1
|
Værøy H, Skar-Fröding R, Hareton E, Fetissov SO. Possible roles of neuropeptide/transmitter and autoantibody modulation in emotional problems and aggression. Front Psychiatry 2024; 15:1419574. [PMID: 39381606 PMCID: PMC11458397 DOI: 10.3389/fpsyt.2024.1419574] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/30/2024] [Indexed: 10/10/2024] Open
Abstract
The theoretical foundations of understanding psychiatric disorders are undergoing changes. Explaining behaviour and neuroendocrine cell communication leaning towards immunology represents a different approach compared to previous models for understanding complex central nervous system processes. One such approach is the study of immunoglobulins or autoantibodies, and their effect on peptide hormones in the neuro-endocrine system. In the present review, we provide an overview of the literature on neuropeptide/transmitter and autoantibody modulation in psychiatric disorders featuring emotional problems and aggression, including associated illness behaviour. Finally, we discuss the role of psycho-immunology as a growing field in the understanding of psychiatric disorders, and that modulation and regulation by IgG autoAbs represent a relatively new subcategory in psycho-immunology, where studies are currently being conducted.
Collapse
Affiliation(s)
- Henning Værøy
- R&D Department, Division of Mental Health Services, Akershus University Hospital, Lørenskog, Norway
| | - Regina Skar-Fröding
- R&D Department, Division of Mental Health Services, Akershus University Hospital, Lørenskog, Norway
| | - Elin Hareton
- Department of Multidiciplinary Laboratory Medicine and Medical Biochemistry, (TLMB), Akershus University Hospital, Lørenskog, Norway
| | - Sergueï O. Fetissov
- Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| |
Collapse
|
2
|
Seitz J, Lahaye E, Andreani NA, Thomas B, Takhlidjt S, Chartrel N, Herpertz-Dahlmann B, Baines JF, Fetissov SO. Long-Term Dynamics of Serum α-MSH and α-MSH-Binding Immunoglobulins with a Link to Gut Microbiota Composition in Patients with Anorexia Nervosa. Neuroendocrinology 2024; 114:907-920. [PMID: 38852579 PMCID: PMC11460951 DOI: 10.1159/000539316] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 05/06/2024] [Indexed: 06/11/2024]
Abstract
INTRODUCTION Immunoglobulins (Ig) reactive with α-melanocyte-stimulating hormone (α-MSH), an anorexigenic neuropeptide, are present in humans and were previously associated with eating disorders. In this longitudinal study involving patients with anorexia nervosa (AN), we determined whether α-MSH in serum is bound to IgG and analyzed long-term dynamics of both α-MSH peptide and α-MSH-reactive Ig in relation to changes in BMI and gut microbiota composition. METHODS The study included 64 adolescents with a restrictive form of AN, whose serum samples were collected at hospital admission, discharge, and during a 1-year follow-up visit and 41 healthy controls, all females. RESULTS We found that in both study groups, approximately 40% of serum α-MSH was reversibly bound to IgG and that levels of α-MSH-reactive IgG but not of α-MSH peptide in patients with AN were low at hospital admission but recovered 1 year later. Total IgG levels were also low at admission. Moreover, BMI-standard deviation score correlated positively with α-MSH IgG in both groups studied but negatively with α-MSH peptide only in controls. Significant correlations between the abundance of specific bacterial taxa in the gut microbiota and α-MSH peptide and IgG levels were found in both study groups, but they were more frequent in controls. CONCLUSION We conclude that IgG in the blood plays a role as an α-MSH-binding protein, whose characteristics are associated with BMI in both patients with AN and controls. Furthermore, the study suggests that low production of α-MSH-reactive IgG during the starvation phase in patients with AN may be related to altered gut microbiota composition.
Collapse
Affiliation(s)
- Jochen Seitz
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University Hospital, RWTH University Aachen, Aachen, Germany
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, LVR-University Hospital, Essen, Germany
| | - Emilie Lahaye
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Nadia Andrea Andreani
- Guest Group Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Benjamin Thomas
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Saloua Takhlidjt
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Nicolas Chartrel
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| | - Beate Herpertz-Dahlmann
- Department of Child and Adolescent Psychiatry, Psychotherapy and Psychosomatics, University Hospital, RWTH University Aachen, Aachen, Germany
| | - John F. Baines
- Guest Group Evolutionary Medicine, Max Planck Institute for Evolutionary Biology, Plön, Germany
- Section of Evolutionary Medicine, Institute for Experimental Medicine, Kiel University, Kiel, Germany
| | - Sergueï O. Fetissov
- Regulatory Peptides – Energy Metabolism and Motivated Behavior Team, Neuroendocrine, Endocrine and Germinal Differentiation and Communication Laboratory, Inserm UMR1239, University of Rouen Normandie, Rouen, France
| |
Collapse
|
3
|
Landini L, Dadson P, Gallo F, Honka MJ, Cena H. Microbiota in anorexia nervosa: potential for treatment. Nutr Res Rev 2023; 36:372-391. [PMID: 35875979 DOI: 10.1017/s0954422422000130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Anorexia nervosa (AN) is characterised by the restriction of energy intake in relation to energy needs and a significantly lowered body weight than normally expected, coupled with an intense fear of gaining weight. Treatment of AN is currently based on psychological and refeeding approaches, but their efficacy remains limited since 40% of patients after 10 years of medical care still present symptoms of AN. The intestine hosts a large community of microorganisms, called the "microbiota", which live in symbiosis with the human host. The gut microbiota of a healthy human is dominated by bacteria from two phyla: Firmicutes and, majorly, Bacteroidetes. However, the proportion in their representation differs on an individual basis and depends on many external factors including medical treatment, geographical location and hereditary, immunological and lifestyle factors. Drastic changes in dietary intake may profoundly impact the composition of the gut microbiota, and the resulting dysbiosis may play a part in the onset and/or maintenance of comorbidities associated with AN, such as gastrointestinal disorders, anxiety and depression, as well as appetite dysregulation. Furthermore, studies have reported the presence of atypical intestinal microbial composition in patients with AN compared with healthy normal-weight controls. This review addresses the current knowledge about the role of the gut microbiota in the pathogenesis and treatment of AN. The review also focuses on the bidirectional interaction between the gastrointestinal tract and the central nervous system (microbiota-gut-brain axis), considering the potential use of the gut microbiota manipulation in the prevention and treatment of AN.
Collapse
Affiliation(s)
- Linda Landini
- S.S.D. Dietetics and Clinical Nutrition ASL 4 Chiavarese Liguria-Sestri Levante Hospital, Sestri Levante, Italy
| | - Prince Dadson
- Turku PET Centre, University of Turku, Turku, Finland
| | - Fabrizio Gallo
- S.S.D. Dietetics and Clinical Nutrition ASL 4 Chiavarese Liguria-Sestri Levante Hospital, Sestri Levante, Italy
| | | | - Hellas Cena
- Dietetics and Clinical Nutrition Laboratory, Department of Public Health, Experimental and Forensic Medicine, University of Pavia, Pavia, Italy
- Clinical Nutrition and Dietetics Service, Unit of Internal Medicine and Endocrinology, ICS Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
4
|
Iervasi E, Strangio A, Greco L, Auricchio R, Saverino D. Circulating anti-hypothalamus antibodies in celiac patients: tissue transglutaminase friend or foe? Immunol Res 2023; 71:839-848. [PMID: 37221348 PMCID: PMC10667380 DOI: 10.1007/s12026-023-09394-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2022] [Accepted: 05/09/2023] [Indexed: 05/25/2023]
Abstract
Celiac disease (CD) is an autoimmune disease with inflammatory characteristics, having a condition of chronic malabsorption, affecting approximately 1% of the population at any age. In recent years, a concrete correlation between eating disorders and CD has emerged. Hypothalamus plays a central role in determining eating behaviour, regulating appetite and, consequently, food intake. One hundred and ten sera from celiac patients (40 active and 70 following a gluten-free diet) were tested for the presence of autoantibodies against primate hypothalamic periventricular neurons by immunofluorescence and by a home-made ELISA assay. In addition, ghrelin was measured by ELISA. As control, 45 blood serums from healthy age matched were analysed. Among active CD, all patients resulted positive for anti-hypothalamus autoantibodies and sera showed significantly higher levels of ghrelin. All of the free-gluten CD were negative for anti-hypothalamus autoantibodies and had low levels of ghrelin, as well as healthy controls. Of interest, anti-hypothalamic autoantibodies directly correlate with anti-tTG amounts and with mucosal damage. In addition, competition assays with recombinant tTG showed a drastically reduction of anti-hypothalamic serum reactivity. Finally, ghrelin levels are increased in CD patients and correlated with anti-tTG autoantibodies and anti-hypothalamus autoantibodies. This study demonstrates for the first time the presence of anti-hypothalamus antibodies and their correlation with the severity of the CD. It also allows us to hypothesize the role of tTG as a putative autoantigen expressed by hypothalamic neurons.
Collapse
Affiliation(s)
- Erika Iervasi
- Department of Experimental Medicine, University of Genoa, Via De Toni, 14, 16132, Genova, Italy
- Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy
| | - Antonella Strangio
- Department of Experimental Medicine, University of Genoa, Via De Toni, 14, 16132, Genova, Italy
| | - Luigi Greco
- Department of Translational Medical Science, University of Naples Federico II, Corso Umberto I, 40, 80138, Napoli, Italy
| | - Renata Auricchio
- Department of Translational Medical Science, University of Naples Federico II, Corso Umberto I, 40, 80138, Napoli, Italy
- European Laboratory for Food-Induced disease (ELFID), University of Naples Federico II, Via Pansini, 5, 80131, Napoli, Italy
| | - Daniele Saverino
- Department of Experimental Medicine, University of Genoa, Via De Toni, 14, 16132, Genova, Italy.
- Ospedale Policlinico San Martino, Largo Rosanna Benzi, 10, 16132, Genova, Italy.
| |
Collapse
|
5
|
Amerio A, Escelsior A, Martino E, Strangio A, Aguglia A, Marcatili M, Conio B, Sukkar SG, Saverino D. The Association between Blood SIRT1 and Ghrelin, Leptin, and Antibody Anti-Hypothalamus: A Comparison in Normal Weight and Anorexia Nervosa. J Pers Med 2023; 13:928. [PMID: 37373917 PMCID: PMC10303472 DOI: 10.3390/jpm13060928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 05/12/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Sirtuin 1 (SIRT1) is a sensor of cell energy availability, regulating metabolic homeostasis as well as leptin and ghrelin, and it could be considered as a potential plasmatic marker. The aim of this study was to assess whether circulating SIRT1 varies consistently with leptin, ghrelin, body mass index (BMI), and IgG reactive to hypothalamic antigens in anorexia nervosa (AN). Fifty-four subjects were evaluated: 32 with AN and 22 normal-weight control subjects. Serum levels of SIRT1, leptin, ghrelin, and IgG reactive to hypothalamic antigens were evaluated by ELISA. Results showed that serum SIRT1 is increased in patients with AN, and the amount is decreased in relation to the duration of the illness. SIRT1 concentration approaches the values obtained for the control group, although the difference is still statistically significant. A negative correlation between serum SIRT1 values and leptin or BMI values has been found. On the contrary, a positive correlation between SIRT1 and ghrelin or IgG specific for hypothalamic antigens is reported. These findings suggest that a peripheral evaluation of SIRT1 could be a possible clinical/biochemical parameter related to AN. In addition, we can assume that SIRT1 is related to autoantibody production and may correlate with the intensity/severity of AN. Thus, reducing the production of autoantibodies specific for hypothalamic cells could be a sign of improvement of the clinical condition.
Collapse
Affiliation(s)
- Andrea Amerio
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), Section of Psychiatry, University of Genoa, 16132 Genoa, Italy; (A.A.); (A.E.); (A.A.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (B.C.); (S.G.S.)
| | - Andrea Escelsior
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), Section of Psychiatry, University of Genoa, 16132 Genoa, Italy; (A.A.); (A.E.); (A.A.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (B.C.); (S.G.S.)
| | - Eleonora Martino
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (B.C.); (S.G.S.)
- Dietetics and Clinical Nutrition Unit, University of Genoa, 16132 Genoa, Italy
| | - Antonella Strangio
- Department of Experimental Medicine (DiMeS), Section of Human Anatomy, University of Genoa, 16132 Genoa, Italy;
| | - Andrea Aguglia
- Department of Neuroscience, Rehabilitation, Ophthalmology, Genetics, Maternal and Child Health (DiNOGMI), Section of Psychiatry, University of Genoa, 16132 Genoa, Italy; (A.A.); (A.E.); (A.A.)
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (B.C.); (S.G.S.)
| | - Matteo Marcatili
- Department of Mental Health, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy;
| | - Benedetta Conio
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (B.C.); (S.G.S.)
| | - Samir Giuseppe Sukkar
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (B.C.); (S.G.S.)
- Dietetics and Clinical Nutrition Unit, University of Genoa, 16132 Genoa, Italy
| | - Daniele Saverino
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (E.M.); (B.C.); (S.G.S.)
