1
|
Vázquez-Pérez LA, Hattori-Hara M, Arankowsky-Sandoval G, Pérez-Mendoza G, Rubi-Castellanos R, Rangel-Méndez JA, Pinto-Escalante D, Canto-Cetina T, González-Herrera L. Association between personality traits, eating behaviors, and the genetic polymorphisms FTO-rs9939609 and MAO-A 30 bp u-VNTR with obesity in Mexican Mayan children. Front Genet 2024; 15:1421870. [PMID: 39130748 PMCID: PMC11310059 DOI: 10.3389/fgene.2024.1421870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 07/09/2024] [Indexed: 08/13/2024] Open
Abstract
Introduction Genetic variants that control dopamine have been associated with obesity in children through loss of control of satiety and impulses, the manifestation of addictive eating behaviors, and specific personality traits. The variants include FTO-rs9939609 and the MAO-A 30 pb u-VNTR low-transcription alleles (LTA). Objective To evaluate the genetic association of FTO-rs9939609 and the MAO-A LTA, along with personality traits and eating behavior with obesity in Mayan children from Mexico. Methods We cross-sectionally evaluated 186 children (70 with obesity and 116 with normal weight) 6-12 years old from Yucatan, Mexico. Nutritional status was defined with body mass index (BMI) percentiles. Personality traits were evaluated with the Conners and TMCQ tests; eating behavior was evaluated with the CEBQ test. Genotyping with real-time PCR and TaqMan probes was used for FTO-rs9939609, whereas PCR amplification was used for MAO-A u-VNTR. Results High-intensity pleasure (p = 0.013) and moderate appetite (p = 0.032) differed according to nutritional status. Heterozygous FTO-rs9939609 T/A children showed higher mean scores of low-intensity pleasure (p = 0.002) and moderate appetite (p = 0.027) than homozygous T/T. Hemizygous boys having MAO-A LTA showed significantly higher mean scores of anxiety (p = 0.001) and impulsivity (p = 0.008). In multivariate models, only LTA alleles of MAO-A explained obesity in boys (OR = 4.44; 95% CI = 1.18-16.63). Conclusion In the present study, MAO-A u-VNTR alleles were associated with obesity in multivariate models only in boys. These alleles might also have a role in personality traits such as anxiety and impulsivity, which secondly contribute to developing obesity in Mayan boys.
Collapse
Affiliation(s)
- Luis Alberto Vázquez-Pérez
- Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autonoma de Yucatan, Mérida, Mexico
| | - Mónica Hattori-Hara
- Secretaría de Educación del Gobierno del Esatdo de Yucatán (SEGEY), Mérida, Mexico
| | - Gloria Arankowsky-Sandoval
- Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autonoma de Yucatan, Mérida, Mexico
| | - Gerardo Pérez-Mendoza
- Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autonoma de Yucatan, Mérida, Mexico
| | - Rodrigo Rubi-Castellanos
- Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autonoma de Yucatan, Mérida, Mexico
| | - Jorge Aarón Rangel-Méndez
- Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autonoma de Yucatan, Mérida, Mexico
| | - Doris Pinto-Escalante
- Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autonoma de Yucatan, Mérida, Mexico
| | - Thelma Canto-Cetina
- Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autonoma de Yucatan, Mérida, Mexico
| | - Lizbeth González-Herrera
- Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autonoma de Yucatan, Mérida, Mexico
| |
Collapse
|
2
|
Anthony DC, Probert F, Gorlova A, Hebert J, Radford-Smith D, Nefedova Z, Umriukhin A, Nedorubov A, Cespuglio R, Shulgin B, Lyundup A, Lesch KP, Strekalova T. Impact of Serotonin Transporter Absence on Brain Insulin Receptor Expression, Plasma Metabolome Changes, and ADHD-like Behavior in Mice fed a Western Diet. Biomolecules 2024; 14:884. [PMID: 39199273 PMCID: PMC11351952 DOI: 10.3390/biom14080884] [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: 06/04/2024] [Revised: 07/02/2024] [Accepted: 07/15/2024] [Indexed: 09/01/2024] Open
Abstract
The impaired function of the serotonin transporter (SERT) in humans has been linked to a higher risk of obesity and type 2 diabetes, especially as people age. Consuming a "Western diet" (WD), which is high in saturated fats, cholesterol, and sugars, can induce metabolic syndrome. Previous research indicated that mice carrying a targeted inactivation of the Sert gene (knockout, KO) and fed a WD display significant metabolic disturbances and behaviors reminiscent of ADHD. These abnormalities might be mediated via a dysfunction in insulin receptor (IR) signaling, which is also associated with adult ADHD. However, the impact of Sert deficiency on IR signaling and systemic metabolic changes has not been thoroughly explored. In this study, we conducted a detailed analysis of locomotor behavior in wild-type (WT) and KO mice fed a WD or control diet. We investigated changes in the blood metabolome and examined, via PCR, the expression of insulin receptor A and B isoforms and key regulators of their function in the brain. Twelve-month-old KO mice and their WT littermates were fed a WD for three weeks. Nuclear magnetic resonance spectroscopy analysis of plasma samples showed that KO mice on a WD had higher levels of lipids and lipoproteins and lower levels of glucose, lactate, alanine, valine, and isoleucine compared to other groups. SERT-KO mice on the control diet exhibited increased brain levels of both IR A and B isoforms, accompanied by a modest increase in the negative regulator ENPP. The KO mice also displayed anxiety-like behavior and reduced exploratory activity in an open field test. However, when the KO animals were fed a WD, the aberrant expression levels of IR isoforms in the KO mice and locomotor behavior were ameliorated indicating a complex interaction between genetic and dietary factors that might contribute to ADHD-like symptoms. Overall, our findings suggest that the lack of Sert leads to a unique metabolic phenotype in aged mice, characterized by dysregulated IR-related pathways. These changes are exacerbated by WD in the blood metabolome and are associated with behavioral abnormalities.
Collapse
Affiliation(s)
- Daniel C. Anthony
- Department of Pharmacology, Oxford University, Oxford OX1 3QT, UK; (D.C.A.); (F.P.); (J.H.); (D.R.-S.)
| | - Fay Probert
- Department of Pharmacology, Oxford University, Oxford OX1 3QT, UK; (D.C.A.); (F.P.); (J.H.); (D.R.-S.)
- Department of Chemistry, Oxford University, Oxford OX1 2JD, UK
| | - Anna Gorlova
- Research and Education Resource Center, Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (A.G.); (R.C.); (A.L.)
| | - Jenna Hebert
- Department of Pharmacology, Oxford University, Oxford OX1 3QT, UK; (D.C.A.); (F.P.); (J.H.); (D.R.-S.)
| | - Daniel Radford-Smith
- Department of Pharmacology, Oxford University, Oxford OX1 3QT, UK; (D.C.A.); (F.P.); (J.H.); (D.R.-S.)
| | - Zlata Nefedova
- Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (Z.N.); (A.U.); (A.N.)
| | - Aleksei Umriukhin
- Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (Z.N.); (A.U.); (A.N.)
| | - Andrey Nedorubov
- Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (Z.N.); (A.U.); (A.N.)
| | - Raymond Cespuglio
- Research and Education Resource Center, Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (A.G.); (R.C.); (A.L.)
| | - Boris Shulgin
- Laboratory of Engineering Profile Physical and Chemical Methods of Analysis, Korkyt Ata Kyzylorda University, Kyzylorda 120014, Kazakhstan;
| | - Aleksey Lyundup
- Research and Education Resource Center, Peoples Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (A.G.); (R.C.); (A.L.)
- Endocrinology Research Centre, Dmitry Ulyanov Str. 19, 117036 Moscow, Russia
| | - Klaus Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University Hospital Würzburg, 97080 Würzburg, Germany;
- Department of Child and Adolescent Psychiatry, Psychosomatics and Psychotherapy, Center of Mental Health, University Hospital Würzburg, 97080 Würzburg, Germany
| | - Tatyana Strekalova
- Department of Pharmacology, Oxford University, Oxford OX1 3QT, UK; (D.C.A.); (F.P.); (J.H.); (D.R.-S.)
| |
Collapse
|
3
|
Hoch J, Burkhard N, Zhang S, Rieder M, Marchini T, Geest V, Krauel K, Zahn T, Schommer N, Hamad MA, Bauer C, Gauchel N, Stallmann D, Normann C, Wolf D, Scharf RE, Duerschmied D, Schanze N. Serotonin transporter-deficient mice display enhanced adipose tissue inflammation after chronic high-fat diet feeding. Front Immunol 2023; 14:1184010. [PMID: 37520561 PMCID: PMC10372416 DOI: 10.3389/fimmu.2023.1184010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 06/13/2023] [Indexed: 08/01/2023] Open
Abstract
Introduction Serotonin is involved in leukocyte recruitment during inflammation. Deficiency of the serotonin transporter (SERT) is associated with metabolic changes in humans and mice. A possible link and interaction between the inflammatory effects of serotonin and metabolic derangements in SERT-deficient mice has not been investigated so far. Methods SERT-deficient (Sert -/-) and wild type (WT) mice were fed a high-fat diet, starting at 8 weeks of age. Metabolic phenotyping (metabolic caging, glucose and insulin tolerance testing, body and organ weight measurements, qPCR, histology) and assessment of adipose tissue inflammation (flow cytometry, histology, qPCR) were carried out at the end of the 19-week high-fat diet feeding period. In parallel, Sert -/- and WT mice received a control diet and were analyzed either at the time point equivalent to high-fat diet feeding or as early as 8-11 weeks of age for baseline characterization. Results After 19 weeks of high-fat diet, Sert -/- and WT mice displayed similar whole-body and fat pad weights despite increased relative weight gain due to lower starting body weight in Sert -/-. In obese Sert -/- animals insulin resistance and liver steatosis were enhanced as compared to WT animals. Leukocyte accumulation and mRNA expression of cytokine signaling mediators were increased in epididymal adipose tissue of obese Sert -/- mice. These effects were associated with higher adipose tissue mRNA expression of the chemokine monocyte chemoattractant protein 1 and presence of monocytosis in blood with an increased proportion of pro-inflammatory Ly6C+ monocytes. By contrast, Sert -/- mice fed a control diet did not display adipose tissue inflammation. Discussion Our observations suggest that SERT deficiency in mice is associated with inflammatory processes that manifest as increased adipose tissue inflammation upon chronic high-fat diet feeding due to enhanced leukocyte recruitment.
Collapse
Affiliation(s)
- Johannes Hoch
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Niklas Burkhard
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Shanshan Zhang
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marina Rieder
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Translational Cardiology, Department of Cardiology, Inselspital, Bern, Switzerland
| | - Timoteo Marchini
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Vincent Geest
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Krystin Krauel
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Timm Zahn
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nicolas Schommer
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Muataz Ali Hamad
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Carolina Bauer
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nadine Gauchel
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Daniela Stallmann
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claus Normann
- Department of Psychiatry and Psychotherapy, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Center for Basics in Neuromodulation, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Dennis Wolf
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Rüdiger Eberhard Scharf
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
- Division of Experimental and Clinical Hemostasis, Hemotherapy, and Transfusion Medicine, Blood and Hemophilia Comprehensive Care Center, Institute of Transplantation Diagnostics and Cell Therapy, Heinrich Heine University Medical Center, Düsseldorf, Germany
| | - Daniel Duerschmied
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- European Center for AngioScience (ECAS) and German Center for Cardiovascular Research (DZHK) Partner Site Heidelberg/Mannheim, Mannheim, Germany
| | - Nancy Schanze
- Cardiology and Angiology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Cardiology, Angiology, Haemostaseology and Medical Intensive Care, University Medical Centre Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
4
|
Bloemendaal M, Veniaminova E, Anthony DC, Gorlova A, Vlaming P, Khairetdinova A, Cespuglio R, Lesch KP, Arias Vasquez A, Strekalova T. Serotonin Transporter (SERT) Expression Modulates the Composition of the Western-Diet-Induced Microbiota in Aged Female Mice. Nutrients 2023; 15:3048. [PMID: 37447374 PMCID: PMC10346692 DOI: 10.3390/nu15133048] [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: 06/09/2023] [Revised: 06/27/2023] [Accepted: 06/29/2023] [Indexed: 07/15/2023] Open
Abstract
Background. The serotonin transporter (SERT), highly expressed in the gut and brain, is implicated in metabolic processes. A genetic variant of the upstream regulatory region of the SLC6A4 gene encoding SERT, the so-called short (s) allele, in comparison with the long (l) allele, results in the decreased function of this transporter, altered serotonergic regulation, an increased risk of psychiatric pathology and type-2 diabetes and obesity, especially in older women. Aged female mice with the complete (Sert-/-: KO) or partial (Sert+/-: HET) loss of SERT exhibit more pronounced negative effects following their exposure to a Western diet in comparison to wild-type (Sert+/+: WT) animals. Aims. We hypothesized that these effects might be mediated by an altered gut microbiota, which has been shown to influence serotonin metabolism. We performed V4 16S rRNA sequencing of the gut microbiota in 12-month-old WT, KO and HET female mice that were housed on a control or Western diet for three weeks. Results. The relative abundance of 11 genera was increased, and the abundance of 6 genera was decreased in the Western-diet-housed mice compared to the controls. There were correlations between the abundance of Streptococcus and Ruminococcaceae_UCG-014 and the expression of the pro-inflammatory marker Toll-like-Receptor 4 (Tlr4) in the dorsal raphe, as well as the expression of the mitochondrial activity marker perixome-proliferator-activated-receptor-cofactor-1b (Ppargc1b) in the prefrontal cortex. Although there was no significant impact of genotype on the microbiota in animals fed with the Control diet, there were significant interactions between diet and genotype. Following FDR correction, the Western diet increased the relative abundance of Intestinimonas and Atopostipes in the KO animals, which was not observed in the other groups. Erysipelatoclostridium abundance was increased by the Western diet in the WT group but not in HET or KO animals. Conclusions. The enhanced effects of a challenge with a Western diet in SERT-deficient mice include the altered representation of several gut genera, such as Intestinimonas, Atopostipes and Erysipelatoclostridium, which are also implicated in serotonergic and lipid metabolism. The manipulation of these genera may prove useful in individuals with the short SERT allele.
