1
|
Hudobenko J, Di Gesù CM, Mooz PR, Petrosino J, Putluri N, Ganesh BP, Rebeles K, Blixt FW, Venna VR, McCullough LD. Maternal dysbiosis produces long-lasting behavioral changes in offspring. Mol Psychiatry 2024:10.1038/s41380-024-02794-0. [PMID: 39443733 DOI: 10.1038/s41380-024-02794-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 10/03/2024] [Accepted: 10/10/2024] [Indexed: 10/25/2024]
Abstract
Advanced maternal age (AMA) is defined as a pregnancy in a woman older than 35 years of age. AMA increases the risk for both maternal and neonatal complications, including miscarriage and stillbirth. AMA has also been linked to neurodevelopmental and neuropsychiatric disorders in the offspring. Recent studies have found that age-associated compositional shifts in the gut microbiota contribute to altered microbial metabolism and enhanced inflammation in the host. We investigated the specific contribution of the maternal microbiome on pregnancy outcomes and offspring behavior by recolonizing young female mice with aged female microbiome prior to pregnancy. We discovered that pre-pregnancy colonization of young dams with microbiome from aged female donors significantly increased fetal loss. There were significant differences in the composition of the gut microbiome in pups born from dams recolonized with aged female biome that persisted through middle age. Offspring born from dams colonized with aged microbiome also had significant changes in levels of neurotransmitters and metabolites in the blood and the brain. Adult offspring from dams colonized with an aged microbiome displayed persistent depressive- and anxiety-like phenotypes. Collectively, these results demonstrate that age-related changes in the composition of the maternal gut microbiome contribute to chronic alterations in the behavior and physiology of offspring. This work highlights the potential of microbiome-targeted approaches, even prior to birth, may reduce the risk of neuropsychiatric disorders.
Collapse
Affiliation(s)
- Jacob Hudobenko
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, CT, USA
| | - Claudia M Di Gesù
- Center for Precision Environmental Health, Baylor College of Medicine, Houston, TX, USA
| | - Patrick R Mooz
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
- UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph Petrosino
- Alkek Center for Metagenomics and Microbiome Research, Department of Molecular Virology & Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Nagireddy Putluri
- Dan L. Duncan Comprehensive Cancer Center, Advanced Technology Core, Alkek Center for Molecular Discovery, Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, TX, USA
| | - Bhanu P Ganesh
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
- UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kristen Rebeles
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
| | - Frank W Blixt
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
| | - Venugopal R Venna
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA
| | - Louise D McCullough
- Department of Neurology, McGovern Medical School at University of Texas Health Science Center Houston, Houston, TX, USA.
- UTHealth Graduate School of Biomedical Sciences, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
2
|
Gustafson KL, Busi SB, McAdams ZL, McCorkle RE, Khodakivskyi P, Bivens NJ, Davis DJ, Raju M, Coghill LM, Goun EA, Amos-Landgraf J, Franklin CL, Wilmes P, Cortese R, Ericsson AC. Fetal programming by the parental microbiome of offspring behavior, and DNA methylation and gene expression within the hippocampus. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589237. [PMID: 39484583 PMCID: PMC11526851 DOI: 10.1101/2024.04.12.589237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Background The microorganisms colonizing the gastrointestinal tract of animals, collectively referred to as the gut microbiome, affect numerous host behaviors dependent on the central nervous system (CNS). Studies comparing germ-free mice to normally colonized mice have demonstrated influences of the microbiome on anxiety-related behaviors, voluntary activity, and gene expression in the CNS. Additionally, there is epidemiologic evidence supporting an intergenerational influence of the maternal microbiome on neurodevelopment of offspring and behavior later in life. There is limited experimental evidence however directly linking the maternal microbiome to long-term neurodevelopmental outcomes, or knowledge regarding mechanisms responsible for such effects. Results Here we show that that the maternal microbiome has a dominant influence on several offspring phenotypes including anxiety-related behavior, voluntary activity, and body weight. Adverse outcomes in offspring were associated with features of the maternal microbiome including bile salt hydrolase activity gene expression (bsh), abundance of certain bile acids, and hepatic expression of Slc10a1. In cross-foster experiments, offspring resembled their birth dam phenotypically, despite faithful colonization in the postnatal period with the surrogate dam microbiome. Genome-wide methylation analysis of hippocampal DNA identified microbiome-associated differences in methylation of 196 loci in total, 176 of which show conserved profiles between mother and offspring. Further, single-cell transcriptional analysis revealed accompanying differences in expression of several differentially methylated genes within certain hippocampal cell clusters, and vascular expression of genes associated with bile acid transport. Inferred cell-to-cell communication in the hippocampus based on coordinated ligand-receptor expression revealed differences in expression of neuropeptides associated with satiety. Conclusions Collectively, these data provide proof-of-principle that the maternal gut microbiome has a dominant influence on the neurodevelopment underlying certain offspring behaviors and activities, and selectively affects genome methylation and gene expression in the offspring CNS in conjunction with that neurodevelopment.
