1
|
Fonseca PAS, Suárez-Vega A, Arranz JJ, Gutiérrez-Gil B. Integration of selective sweeps across the sheep genome: understanding the relationship between production and adaptation traits. Genet Sel Evol 2024; 56:40. [PMID: 38773423 PMCID: PMC11106937 DOI: 10.1186/s12711-024-00910-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 05/07/2024] [Indexed: 05/23/2024] Open
Abstract
BACKGROUND Livestock populations are under constant selective pressure for higher productivity levels for different selective purposes. This pressure results in the selection of animals with unique adaptive and production traits. The study of genomic regions associated with these unique characteristics has the potential to improve biological knowledge regarding the adaptive process and how it is connected to production levels and resilience, which is the ability of an animal to adapt to stress or an imbalance in homeostasis. Sheep is a species that has been subjected to several natural and artificial selective pressures during its history, resulting in a highly specialized species for production and adaptation to challenging environments. Here, the data from multiple studies that aim at mapping selective sweeps across the sheep genome associated with production and adaptation traits were integrated to identify confirmed selective sweeps (CSS). RESULTS In total, 37 studies were used to identify 518 CSS across the sheep genome, which were classified as production (147 prodCSS) and adaptation (219 adapCSS) CSS based on the frequency of each type of associated study. The genes within the CSS were associated with relevant biological processes for adaptation and production. For example, for adapCSS, the associated genes were related to the control of seasonality, circadian rhythm, and thermoregulation. On the other hand, genes associated with prodCSS were related to the control of feeding behaviour, reproduction, and cellular differentiation. In addition, genes harbouring both prodCSS and adapCSS showed an interesting association with lipid metabolism, suggesting a potential role of this process in the regulation of pleiotropic effects between these classes of traits. CONCLUSIONS The findings of this study contribute to a deeper understanding of the genetic link between productivity and adaptability in sheep breeds. This information may provide insights into the genetic mechanisms that underlie undesirable genetic correlations between these two groups of traits and pave the way for a better understanding of resilience as a positive ability to respond to environmental stressors, where the negative effects on production level are minimized.
Collapse
Affiliation(s)
- Pablo A S Fonseca
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana S/N, 24071, León, Spain
| | - Aroa Suárez-Vega
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana S/N, 24071, León, Spain
| | - Juan J Arranz
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana S/N, 24071, León, Spain
| | - Beatriz Gutiérrez-Gil
- Departamento de Producción Animal, Facultad de Veterinaria, Universidad de León, Campus de Vegazana S/N, 24071, León, Spain.
| |
Collapse
|
2
|
Primak A, Bozov K, Rubina K, Dzhauari S, Neyfeld E, Illarionova M, Semina E, Sheleg D, Tkachuk V, Karagyaur M. Morphogenetic theory of mental and cognitive disorders: the role of neurotrophic and guidance molecules. Front Mol Neurosci 2024; 17:1361764. [PMID: 38646100 PMCID: PMC11027769 DOI: 10.3389/fnmol.2024.1361764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Accepted: 03/04/2024] [Indexed: 04/23/2024] Open
Abstract
Mental illness and cognitive disorders represent a serious problem for the modern society. Many studies indicate that mental disorders are polygenic and that impaired brain development may lay the ground for their manifestation. Neural tissue development is a complex and multistage process that involves a large number of distant and contact molecules. In this review, we have considered the key steps of brain morphogenesis, and the major molecule families involved in these process. The review provides many indications of the important contribution of the brain development process and correct functioning of certain genes to human mental health. To our knowledge, this comprehensive review is one of the first in this field. We suppose that this review may be useful to novice researchers and clinicians wishing to navigate the field.
Collapse
Affiliation(s)
- Alexandra Primak
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kirill Bozov
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Kseniya Rubina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Stalik Dzhauari
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Elena Neyfeld
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Federal State Budgetary Educational Institution of the Higher Education “A.I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Maria Illarionova
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Ekaterina Semina
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
| | - Dmitriy Sheleg
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Federal State Budgetary Educational Institution of the Higher Education “A.I. Yevdokimov Moscow State University of Medicine and Dentistry” of the Ministry of Healthcare of the Russian Federation, Moscow, Russia
| | - Vsevolod Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| | - Maxim Karagyaur
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia
- Institute for Regenerative Medicine, Medical Research and Education Center, Lomonosov Moscow State University, Moscow, Russia
| |
Collapse
|
3
|
Underlying Mechanisms Involved in Gambling Disorder Severity: A Pathway Analysis Considering Genetic, Psychosocial, and Clinical Variables. Nutrients 2023; 15:nu15020418. [PMID: 36678289 PMCID: PMC9864492 DOI: 10.3390/nu15020418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 12/29/2022] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Gambling Disorder (GD) has a complex etiology that involves biological and environmental aspects. From a genetic perspective, neurotrophic factors (NTFs) polymorphisms have been associated with the risk of developing GD. The aim of this study was to assess the underlying mechanisms implicated in GD severity by considering the direct and mediational relationship between different variables including genetic, psychological, socio-demographic, and clinical factors. To do so, we used genetic variants that were significantly associated with an increased risk for GD and evaluated its relationship with GD severity through pathway analysis. We found that the interaction between these genetic variants and other different biopsychological features predicted a higher severity of GD. On the one hand, the presence of haplotype block 2, interrelated with haplotype block 3, was linked to a more dysfunctional personality profile and a worse psychopathological state, which, in turn, had a direct link with GD severity. On the other hand, having rs3763614 predicted higher general psychopathology and therefore, higher GD severity. The current study described the presence of complex interactions between biopsychosocial variables previously associated with the etiopathogenesis and severity of GD, while also supporting the involvement of genetic variants from the NTF family.
Collapse
|
4
|
Chitre AS, Hebda-Bauer EK, Blandino P, Bimschleger H, Nguyen KM, Maras P, Li F, Ozel AB, Pan Y, Polesskaya O, Cheng R, Flagel SB, Watson SJ, Li J, Akil H, Palmer AA. Genome-wide association study in a rat model of temperament identifies multiple loci for exploratory locomotion and anxiety-like traits. Front Genet 2023; 13:1003074. [PMID: 36712851 PMCID: PMC9873817 DOI: 10.3389/fgene.2022.1003074] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 10/20/2022] [Indexed: 01/12/2023] Open
Abstract
Common genetic factors likely contribute to multiple psychiatric diseases including mood and substance use disorders. Certain stable, heritable traits reflecting temperament, termed externalizing or internalizing, play a large role in modulating vulnerability to these disorders. To model these heritable tendencies, we selectively bred rats for high and low exploration in a novel environment [bred High Responders (bHR) vs. Low Responders (bLR)]. To identify genes underlying the response to selection, we phenotyped and genotyped 538 rats from an F2 cross between bHR and bLR. Several behavioral traits show high heritability, including the selection trait: exploratory locomotion (EL) in a novel environment. There were significant phenotypic and genetic correlations between tests that capture facets of EL and anxiety. There were also correlations with Pavlovian conditioned approach (PavCA) behavior despite the lower heritability of that trait. Ten significant and conditionally independent loci for six behavioral traits were identified. Five of the six traits reflect different facets of EL that were captured by three behavioral tests. Distance traveled measures from the open field and the elevated plus maze map onto different loci, thus may represent different aspects of novelty-induced locomotor activity. The sixth behavioral trait, number of fecal boli, is the only anxiety-related trait mapping to a significant locus on chromosome 18 within which the Pik3c3 gene is located. There were no significant loci for PavCA. We identified a missense variant in the Plekhf1 gene on the chromosome 1:95 Mb QTL and Fancf and Gas2 as potential candidate genes that may drive the chromosome 1:107 Mb QTL for EL traits. The identification of a locomotor activity-related QTL on chromosome 7 encompassing the Pkhd1l1 and Trhr genes is consistent with our previous finding of these genes being differentially expressed in the hippocampus of bHR vs. bLR rats. The strong heritability coupled with identification of several loci associated with exploratory locomotion and emotionality provide compelling support for this selectively bred rat model in discovering relatively large effect causal variants tied to elements of internalizing and externalizing behaviors inherent to psychiatric and substance use disorders.
Collapse
Affiliation(s)
- Apurva S. Chitre
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Elaine K. Hebda-Bauer
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Peter Blandino
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Hannah Bimschleger
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Khai-Minh Nguyen
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Pamela Maras
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Fei Li
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - A. Bilge Ozel
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Yanchao Pan
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Oksana Polesskaya
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Riyan Cheng
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States
| | - Shelly B. Flagel
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Stanley J. Watson
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Jun Li
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, United States
| | - Huda Akil
- Michigan Neuroscience Institute, University of Michigan, Ann Arbor, MI, United States
| | - Abraham A. Palmer
- Department of Psychiatry, University of California San Diego, La Jolla, CA, United States,Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, United States,*Correspondence: Abraham A. Palmer,
| |
Collapse
|
5
|
Solé-Morata N, Baenas I, Etxandi M, Granero R, Forcales SV, Gené M, Barrot C, Gómez-Peña M, Menchón JM, Ramoz N, Gorwood P, Fernández-Aranda F, Jiménez-Murcia S. The role of neurotrophin genes involved in the vulnerability to gambling disorder. Sci Rep 2022; 12:6925. [PMID: 35484167 PMCID: PMC9051155 DOI: 10.1038/s41598-022-10391-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 04/07/2022] [Indexed: 01/16/2023] Open
Abstract
Evidence about the involvement of genetic factors in the development of gambling disorder (GD) has been assessed. Among studies assessing heritability and biological vulnerability for GD, neurotrophin (NTF) genes have emerged as promising targets, since a growing literature showed a possible link between NTF and addiction-related disorders. Thus, we aimed to explore the role of NTF genes and GD with the hypothesis that some NTF gene polymorphisms could constitute biological risk factors. The sample included 166 patients with GD and 191 healthy controls. 36 single nucleotide polymorphisms (SNPs) from NTFs (NGF, NGFR, NTRK1, BDNF, NTRK2, NTF3, NTRK3, NTF4, CNTF and CNTFR) were selected and genotyped. Linkage disequilibrium (LD) and haplotype constructions were analyzed, in relationship with the presence of GD. Finally, regulatory elements overlapping the identified SNPs variants associated with GD were searched. The between groups comparisons of allele frequencies indicated that 6 SNPs were potentially associated with GD. Single and multiple-marker analyses showed a strong association between both NTF3 and NTRK2 genes, and GD. The present study supports the involvement of the NTF family in the aetiopathogenesis of GD. An altered cross-regulation of different NTF members signalling pathways might be considered as a biological vulnerability factor for GD.