- Department of Experimental Medicine (DiMeS), Section of Human Anatomy, University of Genoa, 16132 Genoa, Italy;
| |
Collapse
|
6
|
Anorexia nervosa and microbiota: systematic review and critical appraisal. Eat Weight Disord 2023; 28:1. [PMID: 36752887 PMCID: PMC9908645 DOI: 10.1007/s40519-023-01529-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 01/04/2023] [Indexed: 02/09/2023] Open
Abstract
PURPOSE Recent studies have reported a gut microbiota imbalance or dysbiosis associated with anorexia nervosa (AN), which has prompted an appraisal of its aetiological role, and the reformulation of AN as a metabo-psychiatric disorder. Thus, the aim of this paper was to critically review the current scientific findings regarding the role of microbiota in anorexia nervosa. METHODS A systematic study of peer-reviewed literature published in four databases between 2009 and 2022 was conducted according to PRISMA guidelines. Both human and animal studies were included. RESULTS A total of 18 studies were included. In animal models, both the preclinical and clinical findings were inconsistent regarding microbiota composition, faecal metabolite concentrations, and the effects of human faecal microbiota transplants. CONCLUSION The methodological limitations, lack of standardisation, and conceptual ambiguity hinder the analysis of microbiota as a key explanatory factor for AN. LEVEL OF EVIDENCE Level I, systematic review.
Collapse
|
7
|
Anti-hypothalamus autoantibodies in anorexia nervosa: a possible new mechanism in neuro-physiological derangement? Eat Weight Disord 2022; 27:2481-2496. [PMID: 35297008 PMCID: PMC9556421 DOI: 10.1007/s40519-022-01388-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 02/26/2022] [Indexed: 12/22/2022] Open
Abstract
PURPOSE Anorexia nervosa (AN) is a serious and complex mental disorder affecting mainly young adult women. AN patients are characterized by low body weight in combination with self-induced starvation, intense fear of gaining weight, and distortion of body image. AN is a multifactorial disease, linked by recent evidence to a dysregulation of the immune system. METHODS In this pilot study, 22 blood serums from AN patients were tested for the presence of autoantibodies against primate hypothalamic periventricular neurons by immunofluorescence and by a home-made ELISA assay. Cellular fluorescence suggests the presence of autoantibodies which are able to recognize these neurons (both to body cell and fiber levels). By means of ELISA, these autoantibodies are quantitatively evaluated. In addition, orexigenic and anorexigenic molecules were measured by ELISA. As control, 18 blood serums from healthy age matched woman were analysed. RESULTS All AN patients showed a reactivity against hypothalamic neurons both by immunofluorescence and ELISA. In addition, ghrelin, pro-opiomelanocortin (POMC), and agouti-related peptide (AGRP) were significantly higher than in control serums (p < 0.0001). In contrast, leptin was significantly lower in AN patients than controls (p < 0.0001). CONCLUSIONS Immunoreaction and ELISA assays on AN blood serum suggest the presence of autoantibodies AN related. However, it is not easy to determine the action of these antibodies in vivo: they could interact with specific ligands expressed by hypothalamic cells preventing their physiological role, however, it is also possible that they could induce an aspecific stimulation in the target cells leading to an increased secretion of anorexigenic molecules. Further studies are needed to fully understand the involvement of the immune system in AN pathogenesis. LEVEL OF EVIDENCE V, descriptive study.
Collapse
|
8
|
İNANÇ Y, TURGUT C, KAYA T. EATİNG ATTİTUDE İN MULTİPLE SCLEROSİS. KAHRAMANMARAŞ SÜTÇÜ İMAM ÜNIVERSITESI TIP FAKÜLTESI DERGISI 2022. [DOI: 10.17517/ksutfd.1132269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023] Open
Abstract
Aim: To examine attitudes to eating in multiple sclerosis (MS) patients.
Material and Method: The study included 64 cases diagnosed with MS according to the 2017 McDonald criteria and a control group of 64 healthy volunteers of similar age, gender, and education level. All the study participants underwent a detailed neurological examination and were then administered the Eating Attitudes Test (EAT), the Beck Anxiety Inventory (BAI), and the Beck Depreession Inventory (BDI).
Results: In the comparisons between the groups of the EAT results, a score of ≥30 was obtained by 24 (37.5%) MS patients and 14 (21.8%) of the control group. When the MS patients were compared in two groups according to the Expanded Disability Status Scale (EDSS) score of ≤3 and ≥4, an EAT score of ≥30 was obtained by 11 (25%) patients with EDSS score ≤ 3 and by 13 (68.5%) patients with EDSS ≥4.
Conclusion:The study results demonstrated that the patients with MS had a worse attitude to eating than the control subjects. When it is considered that some foodstuffs are among MS triggers, the presence of eating disorders or poor eating behaviour becomes important.
Collapse
|
9
|
Grigioni S, Achamrah N, Chan P, Guérin C, Bôle-Feysot C, Delay J, Colange G, Quillard M, Coquard A, Bubenheim M, Jésus P, Tavolacci MP, Déchelotte P, Coëffier M. Intestinal permeability and appetite regulating peptides-reactive immunoglobulins in severely malnourished women with anorexia nervosa. Clin Nutr 2022; 41:1752-1758. [PMID: 35810568 DOI: 10.1016/j.clnu.2022.06.036] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 06/07/2022] [Accepted: 06/24/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND & AIMS In the last decades, the role of microbiota-gut-brain axis has emerged in the regulation of eating behavior and in the pathophysiology of anorexia nervosa (AN) that remains poorly understood. Particularly, a gut-derived dysregulation of immune response has been proposed leading to immunoglobulins directed against appetite-regulating peptides. However, intestinal permeability in patients with anorexia nervosa has been poorly documented. METHODS In the present prospective case-control study, we thus compared intestinal permeability, appetite-regulating peptides and their reactive immunoglobulins measured in severely malnourished women with AN (n = 17; 28 [21-35] y; 14.9 [14.1-15.2] kg/m2) to healthy volunteers (HV, n = 34; 26 [23-35] y; 22.3 [20.6-23.6] kg/m2). RESULTS Patients with AN exhibited an increased urinary lactulose/mannitol ratio, both in 0-5 h (0.033 [0.013-0.116]) and 5-24 h samples (0.115 [0.029-0.582]), when compared to HV (0.02 [0.008-0.045], p = 0.0074 and 0.083 [0.019-0.290], p = 0.0174, respectively), suggesting an increased intestinal permeability. Urinary excretion of sucralose and plasma zonulin were not different. The levels of plasma total ghrelin and desacyl-ghrelin were increased in patients with AN compared to HV, whereas plasma leptin concentration was decreased. In addition, αMSH remained unchanged compared to HV. Finally, we did not observe any modification of the levels of total or free αMSH, leptin or ghrelin-reactive immunoglobulin G and M, as well as for their affinity properties. Only, a weak decrease of the dissociation constant (kd) for acyl-ghrelin-reactive IgG was observed in patients with AN (p = 0.0411). CONCLUSIONS In conclusion, severely malnourished patients with AN show a higher intestinal permeability than HV without evidence of an effect on appetite regulating peptides-reactive immunoglobulins.
Collapse
Affiliation(s)
- Sébastien Grigioni
- Department of Nutrition, Rouen University Hospital, CHU Rouen, France; Université de Rouen Normandie, Inserm UMR1073 « Nutrition, Inflammation and Microbiota-gut-brain Axis », Institute for Research and Innovation in Biomedicine, Rouen, France; Clinical Investigation Center CIC 1404 - Biological Resources Centre, Inserm, Rouen University Hospital, CHU Rouen, France
| | - Najate Achamrah
- Department of Nutrition, Rouen University Hospital, CHU Rouen, France; Université de Rouen Normandie, Inserm UMR1073 « Nutrition, Inflammation and Microbiota-gut-brain Axis », Institute for Research and Innovation in Biomedicine, Rouen, France; Clinical Investigation Center CIC 1404 - Biological Resources Centre, Inserm, Rouen University Hospital, CHU Rouen, France
| | - Philippe Chan
- PISSARO Proteomics Platform, HeRacLeS High-tech Research Infrastructures for Life, UMS 51 - UAR 2026, Inserm, CNRS, Université de Rouen Normandie, Rouen, France
| | - Charlène Guérin
- Department of Nutrition, Rouen University Hospital, CHU Rouen, France; Université de Rouen Normandie, Inserm UMR1073 « Nutrition, Inflammation and Microbiota-gut-brain Axis », Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Christine Bôle-Feysot
- Department of Nutrition, Rouen University Hospital, CHU Rouen, France; Université de Rouen Normandie, Inserm UMR1073 « Nutrition, Inflammation and Microbiota-gut-brain Axis », Institute for Research and Innovation in Biomedicine, Rouen, France
| | - Julie Delay
- Department of Nutrition, Rouen University Hospital, CHU Rouen, France
| | - Guillaume Colange
- Department of Nutrition, Rouen University Hospital, CHU Rouen, France
| | - Muriel Quillard
- Université de Rouen Normandie, Inserm UMR1073 « Nutrition, Inflammation and Microbiota-gut-brain Axis », Institute for Research and Innovation in Biomedicine, Rouen, France; Clinical Investigation Center CIC 1404 - Biological Resources Centre, Inserm, Rouen University Hospital, CHU Rouen, France
| | - Aude Coquard
- Department of Pharmacy, Rouen University Hospital, CHU Rouen, France
| | - Michael Bubenheim
- Department of Clinical Research and Innovation, Rouen University Hospital, CHU Rouen, France
| | - Pierre Jésus
- Nutrition Unit, Limoges University Hospital, Inserm UMR 1094 Tropical Neuro-epidemiology, Limoges, France
| | - Marie-Pierre Tavolacci
- Université de Rouen Normandie, Inserm UMR1073 « Nutrition, Inflammation and Microbiota-gut-brain Axis », Institute for Research and Innovation in Biomedicine, Rouen, France; Clinical Investigation Center CIC 1404 - Biological Resources Centre, Inserm, Rouen University Hospital, CHU Rouen, France
| | - Pierre Déchelotte
- Department of Nutrition, Rouen University Hospital, CHU Rouen, France; Université de Rouen Normandie, Inserm UMR1073 « Nutrition, Inflammation and Microbiota-gut-brain Axis », Institute for Research and Innovation in Biomedicine, Rouen, France; Clinical Investigation Center CIC 1404 - Biological Resources Centre, Inserm, Rouen University Hospital, CHU Rouen, France
| | - Moïse Coëffier
- Department of Nutrition, Rouen University Hospital, CHU Rouen, France; Université de Rouen Normandie, Inserm UMR1073 « Nutrition, Inflammation and Microbiota-gut-brain Axis », Institute for Research and Innovation in Biomedicine, Rouen, France; Clinical Investigation Center CIC 1404 - Biological Resources Centre, Inserm, Rouen University Hospital, CHU Rouen, France.
| |
Collapse
|
10
|
Steinhausen H, Villumsen MD, Hørder K, Winkler LA, Bilenberg N, Støving RK. Increased risk of somatic diseases following anorexia nervosa in a controlled nationwide cohort study. Int J Eat Disord 2022; 55:754-762. [PMID: 35451527 PMCID: PMC9323483 DOI: 10.1002/eat.23718] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 03/19/2022] [Accepted: 04/11/2022] [Indexed: 11/22/2022]
Abstract
OBJECTIVE To assess the risk of somatic diseases in connection with anorexia nervosa (AN). METHOD This matched cohort study was based on Danish registries of all patients born 1961-2008 with a first-time diagnosis of AN in 1994-2018 at age 8-32 and matched controls without an eating disorder. For 13 somatic disease categories, time from inclusion date to time of first somatic diagnosis, accounting for censoring, was studied by use of time-stratified Cox models. RESULTS A total of 9985 AN patients born 1961-2008 and 49,351 controls were followed for a median (interquartile range) of 9.0 (4.4-15.7) years. During the first 2 years after entry there was a 60% higher hazard for any somatic disease among patients with AN than among controls, while the ratio from three to 11 years was reduced to 1.18. Regardless of age at diagnosis, the hazard among patients and controls were no different at approximately a decade after diagnosis of AN and the cumulative risk for patients for 12 of 13 disease categories was always higher or no less that for controls. For all disease categories, the hazard ratio (HR) was higher when close to entry. For most disease categories, age at diagnosis of AN did not modify the effect. DISCUSSION While around 90% of all individuals had any somatic disease at the end of follow-up, the cumulative incidence over time was higher for patients with AN than for controls. Large HRs were seen in the early years after diagnosis during which patients require extensive medical interventions. PUBLIC SIGNIFICANCE Based on Danish registries, a large sample of almost 10,000 patients with AN born 1961-2008 and almost 50,000 matched controls were followed for a median of 9 years. While around 90% of all individuals had any somatic disease at the end of follow-up, the cumulative incidence over time was higher for patients with AN than for controls.
