1
|
Dory EK, Gueta A, Loterstein Y, Moshe L, Matas D, Koren L, Weller A. Intergenerational transfer of binge eating-like behavior: The additive impact of juvenile stress. Appetite 2024; 203:107713. [PMID: 39396762 DOI: 10.1016/j.appet.2024.107713] [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] [Received: 07/18/2024] [Revised: 10/10/2024] [Accepted: 10/11/2024] [Indexed: 10/15/2024]
Abstract
Binge eating (BE) is consuming large amounts of food in a short time, while experiencing loss of control over eating behavior. BE can be hereditary, and juvenile stress (JS) may contribute to its onset. We examined the impact of JS on BE-like behavior, in an animal model of intergenerational BE. Twenty-four female Wistar rats received 2-h access to palatable food (PF) three or five times a week (3 TW or 5 TW) for 4 weeks, followed by the open field test (OFT). At postnatal day (PND)27-29, female offspring either underwent JS (O-JSC) or not (O-CC). At PND51-53, offspring's stress levels were assessed behaviorally. At PND70-85, offspring received 2-h access to PF three times a week to assess their BE-like tendency. Hair samples were collected afterwards. Compared to 5 TW, 3 TW had a greater binge size. In the elevated plus maze and dark\light box, in O-JSC, offspring of 3 TW (O-3TW) spent less time in the open arms and lit area compared to O-5TW. O-3TW consumed more PF than O-5TW. O-JSC consumed more than O-CC. O-3TW-JSC had higher hair CORT levels than O-3TW-CC and O-5TW-JSC. This study highlights the interplay between maternal and offspring experiences, allowing for the study of underlying mechanisms.
Collapse
Affiliation(s)
- Elin Kachuki Dory
- Department of Psychology, Bar Ilan University, Ramat Gan, Israel; Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan, Israel
| | - Avi Gueta
- Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan, Israel
| | - Yoni Loterstein
- Department of Psychology, Bar Ilan University, Ramat Gan, Israel; Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan, Israel
| | - Lital Moshe
- Department of Psychology, Bar Ilan University, Ramat Gan, Israel; Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan, Israel
| | - Devorah Matas
- Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan, Israel; Faculty of Life Sciences, Bar Ilan University, Ramat-Gan, Israel
| | - Lee Koren
- Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan, Israel; Faculty of Life Sciences, Bar Ilan University, Ramat-Gan, Israel
| | - Aron Weller
- Department of Psychology, Bar Ilan University, Ramat Gan, Israel; Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
2
|
Mercante F, Micioni Di Bonaventura E, Pucci M, Botticelli L, Cifani C, D'Addario C, Micioni Di Bonaventura MV. Repeated binge-like eating episodes in female rats alter adenosine A 2A and dopamine D2 receptor genes regulation in the brain reward system. Int J Eat Disord 2024; 57:1433-1446. [PMID: 38650547 DOI: 10.1002/eat.24216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/22/2024] [Accepted: 04/01/2024] [Indexed: 04/25/2024]
Abstract
OBJECTIVE Binge-eating disorder is an eating disorder characterized by recurrent binge-eating episodes, during which individuals consume excessive amounts of highly palatable food (HPF) in a short time. This study investigates the intricate relationship between repeated binge-eating episode and the transcriptional regulation of two key genes, adenosine A2A receptor (A2AAR) and dopamine D2 receptor (D2R), in selected brain regions of rats. METHOD Binge-like eating behavior on HPF was induced through the combination of food restrictions and frustration stress (15 min exposure to HPF without access to it) in female rats, compared to control rats subjected to only restriction or only stress or none of these two conditions. After chronic binge-eating episodes, nucleic acids were extracted from different brain regions, and gene expression levels were assessed through real-time quantitative PCR. The methylation pattern on genes' promoters was investigated using pyrosequencing. RESULTS The analysis revealed A2AAR upregulation in the amygdala and in the ventral tegmental area (VTA), and D2R downregulation in the nucleus accumbens in binge-eating rats. Concurrently, site-specific DNA methylation alterations at gene promoters were identified in the VTA for A2AAR and in the amygdala and caudate putamen for D2R. DISCUSSION The alterations on A2AAR and D2R genes regulation highlight the significance of epigenetic mechanisms in the etiology of binge-eating behavior, and underscore the potential for targeted therapeutic interventions, to prevent the development of this maladaptive feeding behavior. These findings provide valuable insights for future research in the field of eating disorders. PUBLIC SIGNIFICANCE Using an animal model with face, construct, and predictive validity, in which cycles of food restriction and frustration stress evoke binge-eating behavior, we highlight the significance of epigenetic mechanisms on adenosine A2A receptor (A2AAR) and dopamine D2 receptor (D2R) genes regulation. They could represent new potential targets for the pharmacological management of eating disorders characterized by this maladaptive feeding behavior.
Collapse
Affiliation(s)
- Francesca Mercante
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
| | | | - Mariangela Pucci
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
- Department of Biosciences and Nutrition, Karolinska Institute, Huddinge, Sweden
| | - Luca Botticelli
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Carlo Cifani
- Pharmacology Unit, School of Pharmacy, University of Camerino, Camerino, Italy
| | - Claudio D'Addario
- Department of Bioscience and Technology for Food, Agriculture and Environment, University of Teramo, Teramo, Italy
- Department of Clinical Neuroscience, Karolinska Institutet, Stockholm, Sweden
| | | |
Collapse
|
3
|
Al-Daffaie FM, Al-Mudhafar SF, Alhomsi A, Tarazi H, Almehdi AM, El-Huneidi W, Abu-Gharbieh E, Bustanji Y, Alqudah MAY, Abuhelwa AY, Guella A, Alzoubi KH, Semreen MH. Metabolomics and Proteomics in Prostate Cancer Research: Overview, Analytical Techniques, Data Analysis, and Recent Clinical Applications. Int J Mol Sci 2024; 25:5071. [PMID: 38791108 PMCID: PMC11120916 DOI: 10.3390/ijms25105071] [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: 02/12/2024] [Revised: 04/26/2024] [Accepted: 04/26/2024] [Indexed: 05/26/2024] Open
Abstract
Prostate cancer (PCa) is a significant global contributor to mortality, predominantly affecting males aged 65 and above. The field of omics has recently gained traction due to its capacity to provide profound insights into the biochemical mechanisms underlying conditions like prostate cancer. This involves the identification and quantification of low-molecular-weight metabolites and proteins acting as crucial biochemical signals for early detection, therapy assessment, and target identification. A spectrum of analytical methods is employed to discern and measure these molecules, revealing their altered biological pathways within diseased contexts. Metabolomics and proteomics generate refined data subjected to detailed statistical analysis through sophisticated software, yielding substantive insights. This review aims to underscore the major contributions of multi-omics to PCa research, covering its core principles, its role in tumor biology characterization, biomarker discovery, prognostic studies, various analytical technologies such as mass spectrometry and Nuclear Magnetic Resonance, data processing, and recent clinical applications made possible by an integrative "omics" approach. This approach seeks to address the challenges associated with current PCa treatments. Hence, our research endeavors to demonstrate the valuable applications of these potent tools in investigations, offering significant potential for understanding the complex biochemical environment of prostate cancer and advancing tailored therapeutic approaches for further development.
