Yousheng Shu
- State Key Laboratory of Cognitive Neuroscience and Learning and IDG/McGovern Institute for Brain Research, School of Brain and Cognitive Sciences, Beijing Normal University, Beijing, China
| | - Feifan Guo
- Key Laboratory of Nutrition and Metabolism, Institute for Nutritional Sciences, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
153
|
Hinder LM, Murdock BJ, Park M, Bender DE, O'Brien PD, Rumora AE, Hur J, Feldman EL. Transcriptional networks of progressive diabetic peripheral neuropathy in the db/db mouse model of type 2 diabetes: An inflammatory story. Exp Neurol 2018; 305:33-43. [PMID: 29550371 DOI: 10.1016/j.expneurol.2018.03.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Revised: 02/14/2018] [Accepted: 03/13/2018] [Indexed: 12/13/2022]
Abstract
Diabetic peripheral neuropathy is the most common complication of diabetes and a source of considerable morbidity. Numerous molecular pathways are linked to neuropathic progression, but it is unclear whether these pathways are altered throughout the course of disease. Moreover, the methods by which these molecular pathways are analyzed can produce significantly different results; as such it is often unclear whether previously published pathways are viable targets for novel therapeutic approaches. In the current study we examine changes in gene expression patterns in the sciatic nerve (SCN) and dorsal root ganglia (DRG) of db/db diabetic mice at 8, 16, and 24 weeks of age using microarray analysis. Following the collection and verification of gene expression data, we utilized both self-organizing map (SOM) analysis and differentially expressed gene (DEG) analysis to detect pathways that were altered at all time points. Though there was some variability between SOM and DEG analyses, we consistently detected altered immune pathways in both the SCN and DRG over the course of disease. To support these results, we further used multiplex analysis to assess protein changes in the SCN of diabetic mice; we found that multiple immune molecules were upregulated at both early and later stages of disease. In particular, we found that matrix metalloproteinase-12 was highly upregulated in microarray and multiplex data sets suggesting it may play a role in disease progression.
Collapse
Affiliation(s)
- Lucy M Hinder
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Benjamin J Murdock
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Meeyoung Park
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Diane E Bender
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Phillipe D O'Brien
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Amy E Rumora
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Junguk Hur
- Department of Biomedical Sciences, School of Medicine and Health Sciences, University of North Dakota, Grand Forks, ND 58203-9037, USA
| | - Eva L Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA.
| |
Collapse
|
154
|
Sodium butyrate triggers a functional elongation of microglial process via Akt-small RhoGTPase activation and HDACs inhibition. Neurobiol Dis 2018; 111:12-25. [DOI: 10.1016/j.nbd.2017.12.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Revised: 11/24/2017] [Accepted: 12/11/2017] [Indexed: 12/18/2022] Open
|
155
|
Korneev KV, Kondakova AN, Sviriaeva EN, Mitkin NA, Palmigiano A, Kruglov AA, Telegin GB, Drutskaya MS, Sturiale L, Garozzo D, Nedospasov SA, Knirel YA, Kuprash DV. Hypoacylated LPS from Foodborne Pathogen Campylobacter jejuni Induces Moderate TLR4-Mediated Inflammatory Response in Murine Macrophages. Front Cell Infect Microbiol 2018; 8:58. [PMID: 29535976 PMCID: PMC5835049 DOI: 10.3389/fcimb.2018.00058] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2017] [Accepted: 02/12/2018] [Indexed: 11/13/2022] Open
Abstract
Toll-like receptor 4 (TLR4) initiates immune response against Gram-negative bacteria upon specific recognition of lipid A moiety of lipopolysaccharide (LPS), the major component of their cell wall. Some natural differences between LPS variants in their ability to interact with TLR4 may lead to either insufficient activation that may not prevent bacterial growth, or excessive activation which may lead to septic shock. In this study we evaluated the biological activity of LPS isolated from pathogenic strain of Campylobacter jejuni, the most widespread bacterial cause of foodborne diarrhea in humans. With the help of hydrophobic chromatography and MALDI-TOF mass spectrometry we showed that LPS from a C. jejuni strain O2A consists of both hexaacyl and tetraacyl forms. Since such hypoacylation can result in a reduced immune response in humans, we assessed the activity of LPS from C. jejuni in mouse macrophages by measuring its capacity to activate TLR4-mediated proinflammatory cytokine and chemokine production, as well as NFκB-dependent reporter gene transcription. Our data support the hypothesis that LPS acylation correlates with its bioactivity.
Collapse
Affiliation(s)
- Kirill V. Korneev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Immunology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| | - Anna N. Kondakova
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Ekaterina N. Sviriaeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Immunology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| | - Nikita A. Mitkin
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Angelo Palmigiano
- CNR Institute for Polymers Composites and Biomaterials, Catania, Italy
| | - Andrey A. Kruglov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- German Rheumatism Research Center, Leibniz Institute, Berlin, Germany
| | - Georgy B. Telegin
- Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Pushchino, Russia
| | - Marina S. Drutskaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
| | - Luisa Sturiale
- CNR Institute for Polymers Composites and Biomaterials, Catania, Italy
| | - Domenico Garozzo
- CNR Institute for Polymers Composites and Biomaterials, Catania, Italy
| | - Sergei A. Nedospasov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Immunology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow, Russia
- German Rheumatism Research Center, Leibniz Institute, Berlin, Germany
| | - Yuriy A. Knirel
- Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Dmitry V. Kuprash
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, Moscow, Russia
- Department of Immunology, Biological Faculty, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
156
|
Hryhorczuk C, Sheng Z, Décarie-Spain L, Giguère N, Ducrot C, Trudeau LÉ, Routh VH, Alquier T, Fulton S. Oleic Acid in the Ventral Tegmental Area Inhibits Feeding, Food Reward, and Dopamine Tone. Neuropsychopharmacology 2018; 43:607-616. [PMID: 28857071 PMCID: PMC5770761 DOI: 10.1038/npp.2017.203] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 08/23/2017] [Accepted: 08/28/2017] [Indexed: 12/16/2022]
Abstract
Long-chain fatty acids (FAs) act centrally to decrease food intake and hepatic glucose production and alter hypothalamic neuronal activity in a manner that depends on FA type and cellular transport proteins. However, it is not known whether FAs are sensed by ventral tegmental area (VTA) dopamine (DA) neurons to control food-motivated behavior and DA neurotransmission. We investigated the impact of the monounsaturated FA oleate in the VTA on feeding, locomotion, food reward, and DA neuronal activity and DA neuron expression of FA-handling proteins and FA uptake. A single intra-VTA injection of oleate, but not of the saturated FA palmitate, decreased food intake and increased locomotor activity. Furthermore, intra-VTA oleate blunted the rewarding effects of high-fat/sugar food in an operant task and inhibited DA neuronal firing. Using sorted DA neuron preparations from TH-eGFP mice we found that DA neurons express FA transporter and binding proteins, and are capable of intracellular transport of long-chain FA. Finally, we demonstrate that a transporter blocker attenuates FA uptake into DA neurons and blocks the effects of intra-VTA oleate to decrease food-seeking and DA neuronal activity. Together, these results suggest that DA neurons detect FA and that oleate has actions in the VTA to suppress DA neuronal activity and food seeking following cellular incorporation. These findings highlight the capacity of DA neurons to act as metabolic sensors by responding not only to hormones but also to FA nutrient signals to modulate food-directed behavior.
Collapse
Affiliation(s)
- Cecile Hryhorczuk
- CRCHUM and Montreal Diabetes Research Center, Montréal, QC, Canada
- Department of Physiology, Université de Montréal, Montréal, QC, Canada
| | - Zhenyu Sheng
- Rutgers New Jersey Medical School, Department of Pharmacology, Physiology and Neuroscience, Rutgers University, Newark, NJ, USA
| | - Léa Décarie-Spain
- CRCHUM and Montreal Diabetes Research Center, Montréal, QC, Canada
- Department of Neurosciences, Université de Montréal, Montréal, QC, Canada
| | - Nicolas Giguère
- Department of Pharmacology, Université de Montréal, Montréal, QC, Canada
| | - Charles Ducrot
- Department of Pharmacology, Université de Montréal, Montréal, QC, Canada
| | - Louis-Éric Trudeau
- Department of Pharmacology, Université de Montréal, Montréal, QC, Canada
| | - Vanessa H Routh
- Rutgers New Jersey Medical School, Department of Pharmacology, Physiology and Neuroscience, Rutgers University, Newark, NJ, USA
| | - Thierry Alquier
- CRCHUM and Montreal Diabetes Research Center, Montréal, QC, Canada
- Department of Medicine, Université de Montréal, Montréal, QC, Canada
| | - Stephanie Fulton
- CRCHUM and Montreal Diabetes Research Center, Montréal, QC, Canada
- Department of Nutrition, Université de Montréal, Montréal, QC, Canada
| |
Collapse
|
157
|
Carraro RS, Souza GF, Solon C, Razolli DS, Chausse B, Barbizan R, Victorio SC, Velloso LA. Hypothalamic mitochondrial abnormalities occur downstream of inflammation in diet-induced obesity. Mol Cell Endocrinol 2018; 460:238-245. [PMID: 28760600 DOI: 10.1016/j.mce.2017.07.029] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2017] [Revised: 07/27/2017] [Accepted: 07/27/2017] [Indexed: 10/19/2022]
Abstract
Hypothalamic dysfunction is a common feature of experimental obesity. Studies have identified at least three mechanisms involved in the development of hypothalamic neuronal defects in diet-induced obesity: i, inflammation; ii, endoplasmic reticulum stress; and iii, mitochondrial abnormalities. However, which of these mechanisms is activated earliest in response to the consumption of large portions of dietary fats is currently unknown. Here, we used immunoblot, real-time PCR, mitochondrial respiration assays and transmission electron microscopy to evaluate markers of inflammation, endoplasmic reticulum stress and mitochondrial abnormalities in the hypothalamus of Swiss mice fed a high-fat diet for up to seven days. In the present study we show that the expression of the inflammatory chemokine fractalkine was the earliest event detected. Its hypothalamic expression increased as early as 3 h after the introduction of a high-fat diet and was followed by the increase of cytokines. GPR78, an endoplasmic reticulum chaperone, was increased 6 h after the introduction of a high-fat diet, however the actual triggering of endoplasmic reticulum stress was only detected three days later, when IRE-1α was increased. Mitofusin-2, a protein involved in mitochondrial fusion and tethering of mitochondria to the endoplasmic reticulum, underwent a transient reduction 24 h after the introduction of a high-fat diet and then increased after seven days. There were no changes in hypothalamic mitochondrial respiration during the experimental period, however there were reductions in mitochondria/endoplasmic reticulum contact sites, beginning three days after the introduction of a high-fat diet. The inhibition of TNF-α with infliximab resulted in the normalization of mitofusin-2 levels 24 h after the introduction of the diet. Thus, inflammation is the earliest mechanism activated in the hypothalamus after the introduction of a high-fat diet and may play a mechanistic role in the development of mitochondrial abnormalities in diet-induced obesity.