Collapse
Affiliation(s)
- Hans‐Christoph Steinhausen
- Department of Child and Adolescent Mental Health Odense, Mental Health Services in the Region of Southern DenmarkUniversity of Southern DenmarkOdenseDenmark,Child and Adolescent Mental Health Centre, Capital Region PsychiatryCopenhagenDenmark,Department of Child and Adolescent PsychiatryPsychiatric University Hospital of ZurichZurichSwitzerland,Clinical Psychology and Epidemiology, Institute of PsychologyUniversity of BaselBaselSwitzerland
| | - Martin Dalgaard Villumsen
- Department of Epidemiology, Biostatistics, and Biodemography, Institute of Public HealthUniversity of Southern DenmarkOdenseDenmark
| | - Kirsten Hørder
- Department of Child and Adolescent Mental Health Odense, Mental Health Services in the Region of Southern DenmarkUniversity of Southern DenmarkOdenseDenmark
| | - Laura Al‐Dakhiel Winkler
- Center for Eating DisordersOdense University HospitalOdenseDenmark,Psychiatric Services in the Region of Southern DenmarkOdenseDenmark
| | - Niels Bilenberg
- Department of Child and Adolescent Mental Health Odense, Mental Health Services in the Region of Southern DenmarkUniversity of Southern DenmarkOdenseDenmark
| | - René Klinkby Støving
- Center for Eating DisordersOdense University HospitalOdenseDenmark,Psychiatric Services in the Region of Southern DenmarkOdenseDenmark,Endocrine Research UnitOdense University HospitalOdenseDenmark
| |
Collapse
|
11
|
Roubalova R, Prochazkova P, Dvorak J, Hill M, Papezova H, Kreisinger J, Bulant J, Lambertova A, Holanova P, Bilej M, Tlaskalova-Hogenova H. Altered Serum Immunological and Biochemical Parameters and Microbiota Composition in Patients With AN During Realimentation. Front Nutr 2021; 8:680870. [PMID: 34409061 PMCID: PMC8365021 DOI: 10.3389/fnut.2021.680870] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
Anorexia nervosa (AN) is a life-threatening psychiatric disorder with not well-described pathogenesis. Besides the genetic and sociological factors, autoimmunity is also considered to take part in AN pathogenesis. We evaluated general serological factors showing the physiological state of 59 patients with AN at hospital admission and their discharge. We detected the altered levels of some general biochemical and immunological parameters. We also detected decreased levels of appetite-regulating alpha-melanocyte stimulating hormone (α-MSH) in patients at hospital admission. Moreover, elevated anti-α-MSH IgM levels and decreased anti-α-MSH IgA levels were observed in patients with AN. Therefore, we analyzed the gut microbiota composition with special focus on α-MSH antigen-mimetic containing microbes from the Enterobacteriaceae family. We correlated gut bacterial composition with anti-α-MSH Ig levels and detected decreasing IgG levels with increasing alpha diversity. The upregulation of pro-inflammatory cytokines IL-6, IL-17, and TNF-α were detected in patients with AN both prior and after hospitalization. We also evaluated the treatment outcome and improvement was observed in the majority of patients with AN. We provide new data about various serum biochemical parameters and their changes during the patients' hospitalization, with emphasis on the immune system, and its possible participation in AN pathogenesis.
Collapse
Affiliation(s)
- Radka Roubalova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Petra Prochazkova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jiri Dvorak
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Martin Hill
- Department of Steroids and Proteohormones, Institute of Endocrinology, Prague, Czechia
| | - Hana Papezova
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Jakub Kreisinger
- Department of Zoology, Faculty of Science, Charles University, Prague, Czechia
| | - Josef Bulant
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia.,Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Alena Lambertova
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Petra Holanova
- Department of Psychiatry, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Martin Bilej
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Helena Tlaskalova-Hogenova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
12
|
The Microbiota and the Gut-Brain Axis in Controlling Food Intake and Energy Homeostasis. Int J Mol Sci 2021; 22:ijms22115830. [PMID: 34072450 PMCID: PMC8198395 DOI: 10.3390/ijms22115830] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/21/2021] [Accepted: 05/26/2021] [Indexed: 12/12/2022] Open
Abstract
Obesity currently represents a major societal and health challenge worldwide. Its prevalence has reached epidemic proportions and trends continue to rise, reflecting the need for more effective preventive measures. Hypothalamic circuits that control energy homeostasis in response to food intake are interesting targets for body-weight management, for example, through interventions that reinforce the gut-to-brain nutrient signalling, whose malfunction contributes to obesity. Gut microbiota-diet interactions might interfere in nutrient sensing and signalling from the gut to the brain, where the information is processed to control energy homeostasis. This gut microbiota-brain crosstalk is mediated by metabolites, mainly short chain fatty acids, secondary bile acids or amino acids-derived metabolites and subcellular bacterial components. These activate gut-endocrine and/or neural-mediated pathways or pass to systemic circulation and then reach the brain. Feeding time and dietary composition are the main drivers of the gut microbiota structure and function. Therefore, aberrant feeding patterns or unhealthy diets might alter gut microbiota-diet interactions and modify nutrient availability and/or microbial ligands transmitting information from the gut to the brain in response to food intake, thus impairing energy homeostasis. Herein, we update the scientific evidence supporting that gut microbiota is a source of novel dietary and non-dietary biological products that may beneficially regulate gut-to-brain communication and, thus, improve metabolic health. Additionally, we evaluate how the feeding time and dietary composition modulate the gut microbiota and, thereby, the intraluminal availability of these biological products with potential effects on energy homeostasis. The review also identifies knowledge gaps and the advances required to clinically apply microbiome-based strategies to improve the gut-brain axis function and, thus, combat obesity.
Collapse
|
13
|
Anorexia nervosa and juvenile lupus erythematosus in a 16-year-old female patient - common disease origin or random coincidence? Cent Eur J Immunol 2021; 46:127-132. [PMID: 33897295 PMCID: PMC8056349 DOI: 10.5114/ceji.2021.104326] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 10/29/2019] [Indexed: 12/03/2022] Open
Abstract
Adolescence is a period in which eating disorders and juvenile systemic lupus erythematosus are typically diagnosed. The coexistence of both disorders prompts the search for a common aetiology. In this paper, we present a case of a 16-year-old girl with life-threatening anorexia nervosa followed by clinical and immunological manifestations of systemic lupus erythematosus. The severity of the symptoms of anorexia nervosa resulted in significant delay in proper diagnosis of the concomitant systemic disease which had already been active. The administration of immunosuppressive treatment resulted in decreased lupus activity and resolution of the symptoms of anorexia nervosa.Being affected by one severe and chronic disease does not preclude the coexistence of another disease of different aetiology. However, such coexistence may suggest a common pathophysiology. Many authors have indicated a possible link between anorexia nervosa and many autoimmune disorders. Currently, modern genetic techniques have confirmed a significant correlation between these disorders. This issue needs further investigation and may be helpful in arriving at the final diagnosis in similar cases.
Collapse
|
14
|
Türe U, De Bellis A, Harput MV, Bellastella G, Topcuoglu M, Yaltirik CK, Cirillo P, Yola RN, Sav A, Kelestimur F. Hypothalamitis: A Novel Autoimmune Endocrine Disease. A Literature Review and Case Report. J Clin Endocrinol Metab 2021; 106:e415-e429. [PMID: 33104773 DOI: 10.1210/clinem/dgaa771] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Indexed: 12/18/2022]
Abstract
CONTEXT The relationship between the endocrine system and autoimmunity has been recognized for a long time and one of the best examples of autoimmune endocrine disease is autoimmune hypophysitis. A better understanding of autoimmune mechanisms and radiological, biochemical, and immunological developments has given rise to the definition of new autoimmune disorders including autoimmunity-related hypothalamic-pituitary disorders. However, whether hypothalamitis may occur as a distinct entity is still a matter of debate. EVIDENCE ACQUISITION Here we describe a 35-year-old woman with growing suprasellar mass, partial empty sella, central diabetes insipidus, hypopituitarism, and hyperprolactinemia. EVIDENCE SYNTHESIS Histopathologic examination of surgically removed suprasellar mass revealed lymphocytic infiltrate suggestive of an autoimmune disease with hypothalamic involvement. The presence of antihypothalamus antibodies to arginine vasopressin (AVP)-secreting cells (AVPcAb) at high titers and the absence of antipituitary antibodies suggested the diagnosis of isolated hypothalamitis. Some similar conditions have sometimes been reported in the literature but the simultaneous double finding of lymphocytic infiltrate and the presence of AVPcAb so far has never been reported. CONCLUSIONS We think that the hypothalamitis can be considered a new isolated autoimmune disease affecting the hypothalamus while the lymphocytic infundibuloneurohypophysitis can be a consequence of hypothalamitis with subsequent autoimmune involvement of the pituitary. To our knowledge this is the first observation of autoimmune hypothalamic involvement with central diabetes insipidus, partial empty sella, antihypothalamic antibodies and hypopituitarism.
Collapse
Affiliation(s)
- Uğur Türe
- Department of Neurosurgery, Yeditepe University School of Medicine, Istanbul, Turkey
| | - Annamaria De Bellis
- Unit of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Mehmet Volkan Harput
- Department of Neurosurgery, Yeditepe University School of Medicine, Istanbul, Turkey
| | - Giuseppe Bellastella
- Unit of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Melih Topcuoglu
- Department of Radiology, Yeditepe University School of Medicine, Istanbul, Turkey
| | - Cumhur Kaan Yaltirik
- Department of Neurosurgery, Yeditepe University School of Medicine, Istanbul, Turkey
| | - Paolo Cirillo
- Unit of Endocrinology and Metabolic Diseases, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Rima Nur Yola
- Medical Student, Yeditepe University School of Medicine, Istanbul, Turkey
| | - Aydın Sav
- Department of Pathology, Yeditepe University School of Medicine, Istanbul, Turkey
| | - Fahrettin Kelestimur
- Department of Endocrinology, Yeditepe University School of Medicine, Istanbul, Turkey
| |
Collapse
|
15
|
Smitka K, Prochazkova P, Roubalova R, Dvorak J, Papezova H, Hill M, Pokorny J, Kittnar O, Bilej M, Tlaskalova-Hogenova H. Current Aspects of the Role of Autoantibodies Directed Against Appetite-Regulating Hormones and the Gut Microbiome in Eating Disorders. Front Endocrinol (Lausanne) 2021; 12:613983. [PMID: 33953692 PMCID: PMC8092392 DOI: 10.3389/fendo.2021.613983] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Accepted: 03/09/2021] [Indexed: 12/12/2022] Open
Abstract
The equilibrium and reciprocal actions among appetite-stimulating (orexigenic) and appetite-suppressing (anorexigenic) signals synthesized in the gut, brain, microbiome and adipose tissue (AT), seems to play a pivotal role in the regulation of food intake and feeding behavior, anxiety, and depression. A dysregulation of mechanisms controlling the energy balance may result in eating disorders such as anorexia nervosa (AN) and bulimia nervosa (BN). AN is a psychiatric disease defined by chronic self-induced extreme dietary restriction leading to an extremely low body weight and adiposity. BN is defined as out-of-control binge eating, which is compensated by self-induced vomiting, fasting, or excessive exercise. Certain gut microbiota-related compounds, like bacterial chaperone protein Escherichia coli caseinolytic protease B (ClpB) and food-derived antigens were recently described to trigger the production of autoantibodies cross-reacting with appetite-regulating hormones and neurotransmitters. Gut microbiome may be a potential manipulator for AT and energy homeostasis. Thus, the regulation of appetite, emotion, mood, and nutritional status is also under the control of neuroimmunoendocrine mechanisms by secretion of autoantibodies directed against neuropeptides, neuroactive metabolites, and peptides. In AN and BN, altered cholinergic, dopaminergic, adrenergic, and serotonergic relays may lead to abnormal AT, gut, and brain hormone secretion. The present review summarizes updated knowledge regarding the gut dysbiosis, gut-barrier permeability, short-chain fatty acids (SCFA), fecal microbial transplantation (FMT), blood-brain barrier permeability, and autoantibodies within the ghrelin and melanocortin systems in eating disorders. We expect that the new knowledge may be used for the development of a novel preventive and therapeutic approach for treatment of AN and BN.
Collapse
Affiliation(s)
- Kvido Smitka
- First Faculty of Medicine, Institute of Physiology, Charles University, Prague, Czechia
- First Faculty of Medicine, Institute of Pathological Physiology, Charles University, Prague, Czechia
- *Correspondence: Kvido Smitka,
| | - Petra Prochazkova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Radka Roubalova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Jiri Dvorak
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Hana Papezova
- Psychiatric Clinic, Eating Disorder Center, First Faculty of Medicine, Charles University and General University Hospital in Prague, Prague, Czechia
| | - Martin Hill
- Steroid Hormone and Proteofactors Department, Institute of Endocrinology, Prague, Czechia
| | - Jaroslav Pokorny
- First Faculty of Medicine, Institute of Physiology, Charles University, Prague, Czechia
| | - Otomar Kittnar
- First Faculty of Medicine, Institute of Physiology, Charles University, Prague, Czechia
| | - Martin Bilej
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| | - Helena Tlaskalova-Hogenova
- Laboratory of Cellular and Molecular Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
16
|
Carbone EA, D'Amato P, Vicchio G, De Fazio P, Segura-Garcia C. A systematic review on the role of microbiota in the pathogenesis and treatment of eating disorders. Eur Psychiatry 2020; 64:e2. [PMID: 33416044 PMCID: PMC8057489 DOI: 10.1192/j.eurpsy.2020.109] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Background There is growing interest in new factors contributing to the genesis of eating disorders (EDs). Research recently focused on the study of microbiota. Dysbiosis, associated with a specific genetic susceptibility, may contribute to the development of anorexia nervosa (AN), bulimia nervosa, or binge eating disorder, and several putative mechanisms have already been identified. Diet seems to have an impact not only on modification of the gut microbiota, facilitating dysbiosis, but also on its recovery in patients with EDs. Methods This systematic review based on the PICO strategy searching into PubMed, EMBASE, PsychINFO, and Cochrane Library examined the literature on the role of altered microbiota in the pathogenesis and treatment of EDs. Results Sixteen studies were included, mostly regarding AN. Alpha diversity and short-chain fatty acid (SCFA) levels were lower in patients with AN, and affective symptoms and ED psychopathology seem related to changes in gut microbiota. Microbiota-derived proteins stimulated the autoimmune system, altering neuroendocrine control of mood and satiety in EDs. Microbial richness increased in AN after weight regain on fecal microbiota transplantation. Conclusions Microbiota homeostasis seems essential for a healthy communication network between gut and brain. Dysbiosis may promote intestinal inflammation, alter gut permeability, and trigger immune reactions in the hunger/satiety regulation center contributing to the pathophysiological development of EDs. A restored microbial balance may be a possible treatment target for EDs. A better and more in-depth characterization of gut microbiota and gut–brain crosstalk is required. Future studies may deepen the therapeutic and preventive role of microbiota in EDs.