Collapse
Affiliation(s)
- Fatima M. Al-Daffaie
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (F.M.A.-D.); (S.F.A.-M.); (A.A.); (H.T.); (A.M.A.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (W.E.-H.); (E.A.-G.); (A.Y.A.); (K.H.A.)
| | - Sara F. Al-Mudhafar
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (F.M.A.-D.); (S.F.A.-M.); (A.A.); (H.T.); (A.M.A.)
| | - Aya Alhomsi
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (F.M.A.-D.); (S.F.A.-M.); (A.A.); (H.T.); (A.M.A.)
| | - Hamadeh Tarazi
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (F.M.A.-D.); (S.F.A.-M.); (A.A.); (H.T.); (A.M.A.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (W.E.-H.); (E.A.-G.); (A.Y.A.); (K.H.A.)
| | - Ahmed M. Almehdi
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (F.M.A.-D.); (S.F.A.-M.); (A.A.); (H.T.); (A.M.A.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (W.E.-H.); (E.A.-G.); (A.Y.A.); (K.H.A.)
| | - Waseem El-Huneidi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (W.E.-H.); (E.A.-G.); (A.Y.A.); (K.H.A.)
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates;
| | - Eman Abu-Gharbieh
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (W.E.-H.); (E.A.-G.); (A.Y.A.); (K.H.A.)
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Yasser Bustanji
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates;
- School of Pharmacy, The University of Jordan, Amman 11942, Jordan
| | - Mohammad A. Y. Alqudah
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates;
- Department of Clinical Pharmacy, Faculty of Pharmacy, Jordan University of Science and Technology, Irbid 22110, Jordan
| | - Ahmad Y. Abuhelwa
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (W.E.-H.); (E.A.-G.); (A.Y.A.); (K.H.A.)
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates;
| | - Adnane Guella
- Nephrology Department, University Hospital Sharjah, Sharjah 27272, United Arab Emirates;
| | - Karem H. Alzoubi
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (W.E.-H.); (E.A.-G.); (A.Y.A.); (K.H.A.)
- Department of Pharmacy Practice and Pharmacotherapeutics, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates;
| | - Mohammad H. Semreen
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah 27272, United Arab Emirates; (F.M.A.-D.); (S.F.A.-M.); (A.A.); (H.T.); (A.M.A.)
- Research Institute of Medical and Health Sciences, University of Sharjah, Sharjah 27272, United Arab Emirates; (W.E.-H.); (E.A.-G.); (A.Y.A.); (K.H.A.)
| |
Collapse
|
4
|
Schroeder M, Fuenzalida B, Yi N, Shahnawaz S, Gertsch J, Pellegata D, Ontsouka E, Leiva A, Gutiérrez J, Müller M, Brocco MA, Albrecht C. LAT1-dependent placental methionine uptake is a key player in fetal programming of metabolic disease. Metabolism 2024; 153:155793. [PMID: 38295946 DOI: 10.1016/j.metabol.2024.155793] [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: 10/17/2023] [Revised: 12/11/2023] [Accepted: 01/09/2024] [Indexed: 02/06/2024]
Abstract
The Developmental Origins of Health and Disease hypothesis sustains that exposure to different stressors during prenatal development prepares the offspring for the challenges to be encountered after birth. We studied the gestational period as a particularly vulnerable window where different stressors can have strong implications for fetal programming of the offspring's life-long metabolic status via alterations of specific placentally expressed nutrient transporters. To study this mechanism, we used a murine prenatal stress model, human preeclampsia, early miscarriage, and healthy placental tissue samples, in addition to in vitro models of placental cells. In stressed mice, placental overexpression of L-type amino acid transporter 1 (Lat1) and subsequent global placental DNA hypermethylation was accompanied by fetal and adult hypothalamic dysregulation in global DNA methylation and gene expression as well as long-term metabolic abnormalities exclusively in female offspring. In human preeclampsia, early miscarriage, and under hypoxic conditions, placental LAT1 was significantly upregulated, leading to increased methionine uptake and global DNA hypermethylation. Remarkably, subgroups of healthy term placentas with high expression of stress-related genes presented increased levels of placental LAT1 mRNA and protein, DNA and RNA hypermethylation, increased methionine uptake capacity, one-carbon metabolic pathway disruption, higher methionine concentration in the placenta and transport to the fetus specifically in females. Since LAT1 mediates the intracellular accumulation of methionine, global DNA methylation, and one-carbon metabolism in the placenta, our findings hint at a major sex-specific global response to a variety of prenatal stressors affecting placental function, epigenetic programming, and life-long metabolic disease and provide a much-needed insight into early-life factors predisposing females/women to metabolic disorders.
Collapse
Affiliation(s)
- Mariana Schroeder
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland.
| | - Barbara Fuenzalida
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Nan Yi
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Saira Shahnawaz
- Department of Biochemistry, Sargodha Medical College, University of Sargodha, Sargodha, Pakistan; Department of Allied Health Sciences, Sargodha Medical College, University of Sargodha, Sargodha, Pakistan
| | - Jürg Gertsch
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland
| | - Daniele Pellegata
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Edgar Ontsouka
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland
| | - Andrea Leiva
- Faculty of Medicine and Science, Universidad of San Sebastian, Santiago, Chile
| | - Jaime Gutiérrez
- Faculty of Medicine and Science, Universidad of San Sebastian, Santiago, Chile
| | - Martin Müller
- Division of Gynecology and Obstetrics, Lindenhofgruppe, Bern, Switzerland
| | - Marcela A Brocco
- Institute of Biotechnological Research, University of San Martín, Buenos Aires, Argentina
| | - Christiane Albrecht
- Faculty of Medicine, Institute of Biochemistry and Molecular Medicine, University of Bern, Switzerland; Swiss National Centre of Competence in Research, NCCR TransCure, University of Bern, Bern, Switzerland
| |
Collapse
|
5
|
Yoshikawa C, Ariyani W, Kohno D. DNA Methylation in the Hypothalamic Feeding Center and Obesity. J Obes Metab Syndr 2023; 32:303-311. [PMID: 38124554 PMCID: PMC10786209 DOI: 10.7570/jomes23073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/28/2023] [Accepted: 12/19/2023] [Indexed: 12/23/2023] Open
Abstract
Obesity rates have been increasing worldwide for decades, mainly due to environmental factors, such as diet, nutrition, and exercise. However, the molecular mechanisms through which environmental factors induce obesity remain unclear. Several mechanisms underlie the body's response to environmental factors, and one of the main mechanisms involves epigenetic modifications, such as DNA methylation. The pattern of DNA methylation is influenced by environmental factors, and altered DNA methylation patterns can affect gene expression profiles and phenotypes. DNA methylation may mediate the development of obesity caused by environmental factors. Similar to the factors governing obesity, DNA methylation is influenced by nutrients and metabolites. Notably, DNA methylation is associated with body size and weight programming. The DNA methylation levels of proopiomelanocortin (Pomc) and neuropeptide Y (Npy) in the hypothalamic feeding center, a key region controlling systemic energy balance, are affected by diet. Conditional knockout mouse studies of epigenetic enzymes have shown that DNA methylation in the hypothalamic feeding center plays an indispensable role in energy homeostasis. In this review, we discuss the role of DNA methylation in the hypothalamic feeding center as a potential mechanism underlying the development of obesity induced by environmental factors.