Collapse
Affiliation(s)
- Neus Solé-Morata
- Department of Psychiatry, Bellvitge University Hospital, c/Feixa Llarga S/N, Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Isabel Baenas
- Department of Psychiatry, Bellvitge University Hospital, c/Feixa Llarga S/N, Hospitalet de Llobregat, 08907, Barcelona, Spain.,Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain.,Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain
| | - Mikel Etxandi
- Department of Psychiatry, Bellvitge University Hospital, c/Feixa Llarga S/N, Hospitalet de Llobregat, 08907, Barcelona, Spain
| | - Roser Granero
- Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain.,Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain.,Department of Psychobiology and Methodology, Autonomous University of Barcelona, Bellaterra, Spain
| | - Sonia V Forcales
- Serra Húnter Programme, Department of Pathology and Experimental Therapeutics, Faculty of Medicine and Health Sciences, University of Barcelona, Hospitalet de Llobregat, 08907, Spain
| | - Manel Gené
- Genetic Lab, Forensic and Legal Medicine Unit, Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Carme Barrot
- Genetic Lab, Forensic and Legal Medicine Unit, Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Barcelona, Spain
| | - Mónica Gómez-Peña
- Department of Psychiatry, Bellvitge University Hospital, c/Feixa Llarga S/N, Hospitalet de Llobregat, 08907, Barcelona, Spain.,Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain
| | - José M Menchón
- Department of Psychiatry, Bellvitge University Hospital, c/Feixa Llarga S/N, Hospitalet de Llobregat, 08907, Barcelona, Spain.,Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Hospitalet del Llobregat, Spain.,Ciber of Mental Health (CIBERSAM), Instituto de Salud Carlos III, Barcelona, Spain.,Psychiatry and Mental Health Group, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet del Llobregat, Spain
| | - Nicolás Ramoz
- Psychiatry and Mental Health Group, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet del Llobregat, Spain.,Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Team Vulnerability of Psychiatric and Addictive Disorders, Université de Paris, 75014, Paris, France
| | - Philip Gorwood
- Psychiatry and Mental Health Group, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet del Llobregat, Spain.,Institute of Psychiatry and Neuroscience of Paris (IPNP), INSERM U1266, Team Vulnerability of Psychiatric and Addictive Disorders, Université de Paris, 75014, Paris, France
| | - Fernando Fernández-Aranda
- Department of Psychiatry, Bellvitge University Hospital, c/Feixa Llarga S/N, Hospitalet de Llobregat, 08907, Barcelona, Spain.,Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain.,Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain.,Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Hospitalet del Llobregat, Spain
| | - Susana Jiménez-Murcia
- Department of Psychiatry, Bellvitge University Hospital, c/Feixa Llarga S/N, Hospitalet de Llobregat, 08907, Barcelona, Spain. .,Psychoneurobiology of Eating and Addictive Behaviors Group, Neurosciences Program, Bellvitge Biomedical Research Institute (IDIBELL), Hospitalet de Llobregat, Spain. .,Ciber Physiopathology of Obesity and Nutrition (CIBERObn), Instituto de Salud Carlos III, Barcelona, Spain. .,Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona, Hospitalet del Llobregat, Spain.
| |
Collapse
|
6
|
Fontana L, Garzia E, Marfia G, Galiano V, Miozzo M. Epigenetics of functional hypothalamic amenorrhea. Front Endocrinol (Lausanne) 2022; 13:953431. [PMID: 36034425 PMCID: PMC9415998 DOI: 10.3389/fendo.2022.953431] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 07/25/2022] [Indexed: 11/13/2022] Open
Abstract
Functional hypothalamic amenorrhea (FHA) is a temporary infertility characterized by the suppression of the hypothalamic-pituitary-gonadal (HPG) axis, induced by the inhibition of the hypothalamic pulsatile secretion of the gonadotropin-releasing hormone (GnRH), in the presence of stressors, including eating disorders, excessive exercise, and psychological distress. Although the stressful factors that may lead to FHA are well-established, little is known about the inter-individual variability in response to stress and the consequent inhibition of the HPG axis. Not all women, indeed, manifest FHA in presence of stressful conditions. Recent studies highlighted a genetic contribution to FHA. Rare or polymorphic variants in genes that control the development and/or function of GnRH neurons may contribute, indeed, to the adaptability of the reproductive axis to stress factors. Also epigenetic changes have been associated with different pathways involved in the HPG axis and therefore, take part in FHA and confer a personal predisposition to anovulation consequent to a stressful event, or represent biological markers of response to stress. This review summarizes recent advances in the identification of the contribution of (epi)genetics to FHA and to long-term complications of functional amenorrhea, and reports insights into the involvement of additional genetic loci in FHA development on the bases of the clinical and molecular overlap with other gynecological and/or psychological conditions. Finally, we describe the promising application of induced pluripotent stem cells (iPSCs) as a new approach to investigate the molecular pathways involved in FHA.
Collapse
Affiliation(s)
- L. Fontana
- Medical Genetics, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
- Unit of Medical Genetics, ASST Santi Paolo e Carlo, Milan, Italy
| | - E. Garzia
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
- Aerospace Medicine Institute “A. Mosso”, Italian Air Force, Milan, Italy
| | - G. Marfia
- Aerospace Medicine Institute “A. Mosso”, Italian Air Force, Milan, Italy
- Laboratory of Experimental Neurosurgery and Cell Therapy, Neurosurgery Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - V. Galiano
- Reproductive Medicine Unit, Department of Mother and Child, San Paolo Hospital, ASST Santi Paolo e Carlo, Milan, Italy
| | - M. Miozzo
- Medical Genetics, Department of Health Sciences, Università degli Studi di Milano, Milan, Italy
- Unit of Medical Genetics, ASST Santi Paolo e Carlo, Milan, Italy
- *Correspondence: M. Miozzo,
| |
Collapse
|
7
|
Ahamed SK, Barek MA, Roy UK, Kouser M, Reza MS, Mannan AB, Alam MA, Uddin SMN. A review on association and correlation of genetic variants with eating disorders and obesity. FUTURE JOURNAL OF PHARMACEUTICAL SCIENCES 2021. [DOI: 10.1186/s43094-021-00250-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Now, eating disorders and obesity and their correlations are danger signal in worldwide which is caused by multifactor and associated with significant mortality and morbidity.
Main body
Every aspect of a patient’s life is influenced by eating disorders and obesity and their correlations. Due to frequent seeing of obese patients, eating disorders have been included in the review as they can sometimes be associated with obesity. However, it should be noted that most patients having eating disorder are at risk to be obese or overweight. This research explores the risk factors for the two disorders, as well as the assessment of medical complications and treatment recommendations for the disorders. In these two disorders, there is also a correlation. The essential consideration is that eating disorders are impulse-control disorders which are similar to addictive behaviors in some aspects. So it is a crying need to treat a patient with obesity and eating disorders simultaneously to ensure success. Genome-wide association studies (GWASs) have increased our knowledge of the pathophysiology of eating disorders (EDs) and obesity and their correlation.
Conclusion
This review enlightens on the summary of eating disorder, obesity, genotypic traits, molecular relations, interaction, correlation, and effect of eating disorder and obesity which outline potential future directions and clinical implications for patients with EDs and obesity.
Collapse
|
8
|
Barría A, Benzie JAH, Houston RD, De Koning DJ, de Verdal H. Genomic Selection and Genome-wide Association Study for Feed-Efficiency Traits in a Farmed Nile Tilapia ( Oreochromis niloticus) Population. Front Genet 2021; 12:737906. [PMID: 34616434 PMCID: PMC8488396 DOI: 10.3389/fgene.2021.737906] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 08/31/2021] [Indexed: 11/24/2022] Open
Abstract
Nile tilapia is a key aquaculture species with one of the highest production volumes globally. Genetic improvement of feed efficiency via selective breeding is an important goal, and genomic selection may expedite this process. The aims of this study were to 1) dissect the genetic architecture of feed-efficiency traits in a Nile tilapia breeding population, 2) map the genomic regions associated with these traits and identify candidate genes, 3) evaluate the accuracy of breeding value prediction using genomic data, and 4) assess the impact of the genetic marker density on genomic prediction accuracies. Using an experimental video recording trial, feed conversion ratio (FCR), body weight gain (BWG), residual feed intake (RFI) and feed intake (FI) traits were recorded in 40 full-sibling families from the GIFT (Genetically Improved Farmed Tilapia) Nile tilapia breeding population. Fish were genotyped with a ThermoFisher Axiom 65 K Nile tilapia SNP array. Significant heritabilities, ranging from 0.12 to 0.22, were estimated for all the assessed traits using the genomic relationship matrix. A negative but favourable genetic correlation was found between BWG and the feed-efficiency related traits; -0.60 and -0.63 for FCR and RFI, respectively. While the genome-wide association analyses suggested a polygenic genetic architecture for all the measured traits, there were significant QTL identified for BWG and FI on chromosomes seven and five respectively. Candidate genes previously found to be associated with feed-efficiency traits were located in these QTL regions, including ntrk3a, ghrh and eif4e3. The accuracy of breeding value prediction using the genomic data was up to 34% higher than using pedigree records. A SNP density of approximately 5,000 SNPs was sufficient to achieve similar prediction accuracy as the full genotype data set. Our results highlight the potential of genomic selection to improve feed efficiency traits in Nile tilapia breeding programmes.
Collapse
Affiliation(s)
- Agustin Barría
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Easter Bush, Midlothian, United Kingdom
| | - John A. H. Benzie
- WorldFish, Bayan Lepas, Malaysia
- School of Biological Earth and Environmental Sciences, University College Cork, Cork, Ireland
| | - Ross D. Houston
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh Easter Bush, Midlothian, United Kingdom
| | - Dirk-Jan De Koning
- Department of Animal Breeding and Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Hugues de Verdal
- CIRAD, UMR ISEM, Montpellier, France
- ISEM, Univ Montpellier, CNRS, EPHE, IRD, Montpellier, France
- CIRAD, UMR AGAP Institut, Montpellier, France
- UMR AGAP Institut, Univ Montpellier, CIRAD, INRAE, Institut Agro, Montpellier, France
| |
Collapse
|
9
|
Podyma B, Parekh K, Güler AD, Deppmann CD. Metabolic homeostasis via BDNF and its receptors. Trends Endocrinol Metab 2021; 32:488-499. [PMID: 33958275 PMCID: PMC8192464 DOI: 10.1016/j.tem.2021.04.005] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/10/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022]
Abstract
Metabolic disorders result from dysregulation of central nervous system and peripheral metabolic energy homeostatic pathways. To maintain normal energy balance, neural circuits must integrate feedforward and feedback signals from the internal metabolic environment to orchestrate proper food intake and energy expenditure. These signals include conserved meal and adipocyte cues such as glucose and leptin, respectively, in addition to more novel players including brain-derived neurotrophic factor (BDNF). In particular, BDNF's two receptors, tropomyosin related kinase B (TrkB) and p75 neurotrophin receptor (p75NTR), are increasingly appreciated to be involved in whole body energy homeostasis. At times, these two receptors even seem to functionally oppose one another's actions, providing the framework for a potential neurotrophin mediated energy regulatory axis, which we explore further here.