Collapse
Affiliation(s)
- Rodrigo S Carraro
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, 13084-970 Campinas, SP, Brazil
| | - Gabriela F Souza
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, 13084-970 Campinas, SP, Brazil
| | - Carina Solon
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, 13084-970 Campinas, SP, Brazil
| | - Daniela S Razolli
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, 13084-970 Campinas, SP, Brazil
| | - Bruno Chausse
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, 13084-970 Campinas, SP, Brazil; Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, 05508-000 São Paulo, Brazil
| | - Roberta Barbizan
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, 13084-970 Campinas, SP, Brazil
| | - Sheila C Victorio
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, 13084-970 Campinas, SP, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, 13084-970 Campinas, SP, Brazil.
| |
Collapse
|
158
|
Ramalho AF, Bombassaro B, Dragano NR, Solon C, Morari J, Fioravante M, Barbizan R, Velloso LA, Araujo EP. Dietary fats promote functional and structural changes in the median eminence blood/spinal fluid interface-the protective role for BDNF. J Neuroinflammation 2018; 15:10. [PMID: 29316939 PMCID: PMC5761204 DOI: 10.1186/s12974-017-1046-8] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 12/20/2017] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND The consumption of large amounts of dietary fats activates an inflammatory response in the hypothalamus, damaging key neurons involved in the regulation of caloric intake and energy expenditure. It is currently unknown why the mediobasal hypothalamus is the main target of diet-induced brain inflammation. We hypothesized that dietary fats can damage the median eminence blood/spinal fluid interface. METHODS Swiss mice were fed on a high-fat diet, and molecular and structural studies were performed employing real-time PCR, immunoblot, immunofluorescence, transmission electron microscopy, and metabolic measurements. RESULTS The consumption of a high fat diet was sufficient to increase the expression of inflammatory cytokines and brain-derived neurotrophic factor in the median eminence, preceding changes in other circumventricular regions. In addition, it led to an early loss of the structural organization of the median eminence β1-tanycytes. This was accompanied by an increase in the hypothalamic expression of brain-derived neurotrophic factor. The immunoneutralization of brain-derived neurotrophic factor worsened diet-induced functional damage of the median eminence blood/spinal fluid interface, increased diet-induced hypothalamic inflammation, and increased body mass gain. CONCLUSIONS The median eminence/spinal fluid interface is affected at the functional and structural levels early after introduction of a high-fat diet. Brain-derived neurotrophic factor provides an early protection against damage, which is lost upon a persisting consumption of large amounts of dietary fats.
Collapse
Affiliation(s)
- Albina F Ramalho
- LAV, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Bruna Bombassaro
- LAV, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Nathalia R Dragano
- LAV, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Carina Solon
- LAV, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Joseane Morari
- LAV, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Milena Fioravante
- LAV, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Roberta Barbizan
- LAV, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Licio A Velloso
- LAV, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-970, Brazil.
| | - Eliana P Araujo
- Faculty of Nursing, University of Campinas, Campinas, SP, 13084-970, Brazil
| |
Collapse
|
159
|
The effects of overnight nutrient intake on hypothalamic inflammation in a free-choice diet-induced obesity rat model. Appetite 2018; 120:527-535. [PMID: 28988760 DOI: 10.1016/j.appet.2017.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 10/03/2017] [Accepted: 10/03/2017] [Indexed: 12/31/2022]
|
160
|
Zhou YD. Glial Regulation of Energy Metabolism. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2018; 1090:105-121. [PMID: 30390287 DOI: 10.1007/978-981-13-1286-1_6] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
161
|
Abstract
Leptin is an adipocyte-derived hormone, which contributes to the homeostatic regulation of energy balance and metabolism through humoral and neural pathways. Leptin acts on the neurons in certain brain areas such as the hypothalamus, hippocampus, and brain stem to regulate food intake, thermogenesis, energy expenditure, and homeostasis of glucose/lipid metabolism. The pathologically increased circulating leptin is a biomarker of leptin resistance, which is common in obese individuals. Leptin resistance is defined by a reduced sensitivity or a failure in response of the brain to leptin, showing a decrease in the ability of leptin to suppress appetite or enhance energy expenditure, which causes an increased food intake and finally leads to overweight, obesity, cardiovascular diseases, and other metabolic disorders. Leptin resistance is a challenge for clinical treatment or drug discovery of obesity. Until recently, emerging evidence has been showing novel mechanisms of the leptin resistance. Here, we summarized the advances and controversy of leptin resistance and associated diseases, for better understanding the physiology and pathophysiology of leptin as well as the new strategies for treating obesity and metabolic disorders.
Collapse
|
162
|
Ho JM, Ducich NH, Nguyen NQK, Opp MR. Acute sleep disruption- and high-fat diet-induced hypothalamic inflammation are not related to glucose tolerance in mice. Neurobiol Sleep Circadian Rhythms 2018; 4:1-9. [PMID: 29732438 PMCID: PMC5931726 DOI: 10.1016/j.nbscr.2017.09.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Chronic insufficient sleep is a major societal problem and is associated with increased risk of metabolic disease. Hypothalamic inflammation contributes to hyperphagia and weight gain in diet-induced obesity, but insufficient sleep-induced neuroinflammation has yet to be examined in relation to metabolic function. We therefore fragmented sleep of adult male C57BL/6J mice for 18 h daily for 9 days to determine whether sleep disruption elicits inflammatory responses in brain regions that regulate energy balance and whether this relates to glycemic control. To additionally test the hypothesis that exposure to multiple inflammatory factors exacerbates metabolic outcomes, responses were compared in mice exposed to sleep fragmentation (SF), high-fat diet (HFD), both SF and HFD, or control conditions. Three or 9 days of high-fat feeding reduced glucose tolerance but SF alone did not. Transient loss of body mass in SF mice may have affected outcomes. Comparisons of pro-inflammatory cytokine concentrations among central and peripheral metabolic tissues indicate that patterns of liver interleukin-1β concentrations best reflects observed changes in glucose tolerance. However, we demonstrate that SF rapidly and potently increases Iba1 immunoreactivity (-ir), a marker of microglia. After 9 days of manipulations, Iba1-ir remains elevated only in mice exposed to both SF and HFD, indicating a novel interaction between sleep and diet on microglial activation that warrants further investigation.
Collapse
Affiliation(s)
- Jacqueline M. Ho
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Nicole H. Ducich
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Nhat-Quynh K. Nguyen
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
| | - Mark R. Opp
- Department of Anesthesiology & Pain Medicine, University of Washington, Seattle, WA, USA
- Program in Neurobiology and Behavior, University of Washington, Seattle, Washington, USA
- Corresponding author. Present addrss: Department of Integrative Physiology, University of Colorado, UCB 354, 2860 Wilderness Place, 201K, Boulder, CO 80301, USA.
| |
Collapse
|
163
|
Argente-Arizón P, Díaz F, Ros P, Barrios V, Tena-Sempere M, García-Segura LM, Argente J, Chowen JA. The Hypothalamic Inflammatory/Gliosis Response to Neonatal Overnutrition Is Sex and Age Dependent. Endocrinology 2018; 159:368-387. [PMID: 29077836 DOI: 10.1210/en.2017-00539] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 10/20/2017] [Indexed: 01/10/2023]
Abstract
Astrocytes participate in both physiological and pathophysiological responses to metabolic and nutrient signals. Although most studies have focused on the astrocytic response to weight gain due to high-fat/high-carbohydrate intake, surplus intake of a balanced diet also induces excess weight gain. We have accessed the effects of neonatal overnutrition, which has both age- and sex-dependent effects on weight gain, on hypothalamic inflammation/gliosis. Although both male and female Wistar rats accumulate excessive fat mass as early as postnatal day (PND) 10 with neonatal overnutrition, no increase in hypothalamic cytokine levels, markers of astrocytes or microglia, or inflammatory signaling pathways were observed. At PND 50, no effect of neonatal overnutriton was found in either sex, whereas at PND 150, males again weighed significantly more than their controls, and this was coincident with an increase in markers of inflammation and astrogliosis in the hypothalamus. Circulating triglycerides and free fatty acids were also elevated in these males, but not in females or in either sex at PND 10. Thus, the effects of fatty acids and estrogens on astrocytes in vitro were analyzed. Our results indicate that changes in circulating fatty acid levels may be involved in the induction of hypothalamic inflammation/gliosis in excess weight gain, even on a normal diet, and that estrogens could participate in the protection of females from these processes. In conclusion, the interaction of developmental influences, dietary composition, age, and sex determines the central inflammatory response and the associated long-term outcomes of excess weight gain.
Collapse
Affiliation(s)
- Pilar Argente-Arizón
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, Spain
- Department of Pediatrics, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición, Instituto Carlos III, Madrid, Spain
| | - Francisca Díaz
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición, Instituto Carlos III, Madrid, Spain
| | - Purificación Ros
- Department of Pediatrics, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Hospital Universitario Puerto de Hierro-Majadahonda, Madrid, Spain
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición, Instituto Carlos III, Madrid, Spain
| | - Manuel Tena-Sempere
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición, Instituto Carlos III, Madrid, Spain
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, Instituto Maimónides de Investigación Biomédica de Córdoba, Córdoba, Spain
| | - Luis Miguel García-Segura
- Instituto Cajal, Consejo Superior de Investigaciones Científicas, Centro de Investigación Biomédica en Red de Fragilidad y Envejecimiento Saludable, Instituto Carlos III, Madrid, Spain
| | - Jesús Argente
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, Spain
- Department of Pediatrics, Facultad de Medicina, Universidad Autónoma de Madrid, Madrid, Spain
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición, Instituto Carlos III, Madrid, Spain
- Instituto Madrileño de Estudios Svanzados Food Institute, Campus de Excelencia Internacional Universidad Autónoma de Madrid + Consejo Superior de Investigaciones Científicas, Madrid, Spain
| | - Julie A Chowen
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación la Princesa, Madrid, Spain
- Centro de Investigación Biomédica en Red: Fisiopatología de la Obesidad y Nutrición, Instituto Carlos III, Madrid, Spain
| |
Collapse
|
164
|
Woo YS, Bahk WM. The Link Between Obesity and Depression: Exploring Shared Mechanisms. UNDERSTANDING DEPRESSION 2018:203-220. [DOI: 10.1007/978-981-10-6577-4_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
165
|
Kaneko K, Xu P, Cordonier EL, Chen SS, Ng A, Xu Y, Morozov A, Fukuda M. Neuronal Rap1 Regulates Energy Balance, Glucose Homeostasis, and Leptin Actions. Cell Rep 2017; 16:3003-3015. [PMID: 27626668 DOI: 10.1016/j.celrep.2016.08.039] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 06/30/2016] [Accepted: 08/11/2016] [Indexed: 10/21/2022] Open
Abstract
The CNS contributes to obesity and metabolic disease; however, the underlying neurobiological pathways remain to be fully established. Here, we show that the small GTPase Rap1 is expressed in multiple hypothalamic nuclei that control whole-body metabolism and is activated in high-fat diet (HFD)-induced obesity. Genetic ablation of CNS Rap1 protects mice from dietary obesity, glucose imbalance, and insulin resistance in the periphery and from HFD-induced neuropathological changes in the hypothalamus, including diminished cellular leptin sensitivity and increased endoplasmic reticulum (ER) stress and inflammation. Furthermore, pharmacological inhibition of CNS Rap1 signaling normalizes hypothalamic ER stress and inflammation, improves cellular leptin sensitivity, and reduces body weight in mice with dietary obesity. We also demonstrate that Rap1 mediates leptin resistance via interplay with ER stress. Thus, neuronal Rap1 critically regulates leptin sensitivity and mediates HFD-induced obesity and hypothalamic pathology and may represent a potential therapeutic target for obesity treatment.
