1
|
Zhang Y, Wang R, Liu T, Wang R. Exercise as a Therapeutic Strategy for Obesity: Central and Peripheral Mechanisms. Metabolites 2024; 14:589. [PMID: 39590824 PMCID: PMC11596326 DOI: 10.3390/metabo14110589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/28/2024] Open
Abstract
Obesity is a complex, multifactorial condition involving excessive fat accumulation due to an imbalance between energy intake and expenditure, with its global prevalence steadily rising. This condition significantly increases the risk of chronic diseases, including sarcopenia, type 2 diabetes, and cardiovascular diseases, highlighting the need for effective interventions. Exercise has emerged as a potent non-pharmacological approach to combat obesity, targeting both central and peripheral mechanisms that regulate metabolism, energy expenditure, and neurological functions. In the central nervous system, exercise influences appetite, mood, and cognitive functions by modulating the reward system and regulating appetite-controlling hormones to manage energy intake. Concurrently, exercise promotes thermogenesis in adipose tissue and regulates endocrine path-ways and key metabolic organs, such as skeletal muscle and the liver, to enhance fat oxidation and support energy balance. Despite advances in understanding exercise's role in obesity, the precise interaction between the neurobiological and peripheral metabolic pathways remains underexplored, particularly in public health strategies. A better understanding of these interactions could inform more comprehensive obesity management approaches by addressing both central nervous system influences on behavior and peripheral metabolic regulation. This review synthesizes recent insights into these roles, highlighting potential therapeutic strategies targeting both systems for more effective obesity interventions.
Collapse
Affiliation(s)
- Yiyin Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| | - Ruwen Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| | - Tiemin Liu
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | - Ru Wang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, China; (Y.Z.); (R.W.)
| |
Collapse
|
2
|
Dragano NRV, Milbank E, Haddad-Tóvolli R, Garrido-Gil P, Nóvoa E, Fondevilla MF, Capelli V, Zanesco AM, Solon C, Morari J, Pires L, Estevez-Salguero Á, Beiroa D, González-García I, Barca-Mayo O, Diéguez C, Nogueiras R, Labandeira-García JL, Rexen Ulven E, Ulven T, Claret M, Velloso LA, López M. Hypothalamic free fatty acid receptor-1 regulates whole-body energy balance. Mol Metab 2024; 79:101840. [PMID: 38036170 PMCID: PMC10784317 DOI: 10.1016/j.molmet.2023.101840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 11/17/2023] [Indexed: 12/02/2023] Open
Abstract
OBJECTIVE Free fatty acid receptor-1 (FFAR1) is a medium- and long-chain fatty acid sensing G protein-coupled receptor that is highly expressed in the hypothalamus. Here, we investigated the central role of FFAR1 on energy balance. METHODS Central FFAR1 agonism and virogenic knockdown were performed in mice. Energy balance studies, infrared thermographic analysis of brown adipose tissue (BAT) and molecular analysis of the hypothalamus, BAT, white adipose tissue (WAT) and liver were carried out. RESULTS Pharmacological stimulation of FFAR1, using central administration of its agonist TUG-905 in diet-induced obese mice, decreases body weight and is associated with increased energy expenditure, BAT thermogenesis and browning of subcutaneous WAT (sWAT), as well as reduced AMP-activated protein kinase (AMPK) levels, reduced inflammation, and decreased endoplasmic reticulum (ER) stress in the hypothalamus. As FFAR1 is expressed in distinct hypothalamic neuronal subpopulations, we used an AAV vector expressing a shRNA to specifically knockdown Ffar1 in proopiomelanocortin (POMC) neurons of the arcuate nucleus of the hypothalamus (ARC) of obese mice. Our data showed that knockdown of Ffar1 in POMC neurons promoted hyperphagia and body weight gain. In parallel, these mice developed hepatic insulin resistance and steatosis. CONCLUSIONS FFAR1 emerges as a new hypothalamic nutrient sensor regulating whole body energy balance. Moreover, pharmacological activation of FFAR1 could provide a therapeutic advance in the management of obesity and its associated metabolic disorders.
Collapse
Affiliation(s)
- Nathalia R V Dragano
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain; Laboratory of Cell Signaling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil.
| | - Edward Milbank
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Pablo Garrido-Gil
- Department of Morphological Sciences, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Enfermedades Neurodegenerativas (CIBERNED), 28029, Santiago de Compostela, Spain
| | - Eva Nóvoa
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Marcos F Fondevilla
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Valentina Capelli
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Ariane Maria Zanesco
- Laboratory of Cell Signaling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Carina Solon
- Laboratory of Cell Signaling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Joseane Morari
- Laboratory of Cell Signaling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Leticia Pires
- Laboratory of Cell Signaling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Ánxela Estevez-Salguero
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Daniel Beiroa
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Ismael González-García
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Olga Barca-Mayo
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain
| | - Carlos Diéguez
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - Ruben Nogueiras
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain
| | - José L Labandeira-García
- Department of Morphological Sciences, CiMUS, University of Santiago de Compostela, Santiago de Compostela, Spain; CIBER Enfermedades Neurodegenerativas (CIBERNED), 28029, Santiago de Compostela, Spain
| | - Elisabeth Rexen Ulven
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Trond Ulven
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100 Copenhagen, Denmark
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain; CIBER Diabetes y Enfermedades Metabólicas Asociadas (CIBERdem), 08036, Spain; Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain
| | - Licio A Velloso
- Laboratory of Cell Signaling-Obesity and Comorbidities Research Center, University of Campinas, Campinas, Brazil
| | - Miguel López
- Department of Physiology, CiMUS, University of Santiago de Compostela, Santiago de Compostela, 15782, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBEROBN), 15706, Spain.
| |
Collapse
|
3
|
Song JW, Lee KH, Seong H, Shin DM, Shon WJ. Taste receptor type 1 member 3 enables western diet-induced anxiety in mice. BMC Biol 2023; 21:243. [PMID: 37926812 PMCID: PMC10626698 DOI: 10.1186/s12915-023-01723-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Accepted: 10/03/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Accumulating evidence supports that the Western diet (WD), a diet high in saturated fat and sugary drinks, contributes to the pathogenesis of anxiety disorders, which are the most prevalent mental disorders worldwide. However, the underlying mechanisms by which WD causes anxiety remain unclear. Abundant expression of taste receptor type 1 member 3 (TAS1R3) has been identified in the hypothalamus, a key brain area involved in sensing peripheral nutritional signals and regulating anxiety. Thus, we investigated the influence of excessive WD intake on anxiety and mechanisms by which WD intake affects anxiety development using wild-type (WT) and Tas1r3 deficient (Tas1r3-/-) mice fed a normal diet (ND) or WD for 12 weeks. RESULTS WD increased anxiety in male WT mice, whereas male Tas1r3-/- mice were protected from WD-induced anxiety, as assessed by open field (OF), elevated plus maze (EPM), light-dark box (LDB), and novelty-suppressed feeding (NSF) tests. Analyzing the hypothalamic transcriptome of WD-fed WT and Tas1r3-/- mice, we found 1,432 genes significantly up- or down-regulated as a result of Tas1r3 deficiency. Furthermore, bioinformatic analysis revealed that the CREB/BDNF signaling-mediated maintenance of neuronal regeneration, which can prevent anxiety development, was enhanced in WD-fed Tas1r3-/- mice compared with WD-fed WT mice. Additionally, in vitro studies further confirmed that Tas1r3 knockdown prevents the suppression of Creb1 and of CREB-mediated BDNF expression caused by high levels of glucose, fructose, and palmitic acid in hypothalamic neuronal cells. CONCLUSIONS Our results imply that TAS1R3 may play a key role in WD-induced alterations in hypothalamic functions, and that inhibition of TAS1R3 overactivation in the hypothalamus could offer therapeutic targets to alleviate the effects of WD on anxiety.
Collapse
Affiliation(s)
- Jae Won Song
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Keon-Hee Lee
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Hobin Seong
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea
| | - Dong-Mi Shin
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea.
- Research Institute of Human Ecology, Seoul National University, Gwanak-Gu, Seoul, 08826, Republic of Korea.
| | - Woo-Jeong Shon
- Department of Food and Nutrition, Seoul National University College of Human Ecology, Gwanak-Gu, Seoul, 08826, Republic of Korea.
- Research Institute of Human Ecology, Seoul National University, Gwanak-Gu, Seoul, 08826, Republic of Korea.
| |
Collapse
|
4
|
Jiang M, Jang SE, Zeng L. The Effects of Extrinsic and Intrinsic Factors on Neurogenesis. Cells 2023; 12:cells12091285. [PMID: 37174685 PMCID: PMC10177620 DOI: 10.3390/cells12091285] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 04/18/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
In the mammalian brain, neurogenesis is maintained throughout adulthood primarily in two typical niches, the subgranular zone (SGZ) of the dentate gyrus and the subventricular zone (SVZ) of the lateral ventricles and in other nonclassic neurogenic areas (e.g., the amygdala and striatum). During prenatal and early postnatal development, neural stem cells (NSCs) differentiate into neurons and migrate to appropriate areas such as the olfactory bulb where they integrate into existing neural networks; these phenomena constitute the multistep process of neurogenesis. Alterations in any of these processes impair neurogenesis and may even lead to brain dysfunction, including cognitive impairment and neurodegeneration. Here, we first summarize the main properties of mammalian neurogenic niches to describe the cellular and molecular mechanisms of neurogenesis. Accumulating evidence indicates that neurogenesis plays an integral role in neuronal plasticity in the brain and cognition in the postnatal period. Given that neurogenesis can be highly modulated by a number of extrinsic and intrinsic factors, we discuss the impact of extrinsic (e.g., alcohol) and intrinsic (e.g., hormones) modulators on neurogenesis. Additionally, we provide an overview of the contribution of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection to persistent neurological sequelae such as neurodegeneration, neurogenic defects and accelerated neuronal cell death. Together, our review provides a link between extrinsic/intrinsic factors and neurogenesis and explains the possible mechanisms of abnormal neurogenesis underlying neurological disorders.
Collapse
Affiliation(s)
- Mei Jiang
- Department of Human Anatomy, Dongguan Key Laboratory of Stem Cell and Regenerative Tissue Engineering, Dongguan Campus, Guangdong Medical University, Dongguan 523808, China
| | - Se Eun Jang
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
| | - Li Zeng
- Neural Stem Cell Research Lab, Research Department, National Neuroscience Institute, Singapore 308433, Singapore
- Neuroscience and Behavioral Disorders Program, DUKE-NUS Graduate Medical School, Singapore 169857, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technology University, Novena Campus, 11 Mandalay Road, Singapore 308232, Singapore
| |
Collapse
|
5
|
Diéguez E, Nieto-Ruiz A, Martín-Pérez C, Sepúlveda-Valbuena N, Herrmann F, Jiménez J, De-Castellar R, Catena A, García-Santos JA, Bermúdez MG, Campoy C. Association study between hypothalamic functional connectivity, early nutrition, and glucose levels in healthy children aged 6 years: The COGNIS study follow-up. Front Nutr 2022; 9:935740. [PMID: 36313089 PMCID: PMC9597646 DOI: 10.3389/fnut.2022.935740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 09/20/2022] [Indexed: 11/17/2022] Open
Abstract
Breastfeeding (BF) is the gold standard in infant nutrition; knowing how it influences brain connectivity would help understand the mechanisms involved, which would help close the nutritional gap between infant formulas and breast milk. We analyzed potential long-term differences depending on the diet with an experimental infant formula (EF), compared to a standard infant formula (SF) or breastfeeding (BF) during the first 18 months of life on children's hypothalamic functional connectivity (FC) assessed at 6 years old. A total of 62 children participating in the COGNIS randomized clinical trial (Clinical Trial Registration: www.ClinicalTrials.gov, identifier: NCT02094547) were included in this study. They were randomized to receive an SF (n = 22) or a bioactive nutrient-enriched EF (n = 20). BF children were also included as a control study group (BF: n = 20). Brain function was evaluated using functional magnetic resonance imaging (fMRI) and mean glucose levels were collected through a 24-h continuous glucose monitoring (CGM) device at 6 years old. Furthermore, nutrient intake was also analyzed during the first 18 months of life and at 6 years old through 3-day dietary intake records. Groups fed with EF and BF showed lower FC between the medial hypothalamus (MH) and the anterior cingulate cortex (ACC) in comparison with SF-fed children. Moreover, the BF children group showed lower FC between the MH and the left putamen extending to the middle insula, and higher FC between the MH and the inferior frontal gyrus (IFG) compared to the EF-fed children group. These areas are key regions within the salience network, which is involved in processing salience stimuli, eating motivation, and hedonic-driven desire to consume food. Indeed, current higher connectivity found on the MH-IFG network in the BF group was associated with lower simple sugars acceptable macronutrient distribution ranges (AMDRs) at 6 months of age. Regarding linoleic acid intake at 12 months old, a negative association with this network (MH-IFG) only in the BF group was found. In addition, BF children showed lower mean glucose levels compared to SF-fed children at 6 years old. Our results may point out a possible relationship between diet during the first 18 months of life and inclined proclivity for hedonic eating later in life.