Collapse
Affiliation(s)
- Mirjam Bloemendaal
- Departments of Psychiatry & Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (P.V.); (A.A.V.)
| | - Ekaterina Veniaminova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.V.); (A.G.); (A.K.); (R.C.)
| | | | - Anna Gorlova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.V.); (A.G.); (A.K.); (R.C.)
| | - Priscilla Vlaming
- Departments of Psychiatry & Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (P.V.); (A.A.V.)
| | - Adel Khairetdinova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.V.); (A.G.); (A.K.); (R.C.)
| | - Raymond Cespuglio
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, 119991 Moscow, Russia; (E.V.); (A.G.); (A.K.); (R.C.)
- Neuroscience Research Center of Lyon, Claude-Bernard Lyon-1 University, 69500 Bron, France
| | - Klaus Peter Lesch
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, 97080 Würzburg, Germany; (K.P.L.); (T.S.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229 HX Maastricht, The Netherlands
| | - Alejandro Arias Vasquez
- Departments of Psychiatry & Human Genetics, Radboud University Medical Center, 6525 GA Nijmegen, The Netherlands; (P.V.); (A.A.V.)
| | - Tatyana Strekalova
- Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, 97080 Würzburg, Germany; (K.P.L.); (T.S.)
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, 6229 HX Maastricht, The Netherlands
| |
Collapse
|
5
|
Manco L, Machado-Rodrigues AM, Padez C. Association study of common functional genetic polymorphisms in SLC6A4 (5-HTT) and MAOA genes with obesity in portuguese children. Arch Physiol Biochem 2022; 128:1510-1515. [PMID: 32551914 DOI: 10.1080/13813455.2020.1779312] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
OBJECTIVES To investigate the association of polymorphisms in SLC6A4 and MAOA genes with obesity indices in children. MATERIAL AND METHODS A total of 637 Portuguese children (317 girls; 320 boys) aged 3-11 years-old were genotyped for the SLC6A4 polymorphisms, 5-HTTLPR and STin2, and for a MAOA VNTR. Polymorphisms were analysed by PCR-based methods. RESULTS Although non-significant (p = .089), our study revealed the Stin2 10 minor allele with a marked higher frequency in girls with overweight/obesity (0.466) in comparison with controls (0.376). Combining the two SLC6A4 polymorphisms, haplotype S/12 revealed in girls significant or nominally significant protective effects against BMI (β = -0.615; p = .009), BMI Z-score (β = -0.251; p = .006), WC (β = -1.4; p = .02) and WHtR (β = -0.008; p = .04). CONCLUSIONS We found some evidences for the role of SLC6A4 gene in measures of childhood obesity, mainly in girls.
Collapse
Affiliation(s)
- Licínio Manco
- Research Centre for Anthropology and Health (CIAS), Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| | - Aristides M Machado-Rodrigues
- Research Centre for Anthropology and Health (CIAS), Department of Life Sciences, University of Coimbra, Coimbra, Portugal
- High School of Education, Polytechnic Institute of Viseu, Viseu, Portugal
| | - Cristina Padez
- Research Centre for Anthropology and Health (CIAS), Department of Life Sciences, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
6
|
Mavanji V, Pomonis B, Kotz CM. Orexin, serotonin, and energy balance. WIREs Mech Dis 2022; 14:e1536. [PMID: 35023323 PMCID: PMC9286346 DOI: 10.1002/wsbm.1536] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 07/27/2021] [Accepted: 08/23/2021] [Indexed: 12/02/2022]
Abstract
The lateral hypothalamus is critical for the control of ingestive behavior and spontaneous physical activity (SPA), as lesion or stimulation of this region alters these behaviors. Evidence points to lateral hypothalamic orexin neurons as modulators of feeding and SPA. These neurons affect a broad range of systems, and project to multiple brain regions such as the dorsal raphe nucleus, which contains serotoninergic neurons (DRN) important to energy homeostasis. Physical activity is comprised of intentional exercise and SPA. These are opposite ends of a continuum of physical activity intensity and structure. Non‐goal‐oriented behaviors, such as fidgeting, standing, and ambulating, constitute SPA in humans, and reflect a propensity for activity separate from intentional activity, such as high‐intensity voluntary exercise. In animals, SPA is activity not influenced by rewards such as food or a running wheel. Spontaneous physical activity in humans and animals burns calories and could theoretically be manipulated pharmacologically to expend calories and protect against obesity. The DRN neurons receive orexin inputs, and project heavily onto cortical and subcortical areas involved in movement, feeding and energy expenditure (EE). This review discusses the function of hypothalamic orexin in energy‐homeostasis, the interaction with DRN serotonin neurons, and the role of this orexin‐serotonin axis in regulating food intake, SPA, and EE. In addition, we discuss possible brain areas involved in orexin–serotonin cross‐talk; the role of serotonin receptors, transporters and uptake‐inhibitors in the pathogenesis and treatment of obesity; animal models of obesity with impaired serotonin‐function; single‐nucleotide polymorphisms in the serotonin system and obesity; and future directions in the orexin–serotonin field. This article is categorized under:Metabolic Diseases > Molecular and Cellular Physiology
Collapse
Affiliation(s)
- Vijayakumar Mavanji
- Research Service, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| | - Brianna Pomonis
- Research Service, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| | - Catherine M Kotz
- Department of Integrative Biology and Physiology, University of Minnesota, Minneapolis, Minnesota, USA.,Geriatric Research Education and Clinical Center, Minneapolis VA Health Care System, Minneapolis, Minnesota, USA
| |
Collapse
|
7
|
Beecher K, Wang J, Jacques A, Chaaya N, Chehrehasa F, Belmer A, Bartlett SE. Sucrose Consumption Alters Serotonin/Glutamate Co-localisation Within the Prefrontal Cortex and Hippocampus of Mice. Front Mol Neurosci 2021; 14:678267. [PMID: 34262435 PMCID: PMC8273284 DOI: 10.3389/fnmol.2021.678267] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Accepted: 06/08/2021] [Indexed: 01/23/2023] Open
Abstract
The overconsumption of sugar-sweetened food and beverages underpins the current rise in obesity rates. Sugar overconsumption induces maladaptive neuroplasticity to decrease dietary control. Although serotonin and glutamate co-localisation has been implicated in reward processing, it is still unknown how chronic sucrose consumption changes this transmission in regions associated with executive control over feeding—such as the prefrontal cortex (PFC) and dentate gyrus (DG) of the hippocampus. To address this, a total of 16 C57Bl6 mice received either 5% w/v sucrose or water as a control for 12 weeks using the Drinking-In-The-Dark paradigm (n = 8 mice per group). We then examined the effects of chronic sucrose consumption on the immunological distribution of serotonin (5-HT), vesicular glutamate transporter 3 (VGLUT3) and 5-HT+/VGLUT3+ co-localised axonal varicosities. Sucrose consumption over 12 weeks decreased the number of 5-HT–/VGLUT3+ and 5-HT+/VGLUT3+ varicosities within the PFC and DG. The number of 5-HT+/VGLUT3– varicosities remained unchanged within the PFC but decreased in the DG following sucrose consumption. Given that serotonin mediates DG neurogenesis through microglial migration, the number of microglia within the DG was also assessed in both experimental groups. Sucrose consumption decreased the number of DG microglia. Although the DG and PFC are associated with executive control over rewarding activities and emotional memory formation, we did not detect a subsequent change in DG neurogenesis or anxiety-like behaviour or depressive-like behaviour. Overall, these findings suggest that the chronic consumption of sugar alters serotonergic neuroplasticity within neural circuits responsible for feeding control. Although these alterations alone were not sufficient to induce changes in neurogenesis or behaviour, it is proposed that the sucrose consumption may predispose individuals to these cognitive deficits which ultimately promote further sugar intake.
Collapse
Affiliation(s)
- Kate Beecher
- Addiction Neuroscience and Obesity Laboratory, Faculty of Health, School of Clinical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Joshua Wang
- Addiction Neuroscience and Obesity Laboratory, Faculty of Health, School of Clinical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Angela Jacques
- Addiction Neuroscience and Obesity Laboratory, Faculty of Health, School of Clinical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Nicholas Chaaya
- Addiction Neuroscience and Obesity Laboratory, Faculty of Health, School of Clinical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Fatemeh Chehrehasa
- Addiction Neuroscience and Obesity Laboratory, Faculty of Health, School of Biomedical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Arnauld Belmer
- Addiction Neuroscience and Obesity Laboratory, Faculty of Health, School of Clinical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| | - Selena E Bartlett
- Addiction Neuroscience and Obesity Laboratory, Faculty of Health, School of Clinical Sciences, Translational Research Institute, Queensland University of Technology, Brisbane, QLD, Australia
| |
Collapse
|
8
|
Reduced lipolysis in lipoma phenocopies lipid accumulation in obesity. Int J Obes (Lond) 2020; 45:565-576. [PMID: 33235355 PMCID: PMC7906903 DOI: 10.1038/s41366-020-00716-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/21/2020] [Accepted: 11/02/2020] [Indexed: 02/07/2023]
Abstract
BACKGROUND Elucidation of lipid metabolism and accumulation mechanisms is of paramount importance to understanding obesity and unveiling therapeutic targets. In vitro cell models have been extensively used for these purposes, yet, they do not entirely reflect the in vivo setup. Conventional lipomas, characterized by the presence of mature adipocytes and increased adipogenesis, could overcome the drawbacks of cell cultures. Also, they have the unique advantage of easily accessible matched controls in the form of subcutaneous adipose tissue (SAT) from the same individual. We aimed to determine whether lipomas are a good model to understand lipid accumulation. METHODS We histologically compared lipomas and control SAT, followed by assessment of the lipidome using high-resolution 1H NMR spectroscopy and ESI-IT mass spectrometry. RNA-sequencing was used to obtain the transcriptome of lipomas and the matched SAT. RESULTS We found a significant increase of small-size (maximal axis < 70 µm) and very big (maximal axis > 150 µm) adipocytes within lipomas. This suggests both enhanced adipocyte proliferation and increased lipid accumulation. We further show that there is no significant change in the lipid composition compared to matched SAT. To better delineate the pathophysiology of lipid accumulation, we considered two groups with different genetic backgrounds: (1) lipomas with HMGA2 fusions and (2) without gene fusions. To reduce the search space for genes that are relevant for lipid pathophysiology, we focused on the overlapping differentially expressed (DE) genes between the two groups. Gene Ontology analysis revealed that DE genes are enriched in pathways related to lipid accumulation. CONCLUSIONS We show that the common shared lipid accumulation mechanism in lipoma is a reduction in lipolysis, with most gene dysregulations leading to a reduced cAMP in the adipocyte. Superficial lipomas could thus be used as a model for lipid accumulation through altered lipolysis as found in obese patients.
Collapse
|
9
|
Galaviz-Hernández C, Lazalde-Ramos BP, Martínez-Cortés G, Rangel-Villalobos H, Martínez-Aguilar G, Leal-Ugarte E, Peralta-Leal V, González-Rentería S, Rodríguez-Moran M, Jaquez-Chairez F, Guerrero-Romero F, Sosa-Macías M. Association of the 5HTTLPR Polymorphism with Obesity in Mexican Women with High Native American Ancestry. Genet Test Mol Biomarkers 2020; 24:754-758. [PMID: 33050716 DOI: 10.1089/gtmb.2020.0068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Aims: The 5HTT gene has been associated with obesity; this study aimed to determine the association between L- and S-alleles at the 5HTTLPR polymorphism with obesity in indigenous Mexican populations. Materials and Methods: A total of 362 individuals, 289 belonging to eight Native American (NA) groups; 40 Mexican mestizos; and 33 Caucasian Mennonites were enrolled in a cross-sectional study. High (≥90%) and low (<90%) NA ancestry was molecularly determined. A body mass index >30 kg/m2 was considered as obese. The L- and S-alleles of the 5HTTLPR locus were identified by PCR; the association between alleles and obesity was performed by logistic regression analysis. Results: A significantly lower prevalence of obesity (35%) was observed in participants from communities with high NA ancestry (p < 0.005). Under a dominant heritance model the L-allele was associated with obesity in women with high NA ancestry (odds ratio [OR] 7.27; 95% confidence interval [CI] 1.6-32.5; p = 0.009) but not in women with low NA ancestry (OR 0.83; 95% CI 0.3-2.2; p = 0.71); no association was observed in men. Conclusion: Our results suggest that the 5HTTLPR L-allele is a risk factor for developing obesity in Mexican women with high NA ancestry (≥90%).