Collapse
Affiliation(s)
- Kentaro Kaneko
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Pingwen Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Elizabeth L Cordonier
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Siyu S Chen
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Amy Ng
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Yong Xu
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular and Cellular Biology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX 77030, USA
| | - Alexei Morozov
- Unit on Behavioral Genetics, Laboratory of Molecular Pathophysiology, National Institute of Mental Health, National Institutes of Health, Bethesda, MD 20892, USA; Virginia Tech Carilion Research Institute, Roanoke, VA 24016, USA
| | - Makoto Fukuda
- Children's Nutrition Research Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA.
| |
Collapse
|
166
|
Spiljar M, Merkler D, Trajkovski M. The Immune System Bridges the Gut Microbiota with Systemic Energy Homeostasis: Focus on TLRs, Mucosal Barrier, and SCFAs. Front Immunol 2017; 8:1353. [PMID: 29163467 PMCID: PMC5670327 DOI: 10.3389/fimmu.2017.01353] [Citation(s) in RCA: 134] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/03/2017] [Indexed: 12/17/2022] Open
Abstract
The gut microbiota is essential for the development and regulation of the immune system and the metabolism of the host. Germ-free animals have altered immunity with increased susceptibility to immunologic diseases and show metabolic alterations. Here, we focus on two of the major immune-mediated microbiota-influenced components that signal far beyond their local environment. First, the activation or suppression of the toll-like receptors (TLRs) by microbial signals can dictate the tone of the immune response, and they are implicated in regulation of the energy homeostasis. Second, we discuss the intestinal mucosal surface is an immunologic component that protects the host from pathogenic invasion, is tightly regulated with regard to its permeability and can influence the systemic energy balance. The short chain fatty acids are a group of molecules that can both modulate the intestinal barrier and escape the gut to influence systemic health. As modulators of the immune response, the microbiota-derived signals influence functions of distant organs and can change susceptibility to metabolic diseases.
Collapse
Affiliation(s)
- Martina Spiljar
- Faculty of Medicine, Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.,Diabetes Center, Faculty of Medicine, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Doron Merkler
- Faculty of Medicine, Department of Pathology and Immunology, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland
| | - Mirko Trajkovski
- Faculty of Medicine, Department of Cell Physiology and Metabolism, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.,Diabetes Center, Faculty of Medicine, Centre Médical Universitaire, University of Geneva, Geneva, Switzerland.,Institute of Genetics and Genomics in Geneva (iGE3), University of Geneva, Geneva, Switzerland
| |
Collapse
|
167
|
Huang C, Wang P, Xu X, Zhang Y, Gong Y, Hu W, Gao M, Wu Y, Ling Y, Zhao X, Qin Y, Yang R, Zhang W. The ketone body metabolite β-hydroxybutyrate induces an antidepression-associated ramification of microglia via HDACs inhibition-triggered Akt-small RhoGTPase activation. Glia 2017; 66:256-278. [PMID: 29058362 DOI: 10.1002/glia.23241] [Citation(s) in RCA: 111] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2017] [Revised: 09/20/2017] [Accepted: 09/21/2017] [Indexed: 12/27/2022]
Abstract
Direct induction of macrophage ramification has been shown to promote an alternative (M2) polarization, suggesting that the ramified morphology may determine the function of immune cells. The ketone body metabolite β-hydroxybutyrate (BHB) elevated in conditions including fasting and low-carbohydrate ketogenic diet (KD) can reduce neuroinflammation. However, how exactly BHB impacts microglia remains unclear. We report that BHB as well as its producing stimuli fasting and KD induced obvious ramifications of murine microglia in basal and inflammatory conditions in a reversible manner, and these ramifications were accompanied with microglial profile toward M2 polarization and phagocytosis. The protein kinase B (Akt)-small RhoGTPase axis was found to mediate the effect of BHB on microglial shape change, as (i) BHB activated the microglial small RhoGTPase (Rac1, Cdc42) and Akt; (ii) Akt and Rac1-Cdc42 inhibition abolished the pro-ramification effect of BHB; (iii) Akt inhibition prevented the activation of Rac1-Cdc42 induced by BHB treatment. Incubation of microglia with other classical histone deacetylases (HDACs) inhibitors, but not G protein-coupled receptor 109a (GPR109a) activators, also induced microglial ramification and Akt activation, suggesting that the BHB-induced ramification of microglia may be triggered by HDACs inhibition. Functionally, Akt inhibition was found to abrogate the effects of BHB on microglial polarization and phagocytosis. In neuroinflammatory models induced by lipopolysaccharide (LPS) or chronic unpredictable stress (CUS), BHB prevented the microglial process retraction and depressive-like behaviors, and these effects were abolished by Akt inhibition. Our findings for the first time showed that BHB exerts anti-inflammatory actions via promotion of microglial ramification.
Collapse
Affiliation(s)
- Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Peng Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Xing Xu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Yaru Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Yu Gong
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Wenfeng Hu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Minhui Gao
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Yue Wu
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Yong Ling
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Xi Zhao
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| | - Yibin Qin
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Jiangsu Province, #20 Xisi Road, Nantong, Jiangsu Province, 226001, China
| | - Rongrong Yang
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Jiangsu Province, #20 Xisi Road, Nantong, Jiangsu Province, 226001, China
| | - Wei Zhang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong, Jiangsu Province, 226001, China
| |
Collapse
|
168
|
Hotamisligil GS. Foundations of Immunometabolism and Implications for Metabolic Health and Disease. Immunity 2017; 47:406-420. [PMID: 28930657 DOI: 10.1016/j.immuni.2017.08.009] [Citation(s) in RCA: 318] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Revised: 06/06/2017] [Accepted: 08/16/2017] [Indexed: 02/06/2023]
Abstract
Highly ordered interactions between immune and metabolic responses are evolutionarily conserved and paramount for tissue and organismal health. Disruption of these interactions underlies the emergence of many pathologies, particularly chronic non-communicable diseases such as obesity and diabetes. Here, we examine decades of research identifying the complex immunometabolic signaling networks and the cellular and molecular events that occur in the setting of altered nutrient and energy exposures and offer a historical perspective. Furthermore, we describe recent advances such as the discovery that a broad complement of immune cells play a role in immunometabolism and the emerging evidence that nutrients and metabolites modulate inflammatory pathways. Lastly, we discuss how this work may eventually lead to tangible therapeutic advancements to promote health.
Collapse
Affiliation(s)
- Gökhan S Hotamisligil
- Department of Genetics and Complex Diseases and Sabri Ülker Center, Harvard T.H. Chan School of Public Health, Broad Institute of Harvard and MIT, Boston, MA 02115, USA.
| |
Collapse
|
169
|
Dong S, Qi M, Wang Y, Chen L, Weaver JC, Krilis SA, Giannakopoulos B. β2GPI exerts an anti-obesity effect in female mice by inhibiting lipogenesis and promoting lipolysis. Oncotarget 2017; 8:92652-92666. [PMID: 29190946 PMCID: PMC5696212 DOI: 10.18632/oncotarget.21536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2017] [Accepted: 07/12/2017] [Indexed: 02/01/2023] Open
Abstract
In humans, males compared to females have increased visceral adipose tissue which contributes to their increased risk of early death. Mice display analogous sexual diamorphism whereby females are protected from weight gain when fed a high fat diet compared to males. A role has recently been reported for β2-glycoprotein I, an abundant plasma protein, in healthy leanness in humans. In this study we investigated the role of β2-glycoprotein I in fat metabolism in male and female mice fed a normal chow or high fat diet. We have made a number of novel insights into factors contributing to sexual diamorphism in obesity. Female wild type mice are protected from obesity when fed a high fat diet due to down regulation of lipogenesis in the visceral adipose tissues. This down regulation is due to β2-glycoprotein I as female mice deficient in this protein have increased levels of lipogenesis enzymes in their visceral adipose tissues with an accompanying increase in weight compared to female wild type controls. Understanding female specific regulators of obesity may lead to sex specific anti-obesity therapies to address this major health problem.
Collapse
Affiliation(s)
- Shangwen Dong
- Department of Infectious Diseases, Immunology and Sexual Health and Department of Medicine, St George Hospital, University of New South Wales, New South Wales, Sydney, Australia.,Department of Cardiothoracic Surgery, Tianjin Medical University General Hospital, Tianjin Medical University, Tianjin, China
| | - Miao Qi
- Department of Infectious Diseases, Immunology and Sexual Health and Department of Medicine, St George Hospital, University of New South Wales, New South Wales, Sydney, Australia
| | - Ying Wang
- Department of Infectious Diseases, Immunology and Sexual Health and Department of Medicine, St George Hospital, University of New South Wales, New South Wales, Sydney, Australia.,Laboratory of Hormones and Development (Ministry of Health), Metabolic Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - Liming Chen
- Laboratory of Hormones and Development (Ministry of Health), Metabolic Hospital and Tianjin Institute of Endocrinology, Tianjin Medical University, Tianjin, China
| | - James Crofton Weaver
- Department of Cardiology, St George Hospital, New South Wales, Sydney, Australia
| | - Steven Antony Krilis
- Department of Infectious Diseases, Immunology and Sexual Health and Department of Medicine, St George Hospital, University of New South Wales, New South Wales, Sydney, Australia
| | - Bill Giannakopoulos
- Department of Infectious Diseases, Immunology and Sexual Health and Department of Medicine, St George Hospital, University of New South Wales, New South Wales, Sydney, Australia.,Department of Rheumatology, St George Hospital, New South Wales, Sydney, Australia
| |
Collapse
|
170
|
Liu F, Wu J, Gong Y, Wang P, Zhu L, Tong L, Chen X, Ling Y, Huang C. Harmine produces antidepressant-like effects via restoration of astrocytic functions. Prog Neuropsychopharmacol Biol Psychiatry 2017. [PMID: 28625859 DOI: 10.1016/j.pnpbp.2017.06.012] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Depression is a world-wide disease with no effective therapeutic methods. Increasing evidence indicates that astrocytic pathology contributes to the formation of depression. In this study, we investigated the effects of harmine, a natural β-carboline alkaloid and potent hallucinogen, known to modulate astrocytic glutamate transporters, on chronic unpredictable stress (CUS)-induced depressive-like behaviors and astrocytic dysfunctions. Results showed that harmine treatment (10, 20mg/kg) protected the mice against the CUS-induced increases in the immobile time in the tail suspension test (TST) and forced swimming test (FST), and also reversed the reduction in sucrose intake in the sucrose preference experiment. Harmine treatment (20mg/kg) prevented the reductions in brain-derived neurotrophic factor (BDNF) protein levels and hippocampal neurogenesis induced by CUS. In addition, harmine treatment (20mg/kg) increased the protein expression levels of glutamate transporter 1 (GLT-1) and prevented the CUS-induced decreases in glial fibrillary acidic protein (GFAP) protein expressions in the prefrontal cortex and hippocampus, suggesting that restoration of astrocytic functions may be a potential mechanism underlying the antidepressant-like effects of harmine. This opinion was proved by the results that administration of mice with l-Alpha-Aminoadipic Acid (L-AAA), a gliotoxin specific for astrocytes, attenuated the antidepressant-like effects of harmine, and prevented the improvement effects of harmine on BDNF protein levels and hippocampal neurogenesis. These results provide further evidence to confirm that astrocytic dysfunction contributes critically to the development of depression and that harmine exerts antidepressant-like effects likely through restoration of astrocytic functions.
Collapse
Affiliation(s)
- Fengguo Liu
- Department of Neurology, Danyang People's Hospital, #2 Xinmin Western Road, Danyang 212300, Jiangsu, China
| | - Jingjing Wu
- Department of Cardiology, Suzhou Kowloon Hospital of Shanghai Jiaotong University School of Medicine, #118 Wansheng Street, Suzhou 215021, Jiangsu, China
| | - Yu Gong
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Peng Wang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Lei Zhu
- Department of Pharmacy, First People's Hospital of Yancheng, Yulong Western Road, Yancheng 224006, Jiangsu, China
| | - Lijuan Tong
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Xiangfan Chen
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Yong Ling
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China
| | - Chao Huang
- Department of Pharmacology, School of Pharmacy, Nantong University, #19 Qixiu Road, Nantong 226001, Jiangsu, China; Key Laboratory of Inflammation and Molecular Drug Target of Jiangsu Province, #19 Qixiu Road, Nantong 226001, Jiangsu, China.
| |
Collapse
|
171
|
Dodd GT, Tiganis T. Insulin action in the brain: Roles in energy and glucose homeostasis. J Neuroendocrinol 2017; 29. [PMID: 28758251 DOI: 10.1111/jne.12513] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2017] [Revised: 07/05/2017] [Accepted: 07/26/2017] [Indexed: 12/14/2022]
Abstract
A growing body of evidence from research in rodents and humans has identified insulin as an important neuoregulatory peptide in the brain, where it coordinates diverse aspects of energy balance and peripheral glucose homeostasis. This review discusses where and how insulin interacts within the brain and evaluates the physiological and pathophysiological consequences of central insulin signalling in metabolism, obesity and type 2 diabetes.