Collapse
Affiliation(s)
- Estefanía Diéguez
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain,Instituto de Investigación Biosanitaria (ibs.GRANADA), Health Sciences Technological Park, Granada, Spain,EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Ana Nieto-Ruiz
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain,Instituto de Investigación Biosanitaria (ibs.GRANADA), Health Sciences Technological Park, Granada, Spain,EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Cristina Martín-Pérez
- Psychology Department, Faculty of Education, University of Valladolid, Segovia, Spain
| | - Natalia Sepúlveda-Valbuena
- Nutrition and Biochemistry Department, Faculty of Sciences, Pontificia Universidad Javeriana, Bogotá, Colombia
| | - Florian Herrmann
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain,EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Jesús Jiménez
- Ordesa Laboratories, S.L., Sant Boi de Llobregat, Spain
| | | | - Andrés Catena
- Department of Experimental Psychology, School of Psychology, University of Granada, Granada, Spain
| | - José Antonio García-Santos
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain,Instituto de Investigación Biosanitaria (ibs.GRANADA), Health Sciences Technological Park, Granada, Spain,EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Mercedes G. Bermúdez
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain,Instituto de Investigación Biosanitaria (ibs.GRANADA), Health Sciences Technological Park, Granada, Spain,EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain
| | - Cristina Campoy
- Department of Paediatrics, School of Medicine, University of Granada, Granada, Spain,Instituto de Investigación Biosanitaria (ibs.GRANADA), Health Sciences Technological Park, Granada, Spain,EURISTIKOS Excellence Centre for Paediatric Research, Biomedical Research Centre, University of Granada, Granada, Spain,National Network of Research in Epidemiology and Public Health (CIBERESP), Institute of Health Carlos III (Granada's Node), Madrid, Spain,*Correspondence: Cristina Campoy
| |
Collapse
|
6
|
A Short-Term Sucrose Diet Impacts Cell Proliferation of Neural Precursors in the Adult Hypothalamus. Nutrients 2022; 14:nu14132564. [PMID: 35807744 PMCID: PMC9268421 DOI: 10.3390/nu14132564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/15/2022] [Accepted: 06/17/2022] [Indexed: 11/16/2022] Open
Abstract
Radial glia-like cells in the hypothalamus and dorsal vagal complex are neural precursors (NPs) located near subventricular organs: median eminence and area postrema, respectively. Their strategic position can detect blood-borne nutrients, hormones, and mitogenic signals. Hypothalamic NPs increase their proliferation with a mechanism that involves hemichannel (HC) activity. NPs can originate new neurons in response to a short-term high-fat diet as a compensatory mechanism. The effects of high carbohydrate Western diets on adult neurogenesis are unknown. Although sugars are usually consumed as sucrose, more free fructose is now incorporated into food items. Here, we studied the proliferation of both types of NPs in Sprague Dawley rats exposed to a short-term high sucrose diet (HSD) and a control diet. In tanycyte cultures, we evaluated the effects of glucose and fructose and a mix of both hexoses on HC activity. In rats fed an HSD, we observed an increase in the proliferative state of both precursors. Glucose, either in the presence or absence of fructose, but not fructose alone, induced in vitro HC activity. These results should broaden the understanding of the nutrient monitoring capacity of NPs in reacting to changes in feeding behavior, specifically to high sugar western diets.
Collapse
|
7
|
Morari J, Haddad-Tóvolli R, Silva Nogueira PA, Teixeira CJ, Maróstica R, Tobar N, Ramos CD, Velloso LA, Dias Bobbo VC, Anhê GF. Body mass variability in age-matched outbred male Swiss mice is associated to differential control of food intake by ghrelin. Mol Cell Endocrinol 2022; 550:111646. [PMID: 35413387 DOI: 10.1016/j.mce.2022.111646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/05/2022] [Accepted: 04/07/2022] [Indexed: 11/23/2022]
Abstract
Swiss mice belong to an outbred strain of mice largely used as a model for experimental obesity induced by high fat diet (HFD). We have previously demonstrated that a given cohort of age-matched Swiss mice is hallmarked by heterogeneous changes in body weight when exposed to HFD. The reasons underlying such variability, however, are not completely understood. Therefore we aimed to clarify the mechanisms underlying the variability in spontaneous weight gain in age-matched male swiss mice. To achieve that, individuals in a cohort of age-matched male Swiss mice were categorized as prone to body mass gain (PBMG) and resistant to body mass gain (RBMG). PBMG animals had higher caloric intake and body mass gain. RBMG and PBMG mice had a similar reduction in food intake when challenged with leptin but only RBMG exhibited a drop in ghrelin concentrations after refeeding. PBMG also showed increased midbrain levels of ghrelin receptor (Ghsr) and Dopamine receptor d2 (Drd2) mRNAs upon refeeding. Pharmacological blockade of GHSR with JMV3002 failed to reduce food intake in PMBG mice as it did in RBMG. On the other hand, the response to JMV3002 seen in PBMG was hallmarked by singular transcriptional response in the midbrain characterized by a simultaneous increase in both tyrosine hydroxylase (Th) and Proopiomelanocortin (Pomc) expressions. In conclusion, our data show that differences in the expression of genes related to the reward system in the midbrain as well as in ghrelin concentrations in serum correlate with spontaneous variability in body mass and food intake seen in age-matched male Swiss mice.
Collapse
Affiliation(s)
- Joseane Morari
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo, 13083-887, Brazil; Obesity and Comorbidities Research Center, Institute of Biology, University of Campinas, Campinas, Sao Paulo, 13083-864, Brazil; Department of Translational Medicine, School of Medical Sciences, State University of Campinas, Campinas, Sao Paulo, 13083-881, Brazil.
| | - Roberta Haddad-Tóvolli
- Obesity and Comorbidities Research Center, Institute of Biology, University of Campinas, Campinas, Sao Paulo, 13083-864, Brazil
| | - Pedro Augusto Silva Nogueira
- Obesity and Comorbidities Research Center, Institute of Biology, University of Campinas, Campinas, Sao Paulo, 13083-864, Brazil
| | - Caio Jordão Teixeira
- Department of Physiology and Biophysics, Institute of Biomedical Science, University of Sao Paulo, 1524. Prof. Lineu Prestes Ave., ICB1, Sao Paulo, SP, 05508-000, Brazil
| | - Rafael Maróstica
- Obesity and Comorbidities Research Center, Institute of Biology, University of Campinas, Campinas, Sao Paulo, 13083-864, Brazil
| | - Natália Tobar
- Department of Radiology, University of Campinas, Campinas, Sao Paulo, 13084-970, Brazil
| | - Celso Dario Ramos
- Department of Radiology, University of Campinas, Campinas, Sao Paulo, 13084-970, Brazil
| | - Licio Augusto Velloso
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, Campinas, Sao Paulo, 13083-887, Brazil; Obesity and Comorbidities Research Center, Institute of Biology, University of Campinas, Campinas, Sao Paulo, 13083-864, Brazil
| | - Vanessa Cristina Dias Bobbo
- Obesity and Comorbidities Research Center, Institute of Biology, University of Campinas, Campinas, Sao Paulo, 13083-864, Brazil
| | - Gabriel Forato Anhê
- Department of Translational Medicine, School of Medical Sciences, State University of Campinas, Campinas, Sao Paulo, 13083-881, Brazil
| |
Collapse
|
8
|
Bobbo VC, Engel DF, Jara CP, Mendes NF, Haddad-Tovolli R, Prado TP, Sidarta-Oliveira D, Morari J, Velloso LA, Araujo EP. Interleukin-6 actions in the hypothalamus protects against obesity and is involved in the regulation of neurogenesis. J Neuroinflammation 2021; 18:192. [PMID: 34465367 PMCID: PMC8408946 DOI: 10.1186/s12974-021-02242-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 08/18/2021] [Indexed: 01/21/2023] Open
Abstract
Background Interleukin-6 (IL6) produced in the context of exercise acts in the hypothalamus reducing obesity-associated inflammation and restoring the control of food intake and energy expenditure. In the hippocampus, some of the beneficial actions of IL6 are attributed to its neurogenesis-inducing properties. However, in the hypothalamus, the putative neurogenic actions of IL6 have never been explored, and its potential to balance energy intake can be an approach to prevent or attenuate obesity. Methods Wild-type (WT) and IL6 knockout (KO) mice were employed to study the capacity of IL6 to induce neurogenesis. We used cell labeling with Bromodeoxyuridine (BrdU), immunofluorescence, and real-time PCR to determine the expression of markers of neurogenesis and neurotransmitters. We prepared hypothalamic neuroprogenitor cells from KO that were treated with IL6 in order to provide an ex vivo model to further characterizing the neurogenic actions of IL6 through differentiation assays. In addition, we analyzed single-cell RNA sequencing data and determined the expression of IL6 and IL6 receptor in specific cell types of the murine hypothalamus. Results IL6 expression in the hypothalamus is low and restricted to microglia and tanycytes, whereas IL6 receptor is expressed in microglia, ependymocytes, endothelial cells, and astrocytes. Exogenous IL6 reduces diet-induced obesity. In outbred mice, obesity-resistance is accompanied by increased expression of IL6 in the hypothalamus. IL6 induces neurogenesis-related gene expression in the hypothalamus and in neuroprogenitor cells, both from WT as well as from KO mice. Conclusion IL6 induces neurogenesis-related gene expression in the hypothalamus of WT mice. In KO mice, the neurogenic actions of IL6 are preserved; however, the appearance of new fully differentiated proopiomelanocortin (POMC) and neuropeptide Y (NPY) neurons is either delayed or disturbed. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02242-8.