Collapse
Affiliation(s)
| | - Blanca P Lazalde-Ramos
- Laboratorio de Etnofarmacología Biomédica, Unidad Académica de Ciencias Químicas, Universidad Autónoma de Zacatecas, Zacatecas, México
| | - Gabriela Martínez-Cortés
- Instituto de Investigación en Genética Molecular, Centro Universitario de la Ciénega, Universidad de Guadalajara (CUCI-UdeG), Ocotlán, Mexico
| | - Héctor Rangel-Villalobos
- Instituto de Investigación en Genética Molecular, Centro Universitario de la Ciénega, Universidad de Guadalajara (CUCI-UdeG), Ocotlán, Mexico
| | | | - Evelia Leal-Ugarte
- Departamento de Genética Aplicada a la Medicina, Facultad de Medicina e Ingeniería en Sistemas Computacionales, Universidad Autónoma de Tamaulipas, H. Matamoros, México
| | - Valeria Peralta-Leal
- Departamento de Genética Aplicada a la Medicina, Facultad de Medicina e Ingeniería en Sistemas Computacionales, Universidad Autónoma de Tamaulipas, H. Matamoros, México
| | | | | | | | | | - Martha Sosa-Macías
- Instituto Politécnico Nacional, Academia de Genómica, CIIDIR Unidad Durango, Durango, México
| |
Collapse
|
10
|
Patte KA, Davis CA, Levitan RD, Kaplan AS, Carter-Major J, Kennedy JL. A Behavioral Genetic Model of the Mechanisms Underlying the Link Between Obesity and Symptoms of ADHD. J Atten Disord 2020; 24:1425-1436. [PMID: 26794671 DOI: 10.1177/1087054715618793] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Objective: The ADHD-obesity link has been suggested to result from a shared underlying basis of suboptimal dopamine (DA); however, this theory conflicts evidence that an amplified DA signal increases the risk for overeating and weight gain. A model was tested in which ADHD symptoms, predicted by hypodopaminergic functioning in the prefrontal cortex, in combination with an enhanced appetitive drive, predict hedonic eating and, in turn, higher body mass index (BMI). Method: DRD2 and DRD4 markers were genotyped. The model was tested using structural equation modeling in a nonclinical sample (N = 421 adults). Results: The model was a good fit to the data. Controlling for education, all parameter estimates were significant, except for the DRD4-ADHD symptom pathway. The significant indirect effect indicates that overeating mediated the ADHD symptoms-BMI association. Conclusion: Results support the hypothesis that overeating and elevated DA in the ventral striatum-representative of a greater reward response-contribute to the ADHD symptom-obesity relationship.
Collapse
Affiliation(s)
| | - Caroline A Davis
- York University, Toronto, Ontario, Canada.,Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,University of Toronto, Ontario, Canada
| | - Robert D Levitan
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,University of Toronto, Ontario, Canada
| | - Allan S Kaplan
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,University of Toronto, Ontario, Canada
| | | | - James L Kennedy
- Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,University of Toronto, Ontario, Canada
| |
Collapse
|
11
|
Underlying Susceptibility to Eating Disorders and Drug Abuse: Genetic and Pharmacological Aspects of Dopamine D4 Receptors. Nutrients 2020; 12:nu12082288. [PMID: 32751662 PMCID: PMC7468707 DOI: 10.3390/nu12082288] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2020] [Revised: 07/26/2020] [Accepted: 07/27/2020] [Indexed: 02/07/2023] Open
Abstract
The dopamine D4 receptor (DRD4) has a predominant expression in the prefrontal cortex (PFC), brain area strictly involved in the modulation of reward processes related to both food and drug consumption. Additionally, the human DRD4 gene is characterized by a variable number of tandem repeats (VNTR) in the exon 3 and, among the polymorphic variants, the 7-repeat (7R) allele appears as a contributing factor in the neurobiological mechanisms underlying drug abuse, aberrant eating behaviors and related comorbidities. The 7R variant encodes for a receptor with a blunted intracellular response to dopamine, and carriers of this polymorphism might be more tempted to enhance dopamine levels in the brain, through the overconsumption of drugs of abuse or palatable food, considering their reinforcing properties. Moreover, the presence of this polymorphism seems to increase the susceptibility of individuals to engage maladaptive eating patterns in response to negative environmental stimuli. This review is focused on the role of DRD4 and DRD4 genetic polymorphism in these neuropsychiatric disorders in both clinical and preclinical studies. However, further research is needed to better clarify the complex DRD4 role, by using validated preclinical models and novel compounds more selective for DRD4.
Collapse
|
12
|
Harris KM, Halpern CT, Whitsel EA, Hussey JM, Killeya-Jones LA, Tabor J, Dean SC. Cohort Profile: The National Longitudinal Study of Adolescent to Adult Health (Add Health). Int J Epidemiol 2020; 48:1415-1415k. [PMID: 31257425 DOI: 10.1093/ije/dyz115] [Citation(s) in RCA: 277] [Impact Index Per Article: 55.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/29/2019] [Indexed: 01/17/2023] Open
Affiliation(s)
- Kathleen Mullan Harris
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Sociology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Carolyn Tucker Halpern
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Maternal and Child Health, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Eric A Whitsel
- Department of Epidemiology and Department of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Jon M Hussey
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Department of Maternal and Child Health, Gillings School of Public Health, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Ley A Killeya-Jones
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA.,Epidemiology Research Team, Department of Radiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Joyce Tabor
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Sarah C Dean
- Carolina Population Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| |
Collapse
|
13
|
Veniaminova E, Cespuglio R, Chernukha I, Schmitt-Boehrer AG, Morozov S, Kalueff AV, Kuznetsova O, Anthony DC, Lesch KP, Strekalova T. Metabolic, Molecular, and Behavioral Effects of Western Diet in Serotonin Transporter-Deficient Mice: Rescue by Heterozygosity? Front Neurosci 2020; 14:24. [PMID: 32132889 PMCID: PMC7041415 DOI: 10.3389/fnins.2020.00024] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Accepted: 01/10/2020] [Indexed: 12/11/2022] Open
Abstract
Reduced function of the serotonin transporter (SERT) is associated with increased susceptibility to anxiety and depression and with type-2 diabetes, which is especially true in older women. Preference for a "Western diet" (WD), enriched with saturated fat, cholesterol, and sugars, may aggravate these conditions. In previous studies, decreased glucose tolerance, central and peripheral inflammation, dyslipidemia, emotional, cognitive, and social abnormalities were reported in WD-fed young female mice. We investigated the metabolic, molecular, and behavioral changes associated with a 3-week-long dietary regime of either the WD or control diet in 12-month-old female mice with three different Sert genotypes: homozygous (Slc6a4) gene knockout (Sert -/-: KO), heterozygous (Sert +/-: HET), or wild-type mice (Sert +/+: WT). In the WT-WD and KO-WD groups, but not in HET-WD-fed mice, most of changes induced by the WD paralleled those found in the younger mice, including brain overexpression of inflammatory marker Toll-like receptor 4 (Tlr4) and impaired hippocampus-dependent performance in the marble test. However, the 12-month-old female mice became obese. Control diet KO mice exhibited impaired hippocampal-dependent behaviors, increased brain expression of the serotonin receptors Htr2c and Htr1b, as well as increased Tlr4 and mitochondrial regulator, peroxisome proliferator-activated receptor gamma-coactivator-1a (Ppargc1a). Paradoxically, these, and other changes, were reversed in KO-WD mutants, suggesting a complex interplay between Sert deficiency and metabolic factors as well as potential compensatory molecular mechanisms that might be disrupted by the WD exposure. Most, but not all, of the changes in gene expression in the brain and liver of KO mice were not exhibited by the HET mice fed with either diet. Some of the WD-induced changes were similar in the KO-WD and HET-WD-fed mice, but the latter displayed a "rescued" phenotype in terms of diet-induced abnormalities in glucose tolerance, neuroinflammation, and hippocampus-dependent performance. Thus, complete versus partial Sert inactivation in aged mice results in distinct metabolic, molecular, and behavioral consequences in response to the WD. Our findings show that Sert +/- mice are resilient to certain environmental challenges and support the concept of heterosis as evolutionary adaptive mechanism.
Collapse
Affiliation(s)
- Ekaterina Veniaminova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands.,Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia
| | - Raymond Cespuglio
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Faculty of Medicine, Neuroscience Research Center of Lyon, C. Bernard University Lyon 1, Lyon, France
| | - Irina Chernukha
- V.M. Gorbatov Federal Research Center for Food Systems of RAS, Moscow, Russia
| | | | - Sergey Morozov
- Institute of General Pathology and Pathophysiology, Moscow, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, Chongqing, China.,Institute of Translational Biomedicine, St. Petersburg State University, St. Petersburg, Russia.,Ural Federal University, Ekaterinburg, Russia
| | - Oxana Kuznetsova
- V.M. Gorbatov Federal Research Center for Food Systems of RAS, Moscow, Russia
| | - Daniel C Anthony
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Department of Pharmacology, Oxford University, Oxford, United Kingdom
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands.,Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, Netherlands.,Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine, Sechenov First Moscow State Medical University, Moscow, Russia.,Division of Molecular Psychiatry, Center of Mental Health, University of Würzburg, Würzburg, Germany.,Institute of General Pathology and Pathophysiology, Moscow, Russia
| |
Collapse
|
14
|
Veniaminova E, Oplatchikova M, Bettendorff L, Kotenkova E, Lysko A, Vasilevskaya E, Kalueff AV, Fedulova L, Umriukhin A, Lesch KP, Anthony DC, Strekalova T. Prefrontal cortex inflammation and liver pathologies accompany cognitive and motor deficits following Western diet consumption in non-obese female mice. Life Sci 2019; 241:117163. [PMID: 31837337 DOI: 10.1016/j.lfs.2019.117163] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 12/08/2019] [Accepted: 12/09/2019] [Indexed: 12/15/2022]
Abstract
AIMS The high sugar and lipid content of the Western diet (WD) is associated with metabolic dysfunction, non-alcoholic steatohepatitis, and it is an established risk factor for neuropsychiatric disorders. Our previous studies reported negative effects of the WD on rodent emotionality, impulsivity, and sociability in adulthood. Here, we investigated the effect of the WD on motor coordination, novelty recognition, and affective behavior in mice as well as molecular and cellular endpoints in brain and peripheral tissues. MAIN METHODS Female C57BL/6 J mice were fed the WD for three weeks and were investigated for glucose tolerance, insulin resistance, liver steatosis, and changes in motor coordination, object recognition, and despair behavior in the swim test. Lipids and liver injury markers, including aspartate-transaminase, alanine-transaminase and urea were measured in blood. Serotonin transporter (SERT) expression, the density of Iba1-positive cells and concentration of malondialdehyde were measured in brain. KEY FINDINGS WD-fed mice exhibited impaired glucose tolerance and insulin resistance, a loss of motor coordination, deficits in novel object exploration and recognition, increased helplessness, dyslipidemia, as well as signs of a non-alcoholic steatohepatitis (NASH)-like syndrome: liver steatosis and increased liver injury markers. Importantly, these changes were accompanied by decreased SERT expression, elevated numbers of microglia cells and malondialdehyde levels in, and restricted to, the prefrontal cortex. SIGNIFICANCE The WD induces a spectrum of behaviors that are more reminiscent of ADHD and ASD than previously recognized and suggests that, in addition to the impairment of impulsivity and sociability, the consumption of a WD might be expected to exacerbate motor dysfunction that is also known to be associated with adult ADHD and ASD.
Collapse
Affiliation(s)
- Ekaterina Veniaminova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER Maastricht, the Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia
| | - Margarita Oplatchikova
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia
| | - Lucien Bettendorff
- Laboratory of Neurophysiology, GIGA-Neurosciences, University of Liège, Av. Hippocrate 15, 4000 Liège, Belgium
| | - Elena Kotenkova
- V.M. Gorbatov Federal Research Center for Food Systems of RAS, Tallalikhina Str. 26, 109316 Moscow, Russia
| | - Alexander Lysko
- Institute of General Pathology and Pathophysiology, Baltiyskaya Str. 8, 125315 Moscow, Russia
| | - Ekaterina Vasilevskaya
- V.M. Gorbatov Federal Research Center for Food Systems of RAS, Tallalikhina Str. 26, 109316 Moscow, Russia
| | - Allan V Kalueff
- School of Pharmacy, Southwest University, 400715 Chongqing, China; Institute of Translational Biomedicine, St. Petersburg State University, Universitetskaya Nab. 7-9, 199034 St. Petersburg, Russia; Ural Federal University, Mira Str. 19, 620002 Ekaterinburg, Russia
| | - Liliya Fedulova
- V.M. Gorbatov Federal Research Center for Food Systems of RAS, Tallalikhina Str. 26, 109316 Moscow, Russia
| | - Aleksei Umriukhin
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia
| | - Klaus-Peter Lesch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER Maastricht, the Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia; Division of Molecular Psychiatry, Laboratory of Translational Neuroscience, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080 Würzburg, Germany
| | - Daniel C Anthony
- Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia; Department of Pharmacology, Oxford University, Mansfield Road, OX1 3QT Oxford, UK
| | - Tatyana Strekalova
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Universiteitssingel 40, NL 6229ER Maastricht, the Netherlands; Laboratory of Psychiatric Neurobiology, Institute of Molecular Medicine and Department of Normal Physiology, Sechenov First Moscow State Medical University, Trubetskaya Str. 8, 119991 Moscow, Russia; Division of Molecular Psychiatry, Laboratory of Translational Neuroscience, Center of Mental Health, University of Würzburg, Margarete-Höppel-Platz 1, 97080 Würzburg, Germany.
| |
Collapse
|
15
|
Yabut JM, Crane JD, Green AE, Keating DJ, Khan WI, Steinberg GR. Emerging Roles for Serotonin in Regulating Metabolism: New Implications for an Ancient Molecule. Endocr Rev 2019; 40:1092-1107. [PMID: 30901029 PMCID: PMC6624793 DOI: 10.1210/er.2018-00283] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2018] [Accepted: 03/18/2019] [Indexed: 12/12/2022]
Abstract
Serotonin is a phylogenetically ancient biogenic amine that has played an integral role in maintaining energy homeostasis for billions of years. In mammals, serotonin produced within the central nervous system regulates behavior, suppresses appetite, and promotes energy expenditure by increasing sympathetic drive to brown adipose tissue. In addition to these central circuits, emerging evidence also suggests an important role for peripheral serotonin as a factor that enhances nutrient absorption and storage. Specifically, glucose and fatty acids stimulate the release of serotonin from the duodenum, promoting gut peristalsis and nutrient absorption. Serotonin also enters the bloodstream and interacts with multiple organs, priming the body for energy storage by promoting insulin secretion and de novo lipogenesis in the liver and white adipose tissue, while reducing lipolysis and the metabolic activity of brown and beige adipose tissue. Collectively, peripheral serotonin acts as an endocrine factor to promote the efficient storage of energy by upregulating lipid anabolism. Pharmacological inhibition of serotonin synthesis or signaling in key metabolic tissues are potential drug targets for obesity, type 2 diabetes, and nonalcoholic fatty liver disease (NAFLD).