Collapse
Affiliation(s)
- Brandon Podyma
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA; Medical Scientist Training Program, School of Medicine, University of Virginia, Charlottesville, VA 22908-0738, USA.
| | - Kavya Parekh
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | - Ali D Güler
- Department of Biology, University of Virginia, Charlottesville, VA 22903, USA
| | | |
Collapse
|
10
|
Fornaro M, Daray FM, Hunter F, Anastasia A, Stubbs B, De Berardis D, Shin JI, Husain MI, Dragioti E, Fusar-Poli P, Solmi M, Berk M, Vieta E, Carvalho AF. The prevalence, odds and predictors of lifespan comorbid eating disorder among people with a primary diagnosis of bipolar disorders, and vice-versa: Systematic review and meta-analysis. J Affect Disord 2021; 280:409-431. [PMID: 33227671 DOI: 10.1016/j.jad.2020.11.015] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 09/16/2020] [Accepted: 11/07/2020] [Indexed: 12/22/2022]
Abstract
BACKGROUND There are scarce and discrepant data about the prevalence and correlates of co-occurring eating disorders (EDs) among people with a primary diagnosis of bipolar disorder (BD), and vice-versa, compelling a systematic review and meta-analysis on the matter. METHODS MEDLINE/PsycINFO databases were systematically searched for original studies documenting BD⇌ED comorbidity across the lifespan, from inception up until April 20th, 2020. Random-effects meta-analysis and meta-regression analyses were conducted, accounting for multiple moderators. RESULTS Thirty-six studies involved 15,084 primary BD patients. Eleven studies encompassed 15,146 people with primary EDs. Binge eating disorder (BED) occurred in 12.5% (95%C.I.=9.4-16.6%, I2=93.48%) of BDs, while 9.1% (95%C.I.=3.3-22.6%) of BEDs endorsed BD. Bulimia Nervosa (BN) occurred in 7.4% (95%C.I.=6-10%) of people with BD, whereas 6.7% (95%C.I.=12-29.2%) of subjects with BN had a diagnosis of BD. Anorexia Nervosa (AN) occurred in 3.8% (95%C.I.=2-6%) of people with BDs; 2% (95%C.I.=1-2%) of BD patients had a diagnosis of AN. Overall, BD patients with EDs had higher odds of being female vs. non-ED controls. Several moderators yielded statistically significant differences both within- and between different types of BDs and EDs. LIMITATIONS Scant longitudinal studies, especially across different EDs and pediatric samples. High heterogeneity despite subgroup comparisons. Limited discrimination of the quality of the evidence. CONCLUSIONS The rates of BD⇌ED comorbidity vary across different diagnostic groups, more than they do according to the "direction" of BD⇌ED. Further primary studies should focus on the risks, chronology, clinical impact, and management of the onset of intertwined BD⇌ED across different ages, promoting a continuum approach.
Collapse
Affiliation(s)
- Michele Fornaro
- Department of Psychiatry, Federico II University, Naples, Italy; Polyedra Research Foundation, Teramo, Italy.
| | - Federico Manuel Daray
- The University of Buenos Aires, School of Medicine, Institute of Pharmacology, Argentina.
| | - Fernando Hunter
- The University of Buenos Aires, School of Medicine, Institute of Pharmacology, Argentina
| | | | - Brendon Stubbs
- Physiotherapy Department, South London and Maudsley NHS Foundation Trust, London SE5 8AZ, UK; Health Service and Population Research Department and the Department of Psychological, London, UK
| | | | - Jae Il Shin
- Department of Pediatrics, Yonsei University College of Medicine, Seoul, South Korea
| | - Muhammad Ishrat Husain
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction & Mental Health (CAMH), Toronto, ON, Canada
| | - Elena Dragioti
- Pain and Rehabilitation Centre, and Department of Health, Medicine and Caring Sciences, Linköping University, Linköping, Sweden University, Linköping, Sweden; Department of Hygiene and Epidemiology, School of Medicine, University of Ioannina, University Campus, Ioannina, Greece
| | - Paolo Fusar-Poli
- Early Psychosis: Interventions and Clinical-Detection Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK; OASIS Service, South London and Maudsley NHS Foundation Trust, London, UK; Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy; National Institute for Health Research, Maudsley Biomedical Research Centre, South London and Maudsley NHS Foundation Trust, London, UK
| | - Marco Solmi
- Early Psychosis: Interventions and Clinical-Detection Lab, Department of Psychosis Studies, Institute of Psychiatry, Psychology, and Neuroscience, King's College London, UK; Neurosciences Department, University of Padua, Italy; Padua Neuroscience Center, University of Padua, Italy
| | - Michael Berk
- Department of Psychiatry, the University of Melbourne, Parkville, VIC, Australia; Deakin University, IMPACT - the Institute for Mental and Physical Health and Clinical Translation, School of Medicine, Barwon Health, Geelong, Australia; Orygen Youth Health Research Centre, Centre for Youth Mental Health, the University of Melbourne, Parkville, VIC, Australia; The Florey Institute for Neuroscience and Mental Health, the University of Melbourne, Parkville, VIC, Australia
| | - Eduard Vieta
- Bipolar and Depressive Disorders Unit, Institute of Neuroscience, Hospital Clinic, University of Barcelona, IDIBAPS, CIBERSAM, 170 Villarroel st, 12-0, 08036 Barcelona, Catalonia, Spain
| | - André Ferrer Carvalho
- Department of Psychiatry, University of Toronto, Toronto, ON, Canada; Centre for Addiction & Mental Health (CAMH), Toronto, ON, Canada
| |
Collapse
|
11
|
Nilsson IAK, Hökfelt T, Schalling M. The Anorectic Phenotype of the anx/anx Mouse Is Associated with Hypothalamic Dysfunction. NEUROMETHODS 2021:297-317. [DOI: 10.1007/978-1-0716-0924-8_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
12
|
Himmerich H, Bentley J, Kan C, Treasure J. Genetic risk factors for eating disorders: an update and insights into pathophysiology. Ther Adv Psychopharmacol 2019; 9:2045125318814734. [PMID: 30800283 PMCID: PMC6378634 DOI: 10.1177/2045125318814734] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Accepted: 10/15/2018] [Indexed: 12/16/2022] Open
Abstract
Genome-wide-association studies (GWASs), epigenetic, gene-expression and gene-gene interaction projects, nutritional genomics and investigations of the gut microbiota have increased our knowledge of the pathophysiology of eating disorders (EDs). However, compared with anorexia nervosa, genetic studies in patients with bulimia nervosa and binge-eating disorder are relatively scarce, with the exception of a few formal genetic and small-sized candidate-gene-association studies. In this article, we review important findings derived from formal and molecular genetics in order to outline a genetics-based pathophysiological model of EDs. This model takes into account environmental and nutritional factors, genetic factors related to the microbiome, the metabolic and endocrine system, the immune system, and the brain, in addition to phenotypical traits of EDs. Shortcomings and advantages of genetic research in EDs are discussed against the historical background, but also in light of potential future treatment options for patients with EDs.
Collapse
Affiliation(s)
| | - Jessica Bentley
- Department of Psychological Medicine, King’s College London, London, UK
| | - Carol Kan
- Department of Psychological Medicine, King’s College London, London, UK
| | | |
Collapse
|
13
|
Nilsson IAK. The anx/anx Mouse - A Valuable Resource in Anorexia Nervosa Research. Front Neurosci 2019; 13:59. [PMID: 30804742 PMCID: PMC6370726 DOI: 10.3389/fnins.2019.00059] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 01/21/2019] [Indexed: 01/31/2023] Open
Abstract
Animal models are invaluable resources in research concerning the neurobiology of anorexia nervosa (AN), to a large extent since valid clinical samples are rare. None of the existing models can capture all aspects of AN but they are able to mirror the core features of the disorder e.g., elective starvation, emaciation and premature death. The anorectic anx/anx mouse is of particular value for the understanding of the abnormal response to negative energy balance seen in AN. These mice appear normal at birth but gradually develops starvation and emaciation despite full access to food, and die prematurely around three weeks of age. Several changes in hypothalamic neuropeptidergic and -transmitter systems involved in regulating food intake and metabolism have been documented in the anx/anx mouse. These changes are accompanied by signs of inflammation and degeneration in the same hypothalamic regions; including activation of microglia cells and expression of major histocompatibility complex I by microglia and selective neuronal populations. These aberrances are likely related to the dysfunction of complex I (CI) in the oxidative phosphorylation system of the mitochondria, and subsequent increased oxidative stress, which also has been revealed in the hypothalamus of these mice. Interestingly, a similar CI dysfunction has been shown in leukocytes from patients with AN. In addition, a higher expression of the Neurotrophic Receptor Tyrosine Kinase 3 gene has been shown in the anx/anx hypothalamus. This agrees with AN being associated with specific variants of the genes for brain derived neurotrophic factor and Neurotrophic Receptor Tyrosine Kinase 2. The anx/anx mouse is also glucose intolerant and display pancreatic dysfunction related to increased levels of circulating free fatty acids (FFA) and pancreatic inflammation. An increased incidence of eating disorders has been reported for young diabetic women, and as well has increased levels of circulating FFAs in AN. Also similar to individuals with AN, the anx/anx mouse has reduced leptin and increased cholesterol levels in serum. Thus, the anx/anx mouse shares several characteristics with patients with AN, including emaciation, starvation, premature death, diabetic features, increased FFA and low leptin, and is therefore a unique resource in research on the (neuro)biology of AN.
Collapse
Affiliation(s)
- Ida A K Nilsson
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Stockholm, Sweden.,Center for Molecular Medicine, Karolinska University Hospital, Stockholm, Sweden.,Centre for Eating Disorders Innovation, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
14
|
Domínguez-Sanchéz MA, Bustos-Cruz RH, Velasco-Orjuela GP, Quintero AP, Tordecilla-Sanders A, Correa-Bautista JE, Triana-Reina HR, García-Hermoso A, González-Ruíz K, Peña-Guzmán CA, Hernández E, Peña-Ibagon JC, Téllez-T LA, Izquierdo M, Ramírez-Vélez R. Acute Effects of High Intensity, Resistance, or Combined Protocol on the Increase of Level of Neurotrophic Factors in Physically Inactive Overweight Adults: The BrainFit Study. Front Physiol 2018; 9:741. [PMID: 29997519 PMCID: PMC6030369 DOI: 10.3389/fphys.2018.00741] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2017] [Accepted: 05/28/2018] [Indexed: 12/11/2022] Open
Abstract
The purpose of this study was to compare the neurotrophic factor response following one session of high-intensity exercise, resistance training or both in a cohort of physically inactive overweight adults aged 18–30 years old. A randomized, parallel-group clinical trial of 51 men (23.6 ± 3.5 years; 83.5 ± 7.8 kg; 28.0 ± 1.9 kg/m2) who are physically inactive (i.e., < 150 min of moderate-intensity exercise per week or IPAQ score of <600 MET min/week for >6 months) and are either abdominally obese (waist circumference ≥90 cm) or have a body mass index, BMI ≥25 and ≤ 30 kg/m2 were randomized to the following four exercise protocols: high-intensity exercise (4 × 4 min intervals at 85–95% maximum heart rate [HRmax] interspersed with 4 min of recovery at 75–85% HRmax) (n = 14), resistance training (12–15 repetitions per set, at 50–70% of one repetition maximum with 60 s of recovery) (n = 12), combined high-intensity and resistance exercise (n = 13), or non-exercising control (n = 12). The plasma levels of neurotrophin-3 (NT-3), neurotrophin-4 (also known as neurotrophin 4/5; NT-4 or NT-4/5), and brain-derived neurotrophic factor (BDNF) were determined before (pre-exercise) and 1-min post-exercise for each protocol session. Resistance training induced significant increases in NT-3 (+39.6 ng/mL [95% CI, 2.5–76.6; p = 0.004], and NT-4/5 (+1.3 ng/mL [95% CI, 0.3–2.3; p = 0.014]), respectively. Additionally, combined training results in favorable effects on BDNF (+22.0, 95% CI, 2.6–41.5; p = 0.029) and NT-3 (+32.9 ng/mL [95% CI, 12.3–53.4; p = 0.004]), respectively. The regression analysis revealed a significant positive relationship between changes in BDNF levels and changes in NT-4/5 levels from baseline to immediate post-exercise in the combined training group (R2 = 0.345, p = 0.034) but not the other intervention groups. The findings indicate that acute resistance training and combined exercise increase neurotrophic factors in physically inactive overweight adults. Further studies are required to determine the biological importance of changes in neurotrophic responses in overweight men and chronic effects of these exercise protocols. Trial Registration: ClinicalTrials.gov, NCT02915913 (Date: September 22, 2016).