Collapse
Affiliation(s)
- Elvira Anna Carbone
- Department of Health Sciences, University "Magna Graecia", Catanzaro88100, Italy.,Outpatient Service for Clinical Research and Treatment of Eating Disorders, University Hospital Mater Domini, Catanzaro88100, Italy
| | - Pasquale D'Amato
- Department of Medical and Surgical Sciences, University "Magna Graecia", Catanzaro88100, Italy
| | - Giuseppe Vicchio
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Rende87036, Italy
| | - Pasquale De Fazio
- Department of Health Sciences, University "Magna Graecia", Catanzaro88100, Italy
| | - Cristina Segura-Garcia
- Outpatient Service for Clinical Research and Treatment of Eating Disorders, University Hospital Mater Domini, Catanzaro88100, Italy.,Department of Medical and Surgical Sciences, University "Magna Graecia", Catanzaro88100, Italy
| |
Collapse
|
17
|
Galmiche M, Lucas N, Déchelotte P, Deroissart C, Le Solliec MA, Rondeaux J, Azhar S, Grigioni S, Colange G, Delay J, Achamrah N, Folope V, Belmonte L, Lamarre A, Rimbert A, Saillard T, Petit A, Quillard M, Coeffier M, Gillibert A, Lambert G, Legrand R, Tavolacci MP. Plasma Peptide Concentrations and Peptide-Reactive Immunoglobulins in Patients with Eating Disorders at Inclusion in the French EDILS Cohort (Eating Disorders Inventory and Longitudinal Survey). Nutrients 2020; 12:nu12020522. [PMID: 32085628 PMCID: PMC7071399 DOI: 10.3390/nu12020522] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Revised: 02/11/2020] [Accepted: 02/14/2020] [Indexed: 12/12/2022] Open
Abstract
Eating disorders (EDs) are increasingly frequent. Their pathophysiology involves disturbance of peptide signaling and the microbiota–gut–brain axis. This study analyzed peptides and corresponding immunoglobulin (Ig) concentrations in groups of ED. In 120 patients with restrictive (R), bulimic (B), and compulsive (C) ED, the plasma concentrations of leptin, glucagon-like peptide-1 (GLP-1), peptide YY (PYY), and insulin were analyzed by Milliplex and those of acyl ghrelin (AG), des-acyl ghrelin (DAG), and α-melanocyte-stimulating hormone (α-MSH) by ELISA kits. Immunoglobulin G (in response to an antigen) concentrations were analyzed by ELISA, and their affinity for the respective peptide was measured by surface plasmon resonance. The concentrations of leptin, insulin, GLP-1, and PYY were higher in C patients than in R patients. On the contrary, α-MSH, DAG, and AG concentrations were higher in R than in C patients. After adjustment for body mass index (BMI), differences among peptide concentrations were no longer different. No difference in the concentrations of the IgG was found, but the IgG concentrations were correlated with each other. Although differences of peptide concentrations exist among ED subtypes, they may be due to differences in BMI. Changes in the concentration and/or affinity of several anti-peptide IgG may contribute to the physiopathology of ED or may be related to fat mass.
Collapse
Affiliation(s)
- Marie Galmiche
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
| | - Nicolas Lucas
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Pierre Déchelotte
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
- Correspondence: ; Tel.: +06-08-49-66-26
| | - Camille Deroissart
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Marie-Anne Le Solliec
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Julie Rondeaux
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Saida Azhar
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Sébastien Grigioni
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Guillaume Colange
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Julie Delay
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Najate Achamrah
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Vanessa Folope
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Liliana Belmonte
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Adèle Lamarre
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Agnès Rimbert
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Tiphaine Saillard
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - André Petit
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - Muriel Quillard
- CIC-CRB 1404 INSERM, University Hospital of Rouen, 76000 Rouen, France;
| | - Moise Coeffier
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- Nutrition unit, University Hospital of Rouen, 76000 Rouen, France; (G.C.); (J.D.); (A.L.); (T.S.)
| | - André Gillibert
- Department of Biostatistics, Rouen University Hospital, F 76000 Rouen, France;
| | - Grégory Lambert
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Romain Legrand
- TargEDys SA, 91160 Longjumeau, France; (N.L.); (C.D.); (M.-A.L.S.); (J.R.); (S.A.); (G.L.); (R.L.)
| | - Marie-Pierre Tavolacci
- Inserm UMR1073, 76000 Rouen, France; (M.G.); (S.G.); (N.A.); (V.F.); (L.B.); (A.R.); (A.P.); (M.C.); (M.-P.T.)
- Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen, 76000 Rouen, France
- CIC-CRB 1404 INSERM, University Hospital of Rouen, 76000 Rouen, France;
| |
Collapse
|
18
|
Ghenciulescu A, Park RJ, Burnet PWJ. The Gut Microbiome in Anorexia Nervosa: Friend or Foe? Front Psychiatry 2020; 11:611677. [PMID: 33510660 PMCID: PMC7835121 DOI: 10.3389/fpsyt.2020.611677] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
The human gut microbiome is emerging as a key modulator of homeostasis, with far-reaching implications for various multifactorial diseases, including anorexia nervosa (AN). Despite significant morbidity and mortality, the underlying mechanisms of this eating disorder are poorly understood, but the classical view defining AN as a purely psychiatric condition is increasingly being challenged. Accumulating evidence from comparative studies of AN and healthy fecal microbial composition reveals considerable low divergence and altered taxonomic abundance of the AN gut microbiome. When integrated with preclinical data, these findings point to a significant role of the gut microbiome in AN pathophysiology, via effects on host energy metabolism, intestinal permeability, immune function, appetite, and behavior. While complex causal relationships between genetic risk factors, dietary patterns and microbiome, and their relevance for AN onset and perpetuation have not been fully elucidated, preliminary clinical studies support the use of microbiome-based interventions such as fecal microbiota transplants and probiotics as adjuvants to standard AN therapies. Future research should aim to move from observational to mechanistic, as dissecting how specific microbial taxa interact with the host to impact the development of AN could help design novel therapeutic approaches that more effectively address the severe comorbidities and high relapse rate of this serious disorder.
Collapse
Affiliation(s)
- Ana Ghenciulescu
- Oxford Medical School, Medical Sciences Division, University of Oxford, Oxford, United Kingdom
| | - Rebecca J Park
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| | - Philip W J Burnet
- Department of Psychiatry, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
19
|
Cryan JF, O'Riordan KJ, Cowan CSM, Sandhu KV, Bastiaanssen TFS, Boehme M, Codagnone MG, Cussotto S, Fulling C, Golubeva AV, Guzzetta KE, Jaggar M, Long-Smith CM, Lyte JM, Martin JA, Molinero-Perez A, Moloney G, Morelli E, Morillas E, O'Connor R, Cruz-Pereira JS, Peterson VL, Rea K, Ritz NL, Sherwin E, Spichak S, Teichman EM, van de Wouw M, Ventura-Silva AP, Wallace-Fitzsimons SE, Hyland N, Clarke G, Dinan TG. The Microbiota-Gut-Brain Axis. Physiol Rev 2019; 99:1877-2013. [DOI: 10.1152/physrev.00018.2018] [Citation(s) in RCA: 1243] [Impact Index Per Article: 248.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The importance of the gut-brain axis in maintaining homeostasis has long been appreciated. However, the past 15 yr have seen the emergence of the microbiota (the trillions of microorganisms within and on our bodies) as one of the key regulators of gut-brain function and has led to the appreciation of the importance of a distinct microbiota-gut-brain axis. This axis is gaining ever more traction in fields investigating the biological and physiological basis of psychiatric, neurodevelopmental, age-related, and neurodegenerative disorders. The microbiota and the brain communicate with each other via various routes including the immune system, tryptophan metabolism, the vagus nerve and the enteric nervous system, involving microbial metabolites such as short-chain fatty acids, branched chain amino acids, and peptidoglycans. Many factors can influence microbiota composition in early life, including infection, mode of birth delivery, use of antibiotic medications, the nature of nutritional provision, environmental stressors, and host genetics. At the other extreme of life, microbial diversity diminishes with aging. Stress, in particular, can significantly impact the microbiota-gut-brain axis at all stages of life. Much recent work has implicated the gut microbiota in many conditions including autism, anxiety, obesity, schizophrenia, Parkinson’s disease, and Alzheimer’s disease. Animal models have been paramount in linking the regulation of fundamental neural processes, such as neurogenesis and myelination, to microbiome activation of microglia. Moreover, translational human studies are ongoing and will greatly enhance the field. Future studies will focus on understanding the mechanisms underlying the microbiota-gut-brain axis and attempt to elucidate microbial-based intervention and therapeutic strategies for neuropsychiatric disorders.
Collapse
Affiliation(s)
- John F. Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kenneth J. O'Riordan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitlin S. M. Cowan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kiran V. Sandhu
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Thomaz F. S. Bastiaanssen
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcus Boehme
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Martin G. Codagnone
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Sofia Cussotto
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Christine Fulling
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Anna V. Golubeva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Katherine E. Guzzetta
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Minal Jaggar
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Caitriona M. Long-Smith
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joshua M. Lyte
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Jason A. Martin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Alicia Molinero-Perez
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Moloney
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emanuela Morelli
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Enrique Morillas
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Rory O'Connor
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Joana S. Cruz-Pereira
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Veronica L. Peterson
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Kieran Rea
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Nathaniel L. Ritz
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Eoin Sherwin
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Simon Spichak
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Emily M. Teichman
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Marcel van de Wouw
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Ana Paula Ventura-Silva
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Shauna E. Wallace-Fitzsimons
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Niall Hyland
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| | - Timothy G. Dinan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland; and Department of Physiology, University College Cork, Cork, Ireland
| |
Collapse
|
20
|
Rastelli M, Cani PD, Knauf C. The Gut Microbiome Influences Host Endocrine Functions. Endocr Rev 2019; 40:1271-1284. [PMID: 31081896 DOI: 10.1210/er.2018-00280] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Accepted: 04/15/2019] [Indexed: 12/12/2022]
Abstract
The gut microbiome is considered an organ contributing to the regulation of host metabolism. Since the relationship between the gut microbiome and specific diseases was elucidated, numerous studies have deciphered molecular mechanisms explaining how gut bacteria interact with host cells and eventually shape metabolism. Both metagenomic and metabolomic analyses have contributed to the discovery of bacterial-derived metabolites acting on host cells. In this review, we examine the molecular mechanisms by which bacterial metabolites act as paracrine or endocrine factors, thereby regulating host metabolism. We highlight the impact of specific short-chain fatty acids on the secretion of gut peptides (i.e., glucagon-like peptide-1, peptide YY) and other metabolites produced from different amino acids and regulating inflammation, glucose metabolism, or energy homeostasis. We also discuss the role of gut microbes on the regulation of bioactive lipids that belong to the endocannabinoid system and specific neurotransmitters (e.g., γ-aminobutyric acid, serotonin, nitric oxide). Finally, we review the role of specific bacterial components (i.e., ClpB, Amuc_1100) also acting as endocrine factors and eventually controlling host metabolism. In conclusion, this review summarizes the recent state of the art, aiming at providing evidence that the gut microbiome influences host endocrine functions via several bacteria-derived metabolites.
Collapse
Affiliation(s)
- Marialetizia Rastelli
- Université Catholique de Louvain, UCLouvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Brussels, Belgium.,NeuroMicrobiota, European Associated Laboratory (INSERM/UCLouvain), Brussels, Belgium
| | - Patrice D Cani
- Université Catholique de Louvain, UCLouvain, Walloon Excellence in Life Sciences and BIOtechnology (WELBIO), Metabolism and Nutrition Research Group, Louvain Drug Research Institute, Brussels, Belgium.,NeuroMicrobiota, European Associated Laboratory (INSERM/UCLouvain), Brussels, Belgium
| | - Claude Knauf
- NeuroMicrobiota, European Associated Laboratory (INSERM/UCLouvain), Brussels, Belgium.,Institut de Recherche en Santé Digestive et Nutrition (IRSD), Institut National de la Santé et de la Recherche Médicale (INSERM), U1220, Université Paul Sabatier (UPS), Toulouse Cedex 3, France
| |
Collapse
|
21
|
Igudesman D, Sweeney M, Carroll IM, Mayer-Davis EJ, Bulik CM. Gut-Brain Interactions: Implications for a Role of the Gut Microbiota in the Treatment and Prognosis of Anorexia Nervosa and Comparison to Type I Diabetes. Gastroenterol Clin North Am 2019; 48:343-356. [PMID: 31383275 PMCID: PMC6686879 DOI: 10.1016/j.gtc.2019.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Anorexia nervosa has poor prognosis and treatment outcomes and is influenced by genetic, metabolic, and psychological factors. Gut microbes interact with gut physiology to influence metabolism and neurobiology, although potential therapeutic benefits remain unknown. Type 1 diabetes is linked to anorexia nervosa through energy dysregulation, which in both disease states is related to the gut microbiota, disordered eating, and genetics.