Collapse
Affiliation(s)
- Julian M Yabut
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Justin D Crane
- Department of Biology, Northeastern University, Boston, Massachusetts
| | - Alexander E Green
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Damien J Keating
- College of Medicine and Public Health, Flinders University, Bedford Park, South Australia, Australia
| | - Waliul I Khan
- Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.,Farncombe Family Digestive Health Research Institute, McMaster University, Hamilton, Ontario, Canada.,Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| | - Gregory R Steinberg
- Division of Endocrinology and Metabolism, Department of Medicine, McMaster University, Hamilton, Ontario, Canada.,Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada.,Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
16
|
Leite F, Ribeiro L. Dopaminergic Pathways in Obesity-Associated Inflammation. J Neuroimmune Pharmacol 2019; 15:93-113. [PMID: 31317376 DOI: 10.1007/s11481-019-09863-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Accepted: 07/02/2019] [Indexed: 12/11/2022]
|
17
|
Sevilla-González MDR, Quintana-Mendoza BM, Aguilar-Salinas CA. Interaction Between Depression, Obesity, and Type 2 Diabetes: A Complex Picture. Arch Med Res 2018; 48:582-591. [PMID: 29478673 DOI: 10.1016/j.arcmed.2018.02.004] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 02/08/2018] [Indexed: 12/27/2022]
Abstract
Depression plays an important role in the pathogenesis and treatment of obesity and type 2 diabetes (T2D). However, its relevance is frequently unrecognized by clinicians and researchers. The purpose of this review is to present a critical analysis of the evidence linking depression and metabolic disorders and to highlight the practical implications of this complex relationship. Evidence obtained from epidemiological, basic, clinical and controlled studies demonstrate that the association goes beyond a random phenomenon. Epidemiological studies have rendered controversial results due to the lack of control of the confounding variables and the bidirectional relationship that exists between the outcomes and the conditions that modulate the association (i.e. socioeconomic status). Animal and human studies have been useful to define the anatomic substrates and physiologic processes that participate in the association, but, the evidence is preliminary in many areas (i.e gene × environmental interactions). Controlled studies have shown the strong impact that treatment of depression has on body weight and the large effect that has the correction of excess body weight on the depression-related symptoms. Practical implications of the depression-obesity duet include the training of the health providers to assess and treat these conditions in a concomitant manner, the need for translational medicine projects and the application the systems biology approach to fill the existing gaps of knowledge.
Collapse
Affiliation(s)
- Magdalena Del Rocío Sevilla-González
- Unidad de Investigación en Enfermedades Metabólicas. Instituto Nacional de Nutrición Salvador Zubirán and Instituto Tecnológico y de Estudios Superiores de Monterrey Tec Salud, Ciudad de México, México
| | - Brenda Macale Quintana-Mendoza
- Unidad de Investigación en Enfermedades Metabólicas. Instituto Nacional de Nutrición Salvador Zubirán and Instituto Tecnológico y de Estudios Superiores de Monterrey Tec Salud, Ciudad de México, México
| | - Carlos Alberto Aguilar-Salinas
- Unidad de Investigación en Enfermedades Metabólicas. Instituto Nacional de Nutrición Salvador Zubirán and Instituto Tecnológico y de Estudios Superiores de Monterrey Tec Salud, Ciudad de México, México.
| |
Collapse
|
18
|
Abstract
AbstractNegative affect or stress is often found to increase energy intake for high palatable energy-rich foods and hence weight gain. Reduced brain serotonin (5-HT) function is known to increase stress vulnerability and the risk for eating-related disturbances. A short (S) allele polymorphism in the serotonin transporter gene (5-HTTLPR) is associated with a less efficient functioning brain serotonin system and therefore higher stress vulnerability. It has been suggested that this genotype may be directly linked to an increased risk for weight gain and/or obesity. However, a high amount of variability has been apparent in replicating such a direct gene on weight gain relationship. A most recent suggestion is that this gene by weight relationship might be moderated by an additional (cognitive) vulnerability factor involving repetitive negative thinking (rumination). Our objective was to investigate whether the S-allele of 5-HTTLPR contributes to weight gain particularly in high cognitive ruminating individuals. A total of 827 healthy young male and female college students (aged 21·3 (sd 3·0) years; BMI 16–41·7 kg/m2) were genotyped for the 5-HTTLPR polymorphism and assessed for rumination (Event Related Ruminative Index) and body weight. In line with the hypothesis, a hierarchical regression model showed that higher BMI scores were observed in specifically high ruminating S'-carriers (P=0·031, f²=0·022). These results suggest that cognitive rumination may be a critical moderator of the association between 5-HTTLPR and body mass.
Collapse
|
19
|
Avsar O, Kuskucu A, Sancak S, Genc E. Are dopaminergic genotypes risk factors for eating behavior and obesity in adults? Neurosci Lett 2017. [DOI: 10.1016/j.neulet.2017.06.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
20
|
Say YH. The association of insertions/deletions (INDELs) and variable number tandem repeats (VNTRs) with obesity and its related traits and complications. J Physiol Anthropol 2017; 36:25. [PMID: 28615046 PMCID: PMC5471687 DOI: 10.1186/s40101-017-0142-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Accepted: 06/01/2017] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Despite the fact that insertions/deletions (INDELs) are the second most common type of genetic variations and variable number tandem repeats (VNTRs) represent a large portion of the human genome, they have received far less attention than single nucleotide polymorphisms (SNPs) and larger forms of structural variation like copy number variations (CNVs), especially in genome-wide association studies (GWAS) of complex diseases like polygenic obesity. This is exemplified by the vast amount of review papers on the role of SNPs and CNVs in obesity, its related traits (like anthropometric measurements, biochemical variables, and eating behavior), and its related complications (like hypertension, hypertriglyceridemia, hypercholesterolemia, and insulin resistance-collectively known as metabolic syndrome). Hence, this paper reviews the types of INDELs and VNTRs that have been studied for association with obesity and its related traits and complications. These INDELs and VNTRs could be found in the obesity loci or genes from the earliest GWAS and candidate gene association studies, like FTO, genes in the leptin-proopiomelanocortin pathway, and UCP2/3. Given the important role of the brain serotonergic and dopaminergic reward system in obesity susceptibility, the association of INDELs and VNTRs in these neurotransmitters' metabolism and transport genes with obesity is also reviewed. Next, the role of INS VNTR in obesity and its related traits is questionable, since recent large-scale studies failed to replicate the earlier positive associations. As obesity results in chronic low-grade inflammation of the adipose tissue, the proinflammatory cytokine gene IL1RA and anti-inflammatory cytokine gene IL4 have VNTRs that are implicated in obesity. A systemic proinflammatory state in combination with activation of the renin-angiotensin system and decreased nitric oxide bioavailability as found in obesity leads to endothelial dysfunction. This explains why VNTR and INDEL in eNOS and ACE, respectively, could be predisposing factors of obesity. Finally, two novel genes, DOCK5 and PER3, which are involved in the regulation of the Akt/MAPK pathway and circadian rhythm, respectively, have VNTRs and INDEL that might be associated with obesity. SHORT CONCLUSION In conclusion, INDELs and VNTRs could have important functional consequences in the pathophysiology of obesity, and research on them should be continued to facilitate obesity prediction, prevention, and treatment.
Collapse
Affiliation(s)
- Yee-How Say
- Department of Biomedical Science, Faculty of Science, Universiti Tunku Abdul Rahman (UTAR) Kampar Campus, Jalan Universiti, Bandar Barat, 31900, Kampar, Perak, Malaysia.
| |
Collapse
|
21
|
de Luca A, Hankard R, Borys JM, Sinnett D, Marcil V, Levy E. Nutriepigenomics and malnutrition. Epigenomics 2017; 9:893-917. [DOI: 10.2217/epi-2016-0168] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Epigenetics is defined as the modulation of gene expression without changes to the underlying DNA sequence. Epigenetic alterations, as a consequence of in utero malnutrition, may play a role in susceptibility to develop adulthood diseases and inheritance. However, the mechanistic link between epigenetic modifications and abnormalities in nutrition remains elusive. This review provides an update on the association of suboptimal nutritional environment and the high propensity to produce adult-onset chronic illnesses with a particular focus on modifications in genome functions that occur without alterations to the DNA sequence. We will mention the drivers of the phenotype and pattern of epigenetic markers set down during the reprogramming along with novel preventative and therapeutic strategies. New knowledge of epigenetic alterations is opening a gate toward personalized medicine.
Collapse
Affiliation(s)
- Arnaud de Luca
- Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada
- INSERM, U 1069, F-37044 Tours, France
| | - Regis Hankard
- INSERM, U 1069, F-37044 Tours, France
- François Rabelais University, F-37000 Tours, France
| | | | - Daniel Sinnett
- Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada
- Department of Pediatrics, Faculty of Medicine, University of Montreal, Montreal, Quebec H3T 1J4, Canada
| | - Valérie Marcil
- Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada
- Department of Nutrition, Faculty of Medicine, University of Montréal, Montreal, Quebec H3T 1J4, Canada
| | - Emile Levy
- Research Center, Sainte-Justine University Hospital Center, Montreal, Quebec H3T 1C5, Canada
- EPODE International Network, F-75017 Paris, France
- Department of Nutrition, Faculty of Medicine, University of Montréal, Montreal, Quebec H3T 1J4, Canada
| |
Collapse
|
22
|
Miranda RCK, Genro JP, Campagnolo PDB, Mattevi VS, Vitolo MR, Almeida S. Biallelic and triallelic approaches of 5-HTTLPR polymorphism are associated with food intake and nutritional status in childhood. J Nutr Biochem 2017; 43:47-52. [PMID: 28242565 DOI: 10.1016/j.jnutbio.2017.01.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Revised: 12/30/2016] [Accepted: 01/25/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND The 5-HTT gene contains polymorphisms in its promoter region, the insertion/deletion (5-HTTLPR) that creates long (L) or short (S) alleles (biallelic approach) and SNP (rs25531) in L allele (triallelic approach). OBJECTIVES The aim of this study is to investigate the association of the 5-HTTLPR and rs25531 polymorphisms, using bi- and triallelic approach, with dietary intake and anthropometric parameters in children followed until 8 years old. METHODS The sample were 303 children who were recruited at birth and examined at 1, 3 to 4 and 7 to 8 years old. The polymorphisms were analyzed by polymerase-chain-reaction-based methods. RESULTS In the biallelic approach, children with the S/S genotype presented a higher body mass index Z-score in the three developmental stages and higher sum of skinfolds at 3 to 4 and 7 to 8 years old than carriers of the L allele. In the triallelic approach, S/S, Lg/S plus Lg/Lg genotypes were associated with higher energy intake daily at 1 year old and with waist circumference at 3 to 4 years old. CONCLUSIONS In the biallelic approach, the 5-HTTLPR polymorphism is associated with food intake, body mass index Z-score and sum of skinfolds in children, reinforcing the role of the serotonin transporter in childhood obesity. Our data indicate that the biallelic approach is more sensible than the triallelic approach for detected associations with food intake and nutritional status in childhood. Identifying susceptibility genes in early life could provide the foundations for interventions in lifestyle to prevent children to become obese adults.
Collapse
Affiliation(s)
- Raquel C K Miranda
- Laboratório de Biologia Molecular, Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245/Anexo 3 sala 303; CEP 90050-170, Porto Alegre, RS, Brazil
| | - Júlia P Genro
- Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245-sala 309; CEP 90050-170, Porto Alegre, RS, Brazil
| | - Paula D B Campagnolo
- Departamento de Nutrição, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245/Anexo 2-sala 03; CEP 90050-170, Porto Alegre, RS, Brazil
| | - Vanessa S Mattevi
- Laboratório de Biologia Molecular, Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245/Anexo 3 sala 303; CEP 90050-170, Porto Alegre, RS, Brazil; Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245-sala 309; CEP 90050-170, Porto Alegre, RS, Brazil
| | - Márcia R Vitolo
- Departamento de Nutrição, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245/Anexo 2-sala 03; CEP 90050-170, Porto Alegre, RS, Brazil
| | - Silvana Almeida
- Laboratório de Biologia Molecular, Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245/Anexo 3 sala 303; CEP 90050-170, Porto Alegre, RS, Brazil; Departamento de Ciências Básicas da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre, Rua Sarmento Leite, 245-sala 309; CEP 90050-170, Porto Alegre, RS, Brazil.
| |
Collapse
|
23
|
Dalle Molle R, Fatemi H, Dagher A, Levitan RD, Silveira PP, Dubé L. Gene and environment interaction: Is the differential susceptibility hypothesis relevant for obesity? Neurosci Biobehav Rev 2016; 73:326-339. [PMID: 28024828 DOI: 10.1016/j.neubiorev.2016.12.028] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2016] [Revised: 12/09/2016] [Accepted: 12/20/2016] [Indexed: 02/04/2023]
Abstract
The differential susceptibility model states that a given genetic variant is associated with an increased risk of pathology in negative environments but greater than average resilience in enriched ones. While this theory was first implemented in psychiatric-genetic research, it may also help us to unravel the complex ways that genes and environments interact to influence feeding behavior and obesity. We reviewed evidence on gene vs. environment interactions that influence obesity development, aiming to support the applicability of the differential susceptibility model for this condition, and propose that various environmental "layers" relevant for human development should be considered when bearing the differential susceptibility model in mind. Mother-child relationship, socioeconomic status and individual's response are important modifiers of BMI and food intake when interacting with gene variants, "for better and for worse". While only a few studies to date have investigated obesity outcomes using this approach, we propose that the differential susceptibility hypothesis is in fact highly applicable to the study of genetic and environmental influences on feeding behavior and obesity risk.