Collapse
Affiliation(s)
- Kevin L Gustafson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65201, USA
| | - Susheel Bhanu Busi
- UK Centre for Ecology and Hydrology, Wallingford, Oxfordshire, OX10 8BB, UK
| | - Zachary L McAdams
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65201, USA
| | - Rachael E McCorkle
- College of Veterinary Medicine, University of Missouri, Columbia, MO, 65211, USA
| | - Pavlo Khodakivskyi
- Department of Chemistry, College of Arts and Science, University of Missouri, Columbia, MO, 65211, USA
| | - Nathan J Bivens
- University of Missouri Genomics Technology Core, University of Missouri, Columbia, MO, 65211, USA
| | - Daniel J Davis
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65201, USA
| | - Murugesan Raju
- University of Missouri Bioinformatics and Analytics Core, University of Missouri, Columbia, MO, 65211, USA
| | - Lyndon M Coghill
- University of Missouri Bioinformatics and Analytics Core, University of Missouri, Columbia, MO, 65211, USA
| | - Elena A Goun
- Department of Chemistry, College of Arts and Science, University of Missouri, Columbia, MO, 65211, USA
| | - James Amos-Landgraf
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65201, USA
| | - Craig L Franklin
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65201, USA
| | - Paul Wilmes
- Department of Life Sciences and Medicine, Faculty of Science, Technology and Medicine, University of Luxembourg, L-4362 Esch-sur-Alzette, Luxembourg
| | - Rene Cortese
- Department of Child Health & Obstetrics, Gynecology, and Women's Health, School of Medicine, University of Missouri, Columbia, MO, 65212, USA
| | - Aaron C Ericsson
- Department of Veterinary Pathobiology, College of Veterinary Medicine, University of Missouri, Columbia, MO, 65201, USA
| |
Collapse
|
3
|
Bodnar TS, Ainsworth-Cruickshank G, Billy V, Wegener Parfrey L, Weinberg J, Raineki C. Alcohol consumption during pregnancy differentially affects the fecal microbiota of dams and offspring. Sci Rep 2024; 14:16121. [PMID: 38997303 PMCID: PMC11245617 DOI: 10.1038/s41598-024-64313-z] [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: 02/12/2023] [Accepted: 06/07/2024] [Indexed: 07/14/2024] Open
Abstract
Microbiota imbalances are linked to inflammation and disease, as well as neurodevelopmental conditions where they may contribute to behavioral, physiological, and central nervous system dysfunction. By contrast, the role of the microbiota in Fetal Alcohol Spectrum Disorder (FASD), the group of neurodevelopmental conditions that can occur following prenatal alcohol exposure (PAE), has not received similar attention. Here we utilized a rodent model of alcohol consumption during pregnancy to characterize the impact of alcohol on the microbiota of dam-offspring dyads. Overall, bacterial diversity decreased in alcohol-consuming dams and community composition differed from that of controls in alcohol-consuming dams and their offspring. Bacterial taxa and predicted biochemical pathway composition were also altered with alcohol consumption/exposure; however, there was minimal overlap between the changes in dams and offspring. These findings illuminate the potential importance of the microbiota in the pathophysiology of FASD and support investigation into novel microbiota-based interventions.
Collapse
Affiliation(s)
- Tamara S Bodnar
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada.