Collapse
Affiliation(s)
- Daria Igudesman
- Department of Nutrition, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC 27599, USA
| | - Megan Sweeney
- Department of Nutrition, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC 27599, USA
| | - Ian M Carroll
- Department of Nutrition, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC 27599, USA
| | - Elizabeth J Mayer-Davis
- Department of Nutrition, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC 27599, USA
| | - Cynthia M Bulik
- Department of Nutrition, University of North Carolina at Chapel Hill, 135 Dauer Drive, Chapel Hill, NC 27599, USA; Department of Psychiatry, University of North Carolina at Chapel Hill, 101 Manning Drive, Chapel Hill, NC 27599, USA; Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
22
|
On the origin of eating disorders: altered signaling between gut microbiota, adaptive immunity and the brain melanocortin system regulating feeding behavior. Curr Opin Pharmacol 2019; 48:82-91. [PMID: 31430598 DOI: 10.1016/j.coph.2019.07.004] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 07/15/2019] [Indexed: 12/18/2022]
Abstract
Research in the field of gut microbiota - brain axis may contribute to clarifying the origin of anorexia nervosa and bulimia, the two principal forms of eating disorders (ED). The initial key findings in ED patients of plasma immunoglobulins (Ig) that react with α-melanocyte-stimulating hormone (α-MSH), a neuropeptide in the brain signaling satiety, have initiated further studies leading to the discovery of the origin of such autoantibodies and to the understanding their possible functional role. An anorexigenic bacterial protein Escherichia coli caseinolytic protease B was recently found to be responsible for the production of α-MSH-cross-reactive autoantibodies and this protein was also detected in human plasma. Another recent study revealed enhanced activation of appetite-regulating the melanocortin type 4 receptor by immune complexes withα-MSH. Taken together, these data serve to build a pathophysiological model of ED presented in this article.
Collapse
|
23
|
Hedman A, Breithaupt L, Hübel C, Thornton LM, Tillander A, Norring C, Birgegård A, Larsson H, Ludvigsson JF, Sävendahl L, Almqvist C, Bulik CM. Bidirectional relationship between eating disorders and autoimmune diseases. J Child Psychol Psychiatry 2019; 60:803-812. [PMID: 30178543 DOI: 10.1111/jcpp.12958] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/09/2018] [Indexed: 12/20/2022]
Abstract
BACKGROUND Immune system dysfunction may be associated with eating disorders (ED) and could have implications for detection, risk assessment, and treatment of both autoimmune diseases and EDs. However, questions regarding the nature of the relationship between these two disease entities remain. We evaluated the strength of associations for the bidirectional relationships between EDs and autoimmune diseases. METHODS In this nationwide population-based study, Swedish registers were linked to establish a cohort of more than 2.5 million individuals born in Sweden between January 1, 1979 and December 31, 2005 and followed up until December 2013. Cox proportional hazard regression models were used to investigate: (a) subsequent risk of EDs in individuals with autoimmune diseases; and (b) subsequent risk of autoimmune diseases in individuals with EDs. RESULTS We observed a strong, bidirectional relationship between the two illness classes indicating that diagnosis in one illness class increased the risk of the other. In women, the diagnoses of autoimmune disease increased subsequent hazards of anorexia nervosa (AN), bulimia nervosa (BN), and other eating disorders (OED). Similarly, AN, BN, and OED increased subsequent hazards of autoimmune diseases.Gastrointestinal-related autoimmune diseases such as, celiac disease and Crohn's disease showed a bidirectional relationship with AN and OED. Psoriasis showed a bidirectional relationship with OED. The previous occurence of type 1 diabetes increased the risk for AN, BN, and OED. In men, we did not observe a bidirectional pattern, but prior autoimmune arthritis increased the risk for OED. CONCLUSIONS The interactions between EDs and autoimmune diseases support the previously reported associations. The bidirectional risk pattern observed in women suggests either a shared mechanism or a third mediating variable contributing to the association of these illnesses.
Collapse
Affiliation(s)
- Anna Hedman
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - Lauren Breithaupt
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Psychology, George Mason University, Fairfax, VA, USA
| | - Christopher Hübel
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Social, Genetic & Developmental Psychiatry Centre, Institute of Psychiatry, Psychology & Neuroscience, King's College London, London, UK
| | - Laura M Thornton
- Department of Psychiatry, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | - Annika Tillander
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Computer and Information Science, Linköping University, Linköping, Sweden
| | - Claes Norring
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Andreas Birgegård
- Department of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet, & Stockholm Health Care Services, Stockholm County Council, Stockholm, Sweden
| | - Henrik Larsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,School of Medical Sciences, Örebro University, Örebro, Sweden
| | - Jonas F Ludvigsson
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Department of Pediatrics, Örebro University Hospital, Örebro, Sweden
| | - Lars Sävendahl
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Catarina Almqvist
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,Pediatric Allergy and Pulmonology Unit, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Cynthia M Bulik
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden.,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
| |
Collapse
|
24
|
Fetissov SO, Legrand R, Lucas N. Bacterial Protein Mimetic of Peptide Hormone as a New Class of Protein- based Drugs. Curr Med Chem 2019; 26:546-553. [PMID: 28982315 DOI: 10.2174/0929867324666171005110620] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 06/07/2017] [Accepted: 08/27/2017] [Indexed: 01/19/2023]
Abstract
Specific peptide molecules classified as hormones, neuropeptides and cytokines are involved in intercellular signaling regulating various physiological processes in all organs and tissues. This justifies the peptidergic signaling as an attractive pharmacological target. Recently, a protein mimetic of a peptide hormone has been identified in Escherichia coli suggesting the potential use of specific bacterial proteins as a new type of peptide-like drugs. We review the scientific rational and technological approaches leading to the identification of the E. coli caseinolytic protease B (ClpB) homologue protein as a conformational mimetic of α-melanocyte-stimulating hormone (α-MSH), a melanocortin peptide critically involved in the regulation of energy homeostasis in humans and animals. Theoretical and experimental backgrounds for the validation of bacterial ClpB as a potential drug are discussed based on the known E. coli ClpB amino acid sequence homology with α-MSH. Using in silico analysis, we show that other protein sources containing similar to E. coli ClpB α-MSH-like epitopes with potential biological activity may exist in Enterobacteriaceae and in some Brassicaceae. Thus, the original approach leading to the identification of E. coli ClpB as an α-MSH mimetic protein can be applied for the identification of mimetic proteins of other peptide hormones and development of a new type of peptide-like protein-based drugs.
Collapse
Affiliation(s)
- Sergueï O Fetissov
- Inserm UMR1239, 25 rue Lucien Tesniere, 76130, Mont-Saint-Aignan, France.,Institute for Research and Innovation in Biomedicine (IRIB), University of Rouen Normandy, Rouen, 76000, France
| | | | | |
Collapse
|
25
|
Delaney S, Hornig M. Environmental Exposures and Neuropsychiatric Disorders: What Role Does the Gut-Immune-Brain Axis Play? Curr Environ Health Rep 2019; 5:158-169. [PMID: 29423662 DOI: 10.1007/s40572-018-0186-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE OF REVIEW Evidence is growing that environmental exposures-including xenobiotics as well as microbes-play a role in the pathogenesis of many neuropsychiatric disorders. Underlying mechanisms are likely to be complex, involving the developmentally sensitive interplay of genetic/epigenetic, detoxification, and immune factors. Here, we review evidence supporting a role for environmental factors and disrupted gut-immune-brain axis function in some neuropsychiatric conditions. RECENT FINDINGS Studies suggesting the involvement of an altered microbiome in triggering CNS-directed autoimmunity and neuropsychiatric disturbances are presented as an intriguing example of the varied mechanisms by which environmentally induced gut-immune-brain axis dysfunction may contribute to adverse brain outcomes. The gut-immune-brain axis is a burgeoning frontier for investigation of neuropsychiatric illness. Future translational research to define individual responses to exogenous exposures in terms of microbiome-dependent skew of the metabolome, immunity, and brain function may serve as a lens for illumination of pathways involved in the development of CNS disease and fuel discovery of novel interventions.
Collapse
Affiliation(s)
- Shannon Delaney
- Department of Psychiatry, Columbia University Medical Center, New York, NY, 10032, USA
| | - Mady Hornig
- Center for Infection and Immunity, Columbia University Mailman School of Public Health, 722 W 168th St, Rm 1706, New York, NY, 10032, USA.
- Department of Epidemiology, Columbia University Mailman School of Public Health, 722 W 168th St, Rm 1706, New York, NY, 10032, USA.
| |
Collapse
|
26
|
Eating disorders in adolescents with chronic gastrointestinal and endocrine diseases. THE LANCET CHILD & ADOLESCENT HEALTH 2019; 3:181-189. [DOI: 10.1016/s2352-4642(18)30386-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Revised: 11/17/2018] [Accepted: 11/21/2018] [Indexed: 12/19/2022]
|
27
|
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: 9.8] [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
|
28
|
Immunoglobulin G modulation of the melanocortin 4 receptor signaling in obesity and eating disorders. Transl Psychiatry 2019; 9:87. [PMID: 30755592 PMCID: PMC6372612 DOI: 10.1038/s41398-019-0422-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 12/10/2018] [Accepted: 01/02/2019] [Indexed: 12/23/2022] Open
Abstract
Melanocortin 4 receptor (MC4R) plays a key role in regulation of appetite activated by its main ligand α-melanocyte-stimulating hormone (α-MSH) in both central and peripheral targets. α-MSH also binds to circulating immunoglobulins (Igs) but the functional significance of such immune complexes (ICs) in MC4R signaling in normal and pathological conditions of altered appetite has remained unknown. To address this question, we analyzed plasma levels, affinity kinetics, and binding epitopes of α-MSH-reactive IgG extracted from plasma samples of female patients with hyperphagic obesity, anorexia nervosa, bulimia nervosa, binge-eating disorder, and healthy controls. Ability of α-MSH/IgG IC to bind and activate human MC4R were studied in vitro and to influence feeding behavior in vivo in rodents. We found that α-MSH-reactive IgG were low in obese but increased in anorectic and bulimic patients and displayed different epitope and kinetics of IC formation. Importantly, while α-MSH/IgG IC from all subjects were binding and activating MC4R, the receptor binding affinity was decreased in obesity. Additionally, α-MSH/IgG IC had lower MC4R-mediated cAMP activation threshold as compared with α-MSH alone in all but not obese subjects. Furthermore, the cellular internalization rate of α-MSH/IgG IC by MC4R-expressing cells was decreased in obese but increased in patients with anorexia nervosa. Moreover, IgG from obese patients prevented central anorexigenic effect of α-MSH. These findings reveal that MC4R is physiologically activated by IC formed by α-MSH/IgG and that different levels and molecular properties of α-MSH-reactive IgG underlie biological activity of such IC relevant to altered appetite in obesity and eating disorders.
Collapse
|
29
|
Vaeroy H, Schneider F, Fetissov SO. Neurobiology of Aggressive Behavior-Role of Autoantibodies Reactive With Stress-Related Peptide Hormones. Front Psychiatry 2019; 10:872. [PMID: 31866881 PMCID: PMC6904880 DOI: 10.3389/fpsyt.2019.00872] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Accepted: 11/05/2019] [Indexed: 12/31/2022] Open
Abstract
Adrenocorticotropic hormone together with arginine vasopressin and oxytocin, the neuropeptides regulating the stress response and the hypothalamic-pituitary-adrenal axis activity, are known to modulate aggressive behavior. The functional role of the adrenocorticotropic hormone immunoglobulin G autoantibodies in peptidergic signaling and motivated behavior, including aggression, has been shown in experimental and in vitro models. This review summarizes some experimental data implicating autoantibodies reactive with stress-related peptides in aggressive behavior.
Collapse
Affiliation(s)
- Henning Vaeroy
- Department of Psychiatric Research, Akershus University Hospital, Nordbyhagen, Norway
| | - Frida Schneider
- Department of Psychiatric Research, Akershus University Hospital, Nordbyhagen, Norway
| | - Sergueï O Fetissov
- Inserm UMR1239, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, University of Rouen Normandy, Rouen, France
| |
Collapse
|
30
|
Scolnick B. Hypothesis: Clues From Mammalian Hibernation for Treating Patients With Anorexia Nervosa. Front Psychol 2018; 9:2159. [PMID: 30483182 PMCID: PMC6240652 DOI: 10.3389/fpsyg.2018.02159] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 10/19/2018] [Indexed: 12/30/2022] Open
Abstract
This hypothesis is that anorexia nervosa (AN) is a biologically driven disorder, and mammalian hibernation may offer clues to its pathogenesis. Using this approach, this hypothesis offers suggestions for employing heart rate variability as an early diagnostic test for AN; employing the ketogenic diet for refeeding patients, attending to omega 3:6 ratio of polyunsaturated fatty acids (PUFAs) in the refeeding diet; and exploring clinical trials of the endocannabinoid-like agent, palmitoylethanolamde for patients with AN. This hypothesis also explores the role of lipids and autoimmune phenomena in AN, and suggest a lipodomics study to search for antibodies in the serum on patients with AN.
Collapse
Affiliation(s)
- Barbara Scolnick
- Psychology and Brain Science, Boston University, Boston, MA, United States
| |
Collapse
|
31
|
Temko JE, Bouhlal S, Farokhnia M, Lee MR, Cryan JF, Leggio L. The Microbiota, the Gut and the Brain in Eating and Alcohol Use Disorders: A 'Ménage à Trois'? Alcohol Alcohol 2018; 52:403-413. [PMID: 28482009 DOI: 10.1093/alcalc/agx024] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 04/05/2017] [Indexed: 02/06/2023] Open
Abstract
Aims Accumulating evidence for the influence of the gut microbiota on the bidirectional communication along the gut-brain axis suggests a role of the gut microbiota in eating disorders (EDs) and alcohol and substance use disorders. The potential influence of altered gut microbiota (dysbiosis) on behaviors associated with such disorders may have implications for developing therapeutic interventions. Methods A systematic review of preclinical and clinical studies evaluating the gut microbiota, EDs and alcohol and substance use disorders was conducted using MEDLINE, Embase and Web of Science databases with the objective being to examine the role of the gut microbiota in behavioral correlates of these disorders. Original papers focused on the gut microbiota and potential behavioral implications were deemed eligible for consideration. Results The resulting 12 publications were limited to gut microbiota studies related to EDs and alcohol and substance use disorders. Some studies suggest that dysbiosis and gut microbial byproducts may influence the pathophysiology of EDs via direct and indirect interference with peptide hormone signaling. Additionally, dysbiosis was shown to be correlated with alcohol use disorder-related symptoms, i.e. craving, depression and anxiety. Finally, a mouse study suggests that manipulations in the gut microbiota may affect cocaine-related behaviors. Conclusions Promising, albeit preliminary, findings suggest a potential role of the gut microbiota in behavioral correlates of EDs and alcohol and substance use disorders. Short summary Preliminary evidence exists supporting the role of the gut microbiota in eating disorders and alcohol and substance use disorders, although additional investigation is needed to determine what is causative versus epiphenomenological.