Collapse
Affiliation(s)
- G T Dodd
- Metabolic Disease and Obesity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| | - T Tiganis
- Metabolic Disease and Obesity Program, Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
172
|
Barreto-Vianna ARC, Aguila MB, Mandarim-de-Lacerda CA. Beneficial effects of liraglutide (GLP1 analog) in the hippocampal inflammation. Metab Brain Dis 2017; 32:1735-1745. [PMID: 28681199 DOI: 10.1007/s11011-017-0059-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 06/20/2017] [Indexed: 01/08/2023]
Abstract
The brain is very sensitive to metabolic dysfunctions induced by diets high in saturated fatty acids, leading to neuroinflammation. The liraglutide has been found to have neuroprotective effects. However, its neuroprotective action in a model of palmitate-induced neuroinflammation had not yet been evaluated. Mice were intracerebroventricular (ICV) infused with palmitate and received subcutaneous liraglutide. The hippocampal dentate gyrus and CA1 regions were analyzed (morphology and inflammation-related proteins in microglia and astrocyte by confocal microscopy). Also, a real-time PCR was performed to measure the levels of tumor necrosis factor (TNF) alpha and interleukin (IL) 6. Palmitate ICV infusion resulted in pronounced inflammation response in the hippocampus, reactive microgliosis, and astrogliosis, with hypertrophied IBA1 immunoreactive microglia, increased microglial density with ameboid shape, decreased in the number of branches and junctions and increased the major histocompatibility complex (MHC) II expression. Also, we observed in the hippocampus of ICV palmitate infused mice an elevation in the pro-inflammatory cytokine levels TNFalpha and IL6. Liraglutide induced the neuroprotective microglial phenotype, characterized by an increased microglia complexity (enlarged Feret's diameter), an improved number of both cell junctions and processes, and lower circularity, accompanied by a significant reduction in TNFalpha and IL6 expressions. The study provides evidence that liraglutide may be a suitable treatment against the palmitate-induced neuroinflammation, which it is characterized by the reactive microgliosis and astrogliosis, as well as increased pro-inflammatory cytokines, which has been described as one of the primary causes of several pathologies of the central nervous system.
Collapse
Affiliation(s)
- Andre R C Barreto-Vianna
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcia B Aguila
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Carlos A Mandarim-de-Lacerda
- Laboratory of Morphometry, Metabolism, and Cardiovascular Diseases, Biomedical Center, Institute of Biology, State University of Rio de Janeiro, Rio de Janeiro, Brazil.
- Centro Biomedico, Instituto de Biologia, Laboratorio de Morfometria, Metabolismo e doenca Cardiovascular (www.lmmc.uerj.br), Universidade do Estado do Rio de Janeiro, Av 28 de Setembro 87 fds, Rio de Janeiro, RJ, 20551-030, Brazil.
| |
Collapse
|
173
|
Abstract
The hypothalamus is an evolutionarily conserved brain structure that regulates an organism's basic functions, such as homeostasis and reproduction. Several hypothalamic nuclei and neuronal circuits have been the focus of many studies seeking to understand their role in regulating these basic functions. Within the hypothalamic neuronal populations, the arcuate melanocortin system plays a major role in controlling homeostatic functions. The arcuate pro-opiomelanocortin (POMC) neurons in particular have been shown to be critical regulators of metabolism and reproduction because of their projections to several brain areas both in and outside of the hypothalamus, such as autonomic regions of the brain stem and spinal cord. Here, we review and discuss the current understanding of POMC neurons from their development and intracellular regulators to their physiological functions and pathological dysregulation.
Collapse
Affiliation(s)
- Chitoku Toda
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Anna Santoro
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Jung Dae Kim
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Sabrina Diano
- Program in Integrative Cell Signaling and Neurobiology of Metabolism, Yale University School of Medicine, New Haven, Connecticut 06520; .,Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale University School of Medicine, New Haven, Connecticut 06520.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut 06520.,Section of Comparative Medicine, Yale University School of Medicine, New Haven, Connecticut 06520
| |
Collapse
|
174
|
Abstract
Our understanding of adipose tissue as an endocrine organ has been transformed over the last 20 years. During this time, a number of adipocyte-derived factors or adipokines have been identified. This article will review evidence for how adipokines acting via the central nervous system (CNS) regulate normal physiology and disease pathology. The reported CNS-mediated effects of adipokines are varied and include the regulation of energy homeostasis, autonomic nervous system activity, the reproductive axis, neurodevelopment, cardiovascular function, and cognition. Due to the wealth of information available and the diversity of their known functions, the archetypal adipokines leptin and adiponectin will be focused on extensively. Other adipokines with established CNS actions will also be discussed. Due to the difficulties associated with studying CNS function on a molecular level in humans, the majority of our knowledge, and as such the studies described in this paper, comes from work in experimental animal models; however, where possible the relevant data from human studies are also highlighted. © 2017 American Physiological Society. Compr Physiol 7:1359-1406, 2017.
Collapse
Affiliation(s)
- Craig Beall
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Lydia Hanna
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| | - Kate L J Ellacott
- Biomedical Neuroscience Research Group, Institute of Biomedical and Clinical Sciences, University of Exeter Medical School, Exeter, Devon, United Kingdom
| |
Collapse
|
175
|
Fukui H. A new link between innate immunity and hepatic metabolism: attractive unknown place to visit. Hepatobiliary Surg Nutr 2017; 6:264-267. [PMID: 28848750 DOI: 10.21037/hbsn.2017.04.01] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Hiroshi Fukui
- Department of Gastroenterology, Endocrinology and Metabolism, Nara Medical University, 840 Shijo-cho Kashihara, Nara, Japan
| |
Collapse
|
176
|
Chen J, Li J, Yiu JHC, Lam JKW, Wong CM, Dorweiler B, Xu A, Woo CW. TRIF-dependent Toll-like receptor signaling suppresses Scd1 transcription in hepatocytes and prevents diet-induced hepatic steatosis. Sci Signal 2017; 10:10/491/eaal3336. [PMID: 28790196 DOI: 10.1126/scisignal.aal3336] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Nonalcoholic fatty liver disease (NAFLD) includes a spectrum of diseases that ranges in severity from hepatic steatosis to steatohepatitis, the latter of which is a major predisposing factor for liver cirrhosis and cancer. Toll-like receptor (TLR) signaling, which is critical for innate immunity, is generally believed to aggravate disease progression by inducing inflammation. Unexpectedly, we found that deficiency in TIR domain-containing adaptor-inducing interferon-β (TRIF), a cytosolic adaptor that transduces some TLR signals, worsened hepatic steatosis induced by a high-fat diet (HFD) and that such exacerbation was independent of myeloid cells. The aggravated steatosis in Trif-/- mice was due to the increased hepatocyte transcription of the gene encoding stearoyl-coenzyme A (CoA) desaturase 1 (SCD1), the rate-limiting enzyme for lipogenesis. Activation of the TRIF pathway by polyinosinic:polycytidylic acid [poly(I:C)] suppressed the increase in SCD1 abundance induced by palmitic acid or an HFD and subsequently prevented lipid accumulation in hepatocytes. Interferon regulatory factor 3 (IRF3), a transcriptional regulator downstream of TRIF, acted as a transcriptional suppressor by directly binding to the Scd1 promoter. These results suggest an unconventional metabolic function for TLR/TRIF signaling that should be taken into consideration when seeking to pharmacologically inhibit this pathway.
Collapse
Affiliation(s)
- Jing Chen
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong 999077, China.,Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong 999077, China
| | - Jin Li
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong 999077, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong 999077, China.,Department of Endocrinology, Second Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Jensen H C Yiu
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong 999077, China.,Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong 999077, China
| | - Jenny K W Lam
- Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong 999077, China
| | - Chi-Ming Wong
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong 999077, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong 999077, China
| | - Bernhard Dorweiler
- Division of Vascular Surgery, Department of Cardiothoracic and Vascular Surgery, University Medical Center Mainz, Mainz 55131, Germany
| | - Aimin Xu
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong 999077, China. .,Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong 999077, China.,Department of Medicine, Li Ka Shing Faculty of Medicine, University of Hong Kong, Hong Kong 999077, China
| | - Connie W Woo
- State Key Laboratory of Pharmaceutical Biotechnology, University of Hong Kong, Hong Kong 999077, China. .,Department of Pharmacology and Pharmacy, University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
177
|
Knockdown of Tlr4 in the Arcuate Nucleus Improves Obesity Related Metabolic Disorders. Sci Rep 2017; 7:7441. [PMID: 28785099 PMCID: PMC5547063 DOI: 10.1038/s41598-017-07858-6] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 07/03/2017] [Indexed: 02/06/2023] Open
Abstract
High-fat diet-induced hypothalamic metabolic inflammation is emerging as a cause for the development of obesity. It is acknowledged that Toll-like receptor4 (TLR4) signaling plays a crucial role in triggering of the hypothalamic metabolic inflammation during the course of diet-induced obesity. Whether hypothalamic arcuate nucleus (ARC)-restricted TLR4 knockdown improves obesity-related metabolic disorders remains unexplored. In this study, we used TLR4 shRNA lentiviral particles to suppress the TLR4 expression in the hypothalamic ARC of diet-induced obese rat model by stereotaxic injection. Our results demonstrate that ARC-restricted TLR4 knockdown protects obese rats from diet-induced weight gain and energy intake, from diet-induced impaired glucose homeostasis and peripheral insulin resistance, and from high-fat diet-induced hepatic steatosis and adipocyte hypertrophy. Thus, we define ARC-restricted TLR4 knockdown as a potential strategy to combat metabolic disorders associated with obesity.
Collapse
|
178
|
Ono-Moore KD, Zhao L, Huang S, Kim J, Rutkowsky JM, Snodgrass RG, Schneider DA, Quon MJ, Graham JL, Havel PJ, Hwang DH. Transgenic mice with ectopic expression of constitutively active TLR4 in adipose tissues do not show impaired insulin sensitivity. IMMUNITY INFLAMMATION AND DISEASE 2017; 5:526-540. [PMID: 28776958 PMCID: PMC5691308 DOI: 10.1002/iid3.162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 03/17/2017] [Accepted: 03/20/2017] [Indexed: 11/07/2022]
Abstract
INTRODUCTION Chronic low-grade inflammation is associated with obesity and diabetes. However, what causes and mediates chronic inflammation in metabolic disorders is not well understood. Toll-like receptor 4 (TLR4) mediates both infection-induced and sterile inflammation by recognizing pathogen-associated molecular patterns and endogenous molecules, respectively. Saturated fatty acids can activate TLR4, and TLR4-deficient mice were protected from high fat diet (HFD)-induced obesity and insulin resistance, suggesting that TLR4-mediated inflammation may cause metabolic dysfunction, such as obesity and insulin resistance. METHODS We generated two transgenic (TG) mouse lines expressing a constitutively active TLR4 in adipose tissue and determined whether these TG mice would show increased insulin resistance. RESULTS TG mice fed a high fat or a normal chow diet did not exhibit increased insulin resistance compared to their wild-type controls despite increased localized inflammation in white adipose tissue. Furthermore, females of one TG line fed a normal chow diet had improved insulin sensitivity with reduction in both adiposity and body weight when compared with wild-type littermates. There were significant differences between female and male mice in metabolic biomarkers and mRNA expression in proinflammatory genes and negative regulators of TLR4 signaling, regardless of genotype and diet. CONCLUSIONS Together, these results suggest that constitutively active TLR4-induced inflammation in white adipose tissue is not sufficient to induce systemic insulin resistance, and that high fat diet-induced insulin resistance may require other signals in addition to TLR4-mediated inflammation.