Collapse
Affiliation(s)
- Vanessa C Bobbo
- Nursing School, University of Campinas, Campinas, Brazil.,Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Daiane F Engel
- Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Carlos Poblete Jara
- Nursing School, University of Campinas, Campinas, Brazil.,Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Natalia F Mendes
- Nursing School, University of Campinas, Campinas, Brazil.,Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Roberta Haddad-Tovolli
- Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Thais P Prado
- Nursing School, University of Campinas, Campinas, Brazil.,Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Davi Sidarta-Oliveira
- Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Joseane Morari
- Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Licio A Velloso
- Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil
| | - Eliana P Araujo
- Nursing School, University of Campinas, Campinas, Brazil. .,Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP, 13083-877, Brazil.
| |
Collapse
|
9
|
Vinaixa M, Canelles S, González-Murillo Á, Ferreira V, Grajales D, Guerra-Cantera S, Campillo-Calatayud A, Ramírez-Orellana M, Yanes Ó, Frago LM, Valverde ÁM, Barrios V. Increased Hypothalamic Anti-Inflammatory Mediators in Non-Diabetic Insulin Receptor Substrate 2-Deficient Mice. Cells 2021; 10:cells10082085. [PMID: 34440853 PMCID: PMC8391514 DOI: 10.3390/cells10082085] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 08/09/2021] [Accepted: 08/11/2021] [Indexed: 12/11/2022] Open
Abstract
Insulin receptor substrate (IRS) 2 is a key mediator of insulin signaling and IRS-2 knockout (IRS2−/−) mice are a preclinical model to study the development of diabetes, as they develop peripheral insulin resistance and beta-cell failure. The differential inflammatory profile and insulin signaling in the hypothalamus of non-diabetic (ND) and diabetic (D) IRS2−/− mice might be implicated in the onset of diabetes. Because the lipid profile is related to changes in inflammation and insulin sensitivity, we analyzed whether ND IRS2−/− mice presented a different hypothalamic fatty acid metabolism and lipid pattern than D IRS2−/− mice and the relationship with inflammation and markers of insulin sensitivity. ND IRS2−/− mice showed elevated hypothalamic anti-inflammatory cytokines, while D IRS2−/− mice displayed a proinflammatory profile. The increased activity of enzymes related to the pentose-phosphate route and lipid anabolism and elevated polyunsaturated fatty acid levels were found in the hypothalamus of ND IRS2−/− mice. Conversely, D IRS2−/− mice have no changes in fatty acid composition, but hypothalamic energy balance and markers related to anti-inflammatory and insulin-sensitizing properties were reduced. The data suggest that the concurrence of an anti-inflammatory profile, increased insulin sensitivity and polyunsaturated fatty acids content in the hypothalamus may slow down or delay the onset of diabetes.
Collapse
Affiliation(s)
- María Vinaixa
- Metabolomics Platform, IISPV, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, E-43002 Tarragona, Spain; (M.V.); (Ó.Y.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, E-28029 Madrid, Spain; (V.F.); (D.G.)
| | - Sandra Canelles
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.C.); (S.G.-C.); (A.C.-C.); (L.M.F.)
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
| | - África González-Murillo
- Unidad de Terapias Avanzadas, Department of Pediatric Hematology and Oncology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (Á.G.-M.); (M.R.-O.)
| | - Vítor Ferreira
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, E-28029 Madrid, Spain; (V.F.); (D.G.)
- Department of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), E-28029 Madrid, Spain
| | - Diana Grajales
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, E-28029 Madrid, Spain; (V.F.); (D.G.)
- Department of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), E-28029 Madrid, Spain
| | - Santiago Guerra-Cantera
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.C.); (S.G.-C.); (A.C.-C.); (L.M.F.)
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, E-28049 Madrid, Spain
| | - Ana Campillo-Calatayud
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.C.); (S.G.-C.); (A.C.-C.); (L.M.F.)
| | - Manuel Ramírez-Orellana
- Unidad de Terapias Avanzadas, Department of Pediatric Hematology and Oncology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (Á.G.-M.); (M.R.-O.)
| | - Óscar Yanes
- Metabolomics Platform, IISPV, Department of Electronic Engineering (DEEEA), Universitat Rovira i Virgili, E-43002 Tarragona, Spain; (M.V.); (Ó.Y.)
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, E-28029 Madrid, Spain; (V.F.); (D.G.)
| | - Laura M. Frago
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.C.); (S.G.-C.); (A.C.-C.); (L.M.F.)
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Department of Pediatrics, Universidad Autónoma de Madrid, E-28049 Madrid, Spain
| | - Ángela M. Valverde
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, E-28029 Madrid, Spain; (V.F.); (D.G.)
- Department of Metabolism and Cell Signaling, Instituto de Investigaciones Biomédicas Alberto Sols (CSIC-UAM), E-28029 Madrid, Spain
- Correspondence: (Á.M.V.); (V.B.)
| | - Vicente Barrios
- Department of Endocrinology, Hospital Infantil Universitario Niño Jesús, Instituto de Investigación La Princesa, E-28009 Madrid, Spain; (S.C.); (S.G.-C.); (A.C.-C.); (L.M.F.)
- CIBER de Fisiopatología de la Obesidad y Nutrición (CIBEROBN), Instituto de Salud Carlos III, E-28029 Madrid, Spain
- Correspondence: (Á.M.V.); (V.B.)
| |
Collapse
|
10
|
Fatty acids role on obesity induced hypothalamus inflammation: From problem to solution – A review. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2021.03.042] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
11
|
Understanding the appetite modulation pathways: The role of the FFA1 and FFA4 receptors. Biochem Pharmacol 2021; 186:114503. [PMID: 33711286 DOI: 10.1016/j.bcp.2021.114503] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 02/27/2021] [Accepted: 03/02/2021] [Indexed: 11/24/2022]
Abstract
Pharmaconutrition is an area of current interest, especially concerning the advances in the pharmacology of nutrient-sensing receptors, as have been accomplished in the last 20 years. The family of free fatty acid (FFA) receptors is composed of four members, sequentially named as FFA1 to FFA4, which are activated by the short to long-chain fatty acids. The affinity of the FFA1 and FFA4 receptors for the omega-3 polyunsaturated fatty acids prompted pre-clinical and clinical investigations regarding their involvement in metabolic diseases. The main studies have been focused on the receptors' expression analyses, the featuring of knockout mice, and the assessment of selective synthetic ligands. These clearly have indicated a relevant role for FFA1 and FFA4 in the peripheral and central circuits for the regulation of energetic metabolism. This review article aimed to discuss the relevance of the FFA1 and FFA4 receptors in appetite-related complications, mainly related to obesity, cancer cachexia, and anorexia in the elderly, emphasizing whether their pharmacological modulation might be useful for the management of these disorders.
Collapse
|
12
|
Parnova RG. GPR40/FFA1 Free Fatty Acid Receptors and Their Functional Role. NEUROSCIENCE AND BEHAVIORAL PHYSIOLOGY 2021; 51:256-264. [DOI: 10.1007/s11055-021-01064-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 03/08/2020] [Accepted: 03/17/2020] [Indexed: 01/05/2025]
|
13
|
Role of polyunsaturated fatty acids in ischemic stroke - A perspective of specialized pro-resolving mediators. Clin Nutr 2021; 40:2974-2987. [PMID: 33509668 DOI: 10.1016/j.clnu.2020.12.037] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 12/14/2020] [Accepted: 12/26/2020] [Indexed: 12/17/2022]
Abstract
Polyunsaturated fatty acids (PUFAs) have been proposed as beneficial for cardiovascular health. However, results from both epidemiological studies and clinical trials have been inconsistent, whereas most of the animal studies showed promising benefits of PUFAs in the prevention and treatment of ischemic stroke. In recent years, it has become clear that PUFAs are metabolized into various types of bioactive derivatives, including the specialized pro-resolving mediators (SPMs). SPMs exert multiple biofunctions, such as to limit excessive inflammatory responses, regulate lipid metabolism and immune cell functions, decrease production of pro-inflammatory factors, increase anti-inflammatory mediators, as well as to promote tissue repair and homeostasis. Inflammation has been recognised as a key contributor to the pathophysiology of acute ischemic stroke. Owing to their potent pro-resolving actions, SPMs are potential for development of novel anti-stroke therapy. In this review, we will summarize current knowledge of epidemiological studies, basic research and clinical trials concerning PUFAs in stroke prevention and treatment, with special attention to SPMs as the unsung heroes behind PUFAs.
Collapse
|
14
|
Sharif A, Fitzsimons CP, Lucassen PJ. Neurogenesis in the adult hypothalamus: A distinct form of structural plasticity involved in metabolic and circadian regulation, with potential relevance for human pathophysiology. HANDBOOK OF CLINICAL NEUROLOGY 2021; 179:125-140. [PMID: 34225958 DOI: 10.1016/b978-0-12-819975-6.00006-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The adult brain harbors specific niches where stem cells undergo substantial plasticity and, in some regions, generate new neurons throughout life. This phenomenon is well known in the subventricular zone of the lateral ventricles and the subgranular zone of the hippocampus and has recently also been described in the hypothalamus of several rodent and primate species. After a brief overview of preclinical studies illustrating the pathophysiologic significance of hypothalamic neurogenesis in the control of energy metabolism, reproduction, thermoregulation, sleep, and aging, we review current literature on the neurogenic niche of the human hypothalamus. A comparison of the organization of the niche between humans and rodents highlights some common features, but also substantial differences, e.g., in the distribution and extent of the hypothalamic neural stem cells. Exploring the full dynamics of hypothalamic neurogenesis in humans raises a formidable challenge however, given among others, inherent technical limitations. We close with discussing possible functional role(s) of the human hypothalamic niche, and how gaining more insights into this form of plasticity could be relevant for a better understanding of pathologies associated with disturbed hypothalamic function.
Collapse
Affiliation(s)
- Ariane Sharif
- Lille Neuroscience & Cognition, University of Lille, Lille, France.
| | - Carlos P Fitzsimons
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Brain Plasticity Group, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
15
|
Dragano NR, Monfort-Pires M, Velloso LA. Mechanisms Mediating the Actions of Fatty Acids in the Hypothalamus. Neuroscience 2020; 447:15-27. [PMID: 31689488 DOI: 10.1016/j.neuroscience.2019.10.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 10/02/2019] [Accepted: 10/03/2019] [Indexed: 12/12/2022]
|
16
|
Recabal A, Fernández P, López S, Barahona MJ, Ordenes P, Palma A, Elizondo-Vega R, Farkas C, Uribe A, Caprile T, Sáez JC, García-Robles MA. The FGF2-induced tanycyte proliferation involves a connexin 43 hemichannel/purinergic-dependent pathway. J Neurochem 2020; 156:182-199. [PMID: 32936929 PMCID: PMC7894481 DOI: 10.1111/jnc.15188] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/03/2020] [Accepted: 09/08/2020] [Indexed: 11/29/2022]
Abstract
In the adult hypothalamus, the neuronal precursor role is attributed to the radial glia-like cells that line the third-ventricle (3V) wall called tanycytes. Under nutritional cues, including hypercaloric diets, tanycytes proliferate and differentiate into mature neurons that moderate body weight, suggesting that hypothalamic neurogenesis is an adaptive mechanism in response to metabolic changes. Previous studies have shown that the tanycyte glucosensing mechanism depends on connexin-43 hemichannels (Cx43 HCs), purine release, and increased intracellular free calcium ion concentration [(Ca2+ )i ] mediated by purinergic P2Y receptors. Since, Fibroblast Growth Factor 2 (FGF2) causes similar purinergic events in other cell types, we hypothesize that this pathway can be also activated by FGF2 in tanycytes to promote their proliferation. Here, we used bromodeoxyuridine (BrdU) incorporation to evaluate if FGF2-induced tanycyte cell division is sensitive to Cx43 HC inhibition in vitro and in vivo. Immunocytochemical analyses showed that cultured tanycytes maintain the expression of in situ markers. After FGF2 exposure, tanycytic Cx43 HCs opened, enabling release of ATP to the extracellular milieu. Moreover, application of external ATP was enough to induce their cell division, which could be suppressed by Cx43 HC or P2Y1-receptor inhibitors. Similarly, in vivo experiments performed on rats by continuous infusion of FGF2 and a Cx43 HC inhibitor into the 3V, demonstrated that FGF2-induced β-tanycyte proliferation is sensitive to Cx43 HC blockade. Thus, FGF2 induced Cx43 HC opening, triggered purinergic signaling, and increased β-tanycytes proliferation, highlighting some of the molecular mechanisms involved in the cell division response of tanycyte. This article has an Editorial Highlight see https://doi.org/10.1111/jnc.15218.