Collapse
Affiliation(s)
- Jamie E Temko
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, 10 Center Drive (10CRC/15330), Bethesda, MD 20892-1108, USA
| | - Sofia Bouhlal
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, 10 Center Drive (10CRC/15330), Bethesda, MD 20892-1108, USA
| | - Mehdi Farokhnia
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, 10 Center Drive (10CRC/15330), Bethesda, MD 20892-1108, USA
| | - Mary R Lee
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, 10 Center Drive (10CRC/15330), Bethesda, MD 20892-1108, USA
| | - John F Cryan
- APC Microbiome Institute and Department of Anatomy and Neuroscience, University College Cork, Western Gateway Building, Cork, Ireland
| | - Lorenzo Leggio
- Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology, National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, 10 Center Drive (10CRC/15330), Bethesda, MD 20892-1108, USA.,Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences, Brown University, 121 South Main Street, Providence, RI 02903, USA
| |
Collapse
|
32
|
Aurigemma NC, Koltun KJ, VanEvery H, Rogers CJ, De Souza MJ. Linking the Gut Microbiota to Bone Health in Anorexia Nervosa. Curr Osteoporos Rep 2018; 16:65-75. [PMID: 29417446 DOI: 10.1007/s11914-018-0420-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE OF REVIEW The purpose of this review is to examine the anorexia nervosa-microbiota-bone relationship, offering a compilation of the relevant human and animal studies that may contribute to a more comprehensive understanding of potential mechanisms involved. RECENT FINDINGS Recent studies have implicated fermentation by-products of the gut microbiota in bone metabolism. Compromised bone health often accompanies anorexia nervosa due to energy deficiency and hypoestrogenism. The gut microbiome has been implicated as a link between these conditions and impaired bone growth phenotypes. Current research supports decrements in Firmicutes and short-chain fatty acids with increases in Methanobrevibacter smithii and Proteobacteria in anorexia nervosa. A potential mechanism for microbiome-regulated bone growth is through modulation of insulin-like growth factor-1. Future research should aim to examine short-chain fatty acids, probiotics, and prebiotics as alternative therapies to treat low bone density in anorexia nervosa.
Collapse
Affiliation(s)
- Nicole C Aurigemma
- Women's Health and Exercise Lab, Department of Kinesiology, The Pennsylvania State University, University Park, PA, 16802, USA.
| | - Kristen J Koltun
- Women's Health and Exercise Lab, Department of Kinesiology, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Hannah VanEvery
- Departments of Nutritional Sciences and Clinical and Translational Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Connie J Rogers
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Mary Jane De Souza
- Women's Health and Exercise Lab, Department of Kinesiology and Physiology, The Pennsylvania State University, University Park, PA, 16802, USA
| |
Collapse
|
33
|
Zerwas S, Larsen JT, Petersen L, Thornton LM, Quaranta M, Koch SV, Pisetsky D, Mortensen PB, Bulik CM. Eating Disorders, Autoimmune, and Autoinflammatory Disease. Pediatrics 2017; 140:peds.2016-2089. [PMID: 29122972 PMCID: PMC5703777 DOI: 10.1542/peds.2016-2089] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/16/2017] [Indexed: 01/01/2023] Open
Abstract
OBJECTIVES Identifying factors associated with risk for eating disorders is important for clarifying etiology and for enhancing early detection of eating disorders in primary care. We hypothesized that autoimmune and autoinflammatory diseases would be associated with eating disorders in children and adolescents and that family history of these illnesses would be associated with eating disorders in probands. METHODS In this large, nationwide, population-based cohort study of all children and adolescents born in Denmark between 1989 and 2006 and managed until 2012, Danish medical registers captured all inpatient and outpatient diagnoses of eating disorders and autoimmune and autoinflammatory diseases. The study population included 930 977 individuals (48.7% girls). Cox proportional hazards regression models and logistic regression were applied to evaluate associations. RESULTS We found significantly higher hazards of eating disorders for children and adolescents with autoimmune or autoinflammatory diseases: 36% higher hazard for anorexia nervosa, 73% for bulimia nervosa, and 72% for an eating disorder not otherwise specified. The association was particularly strong in boys. Parental autoimmune or autoinflammatory disease history was associated with significantly increased odds for anorexia nervosa (odds ratio [OR] = 1.13, confidence interval [CI] = 1.01-1.25), bulimia nervosa (OR = 1.29; CI = 1.08-1.55) and for an eating disorder not otherwise specified (OR = 1.27; CI = 1.13-1.44). CONCLUSIONS Autoimmune and autoinflammatory diseases are associated with increased risk for eating disorders. Ultimately, understanding the role of immune system disturbance for the etiology and pathogenesis of eating disorders could point toward novel treatment targets.
Collapse
Affiliation(s)
| | - Janne Tidselbak Larsen
- National Centre for Register-Based Research,,The Lundbeck Foundation Initiative for Integrative Psychiatric Research, and
| | - Liselotte Petersen
- National Centre for Register-Based Research,,The Lundbeck Foundation Initiative for Integrative Psychiatric Research, and
| | | | - Michela Quaranta
- Department of Neuroscience, Azienda Ospedaliero-Universitaria San Giovanni Battista and University of Turin, Turin, Italy
| | - Susanne Vinkel Koch
- Faculty of Health Sciences, University of Copenhagen, Copenhagen, Denmark;,Mental Health Centre for Child and Adolescent Psychiatry, Copenhagen, Denmark
| | - David Pisetsky
- Medical Research Service, Durham Veterans Administration Medical Center and Division of Rheumatology and Immunology, Duke University, Durham, North Carolina; and
| | - Preben Bo Mortensen
- National Centre for Register-Based Research,,The Lundbeck Foundation Initiative for Integrative Psychiatric Research, and,Centre for Integrated Register-based Research, Aarhus University, Aarhus, Denmark
| | - Cynthia M. Bulik
- Departments of Psychiatry and,Nutrition, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina;,Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
34
|
The Hypothalamic-Pituitary Axis and Autoantibody Related Disorders. Int J Mol Sci 2017; 18:ijms18112322. [PMID: 29099758 PMCID: PMC5713291 DOI: 10.3390/ijms18112322] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 10/27/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022] Open
Abstract
This review summarized different studies reporting the presence of autoantibodies reacting against cells of the pituitary (APAs) and/or hypothalamus (AHAs). Both APAs and AHAs have been revealed through immunofluorescence using different kinds of substrates. Autoantibodies against gonadotropic cells were mainly found in patients affected by cryptorchidism and hypogonadotropic hypogonadism while those against prolactin cells were found in different kinds of patients, the majority without pituitary abnormalities. APAs to growth hormone (GH) cells have been associated with GH deficiency while those against the adrenocorticotropic cells have distinguished central Cushing's disease patients at risk of incomplete cure after surgical adenoma removal. AHAs to vasopressin cells have identified patients at risk of developing diabetes insipidus. APAs have been also found together with AHAs in patients affected by idiopathic hypopituitarism, but both were also present in different kinds of patients without abnormalities of the hypothalamic-pituitary axis. Despite some data being promising, the clinical use of pituitary and hypothalamus autoantibodies is still limited by the low diagnostic sensitivity, irreproducibility of the results, and the absence of autoantigen/s able to discriminate the autoimmune reaction involving the pituitary or the hypothalamus from the other autoimmune states.
Collapse
|
35
|
Ilzarbe L, Fàbrega M, Quintero R, Bastidas A, Pintor L, García-Campayo J, Gomollón F, Ilzarbe D. Inflammatory Bowel Disease and Eating Disorders: A systematized review of comorbidity. J Psychosom Res 2017; 102:47-53. [PMID: 28992897 DOI: 10.1016/j.jpsychores.2017.09.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Revised: 08/27/2017] [Accepted: 09/13/2017] [Indexed: 12/18/2022]
Abstract
OBJECTIVE Research has shown that there is an association between Inflammatory Bowel Disease, anxiety and mood disorders, however little is known about their association with Eating Disorders. In this paper we will present a case of a young female with a comorbid diagnosis of Inflammatory Bowel Disease and Eating Disorder, and then discuss the results from a systematic review of the literature, describing published cases of patients with the same condition. METHODS A systematized review of the literature was conducted according to MOOSE guidelines. A computerized literature search of MEDLINE, PsycINFO and EMBASE, and a manual search through reference lists of selected original articles were performed to identify all published case-reports, case series and studies of Inflammatory Bowel Disease and Eating Disorders. RESULTS Fourteen articles were included, encompassing 219 cases, including ours. The vast majority were females ranging from 10 to 44years old. Anorexia Nervosa (n=156) and Crohn's Disease (n=129) was the most frequent combination (n=90) reported in the literature. These cases present a poor prognosis because of corticoid refusal, medication abandon and/or deliberate exacerbation of IBD symptoms, in the context of trying to lose weight. CONCLUSION Recent evidence suggests there is a possible association between Inflammatory Bowel Disease and Eating Disorders, although the mechanisms involved in its ethiopathogenesis are still unknown. To be aware of this association is important because a delayed diagnosis of this comorbidity may lead to worse prognosis. Further research and a multidisciplinary approach could facilitate earlier diagnosis and provide therapeutic interventions.
Collapse
Affiliation(s)
- L Ilzarbe
- Faculty of Medicine, Universidad de Zaragoza, Zaragoza, Spain
| | - M Fàbrega
- Department of Child and Adolescent Psychiatry, Imperial College London, London, United Kingdom.
| | - R Quintero
- Psychosomatic and Liason Psychiatry Unit, Department of Psychiatry and Psychology, Neuroscience Institute, Hospital Clínic de Barcelona, Barcelona, Spain.
| | - A Bastidas
- Acute Inpatient Unit, Department of Psychiatry and Psychology, Neuroscience Institute, Hospital Clínic de Barcelona, Barcelona, Spain.
| | - L Pintor
- Psychosomatic and Liason Psychiatry Unit, Department of Psychiatry and Psychology, Neuroscience Institute, Hospital Clínic de Barcelona, Barcelona, Spain.
| | - J García-Campayo
- Faculty of Medicine, Universidad de Zaragoza, Zaragoza, Spain; Department of Psychiatry, Hospital Universitario Miguel Servet, Zaragoza, Spain; Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain; Network for Prevention and Health Promotion in Primary Care (RedIAPP), Madrid, Spain
| | - F Gomollón
- Faculty of Medicine, Universidad de Zaragoza, Zaragoza, Spain; Aragón Health Research Institute (IIS Aragón), Zaragoza, Spain; Inflammatory Bowel Disease Unit, Department of gastroenterology, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain; Centro de Investigación Biomédica en Red, Enfermedades Hepáticas y Digestivas (CIBEREHD), Madrid, Spain
| | - D Ilzarbe
- Department of Child and Adolescent Psychiatry, Institute of Psychiatry, Psychology and Neurosciences, King's College London, London, United Kingdom; Faculty of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
36
|
Lam YY, Maguire S, Palacios T, Caterson ID. Are the Gut Bacteria Telling Us to Eat or Not to Eat? Reviewing the Role of Gut Microbiota in the Etiology, Disease Progression and Treatment of Eating Disorders. Nutrients 2017; 9:nu9060602. [PMID: 28613252 PMCID: PMC5490581 DOI: 10.3390/nu9060602] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Revised: 06/05/2017] [Accepted: 06/07/2017] [Indexed: 02/06/2023] Open
Abstract
Traditionally recognized as mental illnesses, eating disorders are increasingly appreciated to be biologically-driven. There is a growing body of literature that implicates a role of the gut microbiota in the etiology and progression of these conditions. Gut bacteria may act on the gut–brain axis to alter appetite control and brain function as part of the genesis of eating disorders. As the illnesses progress, extreme feeding patterns and psychological stress potentially feed back to the gut ecosystem that can further compromise physiological, cognitive, and social functioning. Given the established causality between dysbiosis and metabolic diseases, an altered gut microbial profile is likely to play a role in the co-morbidities of eating disorders with altered immune function, short-chain fatty acid production, and the gut barrier being the key mechanistic links. Understanding the role of the gut ecosystem in the pathophysiology of eating disorders will provide critical insights into improving current treatments and developing novel microbiome-based interventions that will benefit patients with eating disorders.
Collapse
Affiliation(s)
- Yan Y Lam
- Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia.