Collapse
Affiliation(s)
- Kikumi D Ono-Moore
- Department of Nutrition, University of California, Davis, California.,Western Human Nutrition Research Center, Agricultural Research Service, USDA-ARS, Davis, California
| | - Ling Zhao
- Department of Nutrition, University of Tennessee, Knoxville, Tennessee
| | - Shurong Huang
- Western Human Nutrition Research Center, Agricultural Research Service, USDA-ARS, Davis, California
| | - Jeonga Kim
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, University of Alabama, Birmingham, Alabama
| | - Jennifer M Rutkowsky
- Department of Cardiovascular Medicine, University of California, Davis, California
| | - Ryan G Snodgrass
- Department of Nutrition, University of California, Davis, California.,Western Human Nutrition Research Center, Agricultural Research Service, USDA-ARS, Davis, California
| | - Dina A Schneider
- Western Human Nutrition Research Center, Agricultural Research Service, USDA-ARS, Davis, California
| | - Michael J Quon
- Division of Endocrinology, Diabetes and Nutrition, University of Maryland, School of Medicine, Baltimore, Maryland
| | - James L Graham
- Department of Nutrition, University of California, Davis, California
| | - Peter J Havel
- Department of Nutrition, University of California, Davis, California
| | - Daniel H Hwang
- Department of Nutrition, University of California, Davis, California.,Western Human Nutrition Research Center, Agricultural Research Service, USDA-ARS, Davis, California
| |
Collapse
|
179
|
Paeger L, Pippow A, Hess S, Paehler M, Klein AC, Husch A, Pouzat C, Brüning JC, Kloppenburg P. Energy imbalance alters Ca 2+ handling and excitability of POMC neurons. eLife 2017; 6. [PMID: 28762947 PMCID: PMC5538824 DOI: 10.7554/elife.25641] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 06/29/2017] [Indexed: 01/16/2023] Open
Abstract
Satiety-signaling, pro-opiomelanocortin (POMC)-expressing neurons in the arcuate nucleus of the hypothalamus play a pivotal role in the regulation of energy homeostasis. Recent studies reported altered mitochondrial dynamics and decreased mitochondria- endoplasmic reticulum contacts in POMC neurons during diet-induced obesity. Since mitochondria play a crucial role in Ca2+ signaling, we investigated whether obesity alters Ca2+ handling of these neurons in mice. In diet-induced obesity, cellular Ca2+ handling properties including mitochondrial Ca2+ uptake capacity are impaired, and an increased resting level of free intracellular Ca2+ is accompanied by a marked decrease in neuronal excitability. Experimentally increasing or decreasing intracellular Ca2+ concentrations reproduced electrophysiological properties observed in diet-induced obesity. Taken together, we provide the first direct evidence for a diet-dependent deterioration of Ca2+ homeostasis in POMC neurons during obesity development resulting in impaired function of these critical energy homeostasis-regulating neurons. DOI:http://dx.doi.org/10.7554/eLife.25641.001
Collapse
Affiliation(s)
- Lars Paeger
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Andreas Pippow
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Simon Hess
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Moritz Paehler
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Andreas C Klein
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Andreas Husch
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| | - Christophe Pouzat
- MAP5 - Mathématiques Appliquées à Paris 5, CNRS UMR 8145, Paris, France
| | - Jens C Brüning
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.,Department of Mouse Genetics and Metabolism, Institute for Genetics, Center of Molecular Medicine Cologne, Center for Endocrinology, Diabetes and Preventive Medicine, University Hospital of Cologne, Cologne, Germany.,Max Planck Institute for Metabolism Research, Cologne, Germany
| | - Peter Kloppenburg
- Biocenter, Institute for Zoology, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany
| |
Collapse
|
180
|
Alterations in neuronal control of body weight and anxiety behavior by glutathione peroxidase 4 deficiency. Neuroscience 2017. [DOI: 10.1016/j.neuroscience.2017.05.050] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
181
|
Hypothalamic and inflammatory basis of hypertension. Clin Sci (Lond) 2017; 131:211-223. [PMID: 28057892 DOI: 10.1042/cs20160001] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Revised: 11/07/2016] [Accepted: 11/21/2016] [Indexed: 02/07/2023]
Abstract
Hypertension is a major health problem with great consequences for public health. Despite its role as the primary cause of significant morbidity and mortality associated with cardiovascular disease, the pathogenesis of essential hypertension remains largely unknown. The central nervous system (CNS) in general, and the hypothalamus in particular, are intricately involved in the development and maintenance of hypertension. Over the last several decades, the understanding of the brain's role in the development of hypertension has dramatically increased. This brief review is to summarize the neural mechanisms of hypertension with a focus on neuroendocrine and neurotransmitter involvement, highlighting recent findings that suggest that hypothalamic inflammation disrupts key signalling pathways to affect the central control of blood pressure, and therefore suggesting future development of interventional strategies that exploit recent findings pertaining to the hypothalamic control of blood pressure as well as the inflammatory-sympathetic mechanisms involved in hypertension.
Collapse
|
182
|
Theurich S, Tsaousidou E, Hanssen R, Lempradl AM, Mauer J, Timper K, Schilbach K, Folz-Donahue K, Heilinger C, Sexl V, Pospisilik JA, Wunderlich FT, Brüning JC. IL-6/Stat3-Dependent Induction of a Distinct, Obesity-Associated NK Cell Subpopulation Deteriorates Energy and Glucose Homeostasis. Cell Metab 2017; 26:171-184.e6. [PMID: 28683285 DOI: 10.1016/j.cmet.2017.05.018] [Citation(s) in RCA: 98] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 01/11/2017] [Accepted: 05/30/2017] [Indexed: 12/31/2022]
Abstract
Natural killer (NK) cells contribute to the development of obesity-associated insulin resistance. We demonstrate that in mice obesity promotes expansion of a distinct, interleukin-6 receptor (IL6R)a-expressing NK subpopulation, which also expresses a number of other myeloid lineage genes such as the colony-stimulating factor 1 receptor (Csf1r). Selective ablation of this Csf1r-expressing NK cell population prevents obesity and insulin resistance. Moreover, conditional inactivation of IL6Ra or Stat3 in NK cells limits obesity-associated formation of these myeloid signature NK cells, protecting from obesity, insulin resistance, and obesity-associated inflammation. Also in humans IL6Ra+ NK cells increase in obesity and correlate with markers of systemic low-grade inflammation, and their gene expression profile overlaps with characteristic gene sets of NK cells in obese mice. Collectively, we demonstrate that obesity-associated inflammation and metabolic disturbances depend on interleukin-6/Stat3-dependent formation of a distinct NK population, which may provide a target for the treatment of obesity, metaflammation-associated pathologies, and diabetes.
Collapse
Affiliation(s)
- Sebastian Theurich
- Max-Planck-Institute for Metabolism Research, Gleueler Straße 50, 50931 Cologne, Germany; Department I of Internal Medicine, University Hospital Cologne, 50924 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany
| | - Eva Tsaousidou
- Max-Planck-Institute for Metabolism Research, Gleueler Straße 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50924 Cologne, Germany
| | - Ruth Hanssen
- Max-Planck-Institute for Metabolism Research, Gleueler Straße 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50924 Cologne, Germany
| | - Adelheid M Lempradl
- Max-Planck-Institute of Immunobiology and Epigenetics, 79108 Freiburg, Germany
| | - Jan Mauer
- Max-Planck-Institute for Metabolism Research, Gleueler Straße 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50924 Cologne, Germany
| | - Katharina Timper
- Max-Planck-Institute for Metabolism Research, Gleueler Straße 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50924 Cologne, Germany
| | - Katharina Schilbach
- Max-Planck-Institute for Metabolism Research, Gleueler Straße 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50924 Cologne, Germany
| | - Kat Folz-Donahue
- Max-Planck-Institute for Biology of Ageing, FACS & Imaging Core Facility, 50931 Cologne, Germany
| | - Christian Heilinger
- Max-Planck-Institute for Metabolism Research, Gleueler Straße 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50924 Cologne, Germany
| | - Veronika Sexl
- Institute for Pharmacology and Toxicology, University of Veterinary Medicine, Veterinärplatz 1, 1210 Vienna, Austria
| | | | - F Thomas Wunderlich
- Max-Planck-Institute for Metabolism Research, Gleueler Straße 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50924 Cologne, Germany
| | - Jens C Brüning
- Max-Planck-Institute for Metabolism Research, Gleueler Straße 50, 50931 Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD), University of Cologne, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, 50924 Cologne, Germany.
| |
Collapse
|
183
|
Valdearcos M, Douglass JD, Robblee MM, Dorfman MD, Stifler DR, Bennett ML, Gerritse I, Fasnacht R, Barres BA, Thaler JP, Koliwad SK. Microglial Inflammatory Signaling Orchestrates the Hypothalamic Immune Response to Dietary Excess and Mediates Obesity Susceptibility. Cell Metab 2017; 26:185-197.e3. [PMID: 28683286 PMCID: PMC5569901 DOI: 10.1016/j.cmet.2017.05.015] [Citation(s) in RCA: 317] [Impact Index Per Article: 39.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2016] [Revised: 04/13/2017] [Accepted: 05/29/2017] [Indexed: 01/08/2023]
Abstract
Dietary excess triggers accumulation of pro-inflammatory microglia in the mediobasal hypothalamus (MBH), but the components of this microgliosis and its metabolic consequences remain uncertain. Here, we show that microglial inflammatory signaling determines the immunologic response of the MBH to dietary excess and regulates hypothalamic control of energy homeostasis in mice. Either pharmacologically depleting microglia or selectively restraining microglial NF-κB-dependent signaling sharply reduced microgliosis, an effect that includes prevention of MBH entry by bone-marrow-derived myeloid cells, and greatly limited diet-induced hyperphagia and weight gain. Conversely, forcing microglial activation through cell-specific deletion of the negative NF-κB regulator A20 induced spontaneous MBH microgliosis and cellular infiltration, reduced energy expenditure, and increased both food intake and weight gain even in absence of a dietary challenge. Thus, microglial inflammatory activation, stimulated by dietary excess, orchestrates a multicellular hypothalamic response that mediates obesity susceptibility, providing a mechanistic rationale for non-neuronal approaches to treat metabolic diseases.
Collapse
Affiliation(s)
- Martin Valdearcos
- The Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - John D Douglass
- University of Washington Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Megan M Robblee
- The Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Mauricio D Dorfman
- University of Washington Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Daniel R Stifler
- The Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA
| | - Mariko L Bennett
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Irene Gerritse
- The Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA; Graduate School of Life Sciences, University of Utrecht, 3584 CL Utrecht, the Netherlands
| | - Rachael Fasnacht
- University of Washington Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - Ben A Barres
- Department of Neurobiology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Joshua P Thaler
- University of Washington Diabetes Institute, University of Washington, Seattle, WA 98109, USA; Department of Medicine, University of Washington, Seattle, WA 98109, USA.
| | - Suneil K Koliwad
- The Diabetes Center, University of California San Francisco, San Francisco, CA 94143, USA; Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA.
| |
Collapse
|
184
|
Miura K, Ishioka M, Iijima K. The Roles of the Gut Microbiota and Toll-like Receptors in Obesity and Nonalcoholic Fatty Liver Disease. J Obes Metab Syndr 2017; 26:86-96. [PMID: 31089501 PMCID: PMC6484897 DOI: 10.7570/jomes.2017.26.2.86] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2017] [Revised: 02/28/2017] [Accepted: 03/16/2017] [Indexed: 02/06/2023] Open
Abstract
Obesity is characterized by low-grade chronic inflammation and is closely associated with the cardiovascular diseases, diabetes, and nonalcoholic fatty liver disease. Emerging data demonstrate that the gut microbiota contributes to the development of obesity by regulating the innate immune system, including the Toll-like receptors (TLRs): an altered gut microbiota composition and elevated TLR ligands are observed in obese mice and humans. The changes in the gut microbiota include an increased abundance of Firmicutes phylum and a decreased abundance of Bacteroidetes phylum. The population of beneficial bacteria that function as probiotics is decreased whereas harmful bacteria that can produce lipopolysaccharide, a TLR4 ligand, are increased in the obese state. In addition, the gut permeability is increased in obesity, which allows the delivery of larger amounts of bacterial components to the liver through the portal vein. Immune cells recognize these bacterial components through TLRs and produce diverse cytokines that kill invading pathogens. However, the sustained activation of TLR signaling induces host damage due to chronic exposure to harmful cytokines, which are produced from TLR expressing cells, including monocytes/macrophages. In the obese state, the expression of TLR is increased in several organs, including the adipose tissue and the liver. At the cell level, negative regulators of TLR signaling are suppressed, leading to activation of TLR signaling. These alterations promote inflammation in many organs. Thus, the gut microbiota and TLR signaling are therapeutic targets in patients with obesity and its related diseases.