Collapse
Affiliation(s)
- María A Domínguez-Sanchéz
- Grupo de Investigación Movimiento Corporal Humano, Facultad de Enfermería y Rehabilitación, Universidad de La Sabana, Chía, Colombia
| | - Rosa H Bustos-Cruz
- Evidence-Based Therapeutic Group, Clinical Pharmacology, Universidad de La Sabana, Bogotá, Colombia
| | - Gina P Velasco-Orjuela
- Centro de Estudios en Medición de la Actividad Física, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Andrea P Quintero
- Centro de Estudios en Medición de la Actividad Física, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Alejandra Tordecilla-Sanders
- Centro de Estudios en Medición de la Actividad Física, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Jorge E Correa-Bautista
- Centro de Estudios en Medición de la Actividad Física, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Héctor R Triana-Reina
- Grupo GICAEDS, Programa de Cultura Física, Deporte y Recreación, Universidad Santo Tomás, Bogotá, Colombia
| | - Antonio García-Hermoso
- Laboratorio de Ciencias de la Actividad Física, el Deporte y la Salud, Universidad de Santiago de Chile, Santiago, Chile
| | - Katherine González-Ruíz
- Grupo de Ejercicio Físico y Deportes, Facultad de Salud, Programa de Fisioterapia, Universidad Manuela Beltrán, Bogotá, Colombia
| | - Carlos A Peña-Guzmán
- Facultad de Ingeniería Ambiental, Grupo de Investigación INAM-USTA Universidad Santo Tomás, Bogotá, Colombia
| | - Enrique Hernández
- Centro de Estudios en Medición de la Actividad Física, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Jhonatan C Peña-Ibagon
- Centro de Estudios en Medición de la Actividad Física, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Luis A Téllez-T
- Centro de Estudios en Medición de la Actividad Física, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| | - Mikel Izquierdo
- Department of Health Sciences, Public University of Navarra, Navarrabiomed, CIBER of Frailty and Healthy Aging (CIBERFES) Instituto de Salud Carlos III, Pamplona, Spain
| | - Robinson Ramírez-Vélez
- Centro de Estudios en Medición de la Actividad Física, Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
15
|
Carrera C, Jiménez-Conde J, Derdak S, Rabionet K, Vives-Bauzá C, Soriano-Tárrega C, Giralt-Steinhauer E, Mola-Caminal M, Diaz-Navarro RM, Tur S, Muiño E, Gallego-Fabrega C, Beltran S, Roquer J, Ruiz A, Sotolongo-Grau O, Krupinski J, Lee JM, Cruchaga C, Delgado P, Malik R, Worrall BB, Seshadri S, Montaner J, Fernández-Cadenas I. Whole exome sequencing analysis reveals TRPV3 as a risk factor for cardioembolic stroke/subtitle. Thromb Haemost 2018; 116:1165-1171. [DOI: 10.1160/th16-02-0113] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Accepted: 08/15/2016] [Indexed: 12/12/2022]
Abstract
SummaryGenetic studies suggest that hundreds of genes associated with stroke remain unidentified. Exome sequencing proves useful for finding new genes associated with stroke. We aimed to find new genetic risk factors for cardioembolic stroke by analysing exome sequence data using new strategies. For discovery, we analysed 42 cardioembolic stroke cases and controls with extreme phenotypes (cohort 1), and for replication, 32 cardioembolic stroke cases and controls (cohort 2) using the SeqCapExome capture kit. We then analysed the replicated genes in two new cohorts that comprised 834 cardioembolic strokes and controls (cohort 3) and 64,373 cardioembolic strokes and controls (cohort 4). Transcriptomic in-silico functional analyses were also performed. We found 26 coding regions with a higher frequency of mutations in cardioembolic strokes after correcting for the number of mutations found in the whole exome of every patient. The TRPV3 gene was associated with cardioembolic stroke after replication of exome sequencing analysis (p-value-discovery: 0.018, p-value-replication: 0.014). The analysis of the TRPV3 gene using polymorphisms in cohort 3 and 4 revealed two polymorphisms associated with cardioembolic stroke in both cohorts, the most significant polymorphism being rs151091899 (p-value: 3.1 × 10−05; odds ratio: 5.4) in cohort 3. The genotype of one polymorphism of TRPV3 was associated with a differential expression of genes linked to cardiac malformations. In conclusion, new strategies using exome sequence data have revealed TRPV3 as a new gene associated with cardioembolic stroke. This strategy among others might be useful in finding new genes associated with complex genetic diseases.
Collapse
|
16
|
Bergström U, Lindfors C, Svedberg M, Johansen JE, Häggkvist J, Schalling M, Wibom R, Katz A, Nilsson IAK. Reduced metabolism in the hypothalamus of the anorectic anx/anx mouse. J Endocrinol 2017; 233:15-24. [PMID: 28130409 DOI: 10.1530/joe-16-0383] [Citation(s) in RCA: 8] [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: 01/10/2017] [Accepted: 01/27/2017] [Indexed: 12/13/2022]
Abstract
The anorectic anx/anx mouse exhibits a mitochondrial complex I dysfunction that is related to aberrant expression of hypothalamic neuropeptides and transmitters regulating food intake. Hypothalamic activity, i.e. neuronal firing and transmitter release, is dependent on glucose utilization and energy metabolism. To better understand the role of hypothalamic activity in anorexia, we assessed carbohydrate and high-energy phosphate metabolism, in vivo and in vitro, in the anx/anx hypothalamus. In the fasted state, hypothalamic glucose uptake in the anx/anx mouse was reduced by ~50% of that seen in wild-type (wt) mice (P < 0.05). Under basal conditions, anx/anx hypothalamus ATP and glucose 6-P contents were similar to those in wt hypothalamus, whereas phosphocreatine was elevated (~2-fold; P < 0.001) and lactate was reduced (~35%; P < 0.001). The anx/anx hypothalamus had elevated total AMPK (~25%; P < 0.05) and GLUT4 (~60%; P < 0.01) protein contents, whereas GLUT1 and GLUT3 were similar to that of wt hypothalamus. Interestingly, the activation state of AMPK (ratio of phosphorylated AMPK/total AMPK) was significantly decreased in hypothalamus of the anx/anx mouse (~60% of that in wt; P < 0.05). Finally, during metabolic stress (ischemia), accumulation of lactate (measure of glycolysis) and IMP and AMP (breakdown products of ATP) were ~50% lower in anx/anx vs wt hypothalamus. These data demonstrate that carbohydrate and high-energy phosphate utilization in the anx/anx hypothalamus are diminished under basal and stress conditions. The decrease in hypothalamic metabolism may contribute to the anorectic behavior of the anx/anx mouse, i.e. its inability to regulate food intake in accordance with energy status.
Collapse
Affiliation(s)
- Ulrika Bergström
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Charlotte Lindfors
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Marie Svedberg
- Department of Clinical NeuroscienceCenter for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Jeanette E Johansen
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Jenny Häggkvist
- Department of Clinical NeuroscienceCenter for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Martin Schalling
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| | - Rolf Wibom
- Department of Medical Biochemistry and BiophysicsKarolinska Institutet, Stockholm, Sweden
| | - Abram Katz
- Department of Physical TherapyAriel University, Ariel, Israel
| | - Ida A K Nilsson
- Department of Molecular Medicine and SurgeryKarolinska Institutet, Stockholm, Sweden
- Center for Molecular MedicineKarolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
17
|
Groh A, Jahn K, Burkert A, Neyazi A, Schares L, Janke E, Rehme M, Schuster R, Hillemacher T, Bleich S, Frieling H, Heberlein A. Epigenetic Regulation of the Promotor Region of Vascular Endothelial Growth Factor-A and Nerve Growth Factor in Opioid-Maintained Patients. Eur Addict Res 2017; 23:249-259. [PMID: 29224006 DOI: 10.1159/000485030] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Accepted: 11/07/2017] [Indexed: 12/19/2022]
Abstract
AIMS The nerve growth factor (NGF) and the vascular endothelial growth factor-A (VEGF-A) may be of importance for psychiatric diseases including substance use disorders. The aim of the study was to identify differences in the regulation of both neuropeptides via the DNA-methylation status of the promotor regions of NGF and VEGF-A in different forms of maintenance therapy for opioid dependence and the related stress regulation via the hypothalamic-pituitary-adrenal axis. METHODS We compared methylation levels of opioid-dependent patients receiving treatment with diamorphine (n = 28) or levomethadone (n = 54) and similar levels in a healthy control group (n = 72). RESULTS There was a significantly higher methylation of VEGF-A in opioid-maintained patients with levomethadone compared to that in the control group (estimated marginal means [EMM] [SE]): 0.036 [0.003] vs. 0.020 [0.003]; p < 0.001). We performed a cluster analysis for NGF, splitting up the results in 4 clusters. We found significant changes in methylation rates of the opioid-maintained patients compared to the controls in cluster I ([EMM] [SE]: 0.064 [0.005] vs. 0.084 [0.006]; p = 0.03), cluster II ([EMM] [SE]: 0.133 [0.013] vs. 0.187 [0.014]; p < 0.001) and cluster III ([EMM] [SE]: 0.190 [0.014] vs. 0.128 [0.016]; p < 0.001). CONCLUSIONS The results are of importance, as they indicate that long-term changes in stress regulation regulated by neurotrophines are a crucial part of the symptomatology of opioid dependence, thus influencing drug consumption and the different forms of opioid-maintenance therapies.
Collapse
Affiliation(s)
- Adrian Groh
- Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Molecular Neuroscience Laboratory, Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Kirsten Jahn
- Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Molecular Neuroscience Laboratory, Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Alexandra Burkert
- Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Molecular Neuroscience Laboratory, Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Alexandra Neyazi
- Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Molecular Neuroscience Laboratory, Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Laura Schares
- Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Eva Janke
- Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Marie Rehme
- Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Rilana Schuster
- Department of Addictive Behavior and Addiction Medicine, Central Institute of Mental Health, Medical Faculty Mannheim/Heidelberg University, Mannheim, Germany
| | - Thomas Hillemacher
- Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Stefan Bleich
- Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Helge Frieling
- Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany.,Molecular Neuroscience Laboratory, Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| | - Annemarie Heberlein
- Center for Addiction Research (CARe), Department of Psychiatry, Social Psychiatry and Psychotherapy, Hannover Medical School, Hannover, Germany
| |
Collapse
|
18
|
Kim JY, Jeong JE, Rhee JK, Cho H, Chun JW, Kim TM, Choi SW, Choi JS, Kim DJ. Targeted exome sequencing for the identification of a protective variant against Internet gaming disorder at rs2229910 of neurotrophic tyrosine kinase receptor, type 3 (NTRK3): A pilot study. J Behav Addict 2016; 5:631-638. [PMID: 27826991 PMCID: PMC5370368 DOI: 10.1556/2006.5.2016.077] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Background and aims Internet gaming disorder (IGD) has gained recognition as a potential new diagnosis in the fifth revision of the Diagnostic and Statistical Manual of Mental Disorders, but genetic evidence supporting this disorder remains scarce. Methods In this study, targeted exome sequencing was conducted in 30 IGD patients and 30 control subjects with a focus on genes linked to various neurotransmitters associated with substance and non-substance addictions, depression, and attention deficit hyperactivity disorder. Results rs2229910 of neurotrophic tyrosine kinase receptor, type 3 (NTRK3) was the only single nucleotide polymorphism (SNP) that exhibited a significantly different minor allele frequency in IGD subjects compared to controls (p = .01932), suggesting that this SNP has a protective effect against IGD (odds ratio = 0.1541). The presence of this potentially protective allele was also associated with less time spent on Internet gaming and lower scores on the Young's Internet Addiction Test and Korean Internet Addiction Proneness Scale for Adults. Conclusions The results of this first targeted exome sequencing study of IGD subjects indicate that rs2229910 of NTRK3 is a genetic variant that is significantly related to IGD. These findings may have significant implications for future research investigating the genetics of IGD and other behavioral addictions.