- Department of Biochemistry and Microbiology, School of Environmental and Biological Sciences, Rutgers, The State University of New Jersey, New Brunswick, NJ 08901, USA.
| | - Sarah Maguire
- Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia.
| | - Talia Palacios
- Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia.
| | - Ian D Caterson
- Boden Institute of Obesity, Nutrition, Exercise & Eating Disorders, Charles Perkins Centre, University of Sydney, Sydney, NSW 2006, Australia.
| |
Collapse
|
37
|
Fetissov SO. [Hunger and satiety factors in the regulation of pleasure associated with feeding behavior]. Biol Aujourdhui 2017; 210:259-268. [PMID: 28327283 DOI: 10.1051/jbio/2016025] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Indexed: 11/14/2022]
Abstract
Feeding is an instinctive behavior accompanied by rewarding feeling of pleasure during obtaining and ingesting food, corresponding to the preparatory and consummatory phases of motivated behavior, respectively. Perception of this emotional state together with alternating feelings of hunger and satiety drives the feeding behavior. Because alterations of feeding behavior including either overeating or anorexia may lead to obesity and cachexia, respectively, understanding the neurochemical mechanisms of regulation of feeding pleasure may help to develop new therapies of these diseases. The dopamine (DA) system of the mesolimbic projections plays a key role in behavioral reward in general and is also involved in regulating feeding-associated pleasure in the forebrain including the nucleus accumbens (NAc) and the lateral hypothalamic area (LHA). It suggests that this DA system can be selectively activated by factors specific to different types of motivated behavior including hunger- and satiety- related hormones. Indeed, central administrations of either orexigenic ghrelin or anorexigenic α-melanocyte-stimulating hormone (α-MSH) increase DA release in the NAc. However, DA has also been shown to inhibit food intake when injected into the LHA, historically known as a « hunger center », indicating DA functional involvement in regulation of both appetite and feeding pleasure. Although both NAc and LHA contain neurons expressing melanocortin receptors, only the LHA receives the α-MSH containing nerve terminals from the α-MSH producing neurons of the hypothalamic arcuate nucleus, the main relay of the peripheral hunger and satiety signals to the brain. A recent study showed that α-MSH in the LHA enhances satiety and inhibits feeding pleasure while potently stimulating DA release in this area during both preparatory and consummatory phases of feeding. It suggests that altered signaling by α-MSH to the DA system in the LHA may be involved in the pathophysiology of obesity and anorexia and the possible underlying mechanisms are discussed.
Collapse
|
38
|
A role for intestinal TLR4-driven inflammatory response during activity-based anorexia. Sci Rep 2016; 6:35813. [PMID: 27779218 PMCID: PMC5078809 DOI: 10.1038/srep35813] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 10/05/2016] [Indexed: 12/12/2022] Open
Abstract
Anorexia nervosa (AN) is associated with low-grade systemic inflammation and altered gut microbiota. However, the molecular origin of the inflammation remains unknown. Toll-like receptors are key regulators of innate immune response and their activation seems also to be involved in the control of food intake. We used activity-based anorexia (ABA) model to investigate the role of TLR4 and its contribution in anorexia-associated low-grade inflammation. Here, we found that ABA affected early the intestinal inflammatory status and the hypothalamic response. Indeed, TLR4 was upregulated both on colonic epithelial cells and intestinal macrophages, leading to elevated downstream mucosal cytokine production. These mucosal changes occurred earlier than hypothalamic changes driving to increased levels of IL-1β and IL-1R1 as well as increased levels of plasma corticosterone. Paradoxically, TLR4-deficient mice exhibited greater vulnerability to ABA with increased mortality rate, suggesting a major contribution of TLR4-mediated responses during ABA-induced weight loss.
Collapse
|
39
|
Breton J, Legrand R, Akkermann K, Järv A, Harro J, Déchelotte P, Fetissov SO. Elevated plasma concentrations of bacterial ClpB protein in patients with eating disorders. Int J Eat Disord 2016; 49:805-8. [PMID: 27038326 DOI: 10.1002/eat.22531] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Revised: 02/12/2016] [Accepted: 02/13/2016] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Caseinolytic protease B (ClpB) produced by Enterobacteria, such as Escherichia coli, has been identified as a conformational mimetic of α-melanocyte-stimulating hormone (α-MSH), an anorexigenic and anxiogenic neuropeptide. In mice, ClpB induces α-MSH cross-reactive antibodies and activates anorexigenic brain neurons. In patients with eating disorders (ED), anti-ClpB and anti-α-MSH antibodies correlate with psychopathological traits. However, it is not known if ClpB is present in human plasma including ED patients. METHODS Plasma concentrations of ClpB were measured using a recently developed ClpB immunoassay in female patients with anorexia nervosa, bulimia nervosa, and binge-eating disorder and compared with healthy participants, all characterized by the Eating Disorder Inventory-2 (EDI-2) scale. RESULTS We found that ClpB was readably detectable in plasma of healthy participants and ED patients and that its concentrations were elevated in ED patients, without significant differences in patient's subgroups. Plasma ClpB concentrations correlated with the EDI-2 scores, with α-MSH as well as with plasma levels of anti-ClpB and anti-α-MSH antibodies. DISCUSSION These data revealed that bacterial ClpB is naturally present in human plasma and that its concentrations can be elevated in ED patients and associated with ED-related psychopathological traits. These results support a link between bacterial ClpB and the ED pathophysiology. © 2016 Wiley Periodicals, Inc. (Int J Eat Disord 2016; 49:805-808).
Collapse
Affiliation(s)
- Jonathan Breton
- Nutrition, Gut and Brain Laboratory, Inserm UMR1073, Rouen, 76183, France.,Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, 76000, France
| | - Romain Legrand
- Nutrition, Gut and Brain Laboratory, Inserm UMR1073, Rouen, 76183, France.,Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, 76000, France
| | - Kirsti Akkermann
- Department of Psychology, Estonian Centre of Behavioural and Health Sciences, Tartu, 50409, Estonia
| | - Anu Järv
- Tartu University Clinics, Psychiatric Hospital, University of Tartu, Tartu, 50417, Estonia
| | - Jaanus Harro
- Department of Psychology, Estonian Centre of Behavioural and Health Sciences, Tartu, 50409, Estonia
| | - Pierre Déchelotte
- Nutrition, Gut and Brain Laboratory, Inserm UMR1073, Rouen, 76183, France.,Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, 76000, France.,CHU Charles Nicolle, Rouen University Hospital, Rouen, 76183, France
| | - Sergueï O Fetissov
- Nutrition, Gut and Brain Laboratory, Inserm UMR1073, Rouen, 76183, France.,Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, 76000, France
| |
Collapse
|
40
|
Wotton CJ, James A, Goldacre MJ. Coexistence of eating disorders and autoimmune diseases: Record linkage cohort study, UK. Int J Eat Disord 2016; 49:663-72. [PMID: 27333941 DOI: 10.1002/eat.22544] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/06/2016] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Recent research indicates that eating disorders (ED) are associated with type 1 diabetes and Crohn's disease. The aim of this study was to determine whether, in a hospitalized population, a range of autoimmune diseases (AIDs) occurred more often than expected in people with anorexia nervosa (AN) or bulimia nervosa (BN), and whether AIDs elevated the risk of ED. METHOD Retrospective, record-linkage cohort study using national administrative statistical data on hospital care and mortality in England, 1999-2011. In people admitted when aged 10-44, cohorts of 8,700 females and 651 males with AN, and 4,783 females and 330 males with BN were constructed, along with a control cohort with the same age range. Results were expressed as risk ratios comparing each ED cohort with the control cohort. RESULTS The overall rate ratio for an AID after admission for AN was 2.04 (95% confidence interval 1.81-2.28) in females, and 1.14 (0.37-2.67) in males; and, for BN, 1.83 (1.56-2.14) in females, and 4.41 (2.11-8.10) in males. Rate ratios for AN after admission for an AID were 3.34 (2.94-3.79) in females, 3.76 (2.06-6.53) in males; and those for BN were 2.57 (2.22-2.97) in females, and 3.10 (1.50-5.90) in males. There were significant associations between ED and several specific individual AIDs. DISCUSSION Strong associations between ED and specific AIDs exist, although it is not possible from this study to determine if these are causal. Clinicians should be aware of the co-occurrence of these conditions. © 2016 Wiley Periodicals, Inc.(Int J Eat Disord 2016; 49:663-672).
Collapse
Affiliation(s)
- Clare J Wotton
- Unit of Health-Care Epidemiology, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, United Kingdom
| | - Anthony James
- Highfield Unit, Warneford Hospital, Oxford, OX3 7JX, United Kingdom
| | - Michael J Goldacre
- Unit of Health-Care Epidemiology, Nuffield Department of Population Health, University of Oxford, Old Road Campus, Oxford, OX3 7LF, United Kingdom
| |
Collapse
|
41
|
Gorwood P, Blanchet-Collet C, Chartrel N, Duclos J, Dechelotte P, Hanachi M, Fetissov S, Godart N, Melchior JC, Ramoz N, Rovere-Jovene C, Tolle V, Viltart O, Epelbaum J. New Insights in Anorexia Nervosa. Front Neurosci 2016; 10:256. [PMID: 27445651 PMCID: PMC4925664 DOI: 10.3389/fnins.2016.00256] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 05/23/2016] [Indexed: 12/18/2022] Open
Abstract
Anorexia nervosa (AN) is classically defined as a condition in which an abnormally low body weight is associated with an intense fear of gaining weight and distorted cognitions regarding weight, shape, and drive for thinness. This article reviews recent evidences from physiology, genetics, epigenetics, and brain imaging which allow to consider AN as an abnormality of reward pathways or an attempt to preserve mental homeostasis. Special emphasis is put on ghrelino-resistance and the importance of orexigenic peptides of the lateral hypothalamus, the gut microbiota and a dysimmune disorder of neuropeptide signaling. Physiological processes, secondary to underlying, and premorbid vulnerability factors-the "pondero-nutritional-feeding basements"- are also discussed.
Collapse
Affiliation(s)
- Philip Gorwood
- Centre Hospitalier Sainte-Anne (CMME)Paris, France; UMR-S 894, Institut National de la Santé et de la Recherche Médicale, Centre de Psychiatrie et NeurosciencesParis, France; Université Paris Descartes, Sorbonne Paris CitéParis, France
| | | | - Nicolas Chartrel
- Institut National de la Santé et de la Recherche Médicale U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in BiomedicineRouen, France; Normandy UniversityCaen, France; University of RouenRouen, France
| | - Jeanne Duclos
- Adolescents and Young Adults Psychiatry Department, Institut Mutualiste MontsourisParis, France; CESP, Institut National de la Santé et de la Recherche Médicale, Université Paris-Descartes, USPCParis, France; University Reims, Champagne-Ardenne, Laboratoire Cognition, Santé, Socialisation (C2S)-EA 6291Reims, France
| | - Pierre Dechelotte
- Institut National de la Santé et de la Recherche Médicale U1073 IRIB Normandy UniversityRouen, France; Faculté de Médecine-PharmacieRouen, France
| | - Mouna Hanachi
- Université de Versailles Saint-Quentin-en-Yvelines, Institut National de la Santé et de la Recherche Médicale U1179, équipe Thérapeutiques Innovantes et Technologies Appliquées aux Troubles Neuromoteurs, UFR des Sciences de la Santé Simone VeilMontigny-le-Bretonneux, France; Département de Médecine (Unité de Nutrition), Hôpital Raymond Poincaré, Assistance Publique-Hôpitaux de ParisGarches, France
| | - Serguei Fetissov
- Institut National de la Santé et de la Recherche Médicale U1073 IRIB Normandy University Rouen, France
| | - Nathalie Godart
- Adolescents and Young Adults Psychiatry Department, Institut Mutualiste MontsourisParis, France; CESP, Institut National de la Santé et de la Recherche Médicale, Université Paris-Descartes, USPCParis, France
| | - Jean-Claude Melchior
- Université de Versailles Saint-Quentin-en-Yvelines, Institut National de la Santé et de la Recherche Médicale U1179, équipe Thérapeutiques Innovantes et Technologies Appliquées aux Troubles Neuromoteurs, UFR des Sciences de la Santé Simone VeilMontigny-le-Bretonneux, France; Département de Médecine (Unité de Nutrition), Hôpital Raymond Poincaré, Assistance Publique-Hôpitaux de ParisGarches, France
| | - Nicolas Ramoz
- UMR-S 894, Institut National de la Santé et de la Recherche Médicale, Centre de Psychiatrie et NeurosciencesParis, France; Université Paris Descartes, Sorbonne Paris CitéParis, France
| | - Carole Rovere-Jovene
- Institut de Pharmacologie Moléculaire et Cellulaire, UMR6097, Centre National de la Recherche Scientifique Valbonne, France
| | - Virginie Tolle
- UMR-S 894, Institut National de la Santé et de la Recherche Médicale, Centre de Psychiatrie et NeurosciencesParis, France; Université Paris Descartes, Sorbonne Paris CitéParis, France
| | - Odile Viltart
- Université Lille, Inserm, CHU Lille, UMR-S 1172 - JPArc - Centre de Recherche Jean-Pierre AUBERT Neurosciences et Cancer Lille, France
| | - Jacques Epelbaum
- UMR-S 894, Institut National de la Santé et de la Recherche Médicale, Centre de Psychiatrie et NeurosciencesParis, France; Université Paris Descartes, Sorbonne Paris CitéParis, France
| |
Collapse
|
42
|
Microbes, molecular mimicry and molecules of mood and motivation. Med Hypotheses 2015; 87:40-3. [PMID: 26826639 DOI: 10.1016/j.mehy.2015.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 12/15/2015] [Indexed: 12/18/2022]
Abstract
The hypothesis proposed is that functional disorders, such as irritable bowel syndrome, chronic fatigue syndrome and anorexia nervosa are caused by auto-antibodies to neuronal proteins induced by molecular mimicry with microbial antigens. The age incidence of these conditions, the marked female excess, increase with economic and technological advance, precipitation by infection, and the paucity of histological changes are all consistent with the hypothesis. It can be tested directly using human sera to search for cross reaction with brain proteins in model systems such as Drosophila melanogaster. The conditions might be amenable to treatment using pooled immunoglobulin. Identification and elimination from the microbial flora of the bacteria that express the cross reacting antigens should be possible.