Collapse
Affiliation(s)
- Antonia Recabal
- Departamento de Biología Celular, Universidad de Concepción, Concepción, Chile
| | - Paola Fernández
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago
| | - Sergio López
- Departamento de Biología Celular, Universidad de Concepción, Concepción, Chile
| | - María J Barahona
- Departamento de Biología Celular, Universidad de Concepción, Concepción, Chile
| | - Patricio Ordenes
- Departamento de Biología Celular, Universidad de Concepción, Concepción, Chile
| | - Alejandra Palma
- Departamento de Biología Celular, Universidad de Concepción, Concepción, Chile
| | | | - Carlos Farkas
- Research Institute in Oncology and Hematology, Winnipeg, Manitoba, Canada
| | - Amparo Uribe
- Departamento de Biología Celular, Universidad de Concepción, Concepción, Chile
| | - Teresa Caprile
- Departamento de Biología Celular, Universidad de Concepción, Concepción, Chile
| | - Juan C Sáez
- Departamento de Fisiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago.,Instituto de Neurociencias, Centro Interdisciplinario de Neurociencias de Valparaíso, Universidad de Valparaíso, Valparaíso, Chile
| | | |
Collapse
|
17
|
Jurkowski MP, Bettio L, K. Woo E, Patten A, Yau SY, Gil-Mohapel J. Beyond the Hippocampus and the SVZ: Adult Neurogenesis Throughout the Brain. Front Cell Neurosci 2020; 14:576444. [PMID: 33132848 PMCID: PMC7550688 DOI: 10.3389/fncel.2020.576444] [Citation(s) in RCA: 118] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 08/19/2020] [Indexed: 12/31/2022] Open
Abstract
Convincing evidence has repeatedly shown that new neurons are produced in the mammalian brain into adulthood. Adult neurogenesis has been best described in the hippocampus and the subventricular zone (SVZ), in which a series of distinct stages of neuronal development has been well characterized. However, more recently, new neurons have also been found in other brain regions of the adult mammalian brain, including the hypothalamus, striatum, substantia nigra, cortex, and amygdala. While some studies have suggested that these new neurons originate from endogenous stem cell pools located within these brain regions, others have shown the migration of neurons from the SVZ to these regions. Notably, it has been shown that the generation of new neurons in these brain regions is impacted by neurologic processes such as stroke/ischemia and neurodegenerative disorders. Furthermore, numerous factors such as neurotrophic support, pharmacologic interventions, environmental exposures, and stem cell therapy can modulate this endogenous process. While the presence and significance of adult neurogenesis in the human brain (and particularly outside of the classical neurogenic regions) is still an area of debate, this intrinsic neurogenic potential and its possible regulation through therapeutic measures present an exciting alternative for the treatment of several neurologic conditions. This review summarizes evidence in support of the classic and novel neurogenic zones present within the mammalian brain and discusses the functional significance of these new neurons as well as the factors that regulate their production. Finally, it also discusses the potential clinical applications of promoting neurogenesis outside of the classical neurogenic niches, particularly in the hypothalamus, cortex, striatum, substantia nigra, and amygdala.
Collapse
Affiliation(s)
- Michal P. Jurkowski
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
| | - Luis Bettio
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| | - Emma K. Woo
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
| | - Anna Patten
- Centre for Interprofessional Clinical Simulation Learning (CICSL), Royal Jubilee Hospital, Victoria, BC, Canada
| | - Suk-Yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong
| | - Joana Gil-Mohapel
- Island Medical Program, University of British Columbia, Vancouver, BC, Canada
- Division of Medical Sciences, University of Victoria, Victoria, BC, Canada
| |
Collapse
|
18
|
Mostafa H, Barakat L, Abdo WS, Khalil RM. Omega-3 offers better hypothalamus protection by decreasing POMC expression and elevating ghrelin hormone: a prospective trial to overcome methotrexate-induced anorexia. Endocrine 2020; 69:358-367. [PMID: 32424681 DOI: 10.1007/s12020-020-02342-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 05/05/2020] [Indexed: 12/23/2022]
Abstract
PURPOSE Methotrexate (MTX) therapy is widely used in treatment of different types of diseases including inflammatory diseases, autoimmune disorders, and cancer. However, most of patients respond well to MTX, they suffer from multiple side effects including severe anorexia. Omega-3 fatty acid possesses many beneficial biological activities. Therefore, the objective of our study is to explore the effect of the combined modality of omega-3 (400 mg/kg/day) in MTX-induced anorexia in rats. METHODS The effect of MTX alone and in combination with omega-3 on the body weight, ghrelin hormone level, histopathological findings of taste buds and hypothalamus and POMC gene expression were investigated. RESULTS Interestingly, the capability of omega-3 to overcome the anorexic effect of MTX could be manifested by controlling weight loss, increasing serum HDL, elevating the ghrelin level as well as reducing both lesions within taste buds and hypothalamus and hypothalamic POMC gene expression. CONCLUSIONS our findings revealed that the omega-3 might be used as a complementary supplement during the MTX therapy to ameliorate its anorexic effect.
Collapse
Affiliation(s)
- Heba Mostafa
- Faculty of Science, Biochemistry Department, Port Said University, Port Said, Egypt
| | - Lamia Barakat
- Faculty of Science, Biochemistry Department, Port Said University, Port Said, Egypt
| | - Walied S Abdo
- Faculty of Veterinary medicine, Pathology Department, Kafrelsheik University, Kafr-Elsheik, 33516, Egypt
| | - Rania M Khalil
- Faculty of Pharmacy, Biochemistry department, Delta University for Science and Technology, Gamasa, Egypt.
| |
Collapse
|
19
|
Engel DF, Bobbo VCD, Solon CS, Nogueira GA, Moura-Assis A, Mendes NF, Zanesco AM, Papangelis A, Ulven T, Velloso LA. Activation of GPR40 induces hypothalamic neurogenesis through p38- and BDNF-dependent mechanisms. Sci Rep 2020; 10:11047. [PMID: 32632088 PMCID: PMC7338363 DOI: 10.1038/s41598-020-68110-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Hypothalamic adult neurogenesis provides the basis for renewal of neurons involved in the regulation of whole-body energy status. In addition to hormones, cytokines and growth factors, components of the diet, particularly fatty acids, have been shown to stimulate hypothalamic neurogenesis; however, the mechanisms behind this action are unknown. Here, we hypothesized that GPR40 (FFAR1), the receptor for medium and long chain unsaturated fatty acids, could mediate at least part of the neurogenic activity in the hypothalamus. We show that a GPR40 ligand increased hypothalamic cell proliferation and survival in adult mice. In postnatal generated neurospheres, acting in synergy with brain-derived neurotrophic factor (BDNF) and interleukin 6, GPR40 activation increased the expression of doublecortin during the early differentiation phase and of the mature neuronal marker, microtubule-associated protein 2 (MAP2), during the late differentiation phase. In Neuro-2a proliferative cell-line GPR40 activation increased BDNF expression and p38 activation. The chemical inhibition of p38 abolished GPR40 effect in inducing neurogenesis markers in neurospheres, whereas BDNF immunoneutralization inhibited GPR40-induced cell proliferation in the hypothalamus of adult mice. Thus, GPR40 acts through p38 and BDNF to induce hypothalamic neurogenesis. This study provides mechanistic advance in the understating of how a fatty acid receptor regulates adult hypothalamic neurogenesis.
Collapse
Affiliation(s)
- Daiane F Engel
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil.
| | - Vanessa C D Bobbo
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Carina S Solon
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Guilherme A Nogueira
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Alexandre Moura-Assis
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Natalia F Mendes
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Ariane M Zanesco
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil
| | - Athanasios Papangelis
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Trond Ulven
- Department of Drug Design and Pharmacology, University of Copenhagen, 2100, Copenhagen, Denmark
| | - Licio A Velloso
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP, 13084-970, Brazil.
| |
Collapse
|
20
|
Yamashima T, Ota T, Mizukoshi E, Nakamura H, Yamamoto Y, Kikuchi M, Yamashita T, Kaneko S. Intake of ω-6 Polyunsaturated Fatty Acid-Rich Vegetable Oils and Risk of Lifestyle Diseases. Adv Nutr 2020; 11:1489-1509. [PMID: 32623461 PMCID: PMC7666899 DOI: 10.1093/advances/nmaa072] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 10/30/2019] [Accepted: 05/26/2020] [Indexed: 12/28/2022] Open
Abstract
Although excessive consumption of deep-fried foods is regarded as 1 of the most important epidemiological factors of lifestyle diseases such as Alzheimer's disease, type 2 diabetes, and obesity, the exact mechanism remains unknown. This review aims to discuss whether heated cooking oil-derived peroxidation products cause cell degeneration/death for the occurrence of lifestyle diseases. Deep-fried foods cooked in ω-6 PUFA-rich vegetable oils such as rapeseed (canola), soybean, sunflower, and corn oils, already contain or intrinsically generate "hydroxynonenal" by peroxidation. As demonstrated previously, hydroxynonenal promotes carbonylation of heat-shock protein 70.1 (Hsp70.1), with the resultant impaired ability of cells to recycle damaged proteins and stabilize the lysosomal membrane. Until now, the implication of lysosomal/autophagy failure due to the daily consumption of ω-6 PUFA-rich vegetable oils in the progression of cell degeneration/death has not been reported. Since the "calpain-cathepsin hypothesis" was formulated as a cause of ischemic neuronal death in 1998, its relevance to Alzheimer's neuronal death has been suggested with particular attention to hydroxynonenal. However, its relevance to cell death of the hypothalamus, liver, and pancreas, especially related to appetite/energy control, is unknown. The hypothalamus senses information from both adipocyte-derived leptin and circulating free fatty acids. Concentrations of circulating fatty acid and its oxidized form, especially hydroxynonenal, are increased in obese and/or aged subjects. As overactivation of the fatty acid receptor G-protein coupled receptor 40 (GPR40) in response to excessive or oxidized fatty acids in these subjects may lead to the disruption of Ca2+ homeostasis, it should be evaluated whether GPR40 overactivation contributes to diverse cell death. Here, we describe the molecular implication of ω-6 PUFA-rich vegetable oil-derived hydroxynonenal in lysosomal destabilization leading to cell death. By oxidizing Hsp70.1, both the dietary PUFA- (exogenous) and the membrane phospholipid- (intrinsic) peroxidation product "hydroxynonenal," when combined, may play crucial roles in the occurrence of diverse lifestyle diseases including Alzheimer's disease.
Collapse
Affiliation(s)
| | | | | | | | - Yasuhiko Yamamoto
- Biochemistry and Molecular Vascular Biology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | | | | | | |
Collapse
|
21
|
Sênos Demarco R, Clémot M, Jones DL. The impact of ageing on lipid-mediated regulation of adult stem cell behavior and tissue homeostasis. Mech Ageing Dev 2020; 189:111278. [PMID: 32522455 DOI: 10.1016/j.mad.2020.111278] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 05/05/2020] [Accepted: 06/01/2020] [Indexed: 02/06/2023]
Abstract
Adult stem cells sustain tissue homeostasis throughout life and provide an important reservoir of cells capable of tissue repair in response to stress and tissue damage. Age-related changes to stem cells and/or the specialized niches that house them have been shown to negatively impact stem cell maintenance and activity. In addition, metabolic inputs have surfaced as another crucial layer in the control of stem cell behavior (Chandel et al., 2016; Folmes and Terzic, 2016; Ito and Suda, 2014; Mana et al., 2017; Shyh-Chang and Ng, 2017). Here, we will present a brief review of how lipid metabolism influences adult stem cell behavior under homeostatic conditions and speculate on how changes in lipid metabolism may impact stem cell ageing. This review considers the future of lipid metabolism research in stem cells, with the long-term goal of identifying mechanisms that could be targeted to counter or slow the age-related decline in stem cell function.