Collapse
Affiliation(s)
- Chiharu Yoshikawa
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Winda Ariyani
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| | - Daisuke Kohno
- Metabolic Signal Research Center, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Japan
| |
Collapse
|
6
|
Abstract
Eating behaviours are determined by the integration of interoceptive and environmental inputs. During pregnancy, numerous physiological adaptations take place in the maternal organism to provide an adequate environment for embryonic growth. Among them, whole-body physiological remodelling directly influences eating patterns, commonly causing notable taste perception alterations, food aversions and cravings. Recurrent food cravings for and compulsive eating of highly palatable food can contribute to the development and maintenance of gestational overweight and obesity with potential adverse health consequences for the offspring. Although much is known about how maternal eating habits influence offspring health, the mechanisms that underlie changes in taste perception and food preference during pregnancy (which guide and promote feeding) are only just starting to be elucidated. Given the limited and diffuse understanding of the neurobiology of gestational eating patterns, the aim of this Review is to compile, integrate and discuss the research conducted on this topic in both experimental models and humans. This article sheds light on the mechanisms that drive changes in female feeding behaviours during distinct physiological states. Understanding these processes is crucial to improve gestational parent health and decrease the burden of metabolic and food-related diseases in future generations.
Collapse
Affiliation(s)
- Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
7
|
Deer LK, Su C, Thwaites NA, Davis EP, Doom JR. A framework for testing pathways from prenatal stress-responsive hormones to cardiovascular disease risk. Front Endocrinol (Lausanne) 2023; 14:1111474. [PMID: 37223037 PMCID: PMC10200937 DOI: 10.3389/fendo.2023.1111474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Accepted: 04/10/2023] [Indexed: 05/25/2023] Open
Abstract
Cardiovascular disease (CVD) is a leading cause of death globally, with the prevalence projected to keep rising. Risk factors for adult CVD emerge at least as early as the prenatal period. Alterations in stress-responsive hormones in the prenatal period are hypothesized to contribute to CVD in adulthood, but little is known about relations between prenatal stress-responsive hormones and early precursors of CVD, such as cardiometabolic risk and health behaviors. The current review presents a theoretical model of the relation between prenatal stress-responsive hormones and adult CVD through cardiometabolic risk markers (e.g., rapid catch-up growth, high BMI/adiposity, high blood pressure, and altered blood glucose, lipids, and metabolic hormones) and health behaviors (e.g., substance use, poor sleep, poor diet and eating behaviors, and low physical activity levels). Emerging evidence in human and non-human animal literatures suggest that altered stress-responsive hormones during gestation predict higher cardiometabolic risk and poorer health behaviors in offspring. This review additionally highlights limitations of the current literature (e.g., lack of racial/ethnic diversity, lack of examination of sex differences), and discusses future directions for this promising area of research.
Collapse
Affiliation(s)
- LillyBelle K. Deer
- Department of Psychology, University of Denver, Denver, CO, United States
| | - Chen Su
- Department of Psychology, University of Denver, Denver, CO, United States
| | | | - Elysia Poggi Davis
- Department of Psychology, University of Denver, Denver, CO, United States
- Department of Psychiatry & Human Behavior, University of California, Irvine, Irvine, CA, United States
| | - Jenalee R. Doom
- Department of Psychology, University of Denver, Denver, CO, United States
| |
Collapse
|
8
|
Cao C, Siegel PB, Gilbert ER, Cline MA. Epigenetic modifiers identified as regulators of food intake in a unique hypophagic chicken model. Animal 2022; 16:100549. [PMID: 35679817 DOI: 10.1016/j.animal.2022.100549] [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: 03/08/2021] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 11/01/2022] Open
Abstract
DNA methylation is an epigenetic modification that influences gene transcription; however, the effects of methylation-influencing chemicals on appetite are unknown. We evaluated the effects of single administration of a methyl donor, S-Adenosylmethionine (SAM), or methylation inhibitor, 5-Azacytidine (AZA), on immediate and later-age food intake in an anorexic chick model. The doses of intracerebroventricularly-injected SAM were 0 (vehicle), 0.1, 1, and 10 μg, and of AZA were 0 (vehicle), 1, 5, and 25 μg. When injected on day 5 posthatch, there was no effect of SAM on food intake in either fed or fasted chicks, whereas AZA increased food consumption in the fasted state but decreased it in fed chicks. We then performed a single injection (same doses) at hatch and measured food intake on day 5 in response to neuropeptide Y (NPY; 0.2 μg) injection. Irrespective of NPY, chicks injected with 1 μg of SAM ate more than others on day 5. In contrast, chicks injected with AZA (5 and 25 μg doses) consumed less on day 5. In conclusion, we identified DNA methylation-regulating chemicals as regulators of food intake. AZA but not SAM affected food intake in the short-term, feeding state dependently. Later, both chemicals injected on the day of hatch were associated with food intake changes at a later age, suggesting that feeding pathways might be altered through changes in methylation.
Collapse
Affiliation(s)
- C Cao
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - P B Siegel
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - E R Gilbert
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - M A Cline
- Department of Animal and Poultry Sciences, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA.
| |
Collapse
|
9
|
Resurreccion EP, Fong KW. The Integration of Metabolomics with Other Omics: Insights into Understanding Prostate Cancer. Metabolites 2022; 12:metabo12060488. [PMID: 35736421 PMCID: PMC9230859 DOI: 10.3390/metabo12060488] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/21/2022] [Accepted: 05/24/2022] [Indexed: 02/06/2023] Open
Abstract
Our understanding of prostate cancer (PCa) has shifted from solely caused by a few genetic aberrations to a combination of complex biochemical dysregulations with the prostate metabolome at its core. The role of metabolomics in analyzing the pathophysiology of PCa is indispensable. However, to fully elucidate real-time complex dysregulation in prostate cells, an integrated approach based on metabolomics and other omics is warranted. Individually, genomics, transcriptomics, and proteomics are robust, but they are not enough to achieve a holistic view of PCa tumorigenesis. This review is the first of its kind to focus solely on the integration of metabolomics with multi-omic platforms in PCa research, including a detailed emphasis on the metabolomic profile of PCa. The authors intend to provide researchers in the field with a comprehensive knowledge base in PCa metabolomics and offer perspectives on overcoming limitations of the tool to guide future point-of-care applications.