Collapse
Affiliation(s)
| | - Jo-Eun Jeong
- Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Je-Keun Rhee
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Hyun Cho
- Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Ji-Won Chun
- Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Tae-Min Kim
- Department of Medical Informatics, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Sam-Wook Choi
- Korea Institute on Behavioral Addictions, True Mind Mental Health Clinic, Seoul, Republic of Korea, and Health Care & Information Research Institute, Namseoul University, Cheonan, Republic of Korea
| | - Jung-Seok Choi
- Department of Psychiatry, SMG-SNU Boramae Medical Center, Seoul, Republic of Korea, and Department of Psychiatry and Behavioral Science, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Dai-Jin Kim
- Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea,Corresponding author: Dai-Jin Kim; Department of Psychiatry, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 202 Banpo-daero, Seocho-gu, Seoul 137-701, Republic of Korea; Phone: +82 2 2258 6086; Fax: +82 2 594 3870; E-mail:
| |
Collapse
|
19
|
Klinedinst NJ, Resnick B, Yerges-Armstrong LM, Dorsey SG. The Interplay of Genetics, Behavior, and Pain with Depressive Symptoms in the Elderly. THE GERONTOLOGIST 2016; 55 Suppl 1:S67-77. [PMID: 26055783 DOI: 10.1093/geront/gnv015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
PURPOSE OF STUDY About 25% of older adults suffer from depressive symptoms. Commonly studied candidate genes associated with depression include those that influence serotonin (SLC6A4), dopamine (COMT), or neuroplasticity (BDNF, NTRK3). However, the majority of candidate gene studies do not consider the interplay of genetics, demographic, clinical, and behavioral factors and how they jointly contribute to depressive symptoms among older adults. The purpose of this study was to gain a more comprehensive understanding of depressive symptoms among older adults. DESIGN AND METHODS In this descriptive study, demographic, behavioral, and clinical characteristics (age, gender, comorbidities, volunteering, physical activity, pain, and fear of falling) were obtained via interview of 114 residents in a continuing care retirement community. Peripheral whole blood was collected for DNA extraction. We examined common single nucleotide polymorphisms (SNPs) in the aforementioned genes using path analyses. RESULTS SNPs in the NTRK3 gene, pain, physical activity, and fear of falling were directly associated with depressive symptoms in older adults. Those who had polymorphisms in the NTRK3 gene, pain, fear of falling, and were less physically active were more likely to exhibit depressive symptoms. None of the SNPs in SLC6A4, COMT, or BDNF genes were significantly associated with depressive symptoms. IMPLICATIONS Our use of a path analysis to examine a biopsychosocial model of depressive symptoms provided the opportunity to describe a comprehensive clinical picture of older adults at risk for depressive symptoms. Thus, interventions could be implemented to identify older adults at risk for depressive symptoms.
Collapse
|
20
|
Shih PAB, Woodside DB. Contemporary views on the genetics of anorexia nervosa. Eur Neuropsychopharmacol 2016; 26:663-73. [PMID: 26944296 PMCID: PMC4801707 DOI: 10.1016/j.euroneuro.2016.02.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 02/05/2016] [Accepted: 02/05/2016] [Indexed: 10/22/2022]
Abstract
Anorexia nervosa (AN) is a serious mental illness characterized by severe dietary restriction that leads to high rates of morbidity, chronicity, and mortality. Unfortunately, effective treatment is lacking and few options are available. High rates of familial aggregation and significant heritability suggested that the complex etiology of AN is affected by both genetic and environmental factors. In this paper, we review studies that reported common and rare genetic variation that influence susceptibility of AN through candidate gene studies, genome-wide association studies, and sequencing-based studies. We also discuss gene expression, methylation, imaging genetics, and pharmacogenetics to demonstrate that these studies have collectively advanced our knowledge of how genetic variation contributes to AN susceptibility and clinical course. Lastly, we highlight the importance of gene by environment interactions (G×E) and share our enthusiasm for the use of nutritional genomic approaches to elucidate the interaction among nutrients, metabolic intermediates, and genetic variation in AN. A deeper understanding of how nutrition alters genome stability, how genetic variation influences uptake and metabolism of nutrients, and how response to food components affects disordered eating, will lead to personalized dietary interventions and effective nutraceutical and pharmacological treatments for AN.
Collapse
Affiliation(s)
- Pei-an Betty Shih
- Department of Psychiatry, University of California, San Diego, 9500 Gilman Drive #0664, La Jolla, CA 92093-0664, USA.
| | - D Blake Woodside
- Inpatient Eating Disorders Service, Toronto General Hospital, Canada; Department of Psychiatry, University of Toronto, Canada.
| |
Collapse
|
21
|
Brandys MK, de Kovel CGF, Kas MJ, van Elburg AA, Adan RAH. Overview of genetic research in anorexia nervosa: The past, the present and the future. Int J Eat Disord 2015; 48:814-25. [PMID: 26171770 DOI: 10.1002/eat.22400] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/05/2015] [Indexed: 02/03/2023]
Abstract
BACKGROUND Even though the evidence supporting the presence of a heritable component in the aetiology of anorexia nervosa (AN) is strong, the underlying genetic mechanisms remain poorly understood. The recent publication of a genome-wide association study (GWAS) of AN (Boraska, Mol Psychiatry, 2014) was an important step in genetic research in AN. OBJECTIVE To briefly sum up strengths and weaknesses of candidate-gene and genome-wide approaches, to discuss the genome-wide association studies of AN and to make predictions about the genetic architecture of AN by comparing it to that of schizophrenia (since the diseases share some similarities and genetic research in schizophrenia is more advanced). METHOD Descriptive literature review. RESULTS Despite remarkable efforts, the gene-association studies in AN did not advance our knowledge as much as had been hoped, although some results still await replication. DISCUSSION Continuous effort of participants, clinicians and researchers remains necessary to ensure that genetic research in AN follows a similarly successful path as in schizophrenia. Identification of genetic susceptibility loci provides a basis for follow-up studies.
Collapse
Affiliation(s)
- Marek K Brandys
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Utrecht Research Group for Eating Disorders, Utrecht, The Netherlands
| | - Carolien G F de Kovel
- Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Martien J Kas
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Utrecht Research Group for Eating Disorders, Utrecht, The Netherlands
| | - Annemarie A van Elburg
- Utrecht Research Group for Eating Disorders, Utrecht, The Netherlands.,Department Clinical and Health Psychology, Fac. of Social Sciences, University of Utrecht, Utrecht, The Netherlands.,Rintveld, Center for Eating Disorders, Altrecht Mental Health Institute, Zeist, The Netherlands
| | - Roger A H Adan
- Department of Translational Neuroscience, Brain Center Rudolf Magnus, University Medical Center Utrecht, Utrecht, The Netherlands.,Utrecht Research Group for Eating Disorders, Utrecht, The Netherlands.,Rintveld, Center for Eating Disorders, Altrecht Mental Health Institute, Zeist, The Netherlands
| |
Collapse
|
22
|
Harm avoidance involved in mediating the association between nerve growth factor (NGF) gene polymorphisms and antidepressant efficacy in patients with major depressive disorder. J Affect Disord 2015; 183:187-94. [PMID: 26021968 DOI: 10.1016/j.jad.2015.05.012] [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: 11/06/2014] [Revised: 05/06/2015] [Accepted: 05/07/2015] [Indexed: 12/15/2022]
Abstract
BACKGROUND Antidepressants have variable efficacies in subjects with major depressive disorder (MDD). Nerve growth factor (NGF) has been suggested to play an important role in the pathogenesis of depressive symptoms and the response to antidepressant therapy. The aim of this study was to examine whether NGF gene polymorphisms are associated with the antidepressant therapeutic efficacy in subjects with MDD. METHODS A naturalistic follow-up study was carried out on 557 subjects with MDD. Of the enrolled patients, 304 completed the 8-week open-label antidepressant treatment. Seven single-nucleotide polymorphisms (SNPs) of the NGF gene were genotyped. The 21-item Hamilton Depression Rating Scale was used to assess depressive severity from baseline to endpoint. Tridimensional Personality Questionnaire was used to assess baseline personality traits. Single marker and haplotype analyses were conducted. Binary logistic regression was used to calculate odds ratios of remission. Structural equation modeling was used to analyze the predicted mediation effect. RESULTS A significant difference in genotype frequencies between remitters and non-remitters was observed in three NGF SNPs (rs12760036, rs7523654, and rs17033692). The haplotype analysis revealed that the CCC haplotype (rs2254527-rs6678788-rs12760036) was associated with a higher remission rate, while the CCA haplotype was associated with a lower remission rate. The harm avoidance psychological factor partially mediated the effect of NGF variants on antidepressant efficacy. LIMITATIONS The selected SNPs may not cover whole NGF gene. CONCLUSIONS NGF variants are associated with remission rates after 8-week antidepressant treatment, and harm avoidance partially mediated the effect of NGF variants on treatment outcomes.
Collapse
|
23
|
Yilmaz Z, Kaplan AS, Tiwari AK, Levitan RD, Piran S, Bergen AW, Kaye WH, Hakonarson H, Wang K, Berrettini WH, Brandt HA, Bulik CM, Crawford S, Crow S, Fichter MM, Halmi KA, Johnson CL, Keel PK, Klump KL, Magistretti P, Mitchell JE, Strober M, Thornton LM, Treasure J, Woodside DB, Knight J, Kennedy JL. The role of leptin, melanocortin, and neurotrophin system genes on body weight in anorexia nervosa and bulimia nervosa. J Psychiatr Res 2014; 55:77-86. [PMID: 24831852 PMCID: PMC4191922 DOI: 10.1016/j.jpsychires.2014.04.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/05/2014] [Accepted: 04/04/2014] [Indexed: 12/26/2022]
Abstract
OBJECTIVE Although low weight is a key factor contributing to the high mortality in anorexia nervosa (AN), it is unclear how AN patients sustain low weight compared with bulimia nervosa (BN) patients with similar psychopathology. Studies of genes involved in appetite and weight regulation in eating disorders have yielded variable findings, in part due to small sample size and clinical heterogeneity. This study: (1) assessed the role of leptin, melanocortin, and neurotrophin genetic variants in conferring risk for AN and BN; and (2) explored the involvement of these genes in body mass index (BMI) variations within AN and BN. METHOD Our sample consisted of 745 individuals with AN without a history of BN, 245 individuals with BN without a history of AN, and 321 controls. We genotyped 20 markers with known or putative function among genes selected from leptin, melanocortin, and neurotrophin systems. RESULTS There were no significant differences in allele frequencies among individuals with AN, BN, and controls. AGRP rs13338499 polymorphism was associated with lowest illness-related BMI in those with AN (p = 0.0013), and NTRK2 rs1042571 was associated with highest BMI in those with BN (p = 0.0018). DISCUSSION To our knowledge, this is the first study to address the issue of clinical heterogeneity in eating disorder genetic research and to explore the role of known or putatively functional markers in genes regulating appetite and weight in individuals with AN and BN. If replicated, our results may serve as an important first step toward gaining a better understanding of weight regulation in eating disorders.