Collapse
Affiliation(s)
- Kouichi Miura
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Mitsuaki Ishioka
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| | - Katsunori Iijima
- Department of Gastroenterology, Akita University Graduate School of Medicine, Akita, Japan
| |
Collapse
|
185
|
Maldonado-Avilés JG, Guarnieri DJ, Zhu X, DiLeone RJ. Down-regulation of miRNAs in the brain and development of diet-induced obesity. Int J Dev Neurosci 2017; 64:2-7. [PMID: 28652200 DOI: 10.1016/j.ijdevneu.2017.06.005] [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: 01/25/2017] [Revised: 05/11/2017] [Accepted: 06/22/2017] [Indexed: 01/01/2023] Open
Abstract
Novel therapeutic interventions for obesity and comorbid conditions require knowledge of the molecular elements playing a role in the development of obesity. Chronic low-grade inflammation has been consistently reported in obese individuals. In this study, we first determined whether key molecular modulators of inflammation, microRNA-155 (miR-155) and microRNA-146a (miR-146a), are regulated by an obesogenic diet within brain regions associated with reward, metabolism and energy balance. C57BL/6J mice were chronically exposed to a high-fat diet (HFD) or a standard chow (CTL). Significant reductions in the levels of miR-155 (82%) and miR-146a (41%) levels were observed within the nucleus accumbens of HFD mice compared to CTL. Further analysis of miR-155 regulation showed no significant changes in levels across peripheral tissue (white adipose, spleen, kidney or liver) between HFD and CTL mice. The effect of lower miR-155 on the development of obesity was determined by exposing wild-type (WT) and miR-155 knockout mice (miR-155 KO) to HFD. Male miR-155 KO gained significantly more weight than WT littermates. Metabolic analyses revealed that miR-155 KO significantly ate more HFD compared to WT, without differing in other metabolic measures including energy expenditure. Together, these data show that miR-155 is physiologically down-regulated after intake of an obesogenic diet, and that loss of miR-155 increases intake of an obesogenic diet. Moreover, these findings shed light on a potential miRNA-based mechanism contributing to the development of diet-induced obesity.
Collapse
Affiliation(s)
| | - Douglas J Guarnieri
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA; Department of Biology, Saint Bonaventure University, Saint Bonaventure, NY, USA.
| | - Xianglong Zhu
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - Ralph J DiLeone
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
186
|
New insight into inter-organ crosstalk contributing to the pathogenesis of non-alcoholic fatty liver disease (NAFLD). Protein Cell 2017. [PMID: 28643267 PMCID: PMC5818366 DOI: 10.1007/s13238-017-0436-0] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver dysfunction and a significant global health problem with substantial rise in prevalence over the last decades. It is becoming increasingly clear that NALFD is not only predominantly a hepatic manifestation of metabolic syndrome, but also involves extra-hepatic organs and regulatory pathways. Therapeutic options are limited for the treatment of NAFLD. Accordingly, a better understanding of the pathogenesis of NAFLD is critical for gaining new insight into the regulatory network of NAFLD and for identifying new targets for the prevention and treatment of NAFLD. In this review, we emphasize on the current understanding of the inter-organ crosstalk between the liver and peripheral organs that contributing to the pathogenesis of NAFLD.
Collapse
|
187
|
Inflammation, metaflammation and immunometabolic disorders. Nature 2017; 542:177-185. [PMID: 28179656 DOI: 10.1038/nature21363] [Citation(s) in RCA: 1520] [Impact Index Per Article: 190.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2015] [Accepted: 01/05/2017] [Indexed: 12/11/2022]
Abstract
Proper regulation and management of energy, substrate diversity and quantity, as well as macromolecular synthesis and breakdown processes, are fundamental to cellular and organismal survival and are paramount to health. Cellular and multicellular organization are defended by the immune response, a robust and critical system through which self is distinguished from non-self, pathogenic signals are recognized and eliminated, and tissue homeostasis is safeguarded. Many layers of evolutionarily conserved interactions occur between immune response and metabolism. Proper maintenance of this delicate balance is crucial for health and has important implications for many pathological states such as obesity, diabetes, and other chronic non-communicable diseases.
Collapse
|
188
|
Involvement of CD36 in Modulating the Decrease of NPY and AgRP Induced by Acute Palmitic Acid Stimulation in N1E-115 Cells. Nutrients 2017. [PMID: 28629148 PMCID: PMC5490605 DOI: 10.3390/nu9060626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
Central nervous system (CNS) fatty acid sensing plays an important role in the regulation of food intake, and palmitic acid (PA) is the most important long chain fatty acid (LCFA) in the mammalian diet. To explore the effect of PA on central neuropeptide expression and the role of the cluster of the differentiation of 36 (CD36) in the process, N1E-115 cells were cultured with PA in the presence or absence of sulfosuccinimidyl-oleate (SSO), a CD36 inhibitor. Results showed that 10 μmol/L PA significantly reduced NPY and AgRP mRNA expression after 20 min of exposure, while the expression of CD36 was upregulated. The presence of SSO significantly attenuated the decrease of NPY and AgRP expression that was induced by PA alone, although no notable effect on PA- induced CD36 gene expression was observed. In conclusion, our study suggests the involvement of CD36 in the PA-induced decrease of NPY and AgRP in N1E-115 cells.
Collapse
|
189
|
Ruud J, Steculorum SM, Brüning JC. Neuronal control of peripheral insulin sensitivity and glucose metabolism. Nat Commun 2017; 8:15259. [PMID: 28469281 PMCID: PMC5418592 DOI: 10.1038/ncomms15259] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 03/14/2017] [Indexed: 12/19/2022] Open
Abstract
The central nervous system (CNS) has an important role in the regulation of peripheral insulin sensitivity and glucose homeostasis. Research in this dynamically developing field has progressed rapidly due to techniques allowing targeted transgenesis and neurocircuitry mapping, which have defined the primary responsive neurons, associated molecular mechanisms and downstream neurocircuitries and processes involved. Here we review the brain regions, neurons and molecular mechanisms by which the CNS controls peripheral glucose metabolism, particularly via regulation of liver, brown adipose tissue and pancreatic function, and highlight the potential implications of these regulatory pathways in type 2 diabetes and obesity. The brain controls peripheral glucose metabolism, for example by modulating hepatic gluconeogenesis or by regulating glucose uptake into brown adipose tissue. Here, the authors review the brain regions, neurons and molecular mechanisms involved in these processes, and discuss their relevance to disease.
Collapse
Affiliation(s)
- Johan Ruud
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Sophie M. Steculorum
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
| | - Jens C. Brüning
- Department of Neuronal Control of Metabolism, Max Planck Institute for Metabolism Research, Gleueler Strasse 50, 50931 Cologne, Germany
- Policlinic for Endocrinology, Diabetes and Preventive Medicine (PEDP), University Hospital Cologne, Kerpener Strasse 26, 50924 Cologne, Germany
- Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center for Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Strasse 26, 50931 Cologne, Germany
- National Center for Diabetes Research (DZD), Ingolstädter Land Strasse 1, 85764 Neuherberg, Germany
| |
Collapse
|
190
|
Timper K, Denson JL, Steculorum SM, Heilinger C, Engström-Ruud L, Wunderlich CM, Rose-John S, Wunderlich FT, Brüning JC. IL-6 Improves Energy and Glucose Homeostasis in Obesity via Enhanced Central IL-6 trans-Signaling. Cell Rep 2017; 19:267-280. [PMID: 28402851 DOI: 10.1016/j.celrep.2017.03.043] [Citation(s) in RCA: 174] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 02/03/2017] [Accepted: 03/13/2017] [Indexed: 02/07/2023] Open
Abstract
Interleukin (IL)-6 engages similar signaling mechanisms to leptin. Here, we find that central application of IL-6 in mice suppresses feeding and improves glucose tolerance. In contrast to leptin, whose action is attenuated in obesity, the ability of IL-6 to suppress feeding is enhanced in obese mice. IL-6 suppresses feeding in the absence of neuronal IL-6-receptor (IL-6R) expression in hypothalamic or all forebrain neurons of mice. Conversely, obese mice exhibit increased soluble IL-6R levels in the cerebrospinal fluid. Blocking IL-6 trans-signaling in the CNS abrogates the ability of IL-6 to suppress feeding. Furthermore, gp130 expression is enhanced in the paraventricular nucleus of the hypothalamus (PVH) of obese mice, and deletion of gp130 in the PVH attenuates the beneficial central IL-6 effects on metabolism. Collectively, these experiments indicate that IL-6 trans-signaling is enhanced in the CNS of obese mice, allowing IL-6 to exert its beneficial metabolic effects even under conditions of leptin resistance.
Collapse
Affiliation(s)
- Katharina Timper
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Jesse Lee Denson
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Sophie Marie Steculorum
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Christian Heilinger
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Linda Engström-Ruud
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | - Claudia Maria Wunderlich
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany
| | | | - F Thomas Wunderlich
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany.
| | - Jens Claus Brüning
- Max Planck Institute for Metabolism Research, Department of Neuronal Control of Metabolism, Gleueler Str. 50, 50931 Cologne, Germany; Center for Endocrinology, Diabetes and Preventive Medicine (CEDP), University Hospital Cologne, Kerpener Str. 26, 50924 Cologne, Germany; Excellence Cluster on Cellular Stress Responses in Aging Associated Diseases (CECAD) and Center of Molecular Medicine Cologne (CMMC), University of Cologne, Joseph-Stelzmann-Str. 26, 50931 Cologne, Germany; National Center for Diabetes Research (DZD), Ingolstädter Landstr. 1, 85764 Neuherberg, Germany.
| |
Collapse
|
191
|
Zhou H, Yu M, Roychowdhury S, Sanz-Garcia C, Pollard KA, McMullen MR, Liu X, Li X, Nagy LE. Myeloid-MyD88 Contributes to Ethanol-Induced Liver Injury in Mice Linking Hepatocellular Death to Inflammation. Alcohol Clin Exp Res 2017; 41:719-726. [PMID: 28165624 PMCID: PMC5391031 DOI: 10.1111/acer.13345] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 01/29/2017] [Indexed: 12/18/2022]
Abstract
BACKGROUND Toll-like receptor 4 (TLR4) is critical for ethanol (EtOH)-induced liver injury. TLR4 signaling is mediated by 2 proximal adaptor molecules: myeloid differentiation primary response protein (MyD88) and TLR-domain-containing adaptor-inducing interferon-β (TRIF). Studies utilizing global knockouts of MyD88 and TRIF identified a predominant role for TRIF signaling in the progression of EtOH-induced liver injury. In contrast, IL-1 receptor, which signals solely via the MyD88 pathway, is also known to mediate EtOH-induced liver injury. We postulated that a cell-specific role for MyD88 in myeloid cells might explain these apparently discrepant roles of MyD88. Here we made use of myeloid-specific MyD88-deficient (MyD88LysM-KO ) mice generated by crossing LysM-CRE mice with MyD88fl/fl mice to test this hypothesis. METHODS MyD88LysM-KO and littermate controls were fed a Lieber-DeCarli EtOH-containing diet or pair-fed control diets for 25 days. RESULTS Littermate control, but not MyD88LysM-KO , mice developed early stages of EtOH-induced liver injury including elevated plasma alanine aminotransferase and increased hepatic triglycerides. Lobular inflammation and expression of pro-inflammatory cytokines/chemokines was increased in control but not MyD88LysM-KO . Further, EtOH-induced inflammasome activation, indicated by the presence of cleaved caspase-1 and mature IL-1β protein, was also ameliorated in livers of MyD88LysM-KO mice. In contrast, chronic EtOH-induced apoptosis, assessed via TUNEL staining, was independent of myeloid-MyD88 expression. CONCLUSIONS Collectively, these data demonstrate a cell-specific role for MyD88 in the development of chronic EtOH-induced liver injury. While MyD88LysM-KO still exhibited hepatocellular apoptosis in response to chronic EtOH, the absence of MyD88 on myeloid cells prevented the development of hepatic steatosis and inflammation.