Collapse
Affiliation(s)
- Rafael Sênos Demarco
- Department of Molecular, Cell and Developmental Biology, Los Angeles, CA, 90095, USA
| | - Marie Clémot
- Department of Molecular, Cell and Developmental Biology, Los Angeles, CA, 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - D Leanne Jones
- Department of Molecular, Cell and Developmental Biology, Los Angeles, CA, 90095, USA; Molecular Biology Institute, Los Angeles, CA, 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
22
|
Tonon MC, Vaudry H, Chuquet J, Guillebaud F, Fan J, Masmoudi-Kouki O, Vaudry D, Lanfray D, Morin F, Prevot V, Papadopoulos V, Troadec JD, Leprince J. Endozepines and their receptors: Structure, functions and pathophysiological significance. Pharmacol Ther 2020; 208:107386. [DOI: 10.1016/j.pharmthera.2019.06.008] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 06/20/2019] [Indexed: 02/06/2023]
|
23
|
Clémot M, Sênos Demarco R, Jones DL. Lipid Mediated Regulation of Adult Stem Cell Behavior. Front Cell Dev Biol 2020; 8:115. [PMID: 32185173 PMCID: PMC7058546 DOI: 10.3389/fcell.2020.00115] [Citation(s) in RCA: 49] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Accepted: 02/11/2020] [Indexed: 12/18/2022] Open
Abstract
Adult stem cells constitute an important reservoir of self-renewing progenitor cells and are crucial for maintaining tissue and organ homeostasis. The capacity of stem cells to self-renew or differentiate can be attributed to distinct metabolic states, and it is now becoming apparent that metabolism plays instructive roles in stem cell fate decisions. Lipids are an extremely vast class of biomolecules, with essential roles in energy homeostasis, membrane structure and signaling. Imbalances in lipid homeostasis can result in lipotoxicity, cell death and diseases, such as cardiovascular disease, insulin resistance and diabetes, autoimmune disorders and cancer. Therefore, understanding how lipid metabolism affects stem cell behavior offers promising perspectives for the development of novel approaches to control stem cell behavior either in vitro or in patients, by modulating lipid metabolic pathways pharmacologically or through diet. In this review, we will first address how recent progress in lipidomics has created new opportunities to uncover stem-cell specific lipidomes. In addition, genetic and/or pharmacological modulation of lipid metabolism have shown the involvement of specific pathways, such as fatty acid oxidation (FAO), in regulating adult stem cell behavior. We will describe and compare findings obtained in multiple stem cell models in order to provide an assessment on whether unique lipid metabolic pathways may commonly regulate stem cell behavior. We will then review characterized and potential molecular mechanisms through which lipids can affect stem cell-specific properties, including self-renewal, differentiation potential or interaction with the niche. Finally, we aim to summarize the current knowledge of how alterations in lipid homeostasis that occur as a consequence of changes in diet, aging or disease can impact stem cells and, consequently, tissue homeostasis and repair.
Collapse
Affiliation(s)
- Marie Clémot
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, United States
| | - Rafael Sênos Demarco
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
| | - D. Leanne Jones
- Department of Molecular, Cell and Developmental Biology, University of California, Los Angeles, Los Angeles, CA, United States
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, United States
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, United States
| |
Collapse
|
24
|
Kelley L, Verlezza S, Long H, Loka M, Walker CD. Increased Hypothalamic Projections to the Lateral Hypothalamus and Responses to Leptin in Rat Neonates From High Fat Fed Mothers. Front Neurosci 2020; 13:1454. [PMID: 32082105 PMCID: PMC7005214 DOI: 10.3389/fnins.2019.01454] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 12/30/2019] [Indexed: 12/11/2022] Open
Abstract
The lateral hypothalamus (LHA) is a central hub in the regulation of food intake and metabolism, as it integrates homeostatic and hedonic circuits. During early development, maturing input to and output from the LHA might be particularly sensitive to environmental dietary changes. We examined the effects of a maternal high fat diet (HFD, 60% Kcal in fat) on the density of hypothalamic projections to the orexin (ORX-A) field of the LHA in 10 day-old (PND10) rat pups using retrograde labeling with fluorescent microspheres. We also compared responsiveness of phenotypically identified LHA neurons to leptin administration (3 mg/kg, bw) between pups from control (CD) or high fat (HFD) fed mothers on PND10 and 15-16, at the onset of independent feeding. HFD pups exhibited a higher density of LHA projections (p = 0.05) from the ventromedial hypothalamus (VMH) compared to CD pups and these originated from both SF-1 and BDNF-positive neurons in the VMH. Increased circulating leptin levels in HFD pups, particularly on PND15-16 was consistent with enhanced pSTAT3 responses to leptin in the orexin (ORX-A) field of the LHA, with some of the activated neurons expressing a GABA, but not CART phenotype. ORX-A neurons colocalizing with pERK were significantly higher in PND15-16 HFD pups compared to CD pups, and leptin-induced increase in pERK signaling was only observed in CD pups. There was no significant effect of leptin on pERK in HFD pups. These results suggest that perinatal maternal high fat feeding increases hypothalamic projections to the ORX-A field of the LHA, increases basal activation of ORX-A neurons and direct responsiveness of LHA neurons to leptin. Since these various LHA neuronal populations project quite heavily to Dopamine (DA) neurons in the ventral tegmental area, they might participate in the early dietary programming of mesocorticolimbic reward circuits and food intake.
Collapse
Affiliation(s)
- Lyla Kelley
- Douglas Mental Health University Institute, Montreal, QC, Canada.,Department of Anatomy and Cell Biology, McGill University, Montreal, QC, Canada
| | | | - Hong Long
- Douglas Mental Health University Institute, Montreal, QC, Canada
| | - Mary Loka
- Douglas Mental Health University Institute, Montreal, QC, Canada.,Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Claire-Dominique Walker
- Douglas Mental Health University Institute, Montreal, QC, Canada.,Department of Psychiatry, McGill University, Montreal, QC, Canada
| |
Collapse
|
25
|
Dietary fatty acid quality affects systemic parameters and promotes prostatitis and pre-neoplastic lesions. Sci Rep 2019; 9:19233. [PMID: 31848441 PMCID: PMC6917739 DOI: 10.1038/s41598-019-55882-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Accepted: 12/04/2019] [Indexed: 01/02/2023] Open
Abstract
Environmental and nutritional factors, including fatty acids (FA), are associated with prostatitis, benign prostate hyperplasia and prostate cancer. We hypothesized that different FA in normolipidic diets (7%) affect prostate physiology, increasing the susceptibility to prostate disorders. Thus, we fed male C57/BL6 mice with normolipidic diets based on linseed oil, soybean oil or lard (varying saturated and unsaturated FA contents and ω-3/ω-6 ratios) for 12 or 32 weeks after weaning and examined structural and functional parameters of the ventral prostate (VP) in the systemic metabolic context. Mongolian gerbils were included because they present a metabolic detour for low water consumption (i.e., oxidize FA to produce metabolic water). A linseed oil-based diet (LO, 67.4% PUFAs, ω-3/ω-6 = 3.70) resulted in a thermogenic profile, while a soybean oil-based diet (SO, 52.7% PUFAs, ω-3/ω-6 = 0.11) increased body growth and adiposity. Mice fed lard (PF, 13.1% PUFA, ω-3/ω-6 = 0.07) depicted a biphasic growth, resulting in decreased adiposity in adulthood. SO and PF resulted in hepatic steatosis and steatohepatitis, respectively. PF and SO increased prostate epithelial volume, and lard resulted in epithelial hyperplasia. Animals in the LO group had smaller prostates with predominant atrophic epithelia and inflammatory loci. Inflammatory cells were frequent in the VP of PF mice (predominantly stromal) and LO mice (predominantly luminal). RNAseq after 12 weeks revealed good predictors of a later-onset inflammation. The transcriptome unveiled ontologies related to ER stress after 32 weeks on PF diets. In conclusion, different FA qualities result in different metabolic phenotypes and differentially impact prostate size, epithelial volume, inflammation and gene expression.
Collapse
|
26
|
Cleal JK, Bruce KD, Shearer JL, Thomas H, Plume J, Gregory L, Shepard JN, Spiers-Fitzgerald KL, Mani R, Lewis RM, Lillycrop KA, Hanson MA, Byrne CD, Cagampang FR. Maternal Obesity during Pregnancy Alters Daily Activity and Feeding Cycles, and Hypothalamic Clock Gene Expression in Adult Male Mouse Offspring. Int J Mol Sci 2019; 20:E5408. [PMID: 31671625 PMCID: PMC6862679 DOI: 10.3390/ijms20215408] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Revised: 10/12/2019] [Accepted: 10/28/2019] [Indexed: 02/07/2023] Open
Abstract
An obesogenic diet adversely affects the endogenous mammalian circadian clock, altering daily activity and metabolism, and resulting in obesity. We investigated whether an obese pregnancy can alter the molecular clock in the offspring hypothalamus, resulting in changes to their activity and feeding rhythms. Female mice were fed a control (C, 7% kcal fat) or high fat diet (HF, 45% kcal fat) before mating and throughout pregnancy. Male offspring were fed the C or HF diet postweaning, resulting in four offspring groups: C/C, C/HF, HF/C, and HF/HF. Daily activity and food intake were monitored, and at 15 weeks of age were killed at six time-points over 24 h. The clock genes Clock, Bmal1, Per2, and Cry2 in the suprachiasmatic nucleus (SCN) and appetite genes Npy and Pomc in the arcuate nucleus (ARC) were measured. Daily activity and feeding cycles in the HF/C, C/HF, and HF/HF offspring were altered, with increased feeding bouts and activity during the day and increased food intake but reduced activity at night. Gene expression patterns and levels of Clock, Bmal1, Per2, and Cry2 in the SCN and Npy and Pomc in the ARC were altered in HF diet-exposed offspring. The altered expression of hypothalamic molecular clock components and appetite genes, together with changes in activity and feeding rhythms, could be contributing to offspring obesity.
Collapse
Affiliation(s)
- Jane K Cleal
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Kimberley D Bruce
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Jasmin L Shearer
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Hugh Thomas
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Jack Plume
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Louise Gregory
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - James N Shepard
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Kerry L Spiers-Fitzgerald
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Ravi Mani
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Rohan M Lewis
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Karen A Lillycrop
- Centre for Biological Sciences, Faculty of Natural and Environmental Sciences, University of Southampton, Southampton SO17 1BJ, UK.
| | - Mark A Hanson
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Christopher D Byrne
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| | - Felino R Cagampang
- School of Human Development and Health, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton SO16 6YD, UK.
| |
Collapse
|
27
|
Hypothalamic inflammation and obesity: a mechanistic review. Arch Pharm Res 2019; 42:383-392. [PMID: 30835074 DOI: 10.1007/s12272-019-01138-9] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Accepted: 02/24/2019] [Indexed: 02/08/2023]
Abstract
Obesity is one of the worldwide prevalent disease caused by the imbalance between food intake and energy expenditure. Over a 100 years of research demonstrate that hypothalamus is the critical brain region regulating energy homeostasis, and evidences suggest the participation of non-neuronal populations such as astrocytes and microglia in the regulation of energy homeostasis. Recently, fat-rich diet induced hypothalamic inflammation has been found to deregulate the energy homeostasis, leading to the insulin resistance, glucose intolerance, and obesity. Several underlying mechanisms have been proposed, yet compelling evidences require further elucidations. This review discusses the up to date proposed mechanisms by which fat-rich diet induces hypothalamic inflammation and obesity.