Collapse
Affiliation(s)
- Zeynep Yilmaz
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Clinical Research Department, Centre for Addiction and Mental Health, Toronto, Canada
| | - Allan S Kaplan
- Clinical Research Department, Centre for Addiction and Mental Health, Toronto, Canada; Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada
| | - Arun K Tiwari
- Neurogenetics Section, Centre for Addiction and Mental Health, Toronto, Canada
| | - Robert D Levitan
- Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Mood and Anxiety Program, Centre for Addiction and Mental Health, Toronto, Canada
| | - Sara Piran
- Faculty of Medicine, University of Ottawa, Ottawa, Canada
| | - Andrew W Bergen
- Center for Health Sciences, SRI International, Menlo Park, CA, USA
| | - Walter H Kaye
- Department of Psychiatry, University of California, San Diego, CA, USA
| | - Hakon Hakonarson
- Joseph Stokes Jr. Research Institute, The Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Kai Wang
- Department of Psychiatry, University of Southern California, Los Angeles, CA, USA
| | - Wade H Berrettini
- Department of Psychiatry, Center of Neurobiology and Behavior, University of Pennsylvania, Philadelphia, PA, USA
| | - Harry A Brandt
- Department of Psychiatry, Sheppard Pratt Health System, Towson, MD, USA
| | - Cynthia M Bulik
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA; Department of Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Steven Crawford
- Department of Psychiatry, Sheppard Pratt Health System, Towson, MD, USA
| | - Scott Crow
- Department of Psychiatry, University of Minnesota, Minneapolis, MN, USA
| | - Manfred M Fichter
- Department of Psychiatry, University of Munich (LMU), Munich, Germany; Roseneck Hospital for Behavioral Medicine, Prien, Germany
| | - Katherine A Halmi
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, USA
| | | | - Pamela K Keel
- Department of Psychology, Florida State University, Tallahassee, FL, USA
| | - Kelly L Klump
- Department of Psychology, Michigan State University, East Lansing, MI, USA
| | - Pierre Magistretti
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - James E Mitchell
- Department of Clinical Neuroscience, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, USA; Neuropsychiatric Research Institute, Fargo, ND, USA
| | - Michael Strober
- Department of Psychiatry, Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, University of California, Los Angeles, CA, USA
| | - Laura M Thornton
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Janet Treasure
- Department of Academic Psychiatry, King's College London, Institute of Psychiatry, London, United Kingdom
| | - D Blake Woodside
- Department of Psychiatry, University of Toronto, Toronto, Canada; Eating Disorders Program, Toronto General Hospital, Toronto, Canada
| | - Joanne Knight
- Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Neurogenetics Section, Centre for Addiction and Mental Health, Toronto, Canada
| | - James L Kennedy
- Institute of Medical Science, University of Toronto, Toronto, Canada; Department of Psychiatry, University of Toronto, Toronto, Canada; Neurogenetics Section, Centre for Addiction and Mental Health, Toronto, Canada.
| |
Collapse
|
24
|
Tremolizzo L, Conti E, Bomba M, Uccellini O, Rossi MS, Marfone M, Corbetta F, Santarone ME, Raggi ME, Neri F, Ferrarese C, Nacinovich R. Decreased whole-blood global DNA methylation is related to serum hormones in anorexia nervosa adolescents. World J Biol Psychiatry 2014; 15:327-33. [PMID: 24286295 DOI: 10.3109/15622975.2013.860467] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The one-carbon metabolism, also known as methionine-homocysteine cycle, governs the dynamics of DNA methylation, epigenetically regulating gene expression, and has been reported altered in anorexia nervosa (AN) adult patients. The aim of this study consisted in assessing whole-blood DNA methylation in adolescent AN patients, assessing its significance in relationship to clinical and hormonal variables. METHODS Whole-blood global DNA methylation was measured as incorporation of [(3)H]dCTP following HpaII cut in 32 adolescent females affected by restrictive type AN and compared to 13 healthy controls. Homocysteine, vitamin B12 and folate plasma levels were assessed as well as fasting plasma levels of leptin and steroid hormones. Clinical variables, including severity and associate states and traits, were assessed by means of the EDI-3, CDI and STAI-Y scales. RESULTS We confirm that whole-blood global DNA methylation is modestly albeit significantly reduced in AN adolescents with respect to controls, correlating with plasma leptin and steroid hormone levels. Conversely, clinical traits did not correlate with the outcome variable. CONCLUSIONS A better definition of the epigenetic dysregulation underlying AN pathology or vulnerability might lead to develop useful markers for diagnosis, prognostic classification and tailored therapeutic interventions in these vulnerable patients since the earliest phases of their disease.
Collapse
|
25
|
Abstract
Complex interactions between the brain and peripheral tissues mediate the effective control of energy balance and body weight. Hypothalamic and hindbrain neural circuits integrate peripheral signals informing the nutritional status of the animal and in response regulate nutrient intake and energy utilization. Obesity and its many medical complications emerge from the dysregulation of energy homeostasis. Excessive weight gain might also arise from alterations in reward systems of the brain that drive consumption of calorie dense, palatable foods in the absence of an energy requirement. Several neurotrophins, most notably brain-derived neurotrophic factor, have been implicated in the molecular and cellular processes underlying body weight regulation. Here, we review investigations interrogating their roles in energy balance and reward centers of the brain impacting feeding behavior and energy expenditure.
Collapse
Affiliation(s)
- M Rios
- Department of Neuroscience, Tufts University School of Medicine, Boston, MA, 02111, USA,
| |
Collapse
|
26
|
Dmitrzak-Weglarz M, Skibinska M, Slopien A, Tyszkiewicz M, Pawlak J, Maciukiewicz M, Zaremba D, Rajewski A, Hauser J. Serum neurotrophin concentrations in polish adolescent girls with anorexia nervosa. Neuropsychobiology 2013; 67:25-32. [PMID: 23221923 DOI: 10.1159/000343500] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 09/10/2012] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Several lines of evidence suggest that brain-derived neurotrophic factor (BDNF) plays an important role in weight regulation and eating behaviors as well as in the activity-dependent neuroplasticity underlying learning and memory behaviors involving the hippocampus. In anorexia nervosa (AN) patients, abnormal serum BDNF concentrations, cognitive impairments and specific personality traits have been traditionally observed. This study explores the levels of four serum neurotrophins [BDNF, neurotrophin 3 (NTF3), neurotrophin 4 (NTF4) and glial cell line-derived neurotrophic factor (GDNF)] with respect to their use as potential biomarkers for AN. This study also investigates any associations that might exist between serum neurotrophin levels and neurocognitive impairment or personality traits. METHODS Serum neurotrophin concentrations were measured in 60 AN patients (AN group) and 45 healthy controls (HC group). We correlated the serum levels of the four neurotrophins BDNF, NTF3, NTF4 and GDNF and the clinical type of anorexia. We also analyzed the relationship between serum neurotrophin levels and the Beck Depression Inventory, body mass index, executive functions by the Wisconsin Card Sorting test (WCST) and personality dimensions by the Temperament and Character Inventory (TCI) test. RESULTS Serum NTF4 concentrations were significantly lower when comparing all AN patients (34.7 ± 72.5 pg/ml) or restriction type AN patients (29.1 ± 62.5 pg/ml) with the HC group (58.4 ± 135.8 pg/ml; p = 0.004 and p = 0.005, respectively). A significant correlation (p < 0.005) between BDNF serum levels and patient personality dimensions as measured by the TCI test was observed. Furthermore, significant correlations were observed between NTF4 and GDNF serum levels and executive function as measured by the WCST. CONCLUSIONS These data suggest that NTF4 might serve as a biomarker for AN. Furthermore, BDNF and GDNF serum levels appear to be associated with personality traits and executive function.
Collapse
Affiliation(s)
- Monika Dmitrzak-Weglarz
- Psychiatric Genetics Unit, Department of Psychiatry, Poznan University of Medical Sciences, Poznan, Poland.
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Common genetic background in anorexia nervosa and obsessive compulsive disorder: preliminary results from an association study. J Psychiatr Res 2013; 47:747-54. [PMID: 23337130 DOI: 10.1016/j.jpsychires.2012.12.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/04/2012] [Revised: 10/17/2012] [Accepted: 12/28/2012] [Indexed: 01/11/2023]
Abstract
Several lines of evidence, including psychopathological, neurobiological, pharmacological and epidemiological data, supported the association between Anorexia Nervosa (AN) and Obsessive-Compulsive Disorder (OCD). The aim of the present study is to test the hypothesis of partial common genetic background of both disease, AN and OCD. A total of 116 patients with AN, 74 patients with OCD and 91 controls participated in this study. 213 single-nucleotide polymorphisms (SNPs) in 28 candidate genes were analyzed. Five SNPs achieved 0.004 (the nominal p-value expected by chance), 3 with empirical significant p-values (rs10070190 (CDH9) p = 1 × 10(-3), rs4825476 (GRIA3) p = 4 × 10(-4), and rs1074815 (TPH2) p = 8 × 10(-4)) and 2 additional polymorphisms showing nominal significance (rs2834070 (OLIG2) p = 2 × 10(-3) and rs11783752 (SCL18A1) p = 3 × 10(-3)), were found to be related to both AN and OCD. In addition, rs3825885 (NTRK3, p = 9 × 10(-4)) was identified as an AN risk variant, and rs11179027 (TPH2, p = 2 × 10(-3)) as an OCD marker. The ROC analysis confirmed these results and showed interaction among the significant SNPs. The preliminary results we report here reveal a partial common genetic background in AN and OCD, in agreement with previous clinical findings of common symptomathology between these two diseases and open the field of possible treatments for AN. The interaction observed between the associated polymorphisms, could indicate that there is a biological interaction between the serotonin (TPH2 and SLC18A1) and glutamate (GRIA3) pathways and the factors related to neurogenesis (CDH9, OLIG2 and NTRK3) for the explanation of etiopathophysiology in both diseases. However, the results must be replicated in studies with larger cohorts in order to confirm these associations.
Collapse
|
28
|
|
29
|
|
30
|
Stranahan AM, Martin B, Chadwick W, Park SS, Wang L, Becker KG, WoodIII WH, Zhang Y, Maudsley S. Metabolic context regulates distinct hypothalamic transcriptional responses to antiaging interventions. Int J Endocrinol 2012; 2012:732975. [PMID: 22934110 PMCID: PMC3427989 DOI: 10.1155/2012/732975] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2012] [Accepted: 03/09/2012] [Indexed: 01/19/2023] Open
Abstract
The hypothalamus is an essential relay in the neural circuitry underlying energy metabolism that needs to continually adapt to changes in the energetic environment. The neuroendocrine control of food intake and energy expenditure is associated with, and likely dependent upon, hypothalamic plasticity. Severe disturbances in energy metabolism, such as those that occur in obesity, are therefore likely to be associated with disruption of hypothalamic transcriptomic plasticity. In this paper, we investigated the effects of two well-characterized antiaging interventions, caloric restriction and voluntary wheel running, in two distinct physiological paradigms, that is, diabetic (db/db) and nondiabetic wild-type (C57/Bl/6) animals to investigate the contextual sensitivity of hypothalamic transcriptomic responses. We found that, both quantitatively and qualitatively, caloric restriction and physical exercise were associated with distinct transcriptional signatures that differed significantly between diabetic and non-diabetic mice. This suggests that challenges to metabolic homeostasis regulate distinct hypothalamic gene sets in diabetic and non-diabetic animals. A greater understanding of how genetic background contributes to hypothalamic response mechanisms could pave the way for the development of more nuanced therapeutics for the treatment of metabolic disorders that occur in diverse physiological backgrounds.