Collapse
Affiliation(s)
- Hao Zhou
- Department of Immunology, Cleveland Clinic, Cleveland, OH
| | - Minja Yu
- Department of Immunology, Cleveland Clinic, Cleveland, OH
| | - Sanjoy Roychowdhury
- Department of Pathobiology, Cleveland Clinic, Cleveland, OH
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio
| | | | | | | | - Xiuli Liu
- Department of Anatomic Pathology, Cleveland Clinic, Cleveland, OH
| | - Xiaoxia Li
- Department of Immunology, Cleveland Clinic, Cleveland, OH
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio
| | - Laura E. Nagy
- Department of Pathobiology, Cleveland Clinic, Cleveland, OH
- Department of Gastroenterology/Hepatology, Cleveland Clinic, Cleveland, OH
- Department of Molecular Medicine, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
192
|
Duparc T, Plovier H, Marrachelli VG, Van Hul M, Essaghir A, Ståhlman M, Matamoros S, Geurts L, Pardo-Tendero MM, Druart C, Delzenne NM, Demoulin JB, van der Merwe SW, van Pelt J, Bäckhed F, Monleon D, Everard A, Cani PD. Hepatocyte MyD88 affects bile acids, gut microbiota and metabolome contributing to regulate glucose and lipid metabolism. Gut 2017; 66:620-632. [PMID: 27196572 PMCID: PMC5529962 DOI: 10.1136/gutjnl-2015-310904] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 03/01/2016] [Accepted: 03/03/2016] [Indexed: 12/16/2022]
Abstract
OBJECTIVE To examine the role of hepatocyte myeloid differentiation primary-response gene 88 (MyD88) on glucose and lipid metabolism. DESIGN To study the impact of the innate immune system at the level of the hepatocyte and metabolism, we generated mice harbouring hepatocyte-specific deletion of MyD88. We investigated the impact of the deletion on metabolism by feeding mice with a normal control diet or a high-fat diet for 8 weeks. We evaluated body weight, fat mass gain (using time-domain nuclear magnetic resonance), glucose metabolism and energy homeostasis (using metabolic chambers). We performed microarrays and quantitative PCRs in the liver. In addition, we investigated the gut microbiota composition, bile acid profile and both liver and plasma metabolome. We analysed the expression pattern of genes in the liver of obese humans developing non-alcoholic steatohepatitis (NASH). RESULTS Hepatocyte-specific deletion of MyD88 predisposes to glucose intolerance, inflammation and hepatic insulin resistance independently of body weight and adiposity. These phenotypic differences were partially attributed to differences in gene expression, transcriptional factor activity (ie, peroxisome proliferator activator receptor-α, farnesoid X receptor (FXR), liver X receptors and STAT3) and bile acid profiles involved in glucose, lipid metabolism and inflammation. In addition to these alterations, the genetic deletion of MyD88 in hepatocytes changes the gut microbiota composition and their metabolomes, resembling those observed during diet-induced obesity. Finally, obese humans with NASH displayed a decreased expression of different cytochromes P450 involved in bioactive lipid synthesis. CONCLUSIONS Our study identifies a new link between innate immunity and hepatic synthesis of bile acids and bioactive lipids. This dialogue appears to be involved in the susceptibility to alterations associated with obesity such as type 2 diabetes and NASH, both in mice and humans.
Collapse
Affiliation(s)
- Thibaut Duparc
- WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium,Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Hubert Plovier
- WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium,Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Vannina G Marrachelli
- Fundación de Investigación del Hospital Clínico Universitario de Valencia, INCLIVA, Valencia, Spain
| | - Matthias Van Hul
- WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium,Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Ahmed Essaghir
- de Duve Institute, Université catholique de Louvain, Brussels, Belgium
| | - Marcus Ståhlman
- Wallenberg Laboratory/Sahlgrenska Center for Cardiovascular and Metabolic Research, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Sébastien Matamoros
- WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium,Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Lucie Geurts
- WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium,Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Mercedes M Pardo-Tendero
- Fundación de Investigación del Hospital Clínico Universitario de Valencia, INCLIVA, Valencia, Spain
| | - Céline Druart
- WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium,Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Nathalie M Delzenne
- Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | | | - Schalk W van der Merwe
- Laboratory of Hepatology, University of Leuven (KUL), Belgium,Department of Gastroenterology and Hepatology, Division of Liver and Biliopancreatic Disorders, KUL, Leuven, Belgium
| | - Jos van Pelt
- Laboratory of Hepatology, University of Leuven (KUL), Belgium
| | - Fredrik Bäckhed
- Wallenberg Laboratory/Sahlgrenska Center for Cardiovascular and Metabolic Research, Sahlgrenska University Hospital, Gothenburg, Sweden,Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden,Novo Nordisk Foundation Center for Basic Metabolic Research, Section for Metabolic Receptology and Enteroendocrinology, Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Daniel Monleon
- Fundación de Investigación del Hospital Clínico Universitario de Valencia, INCLIVA, Valencia, Spain
| | - Amandine Everard
- WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium,Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| | - Patrice D Cani
- WELBIO- Walloon Excellence in Life Sciences and BIOtechnology, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium,Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
193
|
Sun X, Luquet S, Small DM. DRD2: Bridging the Genome and Ingestive Behavior. Trends Cogn Sci 2017; 21:372-384. [PMID: 28372879 DOI: 10.1016/j.tics.2017.03.004] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/10/2017] [Accepted: 03/06/2017] [Indexed: 12/26/2022]
Abstract
Recent work highlights the importance of genetic variants that influence brain structure and function in conferring risk for polygenic obesity. The neurotransmitter dopamine (DA) has a pivotal role in energy balance by integrating metabolic signals with circuits supporting cognitive, perceptual, and appetitive functions that guide feeding. It has also been established that diet and obesity alter DA signaling, leading to compulsive-like feeding and neurocognitive impairments. This raises the possibility that genetic variants that influence DA signaling and adaptation confer risk for overeating and cognitive decline. Here, we consider the role of two common gene variants, FTO and TaqIA rs1800497 in driving gene × environment interactions promoting obesity, metabolic dysfunction, and cognitive change via their influence on DA receptor subtype 2 (DRD2) signaling.
Collapse
Affiliation(s)
- Xue Sun
- Department of Psychology, University of Pennsylvania, Philadelphia, PA, USA
| | - Serge Luquet
- Université Paris Diderot, Sorbonne Paris Cité, BFA CNRS UMR 8251, Paris, France; Modern Diet and Physiology Research Center, New Haven, CT, USA
| | - Dana M Small
- Modern Diet and Physiology Research Center, New Haven, CT, USA; The John B. Pierce Laboratory, 290 Congress Avenue, New Haven, CT, USA; Department of Psychiatry, Yale School of Medicine, New Haven, CT, USA; Department of Psychology, Yale University, New Haven, CT, USA.
| |
Collapse
|
194
|
Crowe J, Lumb FE, Harnett MM, Harnett W. Parasite excretory-secretory products and their effects on metabolic syndrome. Parasite Immunol 2017; 39. [PMID: 28066896 DOI: 10.1111/pim.12410] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2016] [Accepted: 01/05/2017] [Indexed: 12/19/2022]
Abstract
Obesity, one of the main causes of metabolic syndrome (MetS), is an increasingly common health and economic problem worldwide, and one of the major risk factors for developing type 2 diabetes and cardiovascular disease. Chronic, low-grade inflammation is associated with MetS and obesity. A dominant type 2/anti-inflammatory response is required for metabolic homoeostasis within adipose tissue: during obesity, this response is replaced by infiltrating, inflammatory macrophages and T cells. Helminths and certain protozoan parasites are able to manipulate the host immune response towards a TH2 immune phenotype that is beneficial for their survival, and there is emerging data that there is an inverse correlation between the incidence of MetS and helminth infections, suggesting that, as with autoimmune and allergic diseases, helminths may play a protective role against MetS disease. Within this review, we will focus primarily on the excretory-secretory products that the parasites produce to modulate the immune system and discuss their potential use as therapeutics against MetS and its associated pathologies.
Collapse
Affiliation(s)
- J Crowe
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, UK
| | - F E Lumb
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| | - M M Harnett
- Institute of Infection, Immunity and Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, UK
| | - W Harnett
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, UK
| |
Collapse
|
195
|
Le Thuc O, Rovère C. [Hypothalamic inflammation and energy balance deregulations: focus on chemokines.]. Biol Aujourdhui 2017; 210:211-225. [PMID: 28327280 DOI: 10.1051/jbio/2016026] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2016] [Indexed: 02/01/2023]
Abstract
The hypothalamus is a key brain region in the regulation of energy balance. It especially controls food intake and both energy storage and expenditure through integration of humoral, neural and nutrient-related signals and cues. Hypothalamic neurons and glial cells act jointly to orchestrate, both spatially and temporally, regulated metabolic functions of the hypothalamus. Thus, the existence of a causal link between hypothalamic inflammation and deregulations of feeding behavior, such as involuntary weight-loss or obesity, has been suggested. Among the inflammatory mediators that could induce deregulations of hypothalamic control of the energy balance, chemokines represent interesting candidates. Indeed, chemokines, primarily known for their chemoattractant role of immune cells to the inflamed site, have also been suggested capable of neuromodulation. Thus, chemokines could disrupt cellular activity together with synthesis and/or secretion of multiple neurotransmitters/mediators that are involved in the maintenance of energy balance. Here, we relate, on one hand, recent results showing the primary role of the central chemokinergic signaling CCL2/CCR2 for metabolic and behavioral adaptation to high-grade inflammation, especially loss of appetite and weight, through its activity on hypothalamic neurons producing the orexigenic peptide Melanin-Concentrating Hormone (MCH) and, on the other hand, results that suggest that chemokines could also deregulate hypothalamic neuropeptidergic circuits to induce an opposite phenotype and eventually participate in the onset/development of obesity. In more details, we will emphasize a study recently showing, in a model of high-grade acute inflammation of LPS injection in mice, that central CCL2/CCR2 signaling is of primary importance for several aspects explaining weight loss associated with inflammation: after LPS injection, animals lose weight, reduce their food intake, increase their fat oxidation (thus energy consumption from fat storage)...These inflammation-induced metabolic and behavioral changes are reduced when central CCR2 signaling is disrupted either pharmacologically (by a specific inhibitor of CCR2) or genetically (in mice deficient for CCR2). This underlines the importance of this signaling in inflammation-related weight loss. We further determined that the LPS-induced and CCR2-mediated weight loss depends on the direct effect of CCR2 activation on MCH neurons activity. Indeed, the MCH neurons express CCR2, and the application of CCL2 on brain slices revealed that activation of CCR2 actually depolarizes MCH neurons and induces delays and/or failures of action potential emission. Furthermore, CCL2 is able to reduce KCl-evoked MCH secretion from hypothalamic explants. Taken together, these results demonstrate the role of the central CCL2/CCR2 signaling in metabolic and behavioral adaptation to inflammation. On the other hand, this first description of how the chemokinergic system can actually modulate the activity of the hypothalamic regulation of energy balance, but also some less advanced studies and some unpublished data, suggest that some other chemokines, such as CCL5, could participate in the development of the opposite phenotype, that is to say obesity.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France - Helmholtz Diabetes Center (HDC) & German Center for Diabetes Research (DZD), Helmholtz Zentrum München & Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Carole Rovère
- Université Côte d'Azur, CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, 06560 Valbonne, France
| |
Collapse
|
196
|
Depurinized milk downregulates rat thymus MyD88/Akt/p38 function, NF-κB-mediated inflammation, caspase-1 activity but not the endonuclease pathway: in vitro/in vivo study. Sci Rep 2017; 7:41971. [PMID: 28176796 PMCID: PMC5296740 DOI: 10.1038/srep41971] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2016] [Accepted: 01/04/2017] [Indexed: 12/17/2022] Open
Abstract
The aim of this study was the evaluation of 15 days dietary regimen of depurinized (DP) milk (obtained using our patented technological procedures) or 1.5% fat UHT milk instead of standard chow diet, on rat thymus and bone marrow MyD88/Akt/p38, NF-κB, caspase-1 and endonuclease pathways, in relation to peripheral blood cell composition. To determine whether the reduced mass of the thymus is a consequence of the direct effect of DP/UHT milk on apoptosis of thymocytes, in vitro Annexin-V-FITC/PI assay was performed. Significant decreases in the thymus wet weight, thymocyte MyD88, Akt-1/phospho-Akt-1 kinase, p38/phospho-p38, NF-κB, caspase-1 activity and CD4+/CD8+ antigen expression were obtained, especially in the DP milk group. The activity of thymocyte alkaline and acid DNase increased in the DP but not in the UHT milk group. The level of IL-6 significantly decreased in DP milk treated group, while the level of total TGF-β and IL-6 increased in UHT milk group. Significant differences in hematological parameters were obtained in commercial milk fed group. Observed results about prevention of experimental diabetes in DP pretreated groups may suggest that purine compounds, uric acid and other volatile toxic compounds of commercial milk may suppress oral tolerance, probably via IL-6 and TGF-β cytokine effects.