- Department of Biological Sciences, University of Calgary, 2500 University Dr NW, Calgary, AB, T2N 1N4, Canada.
| | | | - Vincent Billy
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Laura Wegener Parfrey
- Department of Zoology and Biodiversity Research Centre, University of British Columbia, Vancouver, BC, Canada
- Department of Botany, University of British Columbia, Vancouver, BC, Canada
| | - Joanne Weinberg
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Charlis Raineki
- Department of Cellular and Physiological Sciences, University of British Columbia, Vancouver, BC, Canada
- Department of Psychology, Brock University, St. Catharines, ON, Canada
| |
Collapse
|
4
|
Lu X, Shi Z, Jiang L, Zhang S. Maternal gut microbiota in the health of mothers and offspring: from the perspective of immunology. Front Immunol 2024; 15:1362784. [PMID: 38545107 PMCID: PMC10965710 DOI: 10.3389/fimmu.2024.1362784] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Accepted: 02/28/2024] [Indexed: 04/17/2024] Open
Abstract
Due to the physiological alteration during pregnancy, maternal gut microbiota changes following the metabolic processes. Recent studies have revealed that maternal gut microbiota is closely associated with the immune microenvironment in utero during pregnancy and plays a vital role in specific pregnancy complications, including preeclampsia, gestational diabetes, preterm birth and recurrent miscarriages. Some other evidence has also shown that aberrant maternal gut microbiota increases the risk of various diseases in the offspring, such as allergic and neurodevelopmental disorders, through the immune alignment between mother and fetus and the possible intrauterine microbiota. Probiotics and the high-fiber diet are effective inventions to prevent mothers and fetuses from diseases. In this review, we summarize the role of maternal gut microbiota in the development of pregnancy complications and the health condition of future generations from the perspective of immunology, which may provide new therapeutic strategies for the health management of mothers and offspring.
Collapse
Affiliation(s)
- Xiaowen Lu
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, Hangzhou, China
| | - Zhan Shi
- Department of Obstetrics and Gynecology, The Fourth Affiliated Hospital of Zhejiang University School of Medicine, Yiwu, China
| | - Lingling Jiang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, Hangzhou, China
| | - Songying Zhang
- Assisted Reproduction Unit, Department of Obstetrics and Gynecology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Obstetrics and Gynecology, Key Laboratory of Reproductive Dysfunction, Management of Zhejiang Province, Hangzhou, China
| |
Collapse
|
5
|
Hung LY, Margolis KG. Autism spectrum disorders and the gastrointestinal tract: insights into mechanisms and clinical relevance. Nat Rev Gastroenterol Hepatol 2024; 21:142-163. [PMID: 38114585 DOI: 10.1038/s41575-023-00857-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/11/2023] [Indexed: 12/21/2023]
Abstract
Autism spectrum disorders (ASDs) are recognized as central neurodevelopmental disorders diagnosed by impairments in social interactions, communication and repetitive behaviours. The recognition of ASD as a central nervous system (CNS)-mediated neurobehavioural disorder has led most of the research in ASD to be focused on the CNS. However, gastrointestinal function is also likely to be affected owing to the neural mechanistic nature of ASD and the nervous system in the gastrointestinal tract (enteric nervous system). Thus, it is unsurprising that gastrointestinal disorders, particularly constipation, diarrhoea and abdominal pain, are highly comorbid in individuals with ASD. Gastrointestinal problems have also been repeatedly associated with increased severity of the core symptoms diagnostic of ASD and other centrally mediated comorbid conditions, including psychiatric issues, irritability, rigid-compulsive behaviours and aggression. Despite the high prevalence of gastrointestinal dysfunction in ASD and its associated behavioural comorbidities, the specific links between these two conditions have not been clearly delineated, and current data linking ASD to gastrointestinal dysfunction have not been extensively reviewed. This Review outlines the established and emerging clinical and preclinical evidence that emphasizes the gut as a novel mechanistic and potential therapeutic target for individuals with ASD.
Collapse
Affiliation(s)
- Lin Y Hung
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA
| | - Kara Gross Margolis
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, USA.