Collapse
Affiliation(s)
- Eleazer P. Resurreccion
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA;
| | - Ka-wing Fong
- Department of Toxicology and Cancer Biology, University of Kentucky, Lexington, KY 40506, USA;
- Markey Cancer Center, University of Kentucky, Lexington, KY 40506, USA
- Correspondence: ; Tel.: +1-859-562-3455
| |
Collapse
|
10
|
A Framework for Developing Translationally Relevant Animal Models of Stress-Induced Changes in Eating Behavior. Biol Psychiatry 2022; 91:888-897. [PMID: 34433512 PMCID: PMC8720907 DOI: 10.1016/j.biopsych.2021.06.020] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 06/22/2021] [Accepted: 06/24/2021] [Indexed: 02/08/2023]
Abstract
Stress often affects eating behaviors, leading to increased eating in some individuals and decreased eating in others. Identifying physiological and psychological factors that determine the direction of eating responses to stress has been a major goal of epidemiological and clinical studies. However, challenges of standardizing the stress exposure in humans hinder efforts to uncover the underlying mechanisms. The issue of what determines the direction of stress-induced feeding responses has not been directly addressed in animal models, but assays that combine stress with a feeding-related task are commonly used as readouts of other behaviors, such as anxiety. Sex, estrous cyclicity, circadian cyclicity, caloric restriction, palatable diets, elevated body weight, and properties of the stressors similarly influence feeding behavior in humans and rodent models. Yet, most rodent studies do not use conditions that are most relevant for studying feeding behavior in humans. This review proposes a conceptual framework for incorporating these influences to develop reproducible and translationally relevant assays to study effects of stress on food intake. Such paradigms have the potential to uncover links between emotional eating and obesity as well as to the etiology of eating disorders.
Collapse
|
11
|
Haddad-Tóvolli R, Ramírez S, Muñoz-Moreno E, Milà-Guasch M, Miquel-Rio L, Pozo M, Chivite I, Altirriba J, Obri A, Gómez-Valadés AG, Toledo M, Eyre E, Bortolozzi A, Valjent E, Soria G, Claret M. Food craving-like episodes during pregnancy are mediated by accumbal dopaminergic circuits. Nat Metab 2022; 4:424-434. [PMID: 35379970 DOI: 10.1038/s42255-022-00557-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 02/22/2022] [Indexed: 12/22/2022]
Abstract
Preparation for motherhood requires a myriad of physiological and behavioural adjustments throughout gestation to provide an adequate environment for proper embryonic development1. Cravings for highly palatable foods are highly prevalent during pregnancy2 and contribute to the maintenance and development of gestational overweight or obesity3. However, the neurobiology underlying the distinct ingestive behaviours that result from craving specific foods remain unknown. Here we show that mice, similarly to humans, experience gestational food craving-like episodes. These episodes are associated with a brain connectivity reorganization that affects key components of the dopaminergic mesolimbic circuitry, which drives motivated appetitive behaviours and facilitates the perception of rewarding stimuli. Pregnancy engages a dynamic modulation of dopaminergic signalling through neurons expressing dopamine D2 receptors in the nucleus accumbens, which directly modulate food craving-like events. Importantly, persistent maternal food craving-like behaviour has long-lasting effects on the offspring, particularly in males, leading to glucose intolerance, increased body weight and increased susceptibility to develop eating disorders and anxiety-like behaviours during adulthood. Our results reveal the cognitively motivated nature of pregnancy food cravings and advocates for moderating emotional eating during gestation to prevent deterioration of the offspring's neuropsychological and metabolic health.
Collapse
Affiliation(s)
- Roberta Haddad-Tóvolli
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| | - Sara Ramírez
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Emma Muñoz-Moreno
- Experimental 7T MRI Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Maria Milà-Guasch
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Lluis Miquel-Rio
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | - Macarena Pozo
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Iñigo Chivite
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Jordi Altirriba
- Laboratory of Metabolism, Department of Internal Medicine Specialties, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Arnaud Obri
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Alicia G Gómez-Valadés
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Miriam Toledo
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Elena Eyre
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Analia Bortolozzi
- Institut d'Investigacions Biomèdiques de Barcelona (IIBB), Consejo Superior de Investigaciones Científicas (CSIC), Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- CIBER de Salud Mental (CIBERSAM), Madrid, Spain
| | | | - Guadalupe Soria
- Experimental 7T MRI Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Laboratory of Surgical Neuroanatomy, Faculty of Medicine and Health Sciences, Institute of Neurosciences, Universitat de Barcelona, Barcelona, Spain
- CIBER de Bioingeniera, Biomateriales y Nanomedicina (CIBER-BBN), Barcelona, Spain
| | - Marc Claret
- Neuronal Control of Metabolism (NeuCoMe) Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Barcelona, Spain.
- School of Medicine, Universitat de Barcelona, Barcelona, Spain.
| |
Collapse
|
12
|
Rasmussen JM, Thompson PM, Entringer S, Buss C, Wadhwa PD. Fetal programming of human energy homeostasis brain networks: Issues and considerations. Obes Rev 2022; 23:e13392. [PMID: 34845821 DOI: 10.1111/obr.13392] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/29/2021] [Accepted: 10/24/2021] [Indexed: 02/07/2023]
Abstract
In this paper, we present a transdisciplinary framework and testable hypotheses regarding the process of fetal programming of energy homeostasis brain circuitry. Our model proposes that key aspects of energy homeostasis brain circuitry already are functional by the time of birth (with substantial interindividual variation); that this phenotypic variation at birth is an important determinant of subsequent susceptibility for energy imbalance and childhood obesity risk; and that this brain circuitry exhibits developmental plasticity, in that it is influenced by conditions during intrauterine life, particularly maternal-placental-fetal endocrine, immune/inflammatory, and metabolic processes and their upstream determinants. We review evidence that supports the scientific premise for each element of this formulation, identify future research directions, particularly recent advances that may facilitate a better quantification of the ontogeny of energy homeostasis brain networks, highlight animal and in vitro-based approaches that may better address the determinants of interindividual variation in energy homeostasis brain networks, and discuss the implications of this formulation for the development of strategies targeted towards the primary prevention of childhood obesity.