Collapse
|
28
|
Pawełczyk T, Grancow-Grabka M, Trafalska E, Szemraj J, Żurner N, Pawełczyk A. An increase in plasma brain derived neurotrophic factor levels is related to n-3 polyunsaturated fatty acid efficacy in first episode schizophrenia: secondary outcome analysis of the OFFER randomized clinical trial. Psychopharmacology (Berl) 2019; 236:2811-2822. [PMID: 31098654 PMCID: PMC6695351 DOI: 10.1007/s00213-019-05258-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 04/24/2019] [Indexed: 12/15/2022]
Abstract
RATIONALE N-3 polyunsaturated fatty acids (n-3 PUFA) influence multiple biochemical mechanisms postulated in the pathogenesis of schizophrenia that may influence BDNF synthesis. OBJECTIVES A randomized placebo-controlled study was designed to compare the efficacy of a 26-week intervention composed of either 2.2 g/day of n-3 PUFA or olive oil placebo, with regard to symptom severity in first-episode schizophrenia patients. The secondary outcome measure of the study was to describe the association between n-3 PUFA clinical effect and changes in peripheral BDNF levels. METHODS Seventy-one patients aged 16-35 were enrolled in the study and randomly assigned to the following study arms: 36 to the EPA + DHA group and 35 to the placebo group. Plasma BDNF levels were assessed three times, at baseline and at weeks 8 and 26 of the intervention. BDNF levels were determined in plasma samples using Quantikine Human BDNF ELISA kit. Plasma BDNF level changes were further correlated with changes in the severity of symptoms in different clinical domains. RESULTS A significantly greater increase in plasma BDNF levels was observed in the intervention compared to the placebo group (Cohen's d = 1.54). Changes of BDNF levels inversely correlated with change in depressive symptoms assessed using the Calgary Depression Rating Scale in Schizophrenia (Pearson's r = - 0.195; p = 0.018). CONCLUSIONS The efficacy of a six-month intervention with n-3 PUFA observed in first-episode schizophrenia may be related to an increase in BDNF levels, which may be triggered by the activation of intracellular signaling pathways including transcription factors such as cAMP-reactive element binding protein.
Collapse
Affiliation(s)
- Tomasz Pawełczyk
- Department of Affective and Psychotic Disorders, Medical University of Lodz, ul. Czechoslowacka 8/10, 92-216, Lodz, Poland.
| | - Marta Grancow-Grabka
- 0000 0001 2165 3025grid.8267.bChild and Adolescent Psychiatry Unit, Central Teaching Hospital, Medical University of Lodz, ul. Pomorska 251, 92-213 Lodz, Poland
| | - Elżbieta Trafalska
- 0000 0001 2165 3025grid.8267.bDepartment of Nutrition Hygiene and Epidemiology, Medical University of Lodz, ul. Jaracza 63, 90-251 Lodz, Poland
| | - Janusz Szemraj
- 0000 0001 2165 3025grid.8267.bDepartment of Medical Biochemistry, Medical University of Lodz, ul. Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Natalia Żurner
- 0000 0001 2165 3025grid.8267.bChild and Adolescent Psychiatry Unit, Central Teaching Hospital, Medical University of Lodz, ul. Pomorska 251, 92-213 Lodz, Poland
| | - Agnieszka Pawełczyk
- 0000 0001 2165 3025grid.8267.bDepartment of Affective and Psychotic Disorders, Medical University of Lodz, ul. Czechoslowacka 8/10, 92-216 Lodz, Poland
| |
Collapse
|
29
|
Pimentel LL, Fontes AL, Salsinha AS, Cardoso BB, Gomes AM, Rodríguez-Alcalá LM. Microbiological In Vivo Production of CLNA as a Tool in the Regulation of Host Microbiota in Obesity Control. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2019:369-394. [DOI: 10.1016/b978-0-444-64183-0.00010-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
30
|
Mendes NF, Kim YB, Velloso LA, Araújo EP. Hypothalamic Microglial Activation in Obesity: A Mini-Review. Front Neurosci 2018; 12:846. [PMID: 30524228 PMCID: PMC6262396 DOI: 10.3389/fnins.2018.00846] [Citation(s) in RCA: 68] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 10/29/2018] [Indexed: 01/22/2023] Open
Abstract
Emerging data demonstrate that microglia activation plays a pivotal role in the development of hypothalamic inflammation in obesity. Early after the introduction of a high-fat diet, hypothalamic microglia undergo morphological, and functional changes in response to excessive dietary saturated fats. Initially the resident microglia are affected; however, as diet-induced obesity persists, bone marrow-derived myeloid cells gradually replace resident microglia. Genetic and pharmacological approaches aimed at dampening the inflammatory activity in the hypothalamus of experimental models of obesity have proven beneficial to correct the obese phenotype and improve metabolic abnormalities commonly associated with obesity. These approaches provide an experimental proof-of-concept that hypothalamic inflammation is central to the pathophysiology of obesity; understanding the details of the roles played by microglia in this process may help the development of preventive and therapeutic advances in the field. In this review, we discuss the potential mechanisms underlying hypothalamic microglial activation in high-fat induced obesity.
Collapse
Affiliation(s)
- Natália F Mendes
- School of Nursing, State University of Campinas, Campinas, Brazil.,Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism - Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| | - Young-Bum Kim
- Department of Medicine, Division of Endocrinology, Diabetes, and Metabolism - Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
| | - Lício A Velloso
- Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Rio de Janeiro, Brazil
| | - Eliana P Araújo
- School of Nursing, State University of Campinas, Campinas, Brazil.,Laboratory of Cell Signaling, Obesity and Comorbidities Research Center, State University of Campinas, Campinas, Brazil
| |
Collapse
|
31
|
Abstract
PUFA modulate hypothalamic-pituitary-adrenal (HPA) axis activity and cortisol concentrations and therefore affect physiological stress responses and the regulation of energy balance in the short- and long-term. Especially dietary intake of n-3 PUFA and a lowered n-6:n-3 ratio are highly encouraged due to beneficial and diminishing effects on basal cortisol secretions. However, the time of such effects to occur and how plasma PUFA patterns affect cortisol concentrations in the short-term was rarely investigated. In order to address this, we supplemented forty male and forty female guinea pigs with diets high in the essential PUFA α-linolenic acid (ALA, 18 : 3n-3) and linoleic acid (LA, 18 : 2n-6) for 20 d. Saliva cortisol concentrations in relation to altering plasma PUFA patterns during this time span were analysed in a repeated measurement design both during basal conditions (individual housing) in 5-d intervals and during stressful social confrontations. We detected very fast plasma PUFA accumulation rates, corresponding to the major dietary PUFA, which resulted in plasma PUFA plateau phases after 10 d. ALA negatively and LA positively affected saliva cortisol concentrations throughout the study. A positive effect of the plasma n-6:n-3 ratio on saliva cortisol concentrations was detected during peak plasma PUFA accumulations and social confrontations, while no effects were detected in relation to plasma PUFA plateau phases. These results suggest that the plasma n-6:n-3 ratio diminishes HPA axis activity during altered physiological conditions only and highlights the importance of altering plasma PUFA patterns for HPA axis functions and the control of energy balance and physiological stress.
Collapse
|
32
|
Sona C, Kumar A, Dogra S, Kumar BA, Umrao D, Yadav PN. Docosahexaenoic acid modulates brain-derived neurotrophic factor via GPR40 in the brain and alleviates diabesity-associated learning and memory deficits in mice. Neurobiol Dis 2018; 118:94-107. [DOI: 10.1016/j.nbd.2018.07.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2017] [Revised: 06/22/2018] [Accepted: 07/04/2018] [Indexed: 12/19/2022] Open
|
33
|
Pflimlin E, Bielohuby M, Korn M, Breitschopf K, Löhn M, Wohlfart P, Konkar A, Podeschwa M, Bärenz F, Pfenninger A, Schwahn U, Opatz T, Reimann M, Petry S, Tennagels N. Acute and Repeated Treatment with 5-PAHSA or 9-PAHSA Isomers Does Not Improve Glucose Control in Mice. Cell Metab 2018; 28:217-227.e13. [PMID: 29937376 DOI: 10.1016/j.cmet.2018.05.028] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Revised: 02/13/2018] [Accepted: 05/29/2018] [Indexed: 01/08/2023]
Abstract
Fatty acid esters of hydroxylated fatty acids (FAHFAs) were discovered as a novel class of endogenous mammalian lipids whose profound effects on metabolism have been shown. In the current study, in vitro and in vivo the metabolic effects of two of these FAHFAs, namely palmitic acid-5- (or -9) -hydroxy-stearic acid (5- or 9-PAHSA, respectively) were profiled. In DIO mice fed with differentially composed low- or high-fat diets, acute and subchronic treatment with 5-PAHSA and 9-PAHSA alone, or in combination, did not significantly improve the deranged metabolic status. Neither racemic 5- or 9-PAHSA, nor the enantiomers were able to: (1) increase basal or insulin-stimulated glucose uptake in vitro, (2) stimulate GLP-1 release from GLUTag cells, or (3) induce GSIS in rat, mouse, or human islets or in a human pancreatic β cell line. Therefore, our data do not support the further development of PAHSAs or their derivatives for the control of insulin resistance and hyperglycemia.
Collapse
Affiliation(s)
- Elsa Pflimlin
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Maximilian Bielohuby
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany.
| | - Marcus Korn
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Kristin Breitschopf
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Matthias Löhn
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Paulus Wohlfart
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Anish Konkar
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Michael Podeschwa
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Felix Bärenz
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Anja Pfenninger
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Uwe Schwahn
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Till Opatz
- Institut für Organische Chemie, Johannes Gutenberg Universität Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Marcel Reimann
- Institut für Organische Chemie, Johannes Gutenberg Universität Mainz, Duesbergweg 10-14, 55128 Mainz, Germany
| | - Stefan Petry
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany
| | - Norbert Tennagels
- Sanofi-Aventis Deutschland GmbH, Industriepark Höchst, Bldg. H821, 65926 Frankfurt am Main, Germany.
| |
Collapse
|
34
|
Larrieu T, Layé S. Food for Mood: Relevance of Nutritional Omega-3 Fatty Acids for Depression and Anxiety. Front Physiol 2018; 9:1047. [PMID: 30127751 PMCID: PMC6087749 DOI: 10.3389/fphys.2018.01047] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 07/13/2018] [Indexed: 12/28/2022] Open
Abstract
The central nervous system (CNS) has the highest concentration of lipids in the organism after adipose tissue. Among these lipids, the brain is particularly enriched with polyunsaturated fatty acids (PUFAs) represented by the omega-6 (ω6) and omega-3 (ω3) series. These PUFAs include arachidonic acid (AA) and docosahexaenoic acid (DHA), respectively. PUFAs have received substantial attention as being relevant to many brain diseases, including anxiety and depression. This review addresses an important question in the area of nutritional neuroscience regarding the importance of ω3 PUFAs in the prevention and/or treatment of neuropsychiatric diseases, mainly depression and anxiety. In particular, it focuses on clinical and experimental data linking dietary intake of ω3 PUFAs and depression or anxiety. In particular, we will discuss recent experimental data highlighting how ω3 PUFAs can modulate neurobiological processes involved in the pathophysiology of anxiety and depression. Potential mechanisms involved in the neuroprotective and corrective activity of ω3 PUFAs in the brain are discussed, in particular the sensing activity of free fatty acid receptors and the activity of the PUFAs-derived endocannabinoid system and the hypothalamic-pituitary-adrenal axis.