Collapse
Affiliation(s)
- Roberta Dalle Molle
- Desautels Faculty of Management, McGill Center for the Convergence of Health and Economics, McGill University, Bronfman Building, 1001 Sherbrooke St. W., Montreal, QC H3A 1G5, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada.
| | - Hajar Fatemi
- Desautels Faculty of Management, McGill Center for the Convergence of Health and Economics, McGill University, Bronfman Building, 1001 Sherbrooke St. W., Montreal, QC H3A 1G5, Canada
| | - Alain Dagher
- McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, 3801 University Street, Montreal, QC H3A 2B4, Canada
| | - Robert D Levitan
- Department of Psychiatry, University of Toronto and Centre for Addiction and Mental Health, 100 Stokes Street, Toronto, ON M6J 1H4, Canada
| | - Patricia P Silveira
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Douglas Institute, Perry Pavilion, 6875 Boulevard LaSalle, Montreal, QC H4H 1R3, Canada
| | - Laurette Dubé
- Desautels Faculty of Management, McGill Center for the Convergence of Health and Economics, McGill University, Bronfman Building, 1001 Sherbrooke St. W., Montreal, QC H3A 1G5, Canada
| |
Collapse
|
24
|
Dias H, Muc M, Padez C, Manco L. Association of polymorphisms in 5-HTT (SLC6A4) and MAOA genes with measures of obesity in young adults of Portuguese origin. Arch Physiol Biochem 2016; 122:8-13. [PMID: 26698543 DOI: 10.3109/13813455.2015.1111390] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
OBJECTIVES To investigate the association of polymorphisms in SLC6A4 and MAOA genes with overweight (including obesity). MATERIAL AND METHODS Young adults (n = 535) of Portuguese origin were genotyped for the SLC6A4 polymorphisms 5-HTTLPR and STin2 and a MAOA VNTR. BMI and body fat percentage were measured and a questionnaire was used to assess individual's sport practicing habits. RESULTS In whole study sample, haplotype-based analysis revealed significant association with overweight/obesity for the individual 5-HTTLPR/Stin2 haplotype L10 (p = 0.04). In men, the MAOA 3R genotype was nominally associated with body fat (p = 0.04). In inactive individuals, overweight/obesity was found significantly associated with 5-HTTLPR L-allele (p = 0.01) and nominally associated with STin2 10-allele (p = 0.03). A significant association was also found testing for all haplotype effects (χ(2 )= 8.7; p = 0.03). CONCLUSIONS We found some evidences for the association of SLC6A4 and MAOA genes with measures of obesity. Our results suggest physical inactivity accentuates the influence of SLC6A4 polymorphisms on obesity risk.
Collapse
Affiliation(s)
- Helena Dias
- a Research Centre for Anthropology and Health (CIAS), Department of Life Sciences , University of Coimbra , Portugal
| | - Magdalena Muc
- a Research Centre for Anthropology and Health (CIAS), Department of Life Sciences , University of Coimbra , Portugal
| | - Cristina Padez
- a Research Centre for Anthropology and Health (CIAS), Department of Life Sciences , University of Coimbra , Portugal
| | - Licínio Manco
- a Research Centre for Anthropology and Health (CIAS), Department of Life Sciences , University of Coimbra , Portugal
| |
Collapse
|
25
|
|
26
|
Uzun M, Saglar E, Kucukyildirim S, Erdem B, Unlu H, Mergen H. Association of VNTR polymorphisms in DRD4, 5-HTT and DAT1 genes with obesity. Arch Physiol Biochem 2015; 121:75-9. [PMID: 25687785 DOI: 10.3109/13813455.2014.985686] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
OBJECTIVE To investigate the association between VNTR polymorphisms of DRD4, DAT1 and 5-HTT genes and obesity. MATERIAL AND METHODS Peripheral blood samples of 234 obese (BMI ≥ 30) and 148 healthy individuals (BMI ≤ 25) were objected to PCR to detect the VNTR of the 2nd intron of 5-HTT, 3rd exon of DRD4 and 3'UTR of DAT1 genes. RESULTS The association between obesity and genotype distributions of 5-HTT, DAT1 and DRD4 genes and between obesity and distributions of allele frequencies were tested by Chi Square (χ(2)) test and were not found statistically significant. BMI values for genotype of obese and morbidly obese (BMI > 40) individuals were analyzed by Kruskal-Wallis and not found statistically significant differences between BMI values for the most frequent genotypes of 5-HTT, DAT1 and DRD4 genes. CONCLUSIONS As a conclusion, there was no association between 5-HTT, DAT1 and DRD4 genes VNTR polymorphisms and obesity.
Collapse
Affiliation(s)
- Mustafa Uzun
- Hacettepe University, Faculty of Science, Department of Biology, 06800, Beytepe , Ankara , Turkey and
| | | | | | | | | | | |
Collapse
|
27
|
Hameed A, Ajmal M, Nasir M, Ismail M. Genetic association analysis of serotonin transporter polymorphism (5-HTTLPR) with type 2 diabetes patients of Pakistani population. Diabetes Res Clin Pract 2015; 108:67-71. [PMID: 25680681 DOI: 10.1016/j.diabres.2015.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 12/03/2014] [Accepted: 01/14/2015] [Indexed: 12/01/2022]
Abstract
AIMS It is well established that the serotonergic system contributes to the regulation of glucose homeostasis and feeding and therefore it has been presumed to contribute to the biological susceptibility to type 2 diabetes mellitus (T2DM) and body-mass index (BMI). 5-HTTLPR is a serotonin transporter (5-HTT) gene-linked polymorphic region that regulates the transcriptional activity of 5-HTT. Our aim was to investigate the possible association of 5-HTTLPR polymorphism (L and S alleles) in the promoter region of the serotonin transporter gene with T2DM and/or higher BMI in Pakistani population. METHODS In this study, 574 subjects diagnosed with T2DM and 402 unrelated normal controls from the general Pakistani population were genotyped for 5-HTTLPR polymorphism by PCR amplification and agarose gel electrophoresis. The genotyping data (S/S, S/L and L/L) were recorded and analysed statistically using various software and online available tools. RESULTS In the total sample, patients with type 2 diabetes and controls without diabetes, genotypes were distributed according to Hardy-Weinberg equilibrium, and S allele frequency was 61.52% (0.61). There was no statistical association between 5-HTTLPR polymorphism and the development of T2DM in this Pakistani population (p=0.12). CONCLUSIONS No significant statistical association of 5-HTTLPR polymorphism with type 2 diabetes and obesity in Pakistani population shows that 5-HTTLPR polymorphism is not a major factor in determining type 2 diabetes and obesity in Pakistan.
Collapse
Affiliation(s)
- A Hameed
- Institute of Biomedical and Genetic Engineering, 24-Mauve Area, G-9/1, Islamabad, Pakistan.
| | - M Ajmal
- Institute of Biomedical and Genetic Engineering, 24-Mauve Area, G-9/1, Islamabad, Pakistan
| | - M Nasir
- Institute of Biomedical and Genetic Engineering, 24-Mauve Area, G-9/1, Islamabad, Pakistan
| | - M Ismail
- Institute of Biomedical and Genetic Engineering, 24-Mauve Area, G-9/1, Islamabad, Pakistan
| |
Collapse
|
28
|
Yilmaz Z, Davis C, Loxton NJ, Kaplan AS, Levitan RD, Carter JC, Kennedy JL. Association between MC4R rs17782313 polymorphism and overeating behaviors. Int J Obes (Lond) 2015; 39:114-20. [PMID: 24827639 PMCID: PMC4232480 DOI: 10.1038/ijo.2014.79] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2014] [Revised: 04/10/2014] [Accepted: 04/14/2014] [Indexed: 12/17/2022]
Abstract
BACKGROUND/OBJECTIVES Melanocortins have a crucial role in appetite and weight regulation. Although the melanocortin 4 receptor (MC4R) gene has been repeatedly linked to obesity and antipsychotic-induced weight gain, the mechanism behind how it leads to this effect in still undetermined. The goal of this study was to conduct an in-depth and sophisticated analysis of MC4R polymorphisms, body mass index (BMI), eating behavior and depressed mood. SUBJECTS/METHODS We genotyped 328 individuals of European ancestry on the following MC4R markers based on the relevant literature on obesity and antipsychotic-induced weight gain: rs571312, rs17782313, rs489693, rs11872992, and rs8087522. Height and weight were measured, and information on depressed mood and overeating behaviors was obtained during the in-person assessment. RESULTS BMI was associated with rs17782313 C allele; however, this finding did not survive correction for multiple testing (P = 0.018). Although rs17782313 was significantly associated with depressed mood and overeating behaviors, tests of indirect effects indicated that emotional eating and food cravings, rather than depressed mood, uniquely accounted for the effect of this marker and BMI (n = 152). CONCLUSIONS To our knowledge, this is the first study to investigate the link between MC4R rs17782313, mood and overeating behavior, as well as to demonstrate possible mechanisms behind MC4R's influence on body weight. If replicated in a larger sample, these results may have important clinical implications, including potential for the use of MC4R agonists in the treatment of obesity and disordered eating.
Collapse
Affiliation(s)
- Zeynep Yilmaz
- Center of Excellence for Eating Disorders, University of North Carolina at Chapel Hill, Chapel Hill, NC
- Clinical Research Department, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Caroline Davis
- Clinical Research Department, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Kinesiology & Health Sciences, York University, Toronto, Ontario, Canada
- Eating Disorders Program, Toronto General Hospital, Toronto, Ontario, Canada
| | - Natalie J. Loxton
- School of Psychology, The University of Queensland, Brisbane, Queensland, Australia
| | - Allan S. Kaplan
- Clinical Research Department, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Robert D. Levitan
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Mood and Anxiety Program, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | | | - James L. Kennedy
- Institute of Medical Science, University of Toronto, Toronto, Ontario, Canada
- Department of Psychiatry, University of Toronto, Toronto, Ontario, Canada
- Neurogenetics Section, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| |
Collapse
|
29
|
Yokum S, Marti CN, Smolen A, Stice E. Relation of the multilocus genetic composite reflecting high dopamine signaling capacity to future increases in BMI. Appetite 2014; 87:38-45. [PMID: 25523644 DOI: 10.1016/j.appet.2014.12.202] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2014] [Revised: 11/11/2014] [Accepted: 12/09/2014] [Indexed: 12/22/2022]
Abstract
Because food intake exerts its rewarding effect by increasing dopamine (DA) signaling in reward circuitry, it theoretically follows that individuals with a greater number of genotypes putatively associated with high DA signaling capacity are at increased risk for overeating and subsequent weight gain. We tested the association between the multilocus genetic composite risk score, defined by the total number of genotypes putatively associated with greater DA signaling capacity (i.e. TaqIA A2 allele, DRD2-141C Ins/Del and Del/Del genotypes, DRD4-S allele, DAT1-S allele, and COMT Val/Val genotype), and future increases in Body Mass Index (BMI) in three prospective studies. Participants in Study 1 (N = 30; M age = 15.2; M baseline BMI = 26.9), Study 2 (N = 34; M age = 20.9; M baseline BMI = 28.2), and Study 3 (N = 162; M age = 15.3, M baseline BMI = 20.8) provided saliva samples from which epithelial cells were collected, permitting DNA extraction. The multilocus genetic composite risk score was associated with future increases in BMI in all three studies (Study 1, r = 0.37; Study 2, r = 0.22; Study 3, r = 0.14) and the overall sample (r = 0.19). DRD4-S was associated with increases in BMI in Study 1 (r = 0.42), Study 2 (r = 0.27), and in the overall sample (r = 0.17). DAT1-S was associated with increases in BMI in Study 3 (r = 0.17) and in the overall sample (r = 0.12). There were no associations between the other genotypes (TaqIA, COMT, and DRD2-141C) and change in BMI over 2-year follow-up. Data suggest that individuals with a genetic propensity for greater DA signaling capacity are at risk for future weight gain and that combining alleles that theoretically have a similar function may provide a more reliable method of modeling genetic risk associated with future weight gain than individual genotypes.
Collapse
Affiliation(s)
- Sonja Yokum
- Oregon Research Institute, 1776 Millrace Drive, Eugene, OR 97403, USA.
| | - C Nathan Marti
- Oregon Research Institute, 1776 Millrace Drive, Eugene, OR 97403, USA
| | - Andrew Smolen
- Institute for Behavioral Genetics, University of Colorado, 1480 30th Street, Boulder, CO 80303
| | - Eric Stice
- Oregon Research Institute, 1776 Millrace Drive, Eugene, OR 97403, USA
| |
Collapse
|
30
|
Xie B, Li D, London SJ, Palmer PH, Johnshon CA, Li Y, Shih J, Bergen AW, Nishita D, Swan GE, Ahn R, Conti DV. Gender difference in interactions between MAOA promoter uVNTR polymorphism and negative familial stressors on body mass index among Chinese adolescents. Pediatr Obes 2014; 9:e80-90. [PMID: 23761378 PMCID: PMC4159439 DOI: 10.1111/j.2047-6310.2013.00181.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2012] [Revised: 03/07/2013] [Accepted: 04/29/2013] [Indexed: 11/26/2022]
Abstract
OBJECTIVES Monoamine oxidase A (MAOA) modulates metabolism of serotonin and dopamine metabolism, neurotransmitters involved in regulation of appetite and food intake. The gene coding for MAOA contains a 30-bp tandem repeat (uVNTR) polymorphism in its promoter region that has been previously identified to be associated with obesity with mixed findings in the literature. Our goals were to replicate the population effects of this functional polymorphism on obesity risk, and to further explore gender differences and interaction effects with negative stressors. METHODS Analyses were conducted with data on genotypes, measured weight and height, and self-reported behavioural characteristics among 1101 Chinese adolescents 11-15 years old living in Wuhan, China. RESULTS Girls with the high-activity allele had significantly lower body mass index (BMI; β = -0.25 ± 0.98, P = 0.011) compared to those with the low activity allele. Experience of negative familial stressors (e.g., death or illness of family members, hit or scolded by parents and increased quarrelling with parents, parents argued frequently) significantly weakened this protective genetic effect on BMI (P for interaction = 0.043). Stratified analyses showed a significant protective genetic effect on BMI only within the stratum of low stress level (β = -0.44 ± 0.14, P = 0.002). No similar effect was observed among boys. CONCLUSIONS Our findings confirm the genetic effects of MAOA uVNTR polymorphism on BMI in a Chinese adolescent population and suggest potential genetic interactions with negative familial stressors.