Collapse
|
43
|
Ericson MD, Schnell SM, Freeman KT, Haskell-Luevano C. A fragment of the Escherichia coli ClpB heat-shock protein is a micromolar melanocortin 1 receptor agonist. Bioorg Med Chem Lett 2015; 25:5306-8. [PMID: 26433448 DOI: 10.1016/j.bmcl.2015.09.046] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Revised: 09/15/2015] [Accepted: 09/18/2015] [Indexed: 12/01/2022]
Abstract
The melanocortin system consists of five receptor subtypes (MC1-5R), endogenous agonists derived from the proopiomelanocortin gene transcript, and the antagonists agouti and agouti-related protein. The Escherichia coli heat shock protein ClpB has previously been described as an antigen mimetic to the endogenous melanocortin agonist α-MSH. Herein, we investigated if a fragment of the ClpB protein could directly signal through the melanocortin receptors. We synthesized a complementary fragment of the ClpB protein that partially aligned with α-MSH. Pharmacological assessment of this fragment resulted in no antagonist activity at the MC3R or the MC4R and no agonist activity at the MC4R. Partial receptor activation was observed for the MC3R and MC5R at 100 μM concentrations. This fragment was shown to be a full micromolar MC1R agonist and may serve as a template for future research into selective MC1R ligands.
Collapse
Affiliation(s)
- Mark D Ericson
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA
| | - Sathya M Schnell
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA
| | - Katie T Freeman
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA
| | - Carrie Haskell-Luevano
- Department of Medicinal Chemistry, University of Minnesota, 308 Harvard Street SE, Minneapolis, MN 55455, USA.
| |
Collapse
|
44
|
Dopamine release in the lateral hypothalamus is stimulated by α-MSH in both the anticipatory and consummatory phases of feeding. Psychoneuroendocrinology 2015; 56:79-87. [PMID: 25805178 DOI: 10.1016/j.psyneuen.2015.02.020] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 02/24/2015] [Accepted: 02/24/2015] [Indexed: 12/14/2022]
Abstract
α-Melanocyte-stimulating hormone (α-MSH), is a hypothalamic neuropeptide signaling satiation, but it is not known if α-MSH may stimulate dopamine release in a feeding control brain region of the lateral hypothalamic area (LHA), during the anticipatory and consummatory phases of feeding behavior. To address these questions, dynamics of dopamine release were measured in 15 min microdialysis samples simultaneously from the LHA and the nucleus accumbens (NAc) during consecutive exposure and provision of food and 1% sucrose in Wistar rats after overnight food deprivation. α-MSH was infused via the microdialysis probe either into the LHA or NAc starting before food exposure. Food, sucrose and water intakes were automatically monitored and analyzed concomitantly with microdialysis samples. We found that LHA-α-MSH-infused rats stopped eating earlier and consumed less food and sucrose as compared to control and NAc-α-MSH-infused rats. Exposure to food produced a peak of LHA dopamine in both LHA-α-MSH and NAc-α-MSH-infused rats but not in the controls. During food provision, LHA dopamine levels were strongly elevated in LHA-α-MSH infused rats, while delivery of α-MSH into the NAc induced a less intense increase of dopamine in both NAc and LHA. In all rats, LHA dopamine levels correlated inversely with sucrose intake. In conclusion, our study showed that α-MSH stimulates dopamine release in the LHA during both the anticipatory and consummatory phases of feeding, decreases food intake and inhibits sucrose intake. These data suggest that LHA dopamine release can be involved in α-MSH anorexigenic effects.
Collapse
|
45
|
Microbiome, peptide autoantibodies, and eating disorders: A missing link between gut and brain. Nutrition 2015; 31:544-5. [DOI: 10.1016/j.nut.2015.01.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
46
|
Tennoune N, Legrand R, Ouelaa W, Breton J, Lucas N, Bole-Feysot C, do Rego JC, Déchelotte P, Fetissov SO. Sex-related effects of nutritional supplementation of Escherichia coli: relevance to eating disorders. Nutrition 2014; 31:498-507. [PMID: 25701341 DOI: 10.1016/j.nut.2014.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 11/24/2014] [Accepted: 11/26/2014] [Indexed: 02/09/2023]
Abstract
OBJECTIVES The biological background of sex-related differences in the development of eating disorders (EDs) is unknown. Recent data showed that gut bacteria Escherichia coli induce autoantibodies against anorexigenic α-melanocyte-stimulating hormone (α-MSH) associated with psychopathology in ED. The aim of this study was to compare the effects of E. coli on feeding and autoantibodies against α-MSH and adrenocorticotropic hormone (ACTH), between female and male rats. METHODS Commensal E. coli K12 were given in a culture medium daily to adult Wistar rats by intragastric gavage over a 3-wk period; control rats received culture medium only. RESULTS Before gavage, E. coli K12 DNA was detected in feces of female but not male rats. E. coli provision was accompanied by an increase in body weight gain in females, but a decrease in body weight gain and food intake in males. Independent of E. coli treatment, plasma levels of anti-α-MSH and ACTH immunoglobulin (Ig)G were higher in female than male rats. Females responded to E. coli by increasing α-MSH IgG levels and affinity, but males by increasing α-MSH IgM levels. Affinity of IgG for ACTH was increased in both E. coli-treated females and males, although with different kinetics. IgG from females stimulated more efficiently α-MSH-induced cyclic adenosine monophosphate production by melanocortin 4 receptor-expressing cells compared with IgG from males. DISCUSSION Sex-related response to how E. coli affects feeding and anti-melanocortin hormone antibody production may depend on the presence of these bacteria in the gut before E. coli supplementation. These data suggest that sex-related presence of certain gut bacteria may represent a risk factor for ED development.
Collapse
Affiliation(s)
- Naouel Tennoune
- Inserm UMR1073, Nutrition, Gut and Brain Laboratory, Rouen, France; Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, France
| | - Romain Legrand
- Inserm UMR1073, Nutrition, Gut and Brain Laboratory, Rouen, France; Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, France
| | - Wassila Ouelaa
- Inserm UMR1073, Nutrition, Gut and Brain Laboratory, Rouen, France; Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, France
| | - Jonathan Breton
- Inserm UMR1073, Nutrition, Gut and Brain Laboratory, Rouen, France; Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, France
| | - Nicolas Lucas
- Inserm UMR1073, Nutrition, Gut and Brain Laboratory, Rouen, France; Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, France
| | - Christine Bole-Feysot
- Inserm UMR1073, Nutrition, Gut and Brain Laboratory, Rouen, France; Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, France
| | - Jean-Claude do Rego
- Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, France; Animal behavior platform (SCAC), Rouen, France
| | - Pierre Déchelotte
- Inserm UMR1073, Nutrition, Gut and Brain Laboratory, Rouen, France; Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, France; Rouen University Hospital, CHU Charles Nicolle, Rouen, France
| | - Sergueï O Fetissov
- Inserm UMR1073, Nutrition, Gut and Brain Laboratory, Rouen, France; Institute for Research and Innovation in Biomedicine (IRIB), Rouen University, Normandy University, Rouen, France.
| |
Collapse
|
47
|
Alteration of intestinal barrier function during activity-based anorexia in mice. Clin Nutr 2014; 33:1046-53. [DOI: 10.1016/j.clnu.2013.11.006] [Citation(s) in RCA: 72] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Revised: 11/07/2013] [Accepted: 11/07/2013] [Indexed: 12/12/2022]
|
48
|
Lengweiler S, Kreim S, Barman-Aksözen J, Maurer M, Minder EI. Evaluation of the Immunogenicity of the Synthetic α-Melanocyte-Stimulating Hormone (α-MSH) Analogue Afamelanotide ([Nle4- D-Phe7]-α-MSH, Scenesse®) in Erythropoietic Protoporphyria Patients by ELISA Detecting Both Anti-Afamelanotide and Anti-α-MSH Antibodies. Skin Pharmacol Physiol 2014; 28:103-13. [DOI: 10.1159/000362174] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2013] [Accepted: 03/07/2014] [Indexed: 11/19/2022]
|
49
|
Bacterial ClpB heat-shock protein, an antigen-mimetic of the anorexigenic peptide α-MSH, at the origin of eating disorders. Transl Psychiatry 2014; 4:e458. [PMID: 25290265 PMCID: PMC4350527 DOI: 10.1038/tp.2014.98] [Citation(s) in RCA: 128] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 08/13/2014] [Accepted: 08/21/2014] [Indexed: 12/12/2022] Open
Abstract
The molecular mechanisms at the origin of eating disorders (EDs), including anorexia nervosa (AN), bulimia and binge-eating disorder (BED), are currently unknown. Previous data indicated that immunoglobulins (Igs) or autoantibodies (auto-Abs) reactive with α-melanocyte-stimulating hormone (α-MSH) are involved in regulation of feeding and emotion; however, the origin of such auto-Abs is unknown. Here, using proteomics, we identified ClpB heat-shock disaggregation chaperone protein of commensal gut bacteria Escherichia coli as a conformational antigen mimetic of α-MSH. We show that ClpB-immunized mice produce anti-ClpB IgG crossreactive with α-MSH, influencing food intake, body weight, anxiety and melanocortin receptor 4 signaling. Furthermore, chronic intragastric delivery of E. coli in mice decreased food intake and stimulated formation of ClpB- and α-MSH-reactive antibodies, while ClpB-deficient E. coli did not affect food intake or antibody levels. Finally, we show that plasma levels of anti-ClpB IgG crossreactive with α-MSH are increased in patients with AN, bulimia and BED, and that the ED Inventory-2 scores in ED patients correlate with anti-ClpB IgG and IgM, which is similar to our previous findings for α-MSH auto-Abs. In conclusion, this work shows that the bacterial ClpB protein, which is present in several commensal and pathogenic microorganisms, can be responsible for the production of auto-Abs crossreactive with α-MSH, associated with altered feeding and emotion in humans with ED. Our data suggest that ClpB-expressing gut microorganisms might be involved in the etiology of EDs.
Collapse
|
50
|
Raevuori A, Haukka J, Vaarala O, Suvisaari JM, Gissler M, Grainger M, Linna MS, Suokas JT. The increased risk for autoimmune diseases in patients with eating disorders. PLoS One 2014; 9:e104845. [PMID: 25147950 PMCID: PMC4141740 DOI: 10.1371/journal.pone.0104845] [Citation(s) in RCA: 88] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2014] [Accepted: 07/16/2014] [Indexed: 11/30/2022] Open
Abstract
Objective Research suggests autoimmune processes to be involved in psychiatric disorders. We aimed to address the prevalence and incidence of autoimmune diseases in a large Finnish patient cohort with anorexia nervosa, bulimia nervosa, and binge eating disorder. Methods Patients (N = 2342) treated at the Eating Disorder Unit of Helsinki University Central Hospital between 1995 and 2010 were compared with general population controls (N = 9368) matched for age, sex, and place of residence. Data of 30 autoimmune diseases from the Hospital Discharge Register from 1969 to 2010 were analyzed using conditional and Poisson regression models. Results Of patients, 8.9% vs. 5.4% of control individuals had been diagnosed with one or more autoimmune disease (OR 1.7, 95% CI 1.5–2.0, P<0.001). The increase in endocrinological diseases (OR 2.4, 95% CI 1.8–3.2, P<0.001) was explained by type 1 diabetes, whereas Crohn's disease contributed most to the risk of gastroenterological diseases (OR 1.8, 95% CI 1.4–2.5, P<0.001). Higher prevalence of autoimmune diseases among patients with eating disorders was not exclusively due to endocrinological and gastroenterological diseases; when the two categories were excluded, the increase in prevalence was seen in the patients both before the onset of the eating disorder treatment (OR 1.5, 95% CI 1.1–2.1, P = 0.02) and at the end of the follow-up (OR 1.4, 95% CI 1.1–1.8, P = 0.01). Conclusions We observed an association between eating disorders and several autoimmune diseases with different genetic backgrounds. Our findings support the link between immune-mediated mechanisms and development of eating disorders. Future studies are needed to further explore the risk of autoimmune diseases and immunological mechanisms in individuals with eating disorders and their family members.
Collapse
Affiliation(s)
- Anu Raevuori
- Hjelt Institute, Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Adolescent Psychiatry, Helsinki University Central Hospital, Helsinki, Finland
- Institute of Clinical Medicine, Child Psychiatry, University of Turku, Turku, Finland
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
- * E-mail:
| | - Jari Haukka
- Hjelt Institute, Department of Public Health, University of Helsinki, Helsinki, Finland
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
| | - Outi Vaarala
- Department of Vaccination and Immune Protection, National Institute for Health and Welfare, Helsinki, Finland
| | - Jaana M. Suvisaari
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
- Department of Social Psychiatry, Tampere School of Public Health, Tampere, Finland
| | - Mika Gissler
- Information Department, National Institute for Health and Welfare, Helsinki, Finland
- Nordic School of Public Health, Gothenburg, Sweden
| | - Marjut Grainger
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
| | - Milla S. Linna
- Hjelt Institute, Department of Public Health, University of Helsinki, Helsinki, Finland
| | - Jaana T. Suokas
- Department of Mental Health and Substance Abuse Services, National Institute for Health and Welfare, Helsinki, Finland
- Department of Psychiatry, Helsinki University Central Hospital, Helsinki, Finland
| |
Collapse
|