Collapse
Affiliation(s)
- Thomas Larrieu
- UMR 1286, NutriNeuro: Laboratoire Nutrition et Neurobiologie Intégrée, Institut National de la Recherche Agronomique, Université de Bordeaux, Bordeaux, France
| | - Sophie Layé
- UMR 1286, NutriNeuro: Laboratoire Nutrition et Neurobiologie Intégrée, Institut National de la Recherche Agronomique, Université de Bordeaux, Bordeaux, France
| |
Collapse
|
35
|
Marinho R, Munõz VR, Pauli LSS, Ropelle ECC, Moura LP, Moraes JC, Moura‐Assis A, Cintra DE, da Silva ASR, Ropelle ER, Pauli JR. Endurance training prevents inflammation and apoptosis in hypothalamic neurons of obese mice. J Cell Physiol 2018; 234:880-890. [PMID: 30078194 DOI: 10.1002/jcp.26909] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/12/2018] [Indexed: 12/24/2022]
Affiliation(s)
- Rodolfo Marinho
- Department of Physical Education Institute of Biosciences, São Paulo State University (UNESP) Rio Claro Brazil
| | - Vitor R. Munõz
- School of Applied Sciences, University of Campinas (UNICAMP) Limeira Brazil
| | | | | | - Leandro P. Moura
- Department of Physical Education Institute of Biosciences, São Paulo State University (UNESP) Rio Claro Brazil
- School of Applied Sciences, University of Campinas (UNICAMP) Limeira Brazil
- OCRC—Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas Brazil
- CEPECE—Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP) Limeira Brazil
| | - Juliana C. Moraes
- Faculty of Medical Sciences, State University of Campinas (UNICAMP) Limeira Brazil
| | | | - Dennys E. Cintra
- School of Applied Sciences, University of Campinas (UNICAMP) Limeira Brazil
- OCRC—Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas Brazil
| | - Adelino S. R. da Silva
- School of Physical Education and Sport of Ribeirão Preto, University of São Paulo (USP), Ribeirão Preto São Paulo Brazil
| | - Eduardo R. Ropelle
- School of Applied Sciences, University of Campinas (UNICAMP) Limeira Brazil
- OCRC—Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas Brazil
- CEPECE—Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP) Limeira Brazil
| | - José R. Pauli
- School of Applied Sciences, University of Campinas (UNICAMP) Limeira Brazil
- OCRC—Obesity and Comorbidities Research Center, University of Campinas (UNICAMP), Campinas Brazil
- CEPECE—Center of Research in Sport Sciences. School of Applied Sciences, University of Campinas (UNICAMP) Limeira Brazil
| |
Collapse
|
36
|
Chronic Swimming Exercise Ameliorates Low-Soybean-Oil Diet-Induced Spatial Memory Impairment by Enhancing BDNF-Mediated Synaptic Potentiation in Developing Spontaneously Hypertensive Rats. Neurochem Res 2018; 43:1047-1057. [PMID: 29574667 DOI: 10.1007/s11064-018-2515-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 02/18/2018] [Accepted: 03/20/2018] [Indexed: 12/12/2022]
Abstract
Exercise and low-fat diets are common lifestyle modifications used for the treatment of hypertension besides drug therapy. However, unrestrained low-fat diets may result in deficiencies of low-unsaturated fatty acids and carry contingent risks of delaying neurodevelopment. While aerobic exercise shows positive neuroprotective effects, it is still unclear whether exercise could alleviate the impairment of neurodevelopment that may be induced by certain low-fat diets. In this research, developing spontaneously hypertensive rats (SHR) were treated with chronic swimming exercise and/or a low-soybean-oil diet for 6 weeks. We found that performance in the Morris water maze was reduced and long-term potentiation in the hippocampus was suppressed by the diet, while a combination treatment of exercise and diet alleviated the impairment induced by the specific low-fat diet. Moreover, the combination treatment effectively increased the expression of brain-derived neurotrophic factor (BDNF) and N-methyl-D-aspartic acid receptor (NMDAR), which were both down-regulated by the low-soybean-oil diet in the hippocampus of developing SHR. These findings suggest that chronic swimming exercise can ameliorate the low-soybean-oil diet-induced learning and memory impairment in developing SHR through the up-regulation of BDNF and NMDAR expression.
Collapse
|
37
|
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
|
38
|
Lazutkaite G, Soldà A, Lossow K, Meyerhof W, Dale N. Amino acid sensing in hypothalamic tanycytes via umami taste receptors. Mol Metab 2017; 6:1480-1492. [PMID: 29107294 PMCID: PMC5681271 DOI: 10.1016/j.molmet.2017.08.015] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 08/25/2017] [Accepted: 08/30/2017] [Indexed: 12/15/2022] Open
Abstract
Objective Hypothalamic tanycytes are glial cells that line the wall of the third ventricle and contact the cerebrospinal fluid (CSF). While they are known to detect glucose in the CSF we now show that tanycytes also detect amino acids, important nutrients that signal satiety. Methods Ca2+ imaging and ATP biosensing were used to detect tanycyte responses to l-amino acids. The downstream pathway of the responses was determined using ATP receptor antagonists and channel blockers. The receptors were characterized using mice lacking the Tas1r1 gene, as well as an mGluR4 receptor antagonist. Results Amino acids such as Arg, Lys, and Ala evoke Ca2+ signals in tanycytes and evoke the release of ATP via pannexin 1 and CalHM1, which amplifies the signal via a P2 receptor dependent mechanism. Tanycytes from mice lacking the Tas1r1 gene had diminished responses to lysine and arginine but not alanine. Antagonists of mGluR4 greatly reduced the responses to alanine and lysine. Conclusion Two receptors previously implicated in taste cells, the Tas1r1/Tas1r3 heterodimer and mGluR4, contribute to the detection of a range of amino acids by tanycytes in CSF. Hypothalamic tanycytes can detect amino acids in cerebrospinal fluid. The mechanism is taste receptor-dependent. Tas1r1/Tas1r3 mediates responses to l-arginine and l-lysine. mGluR4 mediates responses to l-alanine and partially those of l-lysine. ATP release from tanycytes evoked by amino acids reaches into the arcuate nucleus.
Collapse
Affiliation(s)
- Greta Lazutkaite
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Alice Soldà
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK
| | - Kristina Lossow
- Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Wolfgang Meyerhof
- Department of Molecular Genetics, German Institute of Human Nutrition Potsdam-Rehbruecke, Arthur-Scheunert-Allee 114-116, 14558, Nuthetal, Germany
| | - Nicholas Dale
- School of Life Sciences, University of Warwick, Coventry, CV4 7AL, UK.
| |
Collapse
|
39
|
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]
|
40
|
Koo BK. Letter: Regulating Hypothalamus Gene Expression in Food Intake: Dietary Composition or Calorie Density? (Diabetes Metab J 2017;41:121-7). Diabetes Metab J 2017; 41:223-224. [PMID: 28657236 PMCID: PMC5489503 DOI: 10.4093/dmj.2017.41.3.223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Affiliation(s)
- Bo Kyung Koo
- Department of Internal Medicine, Seoul Metropolitan Government Seoul National University Boramae Medical Center, Seoul National University College of Medicine, Seoul, Korea.
| |
Collapse
|
41
|
Jang M, Park SY, Kim YW, Jung SP, Kim JY. Response: Regulating Hypothalamus Gene Expression in Food Intake: Dietary Composition or Calorie Density? (Diabetes Metab J 2017;41:121-7). Diabetes Metab J 2017; 41:225-227. [PMID: 28657237 PMCID: PMC5489504 DOI: 10.4093/dmj.2017.41.3.225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Affiliation(s)
- Mi Jang
- Obesity-Diabetes Advanced Research Center, Yeungnam University College of Medicine, Daegu, Korea
| | - So Young Park
- Obesity-Diabetes Advanced Research Center, Yeungnam University College of Medicine, Daegu, Korea
| | - Yong Woon Kim
- Obesity-Diabetes Advanced Research Center, Yeungnam University College of Medicine, Daegu, Korea
| | - Seung Pil Jung
- Obesity-Diabetes Advanced Research Center, Yeungnam University College of Medicine, Daegu, Korea
| | - Jong Yeon Kim
- Obesity-Diabetes Advanced Research Center, Yeungnam University College of Medicine, Daegu, Korea.
| |
Collapse
|
42
|
Ramos-Lobo AM, Donato J. The role of leptin in health and disease. Temperature (Austin) 2017; 4:258-291. [PMID: 28944270 DOI: 10.1080/23328940.2017.1327003] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 04/27/2017] [Accepted: 05/02/2017] [Indexed: 02/07/2023] Open
Abstract
Leptin is a master regulator of energy balance and body adiposity. Additionally, leptin exerts important control on glucose homeostasis, thermogenesis, autonomic nervous system and neuroendocrine axes. In metabolic diseases, such as obesity and diabetes mellitus, leptin signaling may be compromised, indicating the important role of this hormone in the etiology and pathophysiological manifestations of these conditions. In the present manuscript, we reviewed important concepts of leptin signaling, as well as about the effects of leptin on several biologic functions. We also discussed the possible therapeutic use of leptin administration and how our current obesogenic environment contributes to the development of leptin resistance. Our objective was to provide a comprehensive and state-of-the-art review about the importance of leptin to maintain the homeostasis and during pathological conditions.
Collapse
Affiliation(s)
- Angela M Ramos-Lobo
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - Jose Donato
- Department of Physiology and Biophysics, Institute of Biomedical Sciences, University of São Paulo, São Paulo, SP, Brazil
| |
Collapse
|
43
|
Dragano NRV, Solon C, Ramalho AF, de Moura RF, Razolli DS, Christiansen E, Azevedo C, Ulven T, Velloso LA. Polyunsaturated fatty acid receptors, GPR40 and GPR120, are expressed in the hypothalamus and control energy homeostasis and inflammation. J Neuroinflammation 2017; 14:91. [PMID: 28446241 PMCID: PMC5405534 DOI: 10.1186/s12974-017-0869-7] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 04/20/2017] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND The consumption of large amounts of dietary fats is one of the most important environmental factors contributing to the development of obesity and metabolic disorders. GPR120 and GPR40 are polyunsaturated fatty acid receptors that exert a number of systemic effects that are beneficial for metabolic and inflammatory diseases. Here, we evaluate the expression and potential role of hypothalamic GPR120 and GPR40 as targets for the treatment of obesity. METHODS Male Swiss (6-weeks old), were fed with a high fat diet (HFD, 60% of kcal from fat) for 4 weeks. Next, mice underwent stereotaxic surgery to place an indwelling cannula into the right lateral ventricle. intracerebroventricular (icv)-cannulated mice were treated twice a day for 6 days with 2.0 μL saline or GPR40 and GPR120 agonists: GW9508, TUG1197, or TUG905 (2.0 μL, 1.0 mM). Food intake and body mass were measured during the treatment period. At the end of the experiment, the hypothalamus was collected for real-time PCR analysis. RESULTS We show that both receptors are expressed in the hypothalamus; GPR120 is primarily present in microglia, whereas GPR40 is expressed in neurons. Upon intracerebroventricular treatment, GW9508, a non-specific agonist for both receptors, reduced energy efficiency and the expression of inflammatory genes in the hypothalamus. Reducing GPR120 hypothalamic expression using a lentivirus-based approach resulted in the loss of the anti-inflammatory effect of GW9508 and increased energy efficiency. Intracerebroventricular treatment with the GPR120- and GPR40-specific agonists TUG1197 and TUG905, respectively, resulted in milder effects than those produced by GW9508. CONCLUSIONS GPR120 and GPR40 act in concert in the hypothalamus to reduce energy efficiency and regulate the inflammation associated with obesity. The combined activation of both receptors in the hypothalamus results in better metabolic outcomes than the isolated activation of either receptor alone.
Collapse
Affiliation(s)
- Nathalia R. V. Dragano
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP 13084-970 Brazil
| | - Carina Solon
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP 13084-970 Brazil
| | - Albina F. Ramalho
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP 13084-970 Brazil
| | - Rodrigo F. de Moura
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP 13084-970 Brazil
| | - Daniela S. Razolli
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP 13084-970 Brazil
| | - Elisabeth Christiansen
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Carlos Azevedo
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Trond Ulven
- Department of Physics, Chemistry and Pharmacy, University of Southern Denmark, DK-5230 Odense, Denmark
| | - Licio A. Velloso
- Laboratory of Cell Signaling and Obesity and Comorbidities Research Center, University of Campinas, Campinas, SP 13084-970 Brazil
- Laboratory of Cell Signaling, University of Campinas, Rua Cinco de Junho, 350, Cidade Universitária, Campinas, SP 13083-877 Brazil
| |
Collapse
|
44
|
Recabal A, Caprile T, García-Robles MDLA. Hypothalamic Neurogenesis as an Adaptive Metabolic Mechanism. Front Neurosci 2017; 11:190. [PMID: 28424582 PMCID: PMC5380718 DOI: 10.3389/fnins.2017.00190] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Accepted: 03/21/2017] [Indexed: 12/12/2022] Open
Abstract
In the adult brain, well-characterized neurogenic niches are located in the subventricular zone (SVZ) of the lateral ventricles and in the subgranular zone (SGZ) of the hippocampus. In both regions, neural precursor cells (NPCs) share markers of embryonic radial glia and astroglial cells, and in vitro clonal expansion of these cells leads to neurosphere formation. It has also been more recently demonstrated that neurogenesis occurs in the adult hypothalamus, a brain structure that integrates peripheral signals to control energy balance and dietary intake. The NPCs of this region, termed tanycytes, are ependymal-glial cells, which comprise the walls of the infundibular recess of the third ventricle and contact the median eminence. Thus, tanycytes are in a privileged position to detect hormonal, nutritional and mitogenic signals. Recent studies reveal that in response to nutritional signals, tanycytes are capable of differentiating into orexigenic or anorexigenic neurons, suggesting that these cells are crucial for control of feeding behavior. In this review, we discuss evidence, which suggests that hypothalamic neurogenesis may act as an additional adaptive mechanism in order to respond to changes in diet.