Collapse
Affiliation(s)
- Alexis M. Stranahan
- Physiology Department, Georgia Health Sciences University, Augusta, GA 30912, USA
- *Alexis M. Stranahan:
| | - Bronwen Martin
- Metabolism Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Wayne Chadwick
- Receptor Pharmacology Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Sung-Soo Park
- Receptor Pharmacology Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Liyun Wang
- Receptor Pharmacology Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Kevin G. Becker
- Gene Expression and Genomics Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - William H. WoodIII
- Gene Expression and Genomics Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Yongqing Zhang
- Gene Expression and Genomics Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| | - Stuart Maudsley
- Receptor Pharmacology Unit, National Institute on Aging Intramural Research Program, Baltimore, MD 21224-6825, USA
| |
Collapse
|
31
|
Tang C, Zelenak C, Völkl J, Eichenmüller M, Regel I, Fröhlich H, Kempe D, Jimenez L, Le Bellego L, Vergne S, Lang F. Hydration-sensitive gene expression in brain. Cell Physiol Biochem 2011; 27:757-68. [PMID: 21691093 DOI: 10.1159/000330084] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2011] [Indexed: 12/16/2022] Open
Abstract
Dehydration has a profound influence on neuroexcitability. The mechanisms remained, however, incompletely understood. The present study addressed the effect of water deprivation on gene expression in the brain. To this end, animals were exposed to a 24 hours deprivation of drinking water and neuronal gene expression was determined by microarray technology with subsequent confirmation by RT-PCR. As a result, water deprivation was followed by significant upregulation of clathrin (light polypeptide Lcb), serum/glucocorticoid-regulated kinase (SGK) 1, and protein kinase A (PRKA) anchor protein 8-like. Water deprivation led to downregulation of janus kinase and microtubule interacting protein 1, neuronal PAS domain protein 4, thrombomodulin, purinergic receptor P2Y - G-protein coupled 13 gene, gap junction protein beta 1, neurotrophin 3, hyaluronan and proteoglycan link protein 1, G protein-coupled receptor 19, CD93 antigen, forkhead box P1, suppressor of cytokine signaling 3, apelin, immunity-related GTPase family M, serine (or cysteine) peptidase inhibitor clade B member 1a, serine (or cysteine) peptidase inhibitor clade H member 1, glutathion peroxidase 8 (putative), discs large (Drosophila) homolog-associated protein 1, zinc finger and BTB domain containing 3, and H2A histone family member V. Western blotting revealed the downregulation of forkhead box P1, serine (or cysteine) peptidase inhibitor clade H member 1, and gap junction protein beta 1 protein abundance paralleling the respective alterations of transcript levels. In conclusion, water deprivation influences the transcription of a wide variety of genes in the brain, which may participate in the orchestration of brain responses to water deprivation.
Collapse
Affiliation(s)
- Cai Tang
- Department of Physiology, University of Tübingen, Gmelinstr. 5, D-72076 Tübingen
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kindler J, Bailer U, de Zwaan M, Fuchs K, Leisch F, Grün B, Strnad A, Stojanovic M, Windisch J, Lennkh-Wolfsberg C, El-Giamal N, Sieghart W, Kasper S, Aschauer H. No association of the neuropeptide Y (Leu7Pro) and ghrelin gene (Arg51Gln, Leu72Met, Gln90Leu) single nucleotide polymorphisms with eating disorders. Nord J Psychiatry 2011; 65:203-7. [PMID: 21047193 DOI: 10.3109/08039488.2010.525258] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
BACKGROUND Genetic factors likely contribute to the biological vulnerability of eating disorders. AIMS Case-control association study on one neuropeptide Y gene (Leu7Pro) polymorphism and three ghrelin gene (Arg51Gln, Leu72Met and Gln90Leu) polymorphisms. METHODS 114 eating disorder patients (46 with anorexia nervosa, 30 with bulimia nervosa, 38 with binge eating disorder) and 164 healthy controls were genotyped. RESULTS No differences were detected between patients and controls for any of the four polymorphisms in allele frequency and genotype distribution (P > 0.05). Allele frequencies and genotypes had no significant influence on body mass index (P > 0.05) in eating disorder patients. CONCLUSION Positive findings of former case-control studies of associations between ghrelin gene polymorphisms and eating disorders could not be replicated. Neuropeptide Y gene polymorphisms have not been investigated in eating disorders before.
Collapse
Affiliation(s)
- Jochen Kindler
- Department of Psychiatry and Psychotherapy, Division of Biological Psychiatry, Medical University Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Levran O, Peles E, Hamon S, Randesi M, Zhao C, Zhang B, Adelson M, Kreek MJ. Nerve growth factor β polypeptide (NGFB) genetic variability: association with the methadone dose required for effective maintenance treatment. THE PHARMACOGENOMICS JOURNAL 2011; 12:319-27. [PMID: 21358750 PMCID: PMC3130093 DOI: 10.1038/tpj.2011.6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Opioid addiction is a chronic disease with high genetic contribution and a large inter-individual variability in therapeutic response. The goal of this study was to identify pharmacodynamic factors that modulate methadone dose requirement. The neurotrophin family is involved in neural plasticity, learning, memory and behavior and deregulated neural plasticity may underlie the pathophysiology of drug addiction. Brain-derived neurotrophic factor (BDNF) was shown to affect the response to methadone maintenance treatment. This study explores the effects of polymorphisms in the nerve growth factor (β polypeptide) gene, NGFB, on the methadone doses required for successful maintenance treatment for heroin addiction. Genotypes of 14 NGFB polymorphisms were analyzed for association with the stabilizing methadone dose in 72 former severe heroin addicts with no major co-medications. There was significant difference in methadone doses required by subjects with different genotypes of the NGFB intronic single-nucleotide polymorphism rs2239622 (P=0.0002). These results may have clinical importance.
Collapse
Affiliation(s)
- O Levran
- Laboratory of the Biology of Addictive Diseases, The Rockefeller University, New York, NY 10065, USA.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Intron 12 in NTRK3 is associated with bipolar disorder. Psychiatry Res 2011; 185:358-62. [PMID: 20554328 DOI: 10.1016/j.psychres.2010.05.011] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2009] [Revised: 04/19/2010] [Accepted: 05/11/2010] [Indexed: 11/22/2022]
Abstract
Based on the important role of neurotrophic factors in brain development and plasticity and reports of association between schizophrenia and the gene neurotrophic tyrosine kinase receptor 3 (NTRK3), we investigated associations of bipolar disorder with polymorphisms in NTRK3. Recently, our group reported evidence for a possible association of NTRK3 polymorphisms with hippocampal function and schizophrenia. In the present study, we used a homogenous Norwegian case-control sample (the TOP study) consisting of 194 patients diagnosed with bipolar disorder and 336 healthy controls genotyped on the Affymetrix Genome-wide Human SNP Array 6.0. In total 149 markers were investigated for SNP-disease association. Polymorphisms in over 20 markers were nominally associated with bipolar disorder, covering intron 5 to intron 12. Interestingly, our markers appeared to be located close or within the linkage regions reported in schizophrenia, early-onset major depressive disorder and eating disorder, supporting the hypothesis that some genes influence risk beyond traditional diagnostic boundaries.
Collapse
|
35
|
McElroy SL, Frye MA, Hellemann G, Altshuler L, Leverich GS, Suppes T, Keck PE, Nolen WA, Kupka R, Post RM. Prevalence and correlates of eating disorders in 875 patients with bipolar disorder. J Affect Disord 2011; 128:191-8. [PMID: 20674033 DOI: 10.1016/j.jad.2010.06.037] [Citation(s) in RCA: 105] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2010] [Accepted: 06/29/2010] [Indexed: 01/03/2023]
Abstract
OBJECTIVE Relatively little is known about the co-occurrence of bipolar and eating disorders. We therefore assessed the prevalence and clinical correlates of eating disorders in 875 patients with bipolar disorder. METHOD 875 outpatients with DSM-IV bipolar I or II disorder were evaluated with structured diagnostic interviews and clinician- and self-administered questionnaires to determine bipolar and eating disorder diagnoses, other comorbid Axis I disorder diagnoses, and demographic and historical illness characteristics. RESULTS 125 (14.3%) patients met DSM-IV criteria for at least one comorbid lifetime Axis I eating disorder, with binge eating disorder (N=77) being more common than bulimia nervosa (n=42) and anorexia nervosa (N=27). There were no significant eating disorder comorbidity differences between bipolar I and bipolar II patients. Presence of a lifetime comorbid eating disorder was associated with female gender, younger age, earlier age of onset of mood symptoms and of bipolar disorder, presentation in a mixed episode, greater number of prior mood episodes, history of rapid cycling and suicide attempts, greater mean BMI, obesity and severe obesity, and family history of depression, bipolar disorder, alcoholism, and drug abuse. When the three eating disorder groups were compared, lifetime anorexia nervosa was associated with normal weight and a lifetime anxiety disorder, lifetime bulimia nervosa was associated with overweight, and lifetime binge eating disorder was associated with obesity and severe obesity. CONCLUSIONS Patients with bipolar disorder, especially women, not infrequently have comorbid eating disorders, and this comorbidity is associated with an earlier age of onset and more severe course of bipolar illness.
Collapse
Affiliation(s)
- Susan L McElroy
- Craig and Frances Lindner Center of HOPE, Mason, OH 45040, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Caronia LM, Martin C, Welt CK, Sykiotis GP, Quinton R, Thambundit A, Avbelj M, Dhruvakumar S, Plummer L, Hughes VA, Seminara SB, Boepple PA, Sidis Y, Crowley WF, Martin KA, Hall JE, Pitteloud N. A genetic basis for functional hypothalamic amenorrhea. N Engl J Med 2011; 364:215-25. [PMID: 21247312 PMCID: PMC3045842 DOI: 10.1056/nejmoa0911064] [Citation(s) in RCA: 131] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Functional hypothalamic amenorrhea is a reversible form of gonadotropin-releasing hormone (GnRH) deficiency commonly triggered by stressors such as excessive exercise, nutritional deficits, or psychological distress. Women vary in their susceptibility to inhibition of the reproductive axis by such stressors, but it is unknown whether this variability reflects a genetic predisposition to hypothalamic amenorrhea. We hypothesized that mutations in genes involved in idiopathic hypogonadotropic hypogonadism, a congenital form of GnRH deficiency, are associated with hypothalamic amenorrhea. METHODS We analyzed the coding sequence of genes associated with idiopathic hypogonadotropic hypogonadism in 55 women with hypothalamic amenorrhea and performed in vitro studies of the identified mutations. RESULTS Six heterozygous mutations were identified in 7 of the 55 patients with hypothalamic amenorrhea: two variants in the fibroblast growth factor receptor 1 gene FGFR1 (G260E and R756H), two in the prokineticin receptor 2 gene PROKR2 (R85H and L173R), one in the GnRH receptor gene GNRHR (R262Q), and one in the Kallmann syndrome 1 sequence gene KAL1 (V371I). No mutations were found in a cohort of 422 controls with normal menstrual cycles. In vitro studies showed that FGFR1 G260E, FGFR1 R756H, and PROKR2 R85H are loss-of-function mutations, as has been previously shown for PROKR2 L173R and GNRHR R262Q. CONCLUSIONS Rare variants in genes associated with idiopathic hypogonadotropic hypogonadism are found in women with hypothalamic amenorrhea, suggesting that these mutations may contribute to the variable susceptibility of women to the functional changes in GnRH secretion that characterize hypothalamic amenorrhea. Our observations provide evidence for the role of rare variants in common multifactorial disease. (Funded by the Eunice Kennedy Shriver National Institute of Child Health and Human Development and others; ClinicalTrials.gov number, NCT00494169.).