Collapse
|
197
|
Douglass JD, Dorfman MD, Thaler JP. Glia: silent partners in energy homeostasis and obesity pathogenesis. Diabetologia 2017; 60:226-236. [PMID: 27986987 PMCID: PMC5253392 DOI: 10.1007/s00125-016-4181-3] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Accepted: 11/11/2016] [Indexed: 10/20/2022]
Abstract
Body weight stability requires homeostatic regulation to balance energy intake and energy expenditure. Research on this system and how it is affected by obesity has largely focused on the role of hypothalamic neurons as integrators of information about long-term fuel storage, short-term nutrient availability and metabolic demand. Recent studies have uncovered glial cells as additional contributors to energy balance regulation and obesity pathogenesis. Beginning with early work on leptin signalling in astrocytes, this area of research rapidly emerged after the discovery of hypothalamic inflammation and gliosis in obese rodents and humans. Current studies have revealed the involvement of a wide variety of glial cell types in the modulation of neuronal activity, regulation of hormone and nutrient availability, and participation in the physiological regulation of feeding behaviour. In addition, one glial type, microglia, has recently been implicated in susceptibility to diet-induced obesity. Together, these exciting new findings deepen our understanding of energy homeostasis regulation and raise the possibility of identifying novel mechanisms that contribute to the pathogenesis of obesity.
Collapse
Affiliation(s)
- John D Douglass
- UW Diabetes Institute and Department of Medicine, University of Washington, 850 Republican St, S248, Box 358055, Seattle, WA, 98109, USA
| | - Mauricio D Dorfman
- UW Diabetes Institute and Department of Medicine, University of Washington, 850 Republican St, S248, Box 358055, Seattle, WA, 98109, USA
| | - Joshua P Thaler
- UW Diabetes Institute and Department of Medicine, University of Washington, 850 Republican St, S248, Box 358055, Seattle, WA, 98109, USA.
| |
Collapse
|
198
|
Douglass JD, Dorfman MD, Fasnacht R, Shaffer LD, Thaler JP. Astrocyte IKKβ/NF-κB signaling is required for diet-induced obesity and hypothalamic inflammation. Mol Metab 2017; 6:366-373. [PMID: 28377875 PMCID: PMC5369266 DOI: 10.1016/j.molmet.2017.01.010] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 01/18/2017] [Accepted: 01/20/2017] [Indexed: 12/18/2022] Open
Abstract
Objective Obesity and high fat diet (HFD) consumption in rodents is associated with hypothalamic inflammation and reactive gliosis. While neuronal inflammation promotes HFD-induced metabolic dysfunction, the role of astrocyte activation in susceptibility to hypothalamic inflammation and diet-induced obesity (DIO) remains uncertain. Methods Metabolic phenotyping, immunohistochemical analyses, and biochemical analyses were performed on HFD-fed mice with a tamoxifen-inducible astrocyte-specific knockout of IKKβ (GfapCreERIkbkbfl/fl, IKKβ-AKO), an essential cofactor of NF-κB-mediated inflammation. Results IKKβ-AKO mice with tamoxifen-induced IKKβ deletion prior to HFD exposure showed equivalent HFD-induced weight gain and glucose intolerance as Ikbkbfl/fl littermate controls. In GfapCreERTdTomato marker mice treated using the same protocol, minimal Cre-mediated recombination was observed in the mediobasal hypothalamus (MBH). By contrast, mice pretreated with 6 weeks of HFD exposure prior to tamoxifen administration showed substantially increased recombination throughout the MBH. Remarkably, this treatment approach protected IKKβ-AKO mice from further weight gain through an immediate reduction of food intake and increase of energy expenditure. Astrocyte IKKβ deletion after HFD exposure—but not before—also reduced glucose intolerance and insulin resistance, likely as a consequence of lower adiposity. Finally, both hypothalamic inflammation and astrocytosis were reduced in HFD-fed IKKβ-AKO mice. Conclusions These data support a requirement for astrocytic inflammatory signaling in HFD-induced hyperphagia and DIO susceptibility that may provide a novel target for obesity therapeutics. The first direct evidence that astrocyte inflammatory activation promotes obesity. GfapCreER mice given tamoxifen show minimal recombination in MBH astrocytes. GfapCreER mice given tamoxifen after 6 wks of HFD have recombination in the MBH. Astrocyte IKKβ deletion with tamoxifen before HFD has no effect on energy balance. Astrocyte IKKβ deletion with tamoxifen given after HFD reduces DIO susceptibility.
Collapse
Key Words
- ARC, arcuate nucleus
- Agrp, Agouti-related peptide
- Astrocytes
- Bdnf, brain-derived neurotrophic factor
- Cart, cocaine- and amphetamine-regulated transcript
- Ccl2, C–C motif chemokine ligand 2
- DIO, diet-induced obesity
- DMH, dorsomedial hypothalamus
- Energy homeostasis
- GFAP, glial fibrillary acidic protein
- GSIS, glucose-stimulated insulin secretion
- GTT, glucose tolerance test
- HFD, high-fat diet
- Hypothalamus
- IHC, immunohistochemistry
- IKKβ, inhibitor of kappa B kinase beta
- ITT, insulin tolerance test
- Iba1, ionized calcium binding adaptor molecule 1
- Il, interleukin
- Inflammation
- LPS, lipopolysaccharide
- MBH, mediobasal hypothalamus
- Metabolism
- NF-κB, nuclear factor kappa B
- Npy, neuropeptide Y
- Obesity
- Pomc, proopiomelanocortin
- RER, respiratory exchange ratio
- TMX, tamoxifen
- Tnfa, tumor necrosis factor α
- VMN, ventromedial nucleus
- ir, immunoreactivity
Collapse
Affiliation(s)
- J D Douglass
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - M D Dorfman
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - R Fasnacht
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - L D Shaffer
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA 98109, USA
| | - J P Thaler
- Division of Metabolism, Endocrinology & Nutrition, Department of Medicine, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
199
|
Jais A, Brüning JC. Hypothalamic inflammation in obesity and metabolic disease. J Clin Invest 2017; 127:24-32. [PMID: 28045396 DOI: 10.1172/jci88878] [Citation(s) in RCA: 326] [Impact Index Per Article: 40.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Over the last years, hypothalamic inflammation has been linked to the development and progression of obesity and its sequelae. There is accumulating evidence that this inflammation not only impairs energy balance but also contributes to obesity-associated insulin resistance. Elevated activation of key inflammatory mediators such as JNK and IκB kinase (IKK) occurs rapidly upon consumption of a high-fat diet, even prior to significant weight gain. This activation of hypothalamic inflammatory pathways results in the uncoupling of caloric intake and energy expenditure, fostering overeating and further weight gain. In addition, these inflammatory processes contribute to obesity-associated insulin resistance and deterioration of glucose metabolism via altered neurocircuit functions. An understanding of the contributions of different neuronal and non-neuronal cell types to hypothalamic inflammatory processes, and delineation of the differences and similarities between acute and chronic activation of these inflammatory pathways, will be critical for the development of novel therapeutic strategies for the treatment of obesity and metabolic syndrome.
Collapse
|
200
|
Le Thuc O, Stobbe K, Cansell C, Nahon JL, Blondeau N, Rovère C. Hypothalamic Inflammation and Energy Balance Disruptions: Spotlight on Chemokines. Front Endocrinol (Lausanne) 2017; 8:197. [PMID: 28855891 PMCID: PMC5557773 DOI: 10.3389/fendo.2017.00197] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 07/27/2017] [Indexed: 12/20/2022] Open
Abstract
The hypothalamus is a key brain region in the regulation of energy balance as it controls food intake and both energy storage and expenditure through integration of humoral, neural, and nutrient-related signals and cues. Many years of research have focused on the regulation of energy balance by hypothalamic neurons, but the most recent findings suggest that neurons and glial cells, such as microglia and astrocytes, in the hypothalamus actually orchestrate together several metabolic functions. Because glial cells have been described as mediators of inflammatory processes in the brain, the existence of a causal link between hypothalamic inflammation and the deregulations of feeding behavior, leading to involuntary weight loss or obesity for example, has been suggested. Several inflammatory pathways that could impair the hypothalamic control of energy balance have been studied over the years such as, among others, toll-like receptors and canonical cytokines. Yet, less studied so far, chemokines also represent interesting candidates that could link the aforementioned pathways and the activity of hypothalamic neurons. Indeed, chemokines, in addition to their role in attracting immune cells to the inflamed site, have been suggested to be capable of neuromodulation. Thus, they could disrupt cellular activity together with synthesis and/or secretion of multiple neurotransmitters/mediators involved in the maintenance of energy balance. This review discusses the different inflammatory pathways that have been identified so far in the hypothalamus in the context of feeding behavior and body weight control impairments, with a particular focus on chemokines signaling that opens a new avenue in the understanding of the major role played by inflammation in obesity.
Collapse
Affiliation(s)
- Ophélia Le Thuc
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
- Helmholtz Diabetes Center (HDC), German Center for Diabetes Research (DZD), Helmholtz Zentrum München, Neuherberg, Germany
- Division of Metabolic Diseases, Technische Universität München, Munich, Germany
| | - Katharina Stobbe
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Céline Cansell
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Jean-Louis Nahon
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Nicolas Blondeau
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
| | - Carole Rovère
- CNRS, Institut de Pharmacologie Moléculaire et Cellulaire, Université Côte d’Azur, Valbonne, France
- *Correspondence: Carole Rovère,
| |
Collapse
|