Collapse
Affiliation(s)
- Jerod M Rasmussen
- Development, Health and Disease Research Program, University of California, Irvine, California, USA.,Department of Pediatrics, University of California, Irvine, California, USA
| | - Paul M Thompson
- Imaging Genetics Center, Mark and Mary Stevens Institute for Neuroimaging and Informatics, Keck School of Medicine, University of Southern California, Los Angeles, California, USA
| | - Sonja Entringer
- Development, Health and Disease Research Program, University of California, Irvine, California, USA.,Department of Pediatrics, University of California, Irvine, California, USA.,Department of Medical Psychology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Departments of Psychiatry and Human Behavior, Obstetrics and Gynecology, Epidemiology, University of California, Irvine, California, USA
| | - Claudia Buss
- Development, Health and Disease Research Program, University of California, Irvine, California, USA.,Department of Pediatrics, University of California, Irvine, California, USA.,Department of Medical Psychology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany.,Departments of Psychiatry and Human Behavior, Obstetrics and Gynecology, Epidemiology, University of California, Irvine, California, USA
| | - Pathik D Wadhwa
- Development, Health and Disease Research Program, University of California, Irvine, California, USA.,Department of Pediatrics, University of California, Irvine, California, USA.,Departments of Psychiatry and Human Behavior, Obstetrics and Gynecology, Epidemiology, University of California, Irvine, California, USA.,Department of Obstetrics and Gynecology, University of California, Irvine, California, USA.,Department of Epidemiology, University of California, Irvine, California, USA
| |
Collapse
|
13
|
Abstract
Animal and humans exposed to stress early in life are more likely to suffer from long-term behavioral, mental health, metabolic, immune, and cardiovascular health consequences. The hypothalamus plays a nodal role in programming, controlling, and regulating stress responses throughout the life course. Epigenetic reprogramming in the hippocampus and the hypothalamus play an important role in adapting genome function to experiences and exposures during the perinatal and early life periods and setting up stable phenotypic outcomes. Epigenetic programming during development enables one genome to express multiple cell type identities. The most proximal epigenetic mark to DNA is a covalent modification of the DNA itself by enzymatic addition of methyl moieties. Cell-type-specific DNA methylation profiles are generated during gestational development and define cell and tissue specific phenotypes. Programming of neuronal phenotypes and sex differences in the hypothalamus is achieved by developmentally timed rearrangement of DNA methylation profiles. Similarly, other stations in the life trajectory such as puberty and aging involve predictable and scheduled reorganization of DNA methylation profiles. DNA methylation and other epigenetic marks are critical for maintaining cell-type identity in the brain, across the body, and throughout life. Data that have emerged in the last 15 years suggest that like its role in defining cell-specific phenotype during development, DNA methylation might be involved in defining experiential identities, programming similar genes to perform differently in response to diverse experiential histories. Early life stress impact on lifelong phenotypes is proposed to be mediated by DNA methylation and other epigenetic marks. Epigenetic marks, as opposed to genetic mutations, are reversible by either pharmacological or behavioral strategies and therefore offer the potential for reversing or preventing disease including behavioral and mental health disorders. This chapter discusses data testing the hypothesis that DNA methylation modulations of the HPA axis mediate the impact of early life stress on lifelong behavioral and physical phenotypes.
Collapse
Affiliation(s)
- Moshe Szyf
- Department of Pharmacology and Therapeutics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
14
|
Hill EM, Howard CD, Bale TL, Jašarević E. Perinatal exposure to tetracycline contributes to lasting developmental effects on offspring. Anim Microbiome 2021; 3:37. [PMID: 33975649 PMCID: PMC8111738 DOI: 10.1186/s42523-021-00099-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/27/2021] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND For more than 30 years, the tetracycline on/off system of inducible gene expression has been leveraged to study disease mechanisms across many research areas, especially that of metabolism and neuroscience. This system requires acute or chronic exposure to tetracycline derivatives, such as doxycycline, to manipulate gene expression in a temporal and tissue-specific manner, with exposure often being restricted to gestational and early developmental windows. Despite evidence showing that early life antibiotic exposure has adverse effects on gut microbiota, metabolism, physiology, immunity and behavior, little is known regarding the lasting impact of doxycycline treatment on relevant outcomes in experimental offspring. RESULTS To examine the hypothesis that early life doxycycline exposure produces effects on offspring growth, behavior, and gut microbiota, we employed the most commonly used method for tetracycline on/off system by administering a low dose of doxycycline (0.5 mg/ml) in the drinking water to C57Bl/6J and C57BL/6J:129S1/SvImJ dams from embryonic day 15.5 to postnatal day 28. Developmental exposure to low dose doxycycline resulted in significant alterations to growth trajectories and body weight in both strains, which persisted beyond cessation of doxycycline exposure. Developmental doxycycline exposure influenced offspring bacterial community assembly in a temporal and sex-specific manner. Further, gut microbiota composition failed to recover by adulthood, suggesting a lasting imprint of developmental antibiotic exposure. CONCLUSIONS Our results demonstrated that early life doxycycline exposure shifts the homeostatic baseline of prior exposed animals that may subsequently impact responses to experimental manipulations. These results highlight the gut microbiota as an important factor to consider in systems requiring methods of chronic antibiotic administration during pregnancy and critical periods of postnatal development.
Collapse
Affiliation(s)
- Elizabeth M Hill
- Center for Epigenetics Research in Child Health and Brain Development, Department of Pharmacology, University of Maryland School of Medicine, Baltimore, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christopher D Howard
- Center for Epigenetics Research in Child Health and Brain Development, Department of Pharmacology, University of Maryland School of Medicine, Baltimore, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Tracy L Bale
- Center for Epigenetics Research in Child Health and Brain Development, Department of Pharmacology, University of Maryland School of Medicine, Baltimore, USA
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Eldin Jašarević
- Center for Epigenetics Research in Child Health and Brain Development, Department of Pharmacology, University of Maryland School of Medicine, Baltimore, USA.
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
15
|
López-Taboada I, González-Pardo H, Conejo NM. Western Diet: Implications for Brain Function and Behavior. Front Psychol 2020; 11:564413. [PMID: 33329193 PMCID: PMC7719696 DOI: 10.3389/fpsyg.2020.564413] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 10/02/2020] [Indexed: 12/12/2022] Open
Abstract
The Western diet (WD) pattern characterized by high daily intake of saturated fats and refined carbohydrates often leads to obesity and overweight, and it has been linked to cognitive impairment and emotional disorders in both animal models and humans. This dietary pattern alters the composition of gut microbiota, influencing brain function by different mechanisms involving the gut-brain axis. In addition, long-term exposure to highly palatable foods typical of WD could induce addictive-like eating behaviors and hypothalamic-pituitary-adrenal (HPA) axis dysregulation associated with chronic stress, anxiety, and depression. In turn, chronic stress modulates eating behavior, and it could have detrimental effects on different brain regions such as the hippocampus, hypothalamus, amygdala, and several cortical regions. Moreover, obesity and overweight induce neuroinflammation, causing neuronal dysfunction. In this review, we summarize the current scientific evidence about the mechanisms and factors relating WD consumption with altered brain function and behavior. Possible therapeutic interventions and limitations are also discussed, aiming to tackle and prevent this current pandemic.