Collapse
Affiliation(s)
- Bin Xie
- School of Community and Global Health, Claremont Graduate University, Claremont, CA 91711
| | - Dalin Li
- Medical Genetics Institute, Cedars-Sinai Medical Center/University of California at Los Angeles, Los Angeles, CA 90048
| | - Stephanie J. London
- Division of Intramural Research, National Institute of Environmental Health Sciences, National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC 27709
| | - Paula H. Palmer
- School of Community and Global Health, Claremont Graduate University, Claremont, CA 91711
| | - C. Anderson Johnshon
- School of Community and Global Health, Claremont Graduate University, Claremont, CA 91711
| | - Yan Li
- Wuhan City Food and Drug Administration, Wuhan, P. R. China
| | - Jean Shih
- School of Pharmacy, University of Southern California, Los Angeles, CA 90089
| | - Andrew W. Bergen
- Center for Health Sciences, SRI International, Menlo Park, CA 94025
| | - Denise Nishita
- Center for Health Sciences, SRI International, Menlo Park, CA 94025
| | - Gary E. Swan
- Center for Health Sciences, SRI International, Menlo Park, CA 94025
| | - Rosa Ahn
- Joint Science Program, Scripps College, Claremont, CA 91711
| | - David V. Conti
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033
| |
Collapse
|
31
|
van Gestel MA, Kostrzewa E, Adan RAH, Janhunen SK. Pharmacological manipulations in animal models of anorexia and binge eating in relation to humans. Br J Pharmacol 2014; 171:4767-84. [PMID: 24866852 PMCID: PMC4209941 DOI: 10.1111/bph.12789] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2013] [Revised: 03/07/2014] [Accepted: 05/16/2014] [Indexed: 12/22/2022] Open
Abstract
Eating disorders, such as anorexia nervosa (AN), bulimia nervosa (BN) and binge eating disorders (BED), are described as abnormal eating habits that usually involve insufficient or excessive food intake. Animal models have been developed that provide insight into certain aspects of eating disorders. Several drugs have been found efficacious in these animal models and some of them have eventually proven useful in the treatment of eating disorders. This review will cover the role of monoaminergic neurotransmitters in eating disorders and their pharmacological manipulations in animal models and humans. Dopamine, 5-HT (serotonin) and noradrenaline in hypothalamic and striatal regions regulate food intake by affecting hunger and satiety and by affecting rewarding and motivational aspects of feeding. Reduced neurotransmission by dopamine, 5-HT and noradrenaline and compensatory changes, at least in dopamine D2 and 5-HT(2C/2A) receptors, have been related to the pathophysiology of AN in humans and animal models. Also, in disorders and animal models of BN and BED, monoaminergic neurotransmission is down-regulated but receptor level changes are different from those seen in AN. A hypofunctional dopamine system or overactive α2-adrenoceptors may contribute to an attenuated response to (palatable) food and result in hedonic binge eating. Evidence for the efficacy of monoaminergic treatments for AN is limited, while more support exists for the treatment of BN or BED with monoaminergic drugs.
Collapse
Affiliation(s)
- M A van Gestel
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center UtrechtUtrecht, The Netherlands
| | - E Kostrzewa
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center UtrechtUtrecht, The Netherlands
| | - R A H Adan
- Brain Center Rudolf Magnus, Department of Translational Neuroscience, University Medical Center UtrechtUtrecht, The Netherlands
| | - S K Janhunen
- Orion Corporation Orion Pharma, Research and Development, CNS ResearchTurku, Finland
| |
Collapse
|
32
|
Blum K, Thanos PK, Gold MS. Dopamine and glucose, obesity, and reward deficiency syndrome. Front Psychol 2014; 5:919. [PMID: 25278909 PMCID: PMC4166230 DOI: 10.3389/fpsyg.2014.00919] [Citation(s) in RCA: 127] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2014] [Accepted: 08/01/2014] [Indexed: 01/08/2023] Open
Abstract
Obesity as a result of overeating as well as a number of well described eating disorders has been accurately considered to be a world-wide epidemic. Recently a number of theories backed by a plethora of scientifically sound neurochemical and genetic studies provide strong evidence that food addiction is similar to psychoactive drug addiction. Our laboratory has published on the concept known as Reward Deficiency Syndrome (RDS) which is a genetic and epigenetic phenomena leading to impairment of the brain reward circuitry resulting in a hypo-dopaminergic function. RDS involves the interactions of powerful neurotransmitters and results in abnormal craving behavior. A number of important facts which could help translate to potential therapeutic targets espoused in this focused review include: (1) consumption of alcohol in large quantities or carbohydrates binging stimulates the brain’s production of and utilization of dopamine; (2) in the meso-limbic system the enkephalinergic neurons are in close proximity, to glucose receptors; (3) highly concentrated glucose activates the calcium channel to stimulate dopamine release from P12 cells; (4) a significant correlation between blood glucose and cerebrospinal fluid concentrations of homovanillic acid the dopamine metabolite; (5) 2-deoxyglucose (2DG), the glucose analog, in pharmacological doses is associated with enhanced dopamine turnover and causes acute glucoprivation. Evidence from animal studies and fMRI in humans support the hypothesis that multiple, but similar brain circuits are disrupted in obesity and drug dependence and for the most part, implicate the involvement of DA-modulated reward circuits in pathologic eating behaviors. Based on a consensus of neuroscience research treatment of both glucose and drug like cocaine, opiates should incorporate dopamine agonist therapy in contrast to current theories and practices that utilizes dopamine antagonistic therapy. Considering that up until now clinical utilization of powerful dopamine D2 agonists have failed due to chronic down regulation of D2 receptors newer targets based on novel less powerful D2 agonists that up-regulate D2 receptors seems prudent. We encourage new strategies targeted at improving DA function in the treatment and prevention of obesity a subtype of reward deficiency.
Collapse
Affiliation(s)
- Kenneth Blum
- Department of Psychiatry, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville FL, USA ; Department of Addiction Research & Therapy, Malibu Beach Recovery Center Malibu Beach, CA, USA
| | - Panayotis K Thanos
- Behavior Neuropharmacology and Neuroimaging Lab, Department of Psychology, State University of New York Stony Brook, NY, USA
| | - Mark S Gold
- Department of Psychiatry, McKnight Brain Institute, College of Medicine, University of Florida, Gainesville FL, USA ; Department of Addiction Research & Therapy, Malibu Beach Recovery Center Malibu Beach, CA, USA
| |
Collapse
|
33
|
BLUM KENNETH, FEBO MARCELO, MCLAUGHLIN THOMAS, CRONJÉ FRANSJ, HAN DAVID, GOLD SMARK. Hatching the behavioral addiction egg: Reward Deficiency Solution System (RDSS)™ as a function of dopaminergic neurogenetics and brain functional connectivity linking all addictions under a common rubric. J Behav Addict 2014; 3:149-56. [PMID: 25317338 PMCID: PMC4189308 DOI: 10.1556/jba.3.2014.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Revised: 07/04/2014] [Accepted: 07/04/2014] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Following the first association between the dopamine D2 receptor gene polymorphism and severe alcoholism, there has been an explosion of research reports in the psychiatric and behavioral addiction literature and neurogenetics. With this increased knowledge, the field has been rife with controversy. Moreover, with the advent of Whole Genome-Wide Studies (GWAS) and Whole Exome Sequencing (WES), along with Functional Genome Convergence, the multiple-candidate gene approach still has merit and is considered by many as the most prudent approach. However, it is the combination of these two approaches that will ultimately define real, genetic allelic relationships, in terms of both risk and etiology. Since 1996, our laboratory has coined the umbrella term Reward Deficiency Syndrome (RDS) to explain the common neurochemical and genetic mechanisms involved with both substance and non-substance, addictive behaviors. METHODS This is a selective review of peer-reviewed papers primary listed in Pubmed and Medline. RESULTS A review of the available evidence indicates the importance of dopaminergic pathways and resting-state, functional connectivity of brain reward circuits. DISCUSSION Importantly, the proposal is that the real phenotype is RDS and impairments in the brain's reward cascade, either genetically or environmentally (epigenetically) induced, influence both substance and non-substance, addictive behaviors. Understanding shared common mechanisms will ultimately lead to better diagnosis, treatment and prevention of relapse. While, at this juncture, we cannot as yet state that we have "hatched the behavioral addiction egg", we are beginning to ask the correct questions and through an intense global effort will hopefully find a way of "redeeming joy" and permitting homo sapiens live a life, free of addiction and pain.
Collapse
Affiliation(s)
- KENNETH BLUM
- Department of Psychiatry and McKnight Brain Institute, University of Florida, College of Medicine, Gainesville, FL, USA,Department of Clinical Medicine, Malibu Beach Recovery Center, Malibu Beach, CA, USA,Department of Personalized Medicine, IGENE, LLC, Austin, TX, USA,Corresponding author: Kenneth Blum, PhD; Department of Psychiatry and McKnight Brain Institute, University of Florida, College of Medicine, PO Box 103424 Gainesville, Florida, USA, 32610-3424; Phone: +-619-890-2167; Fax: +-352-392-9887; E-mail:
| | - MARCELO FEBO
- Department of Psychiatry and McKnight Brain Institute, University of Florida, College of Medicine, Gainesville, FL, USA
| | | | | | - DAVID HAN
- Department of Management Science and Statistics, University of Texas at San Antonio, Texas, USA
| | - S. MARK GOLD
- Department of Psychiatry and McKnight Brain Institute, University of Florida, College of Medicine, Gainesville, FL, USA,Department of Clinical Medicine, Malibu Beach Recovery Center, Malibu Beach, CA, USA
| |
Collapse
|
34
|
Hesse S, van de Giessen E, Zientek F, Petroff D, Winter K, Dickson JC, Tossici-Bolt L, Sera T, Asenbaum S, Darcourt J, Akdemir UO, Knudsen GM, Nobili F, Pagani M, Vander Borght T, Van Laere K, Varrone A, Tatsch K, Sabri O, Booij J. Association of central serotonin transporter availability and body mass index in healthy Europeans. Eur Neuropsychopharmacol 2014; 24:1240-7. [PMID: 24976619 DOI: 10.1016/j.euroneuro.2014.05.005] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 04/09/2014] [Accepted: 05/09/2014] [Indexed: 01/24/2023]
Abstract
UNLABELLED Serotonin-mediated mechanisms, in particular via the serotonin transporter (SERT), are thought to have an effect on food intake and play an important role in the pathophysiology of obesity. However, imaging studies that examined the correlation between body mass index (BMI) and SERT are sparse and provided contradictory results. The aim of this study was to further test the association between SERT and BMI in a large cohort of healthy subjects. METHODS 127 subjects of the ENC DAT database (58 females, age 52 ± 18 years, range 20-83, BMI 25.2 ± 3.8 kg/m(2), range 18.2-41.1) were analysed using region-of-interest (ROI) and voxel-based approaches to calculate [(123)I]FP-CIT specific-to-nonspecific binding ratios (SBR) in the hypothalamus/thalamus and midbrain/brainstem as SERT-specific target regions. RESULTS In the voxel-based analysis, SERT availability and BMI were positively associated in the thalamus, but not in the midbrain. In the ROI-analysis, the interaction between gender and BMI showed a trend with higher correlation coefficient for men in the midbrain albeit not significant (0.033SBRm(2)/kg, p=0.1). CONCLUSIONS The data are in agreement with previous PET findings of an altered central serotonergic tone depending on BMI, as a probable pathophysiologic mechanism in obesity, and should encourage further clinical studies in obesity targeting the serotonergic system.