Collapse
Affiliation(s)
- Antonia Recabal
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de ConcepciónConcepción, Chile.,Laboratorio de Guía Axonal, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de ConcepciónConcepción, Chile
| | - Teresa Caprile
- Laboratorio de Guía Axonal, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de ConcepciónConcepción, Chile
| | - María de Los Angeles García-Robles
- Laboratorio de Biología Celular, Departamento de Biología Celular, Facultad de Ciencias Biológicas, Universidad de ConcepciónConcepción, Chile
| |
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Dietary intake is a critical regulator of organismal physiology and health. Tissue homeostasis and regeneration are dependent on adult tissue stem cells that self-renew and differentiate into the specialized cell types. As stem cells respond to cues from their environment, dietary signals and nutrients influence tissue biology by altering the function and activity of adult stem cells. In this review, we highlight recent studies that illustrate how diverse diets such as caloric restriction, fasting, high fat diets, and ketogenic diets impact stem cell function and their microenvironments. RECENT FINDINGS Caloric restriction generally exerts positive effects on adult stem cells, notably increasing stem cell functionality in the intestine and skeletal muscle as well as increasing hematopoietic stem cell quiescence. Similarly, fasting confers protection of intestinal, hematopoietic, and neuronal stem cells against injury. High fat diets induce intestinal stem cell niche independence and stem-like properties in intestinal progenitors, while high fat diets impair hematopoiesis and neurogenesis. SUMMARY Caloric restriction and fasting are generally beneficial to adult stem cell function, while high fat diets impair stem cell function or create opportunities for tumorigenesis. However, the effects of each diet on stem cell biology are complex and vary greatly between tissues. Given the recent interest in developing dietary interventions or mimetics as therapeutics, further studies, including on ketogenic diets, will be essential to understand how adult stem cells respond to diet-induced signals and physiology.
Collapse
Affiliation(s)
- Miyeko D. Mana
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, Department of Biology, MIT, Cambridge, MA 02139 USA
| | - Elaine Yih-Shuen Kuo
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, Department of Biology, MIT, Cambridge, MA 02139 USA
| | - Ömer H. Yilmaz
- The David H. Koch Institute for Integrative Cancer Research at MIT, Cambridge, MA 02139, Department of Biology, MIT, Cambridge, MA 02139 USA
- Broad Institute of Harvard and MIT, Cambridge, MA 02142 USA
- Departments of Pathology, Gastroenterology, and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114 USA
| |
Collapse
|
46
|
Pascoal LB, Bombassaro B, Ramalho AF, Coope A, Moura RF, Correa-da-Silva F, Ignacio-Souza L, Razolli D, de Oliveira D, Catharino R, Velloso LA. Resolvin RvD2 reduces hypothalamic inflammation and rescues mice from diet-induced obesity. J Neuroinflammation 2017; 14:5. [PMID: 28086928 PMCID: PMC5234140 DOI: 10.1186/s12974-016-0777-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 12/09/2016] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Diet-induced hypothalamic inflammation is an important mechanism leading to dysfunction of neurons involved in controlling body mass. Studies have shown that polyunsaturated fats can reduce hypothalamic inflammation. Here, we evaluated the presence and function of RvD2, a resolvin produced from docosahexaenoic acid, in the hypothalamus of mice. METHODS Male Swiss mice were fed either chow or a high-fat diet. RvD2 receptor and synthetic enzymes were evaluated by real-time PCR and immunofluorescence. RvD2 was determined by mass spectrometry. Dietary and pharmacological approaches were used to modulate the RvD2 system in the hypothalamus, and metabolic phenotype consequences were determined. RESULTS All enzymes involved in the synthesis of RvD2 were detected in the hypothalamus and were modulated in response to the consumption of dietary saturated fats, leading to a reduction of hypothalamic RvD2. GPR18, the receptor for RvD2, which was detected in POMC and NPY neurons, was also modulated by dietary fats. The substitution of saturated by polyunsaturated fats in the diet resulted in increased hypothalamic RvD2, which was accompanied by reduced body mass and improved glucose tolerance. The intracerebroventricular treatment with docosahexaenoic acid resulted in increased expression of the RvD2 synthetic enzymes, increased expression of anti-inflammatory cytokines and improved metabolic phenotype. Finally, intracerebroventricular treatment with RvD2 resulted in reduced adiposity, improved glucose tolerance and increased hypothalamic expression of anti-inflammatory cytokines. CONCLUSIONS Thus, RvD2 is produced in the hypothalamus, and its receptor and synthetic enzymes are modulated by dietary fats. The improved metabolic outcomes of RvD2 make this substance an attractive approach to treat obesity.
Collapse
Affiliation(s)
- Livia B Pascoal
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Bruna Bombassaro
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Albina F Ramalho
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Andressa Coope
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Rodrigo F Moura
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Felipe Correa-da-Silva
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Leticia Ignacio-Souza
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Daniela Razolli
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil
| | - Diogo de Oliveira
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Rodrigo Catharino
- Faculty of Pharmaceutical Sciences, University of Campinas, Campinas, Brazil
| | - Licio A Velloso
- Obesity and Comorbidities Research Center, Laboratory of Cell Signaling, University of Campinas, Campinas, SP, 13084-761, Brazil.
| |
Collapse
|
47
|
Adult Neurogenesis in the Female Mouse Hypothalamus: Estradiol and High-Fat Diet Alter the Generation of Newborn Neurons Expressing Estrogen Receptor α. eNeuro 2016; 3:eN-NWR-0027-16. [PMID: 27679811 PMCID: PMC5032890 DOI: 10.1523/eneuro.0027-16.2016] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 08/22/2016] [Accepted: 08/23/2016] [Indexed: 11/21/2022] Open
Abstract
Estrogens and leptins act in the hypothalamus to maintain reproduction and energy homeostasis. Neurogenesis in the adult mammalian hypothalamus has been implicated in the regulation of energy homeostasis. Recently, high-fat diet (HFD) and estradiol (E2) have been shown to alter cell proliferation and the number of newborn leptin-responsive neurons in the hypothalamus of adult female mice. The current study tested the hypothesis that new cells expressing estrogen receptor α (ERα) are generated in the arcuate nucleus (ARC) and the ventromedial nucleus of the hypothalamus (VMH) of the adult female mouse, hypothalamic regions that are critical in energy homeostasis. Adult mice were ovariectomized and implanted with capsules containing E2 or oil. Within each hormone group, mice were fed an HFD or standard chow for 6 weeks and treated with BrdU to label new cells. Newborn cells that respond to estrogens were identified in the ARC and VMH, of which a subpopulation was leptin sensitive, indicating that the subpopulation consists of neurons. Moreover, there was an interaction between diet and hormone with an effect on the number of these newborn ERα-expressing neurons that respond to leptin. Regardless of hormone treatment, HFD increased the number of ERα-expressing cells in the ARC and VMH. E2 decreased hypothalamic fibroblast growth factor 10 (Fgf10) gene expression in HFD mice, suggesting a role for Fgf10 in E2 effects on neurogenesis. These findings of newly created estrogen-responsive neurons in the adult brain provide a novel mechanism by which estrogens can act in the hypothalamus to regulate energy homeostasis in females.
Collapse
|
48
|
Araujo EP, Moraes JC, Cintra DE, Velloso LA. MECHANISMS IN ENDOCRINOLOGY: Hypothalamic inflammation and nutrition. Eur J Endocrinol 2016; 175:R97-R105. [PMID: 27006108 DOI: 10.1530/eje-15-1207] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 03/22/2016] [Indexed: 12/29/2022]
Abstract
Selected subpopulations of hypothalamic neurons play important roles in the regulation of whole body energy homeostasis. Studies have shown that the saturated fats present in large amounts in western diets can activate an inflammatory response in the hypothalamus, affecting the capacity of such neurons to respond appropriately to satiety and adipostatic signals. In the first part of this review, we will explore the mechanisms behind saturated fatty acid-induced hypothalamic dysfunction. Next, we will present and discuss recent studies that have identified the mechanisms that mediate some of the anti-inflammatory actions of unsaturated fatty acids in the hypothalamus and the potential for exploring these mechanisms to prevent or treat obesity.
Collapse
Affiliation(s)
- Eliana P Araujo
- Laboratory of Cell SignalingUniversity of Campinas, Campinas, São Paulo, Brazil Faculty of NursingUniversity of Campinas, Campinas, São Paulo, Brazil
| | - Juliana C Moraes
- Laboratory of Cell SignalingUniversity of Campinas, Campinas, São Paulo, Brazil
| | - Dennys E Cintra
- Laboratory of Cell SignalingUniversity of Campinas, Campinas, São Paulo, Brazil Faculty of Applied SciencesUniversity of Campinas, Campinas, São Paulo, Brazil
| | - Licio A Velloso
- Laboratory of Cell SignalingUniversity of Campinas, Campinas, São Paulo, Brazil
| |
Collapse
|
49
|
Fan C, Fu H, Dong H, Lu Y, Lu Y, Qi K. Maternal n-3 polyunsaturated fatty acid deprivation during pregnancy and lactation affects neurogenesis and apoptosis in adult offspring: associated with DNA methylation of brain-derived neurotrophic factor transcripts. Nutr Res 2016; 36:1013-1021. [PMID: 27632922 DOI: 10.1016/j.nutres.2016.06.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 05/24/2016] [Accepted: 06/01/2016] [Indexed: 01/06/2023]
Abstract
In this study, we hypothesized that n-3 polyunsaturated fatty acid (PUFA) deficiency during pregnancy and lactation will make a lasting impact on brain neurogenesis and apoptosis of the adult offspring and that these harmful effects cannot be reversed by n-3 PUFA supplementation after weaning. Moreover, the underlying mechanisms may be attributable to the epigenetic changes of brain-derived neurotrophic factor (BDNF). C57BL/6J female mice were fed with n-3 PUFA-deficient diet (n-3 def) or n-3 PUFA-adequate diet (n-3 adq) throughout pregnancy and lactation. At postnatal 21 days, equal numbers of male pups from both groups were fed the opposite diet, and the remaining male pups were fed with the same diets as their mothers until 3 months of age. Feeding the n-3 adq diet to pups from the maternal n-3 def group significantly increased the n-3 PUFA concentration but did not change expressions of calretinin, Bcl2, and Bax in the hippocampus. Feeding the n-3 def diet to pups from the maternal n-3 adq group significantly reduced the n-3 PUFA concentration but did not reduce expressions of calretinin and Bcl2. Similarly, BDNF levels, especially mRNA expressions of BDNF transcripts IV and IX, were also reduced by maternal n-3 def and not reversed by n-3 PUFA supplementation after weaning. The decrease in BDNF expression by maternal n-3 def diet was associated with greater DNA methylation at special CpG sites. These results suggested that the maternal n-3 PUFA deficiency during pregnancy and lactation imprints long-term changes of brain development in adult offspring.
Collapse
Affiliation(s)
- Chaonan Fan
- Nutrition Research Unit, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Huicong Fu
- Nutrition Research Unit, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Hua Dong
- Nutrition Research Unit, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yuanyuan Lu
- Nutrition Research Unit, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Yanfei Lu
- Nutrition Research Unit, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Kemin Qi
- Nutrition Research Unit, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|