Collapse
Affiliation(s)
| | | | - Nélida María Conejo
- Laboratory of Neuroscience, Department of Psychology, Instituto de Neurociencias del Principado de Asturias (INEUROPA), University of Oviedo, Oviedo, Spain
| |
Collapse
|
16
|
Cirulli F, Musillo C, Berry A. Maternal Obesity as a Risk Factor for Brain Development and Mental Health in the Offspring. Neuroscience 2020; 447:122-135. [PMID: 32032668 DOI: 10.1016/j.neuroscience.2020.01.023] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 01/10/2020] [Accepted: 01/14/2020] [Indexed: 12/30/2022]
Abstract
Maternal obesity plays a key role in the health trajectory of the offspring. Although research on this topic has largely focused on the potential of this condition to increase the risk for child obesity, it is becoming more and more evident that it can also significantly impact cognitive function and mental health. The mechanisms underlying these effects are starting to be elucidated and point to the placenta as a critical organ that may mediate changes in the response to stress, immune function and oxidative stress. Long-term effects of maternal obesity may rely upon epigenetic changes in selected genes that are involved in metabolic and trophic regulations of the brain. More recent evidence also indicates the gut microbiota as a potential mediator of these effects. Overall, understanding cause-effect relationships can allow the development of preventive measures that could rely upon dietary changes in the mother and the offspring. Addressing diets appears more feasible than developing new pharmacological targets and has the potential to affect the multiple interconnected physiological pathways engaged by these complex regulations, allowing prevention of both metabolic and mental disorders.
Collapse
Affiliation(s)
- Francesca Cirulli
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy.
| | - Chiara Musillo
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| | - Alessandra Berry
- Center for Behavioral Sciences and Mental Health, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
17
|
Glucocorticoid exposure during hippocampal neurogenesis primes future stress response by inducing changes in DNA methylation. Proc Natl Acad Sci U S A 2019; 117:23280-23285. [PMID: 31399550 DOI: 10.1073/pnas.1820842116] [Citation(s) in RCA: 116] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Prenatal stress exposure is associated with risk for psychiatric disorders later in life. This may be mediated in part via enhanced exposure to glucocorticoids (GCs), which are known to impact neurogenesis. We aimed to identify molecular mediators of these effects, focusing on long-lasting epigenetic changes. In a human hippocampal progenitor cell (HPC) line, we assessed the short- and long-term effects of GC exposure during neurogenesis on messenger RNA (mRNA) expression and DNA methylation (DNAm) profiles. GC exposure induced changes in DNAm at 27,812 CpG dinucleotides and in the expression of 3,857 transcripts (false discovery rate [FDR] ≤ 0.1 and absolute fold change [FC] expression ≥ 1.15). HPC expression and GC-affected DNAm profiles were enriched for changes observed during human fetal brain development. Differentially methylated sites (DMSs) with GC exposure clustered into 4 trajectories over HPC differentiation, with transient as well as long-lasting DNAm changes. Lasting DMSs mapped to distinct functional pathways and were selectively enriched for poised and bivalent enhancer marks. Lasting DMSs had little correlation with lasting expression changes but were associated with a significantly enhanced transcriptional response to a second acute GC challenge. A significant subset of lasting DMSs was also responsive to an acute GC challenge in peripheral blood. These tissue-overlapping DMSs were used to compute a polyepigenetic score that predicted exposure to conditions associated with altered prenatal GCs in newborn's cord blood DNA. Overall, our data suggest that early exposure to GCs can change the set point of future transcriptional responses to stress by inducing lasting DNAm changes. Such altered set points may relate to differential vulnerability to stress exposure later in life.
Collapse
|
18
|
Lebow MA, Schroeder M, Tsoory M, Holzman-Karniel D, Mehta D, Ben-Dor S, Gil S, Bradley B, Smith AK, Jovanovic T, Ressler KJ, Binder EB, Chen A. Glucocorticoid-induced leucine zipper "quantifies" stressors and increases male susceptibility to PTSD. Transl Psychiatry 2019; 9:178. [PMID: 31346158 PMCID: PMC6658561 DOI: 10.1038/s41398-019-0509-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Accepted: 05/24/2019] [Indexed: 12/04/2022] Open
Abstract
Post-traumatic stress disorder (PTSD) selectively develops in some individuals exposed to a traumatic event. Genetic and epigenetic changes in glucocorticoid pathway sensitivity may be essential for understanding individual susceptibility to PTSD. This study focuses on PTSD markers in the glucocorticoid pathway, spotlighting glucocorticoid-induced leucine zipper (GILZ), a transcription factor encoded by the gene Tsc22d3 on the X chromosome. We propose that GILZ uniquely "quantifies" exposure to stressors experienced from late gestation to adulthood and that low levels of GILZ predispose individuals to PTSD in males only. GILZ mRNA and methylation were measured in 396 male and female human blood samples from the Grady Trauma Project cohort (exposed to multiple traumatic events). In mice, changes in glucocorticoid pathway genes were assessed following exposure to stressors at distinct time points: (i) CRF-induced prenatal stress (CRF-inducedPNS) with, or without, additional exposure to (ii) PTSD induction protocol in adulthood, which induces PTSD-like behaviors in a subset of mice. In humans, the number of traumatic events correlated negatively with GILZ mRNA levels and positively with % methylation of GILZ in males only. In male mice, we observed a threefold increase in the number of offspring exhibiting PTSD-like behaviors in those exposed to both CRF-inducedPNS and PTSD induction. This susceptibility was associated with reduced GILZ mRNA levels and epigenetic changes, not found in females. Furthermore, virus-mediated shRNA knockdown of amygdalar GILZ increased susceptibility to PTSD. Mouse and human data confirm that dramatic alterations in GILZ occur in those exposed to a stressor in early life, adulthood or both. Therefore, GILZ levels may help identify at-risk populations for PTSD prior to additional traumatic exposures.