Collapse
Affiliation(s)
- Swen Hesse
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany; Leipzig University Medical Center, Integrated Research and Treatment Centre (IFB) Adiposity Diseases, Leipzig, Germany.
| | - Elsmarieke van de Giessen
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Franziska Zientek
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany
| | - David Petroff
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany; Centre for Clinical Studies, Leipzig, Germany
| | - Karsten Winter
- Translational Centre for Regenerative Medicine (TRM) Leipzig, Germany
| | - John C Dickson
- Institute of Nuclear Medicine, University College London Hospital, London, UK
| | - Livia Tossici-Bolt
- Department of Medical Physics and Bioengineering, Southampton University Hospitals NHS Trust, Southampton, UK
| | - Terez Sera
- University of Szeged, Department of Nuclear Medicine and Euromedic Szeged, Szeged, Hungary
| | - Susanne Asenbaum
- Department of Nuclear Medicine, Medical University of Vienna, Austria
| | - Jacques Darcourt
- Nuclear Medicine Department, Centre Antoine Lacassagne, University of Nice-Sophia Antipolis, Nice, France
| | - Umit O Akdemir
- Department of Nuclear Medicine, Gazi University, Faculty of Medicine, Ankara, Turkey
| | - Gitte M Knudsen
- Neurobiology Research Unit, Rigshospitalet - University of Copenhagen, Copenhagen, Denmark
| | - Flavio Nobili
- Clinical Neurophysiology Unit, Department of Neuroscience, Ophthalmology and Genetics, San Martino Hospital, University of Genoa, Genoa, Italy
| | - Marco Pagani
- Department of Nuclear Medicine, Karolinska University Hospital, Stockholm, Sweden; Institute of Cognitive Sciences and Technologies, CNR, Rome & Padua, Italy
| | - Thierry Vander Borght
- Nuclear Medicine Division, Université Catholique de Louvain, Mont-Godinne Medical Center, Louvain-la-Neuve, Belgium
| | - Koen Van Laere
- Nuclear Medicine, University Hospital, K.U. Leuven, Leuven, Belgium
| | - Andrea Varrone
- Karolinska Institutet, Department of Clinical Neuroscience, Stockholm, Sweden
| | - Klaus Tatsch
- Department of Nuclear Medicine, University of Munich, Munich, Germany; Department of Nuclear Medicine, Municipal Hospital Karlsruhe Inc., Karlsruhe, Germany
| | - Osama Sabri
- Department of Nuclear Medicine, University of Leipzig, Leipzig, Germany; Leipzig University Medical Center, Integrated Research and Treatment Centre (IFB) Adiposity Diseases, Leipzig, Germany
| | - Jan Booij
- Department of Nuclear Medicine, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
35
|
Calzadilla P, Gómez-Serrano M, García-Santos E, Schiappacasse A, Abalde Y, Calvo JC, Peral B, Guerra LN. N-Acetylcysteine affects obesity-related protein expression in 3T3-L1 adipocytes. Redox Rep 2014; 18:210-8. [PMID: 24112955 DOI: 10.1179/1351000213y.0000000066] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES Oxidative stress plays critical roles in the pathogeneses of diabetes, hypertension, and atherosclerosis, but its effect on fat accumulation is still unclear. In this study, we analyzed the role of the well-known antioxidant and a glutathione (GSH) precursor N-acetylcysteine (NAC) in fat accumulation and the expression of obesity-associated proteins. METHODS We studied the effects of 10 µM NAC on obesity-related protein expression in cultured 3T3-L1 preadipocytes, which are able to differentiate into mature adipocytes and accumulate lipids. RESULTS NAC treatment inhibited fat accumulation and reduced the expression of obesity-related proteins, including monoamine oxidase A, heat shock protein 70 (HSP70), aminoacylase -1 (ACY-1), and transketolase. DISCUSSION Our results suggest that the effects of NAC on triglycerides (Tgs) and protein expression are correlated. In support of this, we showed that NAC treatment affected both the Tg synthesis pathway and the expression levels of proteins implicated in human obesity.
Collapse
Affiliation(s)
- P Calzadilla
- Universidad de Buenos Aires - IQUIBICEN, CONICET. Intendente Güiraldes 2160, Pabellón 2 (1428), Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Leptin, adiponectin and serotonin levels in lean and obese dogs. BMC Vet Res 2014; 10:113. [PMID: 24886049 PMCID: PMC4030042 DOI: 10.1186/1746-6148-10-113] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Accepted: 05/08/2014] [Indexed: 01/08/2023] Open
Abstract
Background Serotonin (5-hydroytryptamine or 5HT) is associated with numerous behavioral and psychological factors and is a biochemical marker of mood. 5HT is involved in the hypothalamic regulation of energy consumption. 5HT controls appetite in the central nerve system (CNS) and stimulates intestinal mobility. There are few studies looking at the role of 5HT and the relationship between peripheral circulating serotonin and obesity. The aim of this study was to find any differences in leptin, adiponectin, and 5HT between lean and obese dogs and to identify correlations among these factors. Results Leptin, triglyceride (TG) and cholesterol levels were higher in the obese group (all p < 0.01). Adiponectin and 5HT levels were higher in the lean group compared to the obese group (p < 0.01). Leptin (r = 0.628, p < 0.01), TG (r = 0.491, p < 0.01) and cholesterol (r = 0.419, p < 0.01) were positively correlated with body condition score (BCS), and adiponectin (r = -0.446, p < 0.01) and 5HT (r = -0.490, p < 0.01) were negatively correlated with BCS. Leptin was negatively correlated with adiponectin (r = -0.294, p < 0.01) and 5HT (r = -0.343, p < 0.01). 5HT was negatively correlated with leptin (r = -0.343, p < 0.01), TG (r = -0.268, p < 0.05) and cholesterol (r = -0.357, p < 0.05). Conclusions 5HT is an important appetite control neurotransmitter, but there are limited studies for 5HT levels related to obesity in dogs. To the best of our knowledge, this is the first study to evaluate peripheral 5HT levels in obese dogs. From this research, we can assume that 5HT may be correlated with canine obesity. Further studies will be needed to further elucidate the role of low serum 5HT levels in canine obesity.
Collapse
|
37
|
Differential influence of the 5-HTTLPR genotype, neuroticism and real-life acute stress exposure on appetite and energy intake. Appetite 2014; 77:83-93. [PMID: 24630938 DOI: 10.1016/j.appet.2014.03.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2013] [Revised: 02/21/2014] [Accepted: 03/03/2014] [Indexed: 12/29/2022]
Abstract
Stress or negative mood often promotes energy intake and overeating. Since the serotonin transporter-linked polymorphic region (5-HTTLPR) is found to mediate stress vulnerability as well as to influence energy intake, this gene may also influence the negative effects of stress exposure on overeating. Moreover, since stress proneness also reflects cognitive stress vulnerability - as often defined by trait neuroticism - this may additionally predispose for stress-induced overeating. In the present study it was investigated whether the 5-HTTLPR genotype interacted with neuroticism on changes in mood, appetite and energy intake following exposure to a real-life academic examination stressor. In a balanced-experimental design, homozygous S-allele and L-allele carriers (N = 94) with the lowest and highest neuroticism scores were selected from a large database of 5-HTTLPR genotyped students. Mood, appetite and energy intake were measured before and after a 2-hour academic examination and compared with a control day. Examination influenced appetite for particular sweet snacks differently depending on 5-HTTLPR genotype and neuroticism. S/S compared with L/L subjects reported greater examination stress, and this was accompanied by a more profound post-stress increase in appetite for sweet snacks. Data also revealed a 5-HTTLPR genotype by trait neuroticism interaction on energy intake, regardless of examination. These results consolidate previous assumptions of 5-HTTLPR involvement in stress vulnerability and suggest 5-HTTLPR and neuroticism may influence stress-induced overeating depending on the type of food available. These findings furthermore link previous findings of increased risk for weight gain in S/S-allele carriers, particularly with high scores on trait neuroticism, to increased energy intake.
Collapse
|
38
|
Roth CL, Hinney A, Schur EA, Elfers CT, Reinehr T. Association analyses for dopamine receptor gene polymorphisms and weight status in a longitudinal analysis in obese children before and after lifestyle intervention. BMC Pediatr 2013; 13:197. [PMID: 24283216 PMCID: PMC4219494 DOI: 10.1186/1471-2431-13-197] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 11/22/2013] [Indexed: 01/09/2023] Open
Abstract
Background Dopamine receptors are involved in midbrain reward circuit activation. Polymorphisms in two dopamine receptor genes, DRD2 and DRD4, have been associated with altered perception of food reward and weight gain. The objective of this study was to determine whether the same risk alleles were associated with overweight/obesity and with lower reduction of overweight after a 1-year lifestyle intervention. Methods In a longitudinal study the association of polymorphisms in DRD2 (rs18000497, risk allele: T, formerly A1 allele at the TaqI A1 polymorphism) and DRD4 (variable number of tandem repeats (VNTR); 48 bp repeat in exon III; risk alleles: 7 repeats or longer: 7R+) was tested on weight loss success following a 1-year lifestyle childhood obesity intervention (OBELDICKS). An additional exploratory cross-sectional case-control study was performed to compare the same DRD polymorphisms in these overweight/obese children and adolescents versus lean adult controls. Subjects were 423 obese and 28 overweight children participating in lifestyle intervention (203 males), age median 12.0 (interquartile range 10.0–13.7) years, body mass index - standard deviation score (BMI-SDS) 2.4 ± 0.5; 583 lean adults (232 males); age median 25.3 (interquartile range 22.5–26.8) years, BMI 19.1 ± 1.9 kg/m2. BMI, BMI-SDS and skinfold thickness measures were assessed at baseline and after 1 year; genotyping was performed for DRD2 risk variant rs1800497 and DRD4 exon III VNTR. Results The DRD2 genotype had a nominal effect on success in the weight loss intervention. The weakest BMI-SDS reduction was in children homozygous for two rs1800497 T-alleles (n = 11) compared to the combined group with zero (n = 308) or one (n = 132) rs1800497 T-allele (-0.08 ± 0.36 vs. -0.28 ± 0.34; p < 0.05). There was no association between the DRD4 VNTR alleles and genotypes and success in the weight loss intervention. No associations of the risk alleles of the DRD2 and DRD4 polymorphisms and obesity were observed in the cross-sectional part of the study. Conclusions We did not find association between polymorphisms in DRD2 and DRD4 genes and weight status. However, obese carriers of two DRD2 rs1800497 T-alleles may be at risk for weak responses to lifestyle interventions aimed at weight reduction. Trial registration Obesity intervention program “Obeldicks” is registered at clinicaltrials.gov (NCT00435734).
Collapse
Affiliation(s)
- Christian L Roth
- Department of Pediatrics, University of Washington, Seattle Children's Research Institute, 1900 Ninth Ave, Seattle, WA 98101, USA.
| | | | | | | | | |
Collapse
|
39
|
Giannaccini G, Betti L, Palego L, Marsili A, Santini F, Pelosini C, Fabbrini L, Schmid L, Giusti L, Maffei M, Lanza M, Cristofaro M, Baroni S, Mauri M, Vitti P, Fierabracci P, Lucacchini A. The expression of platelet serotonin transporter (SERT) in human obesity. BMC Neurosci 2013; 14:128. [PMID: 24138674 PMCID: PMC4016247 DOI: 10.1186/1471-2202-14-128] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2013] [Accepted: 10/09/2013] [Indexed: 01/20/2023] Open
Abstract
Background Serotonin (5-HT) is a well-known modulator of eating behavior. However, the molecular mechanisms linking its action to body weight balance have been only partially elucidated. Since platelets are a suitable peripheral model to study 5-HT transport, metabolism and release, we herein evaluated the expression of the platelet 5-HT re-uptake system (SERT) by [3H]-paroxetine binding assay. A cohort of 114 unrelated individuals (34 males, 80 females; age, mean ± SD: 38.57 ± 12.47 years) without major psychiatric disorders, was recruited following a naturalistic design regarding age or gender and classified accordingly to their body mass index (BMI). Subjects were divided into 5 groups: normal-weight (NW), overweight (OW) and grade I-III obese (OB) individuals. For gender analyses, data were transformed into [3H]-paroxetine density (Bmax)/BMI ratios to overcome both the disparity of women vs. men number and anthropometric differences between sexes. Results [3H]-paroxetine Bmax (SERT density, fmol/mg proteins) was reduced in platelet membranes of grade II (p < 0.01) and III (p < 0.001) obese subjects vs. controls and in overweight subjects (p < 0.05) vs. grade III obese individuals. Considering all patients together, a strong negative correlation between Bmax and BMI (r = −0.449; P < 0.0001) was demonstrated. Conversely, [3H]-paroxetine KD (dissociation constant, nM) did not differ among groups. No gender-related variation concerning Bmax/BMI ratios was observed in this cohort of subjects. Conclusions The down-regulation of SERT in platelet membranes of severe human obesity (BMI > 35 Kg/m2) confirms the involvement of 5-HT system in body weight gain. Moreover, this findings may help to elucidate those monoamine-endocrine networks acting on fat storage, adipocyte signaling and energy balance. Targeting 5-HT/5-HT-related markers will possibly uncover the existence of human obesity subtypes.
Collapse
Affiliation(s)
- Gino Giannaccini
- Department of Pharmacy, University of Pisa, via Bonanno 6, Pisa 56126-I, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Shinozaki G, Kumar Y, Rosen BH, Rundell JR, Mrazek DA, Kung S. "Diminished" association between the serotonin transporter linked polymorphism (5HTTLPR) and body mass index in a large psychiatric sample. J Affect Disord 2013; 151:397-400. [PMID: 23838390 DOI: 10.1016/j.jad.2013.06.021] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 06/14/2013] [Indexed: 01/05/2023]
Abstract
BACKGROUND The role of the promoter polymorphism (5HTTLPR) of the serotonin transporter gene (SLC6A4) in psychiatric illnesses has been studied extensively. Serotonergic function also regulates many central nervous system, including appetite and feeding behaviors. The 5HTTLPR short allele was found to be associated with increased body mass index and obesity risk among the general population. No data is available to support generalizability of such association among psychiatric population. METHODS We examined the relationship between BMI and the 5HTTLPR genotype in a large sample of 1831 psychiatric patients at Mayo Clinic, Rochester, Minnesota, using a retrospective chart review. RESULTS Average BMI among groups with the short/short (28.29 ± 7.27 kg/m(2)), the short/long (28.07 ± 6.45 kg/m(2)) and the long/long (28.15 ± 7.51 kg/m(2)) genotypes of 5HTTLPR were not statistically different. This negative association persisted even with the sub-analysis of the Caucasians. However, we observed an increased rate of obesity among our psychiatric patient sample compared to the general population of Minnesota (36.6% versus 27.6%, p=0.0001 for males, 30.3% versus 24.4%, p=0.0001 for females). Also, sub-analysis showed female inpatients to have a significantly higher average BMI than outpatients (28.64 ± 8.08 kg/m(2) versus 27.13 ± 6.92 kg/m(2), p=0.026). This confirmed a significant association between mental health disorder and BMI. LIMITATIONS Retrospective study design with limited control for potential confounders. CONCLUSIONS In this large sample of psychiatric patients we found no significant association between 5HTTLPR genotype and BMI, which is different from the case with general population reported in the literature.