Collapse
Affiliation(s)
- Lisa M Caronia
- Harvard Center for Reproductive Endocrine Sciences and Reproductive Endocrine Unit and the Department of Medicine, Massachusetts General Hospital, Boston, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Gratacòs M, Escaramís G, Bustamante M, Saus E, Agüera Z, Bayés M, Cellini E, de Cid R, Fernández-Aranda F, Forcano L, González JR, Gorwood P, Hebebrand J, Hinney A, Mercader JM, Nacmias B, Ramoz N, Ribasés M, Ricca V, Romo L, Sorbi S, Versini A, Estivill X. Role of the neurotrophin network in eating disorders' subphenotypes: body mass index and age at onset of the disease. J Psychiatr Res 2010; 44:834-40. [PMID: 20219210 DOI: 10.1016/j.jpsychires.2010.01.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2009] [Revised: 01/15/2010] [Accepted: 01/25/2010] [Indexed: 01/06/2023]
Abstract
Eating disorders (ED) are severe psychiatric diseases that most likely result from, and are sustained by socio-cultural, psychological and biological factors. We explored whether members of the neurotrophin family are disease-modifying factors of quantitative traits, potentially contributing to the outcome or prognosis of the disease. We studied lifetime minimum and maximum body mass index (minBMI and maxBMI) and age at onset of the disease in a sample of 991 ED patients from France, Germany, Italy and Spain and analysed 183 genetic variants located in 10 candidate genes encoding different neurotrophins and their receptors. We used a hierarchical model approach to include prior genetic knowledge of the specific and found that variants in CNTF, in its receptor CNTFR, and in NTRK2 were significantly associated with a lower age at onset of the ED. In addition, one variant in NTRK1 was associated with a higher minBMI. The results suggest that for these two subphenotypes, CNTF, CNTFR, NTRK1 and NTRK2 might act as disease-modifying factors and add preliminary evidence to the global hypothesis that EDs are the result of complex interactions and reciprocal controls between the immune, endocrine and central nervous systems.
Collapse
Affiliation(s)
- Mònica Gratacòs
- CIBER en Epidemiología y Salud Pública (CIBERESP), Genes and Disease Program, Center for Genomic Regulation (CRG-UPF), Barcelona, Catalonia, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Mercader JM, Fernández-Aranda F, Gratacòs M, Aguera Z, Forcano L, Ribasés M, Villarejo C, Estivill X. Correlation of BDNF blood levels with interoceptive awareness and maturity fears in anorexia and bulimia nervosa patients. J Neural Transm (Vienna) 2010; 117:505-12. [DOI: 10.1007/s00702-010-0377-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2009] [Accepted: 01/27/2010] [Indexed: 01/15/2023]
|
39
|
Susceptibility to stress in transgenic mice overexpressing TrkC, a model of panic disorder. J Psychiatr Res 2010; 44:157-67. [PMID: 19698958 DOI: 10.1016/j.jpsychires.2009.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2009] [Revised: 07/24/2009] [Accepted: 07/27/2009] [Indexed: 12/21/2022]
Abstract
Stressful life events increase the susceptibility for subsequent onset of psychiatric disorders in humans. Previous research has implicated neurotrophins in the onset of some stress-related diseases, such as major depression disorder, post-traumatic stress disorder or panic disorder. We have tested the hypothesis that the neurotrophin-3 (NT-3)/TrkC system is a genetic interface mediating the deleterious effects of stress on the initiation of panic disorder and other pathologies. To this aim, we have analyzed the functionality of HPA axis and the behavioral consequences of different types of stressful conditions in a mouse model of panic disorder, which overexpresses TrkC, the high affinity-receptor for NT-3 (TgNTRK3). Our results reveal that TgNTRK3 mice exhibit an altered circadian corticosterone rhythm that is reversed by clonidine treatment, but normal expression of genes involved in the control of the hypothalamus-pituitary-adrenal (HPA) axis (CRH, GR) and normal corticosterone response to acute and chronic stressors. In contrast, they exhibit an altered pattern of activation of stress-related brain areas and showed enhanced anxiety-related behavior and more passive strategies than wild types under some chronic stress conditions. We conclude that TgNTRK3 mice present differences in their response to stress characterized by subtle changes in the HPA axis, marked changes in acute stress-induced brain activation and altered coping strategies, suggesting a key role of TrkC receptor in the stress neural circuitry and in the behavioral consequences of chronic stress.
Collapse
|
40
|
Eating disorders: the current status of molecular genetic research. Eur Child Adolesc Psychiatry 2010; 19:211-26. [PMID: 20033240 PMCID: PMC2839487 DOI: 10.1007/s00787-009-0085-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2009] [Accepted: 12/04/2009] [Indexed: 12/31/2022]
Abstract
Anorexia nervosa (AN) and bulimia nervosa (BN) are complex disorders characterized by disordered eating behavior where the patient's attitude towards weight and shape, as well as their perception of body shape, are disturbed. Formal genetic studies on twins and families suggested a substantial genetic influence for AN and BN. Candidate gene studies have initially focused on the serotonergic and other central neurotransmitter systems and on genes involved in body weight regulation. Hardly any of the positive findings achieved in these studies were unequivocally confirmed or substantiated in meta-analyses. This might be due to too small sample sizes and thus low power and/or the genes underlying eating disorders have not yet been analyzed. However, some studies that also used subphenotypes (e.g., restricting type of AN) led to more specific results; however, confirmation is as yet mostly lacking. Systematic genome-wide linkage scans based on families with at least two individuals with an eating disorder (AN or BN) revealed initial linkage regions on chromosomes 1, 3 and 4 (AN) and 10p (BN). Analyses on candidate genes in the chromosome 1 linkage region led to the (as yet unconfirmed) identification of certain variants associated with AN. Genome-wide association studies are under way and will presumably help to identify genes and pathways involved in these eating disorders. The elucidation of the molecular mechanisms underlying eating disorders might improve therapeutic approaches.
Collapse
|
41
|
Hinney A, Scherag S, Hebebrand J. Genetic findings in anorexia and bulimia nervosa. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2010; 94:241-70. [PMID: 21036328 DOI: 10.1016/b978-0-12-375003-7.00009-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Anorexia nervosa (AN) and bulimia nervosa (BN) are complex disorders associated with disordered eating behavior. Heritability estimates derived from twin and family studies are high, so that substantial genetic influences on the etiology can be assumed for both. As the monoaminergic neurotransmitter systems are involved in eating disorders (EDs), candidate gene studies have centered on related genes; additionally, genes relevant for body weight regulation have been considered as candidates. Unfortunately, this approach has yielded very few positive results; confirmed associations or findings substantiated in meta-analyses are scant. None of these associations can be considered unequivocally validated. Systematic genome-wide approaches have been performed to identify genes with no a priori evidence for their relevance in EDs. Family-based scans revealed linkage peaks in single chromosomal regions for AN and BN. Analyses of candidate genes in one of these regions led to the identification of genetic variants associated with AN. Currently, an international consortium is conducting a genome-wide association study for AN, which will hopefully lead to the identification of the first genome-wide significant markers.
Collapse
Affiliation(s)
- Anke Hinney
- Department of Child and Adolescent Psychiatry and Psychotherapy, University of Duisburg-Essen, Germany
| | | | | |
Collapse
|
42
|
Functional variants of the serotonin receptor type 3A and B gene are associated with eating disorders. Pharmacogenet Genomics 2009; 19:790-9. [PMID: 19741568 DOI: 10.1097/fpc.0b013e32833132b3] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
OBJECTIVE As a key player in modulating both human physiological and behavioural functions including anxiety, perception and in particular appetite, serotonin (5-hydroxytryptamine, 5-HT) is likely to be involved in the aetiology of eating disorders. Studies showing serotonin receptor type 3 (5-HT3) receptors to mediate food intake depression (anorexic response) have triggered our interest in investigating the putative role of variants in the 5-HT3 receptor genes, HTR3A and HTR3B, in the susceptibility to anorexia nervosa (AN) and bulimia nervosa (BN). METHODS Two hundred and sixty-five patients with AN and 91 patients with BN as well as 191 healthy controls served as a pilot study group for mutational analysis by direct sequencing. Variants showing a significant association were subsequently genotyped in an independent Spanish cohort of 78 patients with AN and 119 patients with BN as well as 331 healthy controls for replication purposes. RESULTS In the pilot study, we found the coding HTR3B variant, p.Y129S, (rs1176744, P = 0.004, odds ratio = 2.06) to be associated with the restrictive subtype of AN. The association was confirmed in the Spanish study group (P = 0.034, odds ratio = 2.26). CONCLUSION Our study provides first evidence for an involvement of 5-HT3 variants in the aetiopathology of eating disorders in humans.
Collapse
|
43
|
de Krom M, Bauer F, Collier D, Adan RAH, la Fleur SE. Genetic variation and effects on human eating behavior. Annu Rev Nutr 2009; 29:283-304. [PMID: 19400703 DOI: 10.1146/annurev-nutr-080508-141124] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Feeding is a physiological process, influenced by genetic factors and the environment. In recent years, many studies have been performed to unravel the involvement of genetics in both eating behavior and its pathological forms: eating disorders and obesity. In this review, we provide a condensed introduction on the neurological aspects of eating and we describe the current status of research into the genetics of eating behavior, primarily focused on specific traits such as taste, satiation, and hunger. This is followed by an overview on the genetic studies done to unravel the heritable background of obesity and eating disorders. We examine the discussion currently taking place in the field of genetics of complex disorders and phenotypes on how to perform good and powerful studies, with the use of large-scale whole-genome association studies as one of the possible solutions. In the final part of this review, we give our view on the latest developments, including endophenotype approaches and animal studies. Studies of endophenotypes of eating behavior may help to identify core traits that are genetically influenced. Such studies would yield important knowledge on the underlying biological scaffold on which diagnostic criteria for eating disorders could be based and would provide information to influence eating behavior toward healthier living.
Collapse
Affiliation(s)
- Mariken de Krom
- Rudolf Magnus Institute of Neuroscience, Department of Neuroscience & Pharmacology, UMC Utrecht, Utrecht, The Netherlands.
| | | | | | | | | |
Collapse
|
44
|
Allele variants in functional MicroRNA target sites of the neurotrophin-3 receptor gene (NTRK3) as susceptibility factors for anxiety disorders. Hum Mutat 2009; 30:1062-71. [DOI: 10.1002/humu.21005] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
45
|
Komaki G, Moriguchi Y, Ando T, Yoshiuchi K, Nakao M. Prospects of psychosomatic medicine. Biopsychosoc Med 2009; 3:1. [PMID: 19161633 PMCID: PMC2642858 DOI: 10.1186/1751-0759-3-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2009] [Accepted: 01/22/2009] [Indexed: 11/10/2022] Open
Affiliation(s)
- Gen Komaki
- Department of Psychosomatic Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Yoshiya Moriguchi
- Department of Psychosomatic Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
- Department of Psychology, Boston College, Boston, USA
| | - Tetsuya Ando
- Department of Psychosomatic Research, National Institute of Mental Health, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kazuhiro Yoshiuchi
- Department of Stress Sciences and Psychosomatic Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mutsuhiro Nakao
- Department of Hygiene and Public Health, Teikyo University School of Medicine, Itabashi, Tokyo, Japan
- Division of Psychosomatic Medicine, Teikyo University Hospital, Itabashi, Tokyo, Japan
| |
Collapse
|