Collapse
Affiliation(s)
- Maya A. Lebow
- 0000 0004 0604 7563grid.13992.30Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel ,0000 0000 9497 5095grid.419548.5Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Mariana Schroeder
- 0000 0004 0604 7563grid.13992.30Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel ,0000 0000 9497 5095grid.419548.5Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Michael Tsoory
- 0000 0004 0604 7563grid.13992.30Department of Veterinary Resources, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Dorin Holzman-Karniel
- 0000 0004 0604 7563grid.13992.30Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Divya Mehta
- 0000 0000 9497 5095grid.419548.5Department of Translational Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany
| | - Shifra Ben-Dor
- 0000 0004 0604 7563grid.13992.30Department of Biological Services, Bioinformatics and Biological Computing Unit, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Shosh Gil
- 0000 0004 0604 7563grid.13992.30Department of Neurobiology, Weizmann Institute of Science, 76100 Rehovot, Israel
| | - Bekh Bradley
- 0000 0004 0419 4084grid.414026.5Atlanta Veterans Affairs Medical Center, Decatur, GA USA ,0000 0001 0941 6502grid.189967.8Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Alicia K. Smith
- 0000 0001 0941 6502grid.189967.8Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Tanja Jovanovic
- 0000 0001 0941 6502grid.189967.8Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Kerry J. Ressler
- 0000 0001 0941 6502grid.189967.8Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Elisabeth B. Binder
- 0000 0000 9497 5095grid.419548.5Department of Translational Psychiatry, Max Planck Institute of Psychiatry, 80804 Munich, Germany ,0000 0001 0941 6502grid.189967.8Department of Psychiatry and Behavioral Sciences, Emory University School of Medicine, Atlanta, GA 30322 USA
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, 76100, Rehovot, Israel. .,Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, 80804, Munich, Germany.
| |
Collapse
|
19
|
Biological underpinnings from psychosocial stress towards appetite and obesity during youth: research implications towards metagenomics, epigenomics and metabolomics. Nutr Res Rev 2019; 32:282-293. [PMID: 31298176 DOI: 10.1017/s0954422419000143] [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] [Indexed: 12/25/2022]
Abstract
Psychosocial stress, uncontrolled eating and obesity are three interrelated epidemiological phenomena already present during youth. This broad narrative conceptual review summarises main biological underpinnings of the stress-diet-obesity pathway and how new techniques can further knowledge. Cortisol seems the main biological factor from stress towards central adiposity; and diet, physical activity and sleep are the main behavioural pathways. Within stress-diet, the concepts of comfort food and emotional eating are highlighted, as cortisol affects reward pathways and appetite brain centres with a role for insulin, leptin, neuropeptide Y (NPY), endocannabinoids, orexin and gastrointestinal hormones. More recently researched biological underpinnings are microbiota, epigenetic modifications and metabolites. First, the gut microbiota reaches the stress-regulating and appetite-regulating brain centres via the gut-brain axis. Second, epigenetic analyses are recommended as diet, obesity, stress and gut microbiota can change gene expression which then affects appetite, energy homeostasis and stress reactivity. Finally, metabolomics would be a good technique to disentangle stress-diet-obesity interactions as multiple biological pathways are involved. Saliva might be an ideal biological matrix as it allows metagenomic (oral microbiota), epigenomic and metabolomic analyses. In conclusion, stress and diet/obesity research should be combined in interdisciplinary collaborations with implementation of several -omics analyses.
Collapse
|
20
|
Obri A, Claret M. The role of epigenetics in hypothalamic energy balance control: implications for obesity. Cell Stress 2019; 3:208-220. [PMID: 31309172 PMCID: PMC6612891 DOI: 10.15698/cst2019.07.191] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Despite enormous social and scientific efforts, obesity rates continue to increase worldwide. While genetic factors contribute to obesity development, genetics alone cannot explain the current epidemic. Obesity is essentially the consequence of complex genetic-environmental interactions. Evidence suggests that contemporary lifestyles trigger epigenetic changes, which can dysregulate energy balance and thus contribute to obesity. The hypothalamus plays a pivotal role in the regulation of body weight, through a sophisticated network of neuronal systems. Alterations in the activity of these neuronal pathways have been implicated in the pathophysiology of obesity. Here, we review the current knowledge on the central control of energy balance with a focus on recent studies linking epigenetic mechanisms in the hypothalamus to the development of obesity and metabolic disorders.
Collapse
Affiliation(s)
- Arnaud Obri
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Marc Claret
- Neuronal Control of Metabolism Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain.,Centro de Investigación Biomédica en Red (CIBER) de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 08036 Barcelona, Spain
| |
Collapse
|
21
|
Balsevich G, Abizaid A, Chen A, Karatsoreos IN, Schmidt MV. Stress and glucocorticoid modulation of feeding and metabolism. Neurobiol Stress 2019; 11:100171. [PMID: 31193462 PMCID: PMC6529856 DOI: 10.1016/j.ynstr.2019.100171] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 05/07/2019] [Accepted: 05/14/2019] [Indexed: 12/04/2022] Open
Abstract
This perspective highlights research presented as part of the symposium entitled, “Stress and Glucocorticoid Modulation of Feeding and Metabolism” at the 2018 Neurobiology of Stress Workshop held in Banff, AB, Canada. The symposium comprised five researchers at different career stages who each study different aspects of the interaction between the stress response and metabolic control. Their collective results reveal the complexity of this relationship in terms of behavioural and physiological outcomes. Their work emphasizes the need to consider the level of interaction (cellular, tissue, systems) as well as the timing and context in which the interaction is studied. Rather than a comprehensive review on the work presented at the Symposium, here we discuss recurring themes that emerged at the biennial workshop, which address new avenues of research that will drive the field forward.
Collapse
Affiliation(s)
- G Balsevich
- Hotchkiss Brain Institute, University of Calgary, 3330 Hospital Dr NW, Calgary, AB, T2N 4N1, Canada
| | - A Abizaid
- Institute of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON, K1S 5B6, Canada
| | - A Chen
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstrasse 2 - 10, Munich, 80804, Germany
| | - I N Karatsoreos
- Department of Integrative Physiology and Neuroscience, Washington State University, 1815 Ferdinand's Lane, Pullman, WA, 99164, United States
| | - M V Schmidt
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Kraepelinstrasse 2 - 10, Munich, 80804, Germany
| |
Collapse
|
22
|
Itoga CA, Chen Y, Fateri C, Echeverry PA, Lai JM, Delgado J, Badhon S, Short A, Baram TZ, Xu X. New viral-genetic mapping uncovers an enrichment of corticotropin-releasing hormone-expressing neuronal inputs to the nucleus accumbens from stress-related brain regions. J Comp Neurol 2019; 527:2474-2487. [PMID: 30861133 DOI: 10.1002/cne.24676] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2018] [Revised: 02/14/2019] [Accepted: 03/04/2019] [Indexed: 12/18/2022]
Abstract
Corticotropin-releasing hormone (CRH) is an essential, evolutionarily-conserved stress neuropeptide. In addition to hypothalamus, CRH is expressed in brain regions including amygdala and hippocampus where it plays crucial roles in modulating the function of circuits underlying emotion and cognition. CRH+ fibers are found in nucleus accumbens (NAc), where CRH modulates reward/motivation behaviors. CRH actions in NAc may vary by the individual's stress history, suggesting roles for CRH in neuroplasticity and adaptation of the reward circuitry. However, the origin and extent of CRH+ inputs to NAc are incompletely understood. We employed viral genetic approaches to map both global and CRH+ projection sources to NAc in mice. We injected into NAc variants of a new designer adeno-associated virus that permits robust retrograde access to NAc-afferent projection neurons. Cre-dependent viruses injected into CRH-Cre mice enabled selective mapping of CRH+ afferents. We employed anterograde AAV1-directed axonal tracing to verify NAc CRH+ fiber projections and established the identity of genetic reporter-labeled cells via validated antisera against native CRH. We quantified the relative contribution of CRH+ neurons to total NAc-directed projections. Combined retrograde and anterograde tracing identified the paraventricular nucleus of the thalamus, bed nucleus of stria terminalis, basolateral amygdala, and medial prefrontal cortex as principal sources of CRH+ projections to NAc. CRH+ NAc afferents were selectively enriched in NAc-projecting brain regions involved in diverse aspects of the sensing, processing and memory of emotionally salient events. These findings suggest multiple, complex potential roles for the molecularly-defined, CRH-dependent circuit in modulation of reward and motivation behaviors.