Collapse
Affiliation(s)
- Gen Shinozaki
- Sioux Falls VA Health Care System, Sioux Falls, SD, USA.
| | | | | | | | | | | |
Collapse
|
41
|
Early community context, genes, and youth body mass index trajectories: an investigation of gene-community interplay over early life course. J Adolesc Health 2013; 53:328-34. [PMID: 23711506 DOI: 10.1016/j.jadohealth.2013.04.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2013] [Revised: 03/30/2013] [Accepted: 04/02/2013] [Indexed: 01/06/2023]
Abstract
PURPOSE To investigate additive and interactive influences of community adversity and cumulative genetic sensitivity on youth body mass index (BMI) trajectories over adolescence and young adulthood. METHODS We used latent growth curve modeling to examine BMI trajectories over three waves (1995, 2001, and 2008) of the National Longitudinal Study of Adolescent Health (n = 14,563). We measured genetic sensitivity by a cumulative index of genes associated with serotonin and dopamine functions. RESULTS Community adversity was positively associated with the initial level and rate of change in BMI trajectories over time. Adolescents experiencing community adversity had a higher BMI at Wave 1 and gained weight more quickly than those who did not live in adverse communities. Community adversity interacted with cumulative genetic sensitivity to explain variation in the rate of change in BMI trajectories. The influence of community adversity was greater for those with more sensitivity alleles than those with fewer sensitivity alleles. Gender, race/ethnicity, and family contexts were also associated with youth BMI trajectories. CONCLUSIONS Community adversity in early adolescence, and its interaction with genes, has far-reaching consequences, including the rate of change in BMI trajectories extending into adulthood. This work has practical implications for future intervention/prevention programs.
Collapse
|
42
|
Clum GA, Rice JC, Broussard M, Johnson CC, Webber LS. Associations between depressive symptoms, self-efficacy, eating styles, exercise and body mass index in women. J Behav Med 2013; 37:577-86. [DOI: 10.1007/s10865-013-9526-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Accepted: 07/16/2013] [Indexed: 11/28/2022]
|
43
|
Abstract
OBJECTIVE A functional promoter polymorphism in the monoamine oxidase A (MAOA) gene has been implicated in neuropsychiatric disorders and also moderates the association between early-life stress and mental disorders, which often co-occur with cardiovascular disease. No study has examined the relationship between MAOA genotype, childhood trauma, and subclinical atherosclerosis. The objective of this investigation was to examine whether childhood trauma moderates the association between MAOA genotype and subclinical atherosclerosis. METHODS A sample including 289 middle-aged male twin pairs was studied. Subclinical atherosclerosis was assessed by brachial flow-mediated dilation (FMD) using ultrasound. Childhood trauma, before age 18 years, was measured with the Early Trauma Inventory and included physical, emotional, and sexual abuse as well as general trauma. Generalized estimating equation models were used to test the main and interactive effects of the MAOA genotype and each domain of childhood trauma on FMD, adjusting for known risk factors. RESULTS General trauma was the most prevalent childhood trauma (28.4%), followed by physical abuse (25.0%), emotional abuse (19.4%), and sexual abuse (11.6%). MAOA genotype was not associated with any domain of childhood trauma. There was no significant evidence for a main effect for the MAOA genotype (β = .02, p = .82) or childhood trauma (.005 < β < .10, p > .54) FMD. However, a significant interaction was observed between MAOA genotype and physical (β interaction = .37, p = .026) or emotional abuse (β interaction = .43, p = .025) on subclinical atherosclerosis. CONCLUSIONS Childhood trauma modulates the impact of MAOA variant on subclinical atherosclerosis, independent of traditional cardiovascular risk factors.
Collapse
|
44
|
Goldfield GS, Dowler LM, Walker M, Cameron JD, Ferraro ZM, Doucet E, Adamo KB. Are dopamine-related genotypes risk factors for excessive gestational weight gain? Int J Womens Health 2013; 5:253-9. [PMID: 23723720 PMCID: PMC3665497 DOI: 10.2147/ijwh.s43935] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Background Excessive gestational weight gain is associated with postpartum weight retention and downstream child obesity. Dopamine plays a critical role in the regulation of energy intake and body weight. The purpose of this study was to examine the relationship between excessive gestational weight gain and dopamine pathway-related polymorphisms, namely the variable nucleotide tandem repeat in the 3′untranslated region (UTR) region of the SLC6A3 (DAT-1) dopamine transporter gene and the 30-base pair variable nucleotide tandem repeat polymorphism of the 5′UTR of the monoamine oxidase-A (MAO-A) gene. Methods Ninety-three women of mean age 31.7 ± 4.2 years were recruited from the Ottawa and Kingston birth cohort and assessed at 12–20 weeks’ gestation. Mean body mass index was 22.7 ± 2.5 kg/m2. Excessive gestational weight gain was defined according to the 2009 Institute of Medicine guidelines based on body mass index. Genotype analyses were performed using polymerase chain reaction and agarose gel electrophoresis. Results There was no relationship between the prevalence or magnitude of excessive gestational weight gain among women with the 3′ UTR single nucleotide polymorphism of the DAT-1 gene. However, 70% (19 of 27) of women carrying the MAO-A 4/4 (high activity) allele exceeded recommendations for gestational weight gain compared with 48% (32 of 60) of those with the pooled 3/3, 3/4, and 3/3.5 (low activity) alleles (P < 0.05). Similarly, those with the MAO-A 4/4 allele had significantly greater gestational weight gain than those with the 3/3, 3/4, or 3/3.5 pooled genotypes (19.3 ± 4.1 versus 17.0 ± 5.0 kg, P = 0.03). Conclusion Carriers of the 4/4 variants of the MAO-A gene may be at increased risk for excessive gestational weight gain.
Collapse
Affiliation(s)
- Gary S Goldfield
- Healthy Active Living and Obesity Research Group, Children's Hospital of Eastern Ontario Research Institute, Ottawa, ON, Canada ; Department of Paediatrics, University of Ottawa, Ottawa, ON, Canada ; School of Human Kinetics, University of Ottawa, Ottawa, ON, Canada ; School of Psychology, University of Ottawa, Ottawa, ON, Canada ; Department of Psychology, Carleton University, Ottawa, ON, Canada
| | | | | | | | | | | | | |
Collapse
|
45
|
Abstract
Several lines of research have found that genes in the serotonergic system may cause susceptibility to eating disorders (EDs). In particular, functional polymorphisms of the serotonin transporter gene (5-HTT) have been suspected to play a role in the pathogenesis of eating disorders. Several studies have examined the association between the 5-HTTLPR polymorphism and bulimia nervosa (BN). The results of these investigations have been unclear. The aims of this meta-analysis were to clarify the association between BN and 5-HTTLPR using statistical models not used by previous meta-analyses, and extend upon previous meta-analyses by including new samples. PsychINFO, ISI, and PubMed databases were searched for studies published up to May 2011. Ultimately, six case-control samples were included. Data were pooled using dominant and additive models. Both models showed a nonsignificant association between the 5-HTTLPR polymorphism and BN. However, this does not detract from recent research suggesting that the 5-HTTLPR polymorphism may be responsible for the phenotypic variability in the psychopathological symptoms observed in patients with BN. Future research should examine the association of BN with 5-HTTLPR using the recently proposed triallelic model.
Collapse
|
46
|
Chen C, Chen W, Chen C, Moyzis R, He Q, Lei X, Li J, Wang Y, Liu B, Xiu D, Zhu B, Dong Q. Genetic variations in the serotoninergic system contribute to body-mass index in Chinese adolescents. PLoS One 2013; 8:e58717. [PMID: 23554917 PMCID: PMC3598805 DOI: 10.1371/journal.pone.0058717] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2012] [Accepted: 02/05/2013] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE Obesity has become a worldwide health problem in the past decades. Human and animal studies have implicated serotonin in appetite regulation, and behavior genetic studies have shown that body mass index (BMI) has a strong genetic component. However, the roles of genes related to the serotoninergic (5-hydroxytryptamine,5-HT) system in obesity/BMI are not well understood, especially in Chinese subjects. SUBJECTS AND DESIGN With a sample of 478 healthy Chinese volunteers, this study investigated the relation between BMI and genetic variations of the serotoninergic system as characterized by 136 representative polymorphisms. We used a system-level approach to identify SNPs associated with BMI, then estimated their overall contribution to BMI by multiple regression and verified it by permutation. RESULTS We identified 12 SNPs that made statistically significant contributions to BMI. After controlling for gender and age, four of these SNPs accounted for 7.7% additional variance of BMI. Permutation analysis showed that the probability of obtaining these findings by chance was low (p = 0.015, permuted for 1000 times). CONCLUSION These results showed that genetic variations in the serotoninergic system made a moderate contribution to individual differences in BMI among a healthy Chinese sample, suggesting that a similar approach can be used to study obesity.
Collapse
Affiliation(s)
- Chunhui Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Wen Chen
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Chuansheng Chen
- Department of Psychology and Social Behavior, University of California Irvine, Irvine, California, United States of America
| | - Robert Moyzis
- Department of Biological Chemistry and Institute of Genomics and Bioinformatics, University of California Irvine, Irvine, California, United States of America
| | - Qinghua He
- Department of Psychology, University of Southern California, Los Angeles, California, United States of America
| | - Xuemei Lei
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Jin Li
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Yunxin Wang
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Bin Liu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Daiming Xiu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Bi Zhu
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| | - Qi Dong
- State Key Laboratory of Cognitive Neuroscience and Learning, Beijing Normal University, Beijing, China
| |
Collapse
|
47
|
Abstract
Longevity is influenced by genetic and environmental factors. The brain's dopamine system may be particularly relevant, since it modulates traits (e.g., sensitivity to reward, incentive motivation, sustained effort) that impact behavioral responses to the environment. In particular, the dopamine D4 receptor (DRD4) has been shown to moderate the impact of environments on behavior and health. We tested the hypothesis that the DRD4 gene influences longevity and that its impact is mediated through environmental effects. Surviving participants of a 30-year-old population-based health survey (N = 310; age range, 90-109 years; the 90+ Study) were genotyped/resequenced at the DRD4 gene and compared with a European ancestry-matched younger population (N = 2902; age range, 7-45 years). We found that the oldest-old population had a 66% increase in individuals carrying the DRD4 7R allele relative to the younger sample (p = 3.5 × 10(-9)), and that this genotype was strongly correlated with increased levels of physical activity. Consistent with these results, DRD4 knock-out mice, when compared with wild-type and heterozygous mice, displayed a 7-9.7% decrease in lifespan, reduced spontaneous locomotor activity, and no lifespan increase when reared in an enriched environment. These results support the hypothesis that DRD4 gene variants contribute to longevity in humans and in mice, and suggest that this effect is mediated by shaping behavioral responses to the environment.
Collapse
|
48
|
Markus CR, Capello AE. Contribution of the 5-HTTLPR gene by neuroticism on weight gain in male and female participants. Psychiatr Genet 2012; 22:279-85. [DOI: 10.1097/ypg.0b013e328358632a] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
49
|
Abstract
AbstractThe science of genetics is undergoing a paradigm shift. Recent discoveries, including the activity of retrotransposons, the extent of copy number variations, somatic and chromosomal mosaicism, and the nature of the epigenome as a regulator of DNA expressivity, are challenging a series of dogmas concerning the nature of the genome and the relationship between genotype and phenotype. According to three widely held dogmas, DNA is the unchanging template of heredity, is identical in all the cells and tissues of the body, and is the sole agent of inheritance. Rather than being an unchanging template, DNA appears subject to a good deal of environmentally induced change. Instead of identical DNA in all the cells of the body, somatic mosaicism appears to be the normal human condition. And DNA can no longer be considered the sole agent of inheritance. We now know that the epigenome, which regulates gene expressivity, can be inherited via the germline. These developments are particularly significant for behavior genetics for at least three reasons: First, epigenetic regulation, DNA variability, and somatic mosaicism appear to be particularly prevalent in the human brain and probably are involved in much of human behavior; second, they have important implications for the validity of heritability and gene association studies, the methodologies that largely define the discipline of behavior genetics; and third, they appear to play a critical role in development during the perinatal period and, in particular, in enabling phenotypic plasticity in offspring. I examine one of the central claims to emerge from the use of heritability studies in the behavioral sciences, the principle of minimal shared maternal effects, in light of the growing awareness that the maternal perinatal environment is a critical venue for the exercise of adaptive phenotypic plasticity. This consideration has important implications for both developmental and evolutionary biology.
Collapse
|
50
|
van de Giessen E, Hesse S, Caan MWA, Zientek F, Dickson JC, Tossici-Bolt L, Sera T, Asenbaum S, Guignard R, Akdemir UO, Knudsen GM, Nobili F, Pagani M, Vander Borght T, Van Laere K, Varrone A, Tatsch K, Booij J, Sabri O. No association between striatal dopamine transporter binding and body mass index: a multi-center European study in healthy volunteers. Neuroimage 2012; 64:61-7. [PMID: 22982354 DOI: 10.1016/j.neuroimage.2012.09.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2012] [Revised: 08/30/2012] [Accepted: 09/02/2012] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION Dopamine is one among several neurotransmitters that regulate food intake and overeating. Thus, it has been linked to the pathophysiology of obesity and high body mass index (BMI). Striatal dopamine D(2) receptor availability is lower in obesity and there are indications that striatal dopamine transporter (DAT) availability is also decreased. In this study, we tested whether BMI and striatal DAT availability are associated. METHODS The study included 123 healthy individuals from a large European multi-center database. They had a BMI range of 18.2-41.1 kg/m(2) and were scanned using [(123)I]FP-CIT SPECT imaging. Scans were analyzed with both region-of-interest and voxel-based analysis to determine the binding potential for DAT availability in the caudate nucleus and putamen. A direct relation between BMI and DAT availability was assessed and groups with high and low BMI were compared for DAT availability. RESULTS No association between BMI and striatal DAT availability was found. CONCLUSION The lack of an association between BMI and striatal DAT availability suggests that the regulation of striatal synaptic dopamine levels by DAT plays no or a limited role in the pathophysiology of overweight and obesity.
Collapse
Affiliation(s)
- Elsmarieke van de Giessen
- Department of Nuclear Medicine, Academic Medical Center University of Amsterdam, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|