Collapse
Affiliation(s)
- Christy A Itoga
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Yuncai Chen
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California.,Department of Pediatrics, School of Medicine, University of California-Irvine, Irvine, California
| | - Cameron Fateri
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Paula A Echeverry
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Jennifer M Lai
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Jasmine Delgado
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Shapatur Badhon
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California
| | - Annabel Short
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California.,Department of Pediatrics, School of Medicine, University of California-Irvine, Irvine, California
| | - Tallie Z Baram
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California.,Department of Pediatrics, School of Medicine, University of California-Irvine, Irvine, California
| | - Xiangmin Xu
- Department of Anatomy and Neurobiology, School of Medicine, University of California-Irvine, Irvine, California.,Department of Biomedical Engineering, University of California, Irvine, California.,Department of Microbiology and Molecular Genetics, University of California, Irvine, California
| |
Collapse
|
23
|
Schroeder M, Jakovcevski M, Polacheck T, Drori Y, Ben-Dor S, Röh S, Chen A. Sex dependent impact of gestational stress on predisposition to eating disorders and metabolic disease. Mol Metab 2018; 17:1-16. [PMID: 30174229 PMCID: PMC6197785 DOI: 10.1016/j.molmet.2018.08.005] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 08/08/2018] [Accepted: 08/14/2018] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE Vulnerability to eating disorders (EDs) is broadly assumed to be associated with early life stress. However, a careful examination of the literature shows that susceptibility to EDs may depend on the type, severity and timing of the stressor and the sex of the individual. We aimed at exploring the link between chronic prenatal stress and predisposition to EDs and metabolic disease. METHODS We used a chronic variable stress protocol during gestation to explore the metabolic response of male and female offspring to food restriction (FR), activity-based anorexia (ABA), binge eating (BE) and exposure to high fat (HF) diet. RESULTS Contrary to controls, prenatally stressed (PNS) female offspring showed resistance to ABA and BE and displayed a lower metabolic rate leading to hyperadiposity and obesity on HF diet. Male PNS offspring showed healthy responses to FR and ABA, increased propensity to binge and improved coping with HF compared to controls. We found that long-lasting abnormal responses to metabolic challenge are linked to fetal programming and adult hypothalamic dysregulation in PNS females, resulting from sexually dimorphic adaptations in placental methylation and gene expression. CONCLUSIONS Our results show that maternal stress may have variable and even opposing effects on ED risk, depending on the ED and the sex of the offspring.
Collapse
Affiliation(s)
- Mariana Schroeder
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, 80804, Germany.
| | - Mira Jakovcevski
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, 80804, Germany
| | - Tamar Polacheck
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Yonat Drori
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Shifra Ben-Dor
- Bioinformatics and Biological Computing Unit, Biological Services, Weizmann Institute of Science, Rehovot, 76100, Israel
| | - Simone Röh
- Department of Translational Research in Psychiatry, Max Planck Institute of Psychiatry, Munich, 80804, Germany
| | - Alon Chen
- Department of Neurobiology, Weizmann Institute of Science, Rehovot, 76100, Israel; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, 80804, Germany.
| |
Collapse
|
24
|
Placental miR-340 mediates vulnerability to activity based anorexia in mice. Nat Commun 2018; 9:1596. [PMID: 29686286 PMCID: PMC5913294 DOI: 10.1038/s41467-018-03836-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Accepted: 03/15/2018] [Indexed: 12/20/2022] Open
Abstract
Anorexia nervosa (AN) is a devastating eating disorder characterized by self-starvation that mainly affects women. Its etiology is unknown, which impedes successful treatment options leading to a limited chance of full recovery. Here, we show that gestation is a vulnerable window that can influence the predisposition to AN. By screening placental microRNA expression of naive and prenatally stressed (PNS) fetuses and assessing vulnerability to activity-based anorexia (ABA), we identify miR-340 as a sexually dimorphic regulator involved in prenatal programming of ABA. PNS caused gene-body hypermethylation of placental miR-340, which is associated with reduced miR-340 expression and increased protein levels of several target transcripts, GR, Cry2 and H3F3b. MiR-340 is linked to the expression of several nutrient transporters both in mice and human placentas. Using placenta-specific lentiviral transgenes and embryo transfer, we demonstrate the key role miR-340 plays in the mechanism involved in early life programming of ABA. Anorexia nervosa is characterised by self-starvation but its etiology is not completely understood. Here the authors describe how prenatal stress can induce activity-based anorexia in the offspring during early adulthood by upregulating miR-340 expression in the placenta that affects expression of nutrient transporters.
Collapse
|
25
|
Novelle MG, Diéguez C. Food Addiction and Binge Eating: Lessons Learned from Animal Models. Nutrients 2018; 10:E71. [PMID: 29324652 PMCID: PMC5793299 DOI: 10.3390/nu10010071] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 12/26/2017] [Accepted: 01/09/2018] [Indexed: 01/10/2023] Open
Abstract
The feeding process is required for basic life, influenced by environment cues and tightly regulated according to demands of the internal milieu by regulatory brain circuits. Although eating behaviour cannot be considered "addictive" under normal circumstances, people can become "addicted" to this behaviour, similarly to how some people are addicted to drugs. The symptoms, cravings and causes of "eating addiction" are remarkably similar to those experienced by drug addicts, and both drug-seeking behaviour as eating addiction share the same neural pathways. However, while the drug addiction process has been highly characterised, eating addiction is a nascent field. In fact, there is still a great controversy over the concept of "food addiction". This review aims to summarize the most relevant animal models of "eating addictive behaviour", emphasising binge eating disorder, that could help us to understand the neurobiological mechanisms hidden under this behaviour, and to improve the psychotherapy and pharmacological treatment in patients suffering from these pathologies.
Collapse
Affiliation(s)
- Marta G Novelle
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria (IDIS), CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 15786 Santiago de Compostela, Spain.
| | - Carlos Diéguez
- Department of Physiology, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela-Instituto de Investigación Sanitaria (IDIS), CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Instituto de Salud Carlos III, 15786 Santiago de Compostela, Spain.
| |
Collapse
|