- Department of Cell Biology, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
- Department of Pediatrics, NYU Grossman School of Medicine and Langone Medical Center, New York, NY, USA.
| |
Collapse
|
6
|
Nohesara S, Abdolmaleky HM, Zhou JR, Thiagalingam S. Microbiota-Induced Epigenetic Alterations in Depressive Disorders Are Targets for Nutritional and Probiotic Therapies. Genes (Basel) 2023; 14:2217. [PMID: 38137038 PMCID: PMC10742434 DOI: 10.3390/genes14122217] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Major depressive disorder (MDD) is a complex disorder and a leading cause of disability in 280 million people worldwide. Many environmental factors, such as microbes, drugs, and diet, are involved in the pathogenesis of depressive disorders. However, the underlying mechanisms of depression are complex and include the interaction of genetics with epigenetics and the host immune system. Modifications of the gut microbiome and its metabolites influence stress-related responses and social behavior in patients with depressive disorders by modulating the maturation of immune cells and neurogenesis in the brain mediated by epigenetic modifications. Here, we discuss the potential roles of a leaky gut in the development of depressive disorders via changes in gut microbiota-derived metabolites with epigenetic effects. Next, we will deliberate how altering the gut microbiome composition contributes to the development of depressive disorders via epigenetic alterations. In particular, we focus on how microbiota-derived metabolites such as butyrate as an epigenetic modifier, probiotics, maternal diet, polyphenols, drugs (e.g., antipsychotics, antidepressants, and antibiotics), and fecal microbiota transplantation could positively alleviate depressive-like behaviors by modulating the epigenetic landscape. Finally, we will discuss challenges associated with recent therapeutic approaches for depressive disorders via microbiome-related epigenetic shifts, as well as opportunities to tackle such problems.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Hamid Mostafavi Abdolmaleky
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boson, MA 02215, USA;
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boson, MA 02215, USA;
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
7
|
Zhou GQ, Huang MJ, Yu X, Zhang NN, Tao S, Zhang M. Early life adverse exposures in irritable bowel syndrome: new insights and opportunities. Front Pediatr 2023; 11:1241801. [PMID: 37732013 PMCID: PMC10507713 DOI: 10.3389/fped.2023.1241801] [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: 06/17/2023] [Accepted: 08/22/2023] [Indexed: 09/22/2023] Open
Abstract
Irritable bowel syndrome (IBS) is a prevalent functional gastrointestinal disorder worldwide. Extensive research has identified multiple factors contributing to its development, including genetic predisposition, chronic infection, gut dysbiosis, aberrant serotonin metabolism, and brain dysfunction. Recent studies have emphasized the critical role of the early life stage as a susceptibility window for IBS. Current evidence suggests that diet can heighten the risk of IBS in offspring by influencing the microbiota composition, intestinal epithelium structure, gene expression, and brain-gut axis. The use of antibiotics during pregnancy and the neonatal period disrupts the normal gut microbiota structure, aligning it with the characteristics observed in IBS patients. Additionally, early life stress impacts susceptibility to IBS by modulating TLR4, NK1, and the hypothalamic-pituitary-adrenal (HPA) axis while compromising the offspring's immune system. Formula feeding facilitates the colonization of pathogenic bacteria in the intestines, concurrently reducing the presence of probiotics. This disruption of the Th1 and Th2 cell balance in the immune system weakens the intestinal epithelial barrier. Furthermore, studies suggest that delivery mode influences the occurrence of IBS by altering the composition of gut microbes. This review aims to provide a comprehensive summary of the existing evidence regarding the impact of adverse early life exposures on IBS during pregnancy, intrapartum, and neonatal period. By consolidating this knowledge, the review enhances our understanding of the direct and indirect mechanisms underlying early life-related IBS and offers new insights and research directions from childhood to adulthood.
Collapse
Affiliation(s)
| | | | | | | | | | - Ming Zhang
- Department of General Practice, Honghui Hospital, Xi'an Jiaotong University, Xi’an, China
| |
Collapse
|
8
|
Vieira ACA, Pinheiro RO, Soares NL, Bezerra MLR, Nascimento DDS, Alves AF, Sousa MCDP, Dutra MLDV, Lima MDS, Donato NR, Aquino JDS. Maternal high-fat diet alters the neurobehavioral, biochemical and inflammatory parameters of their adult female rat offspring. Physiol Behav 2023; 266:114180. [PMID: 37037382 DOI: 10.1016/j.physbeh.2023.114180] [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: 12/30/2022] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 04/12/2023]
Abstract
BACKGROUND Lipid metabolism dysregulations have been associated with depressive and anxious behaviors which can affect pregnant and lactating individuals, with indications that such changes extend to the offspring. Therefore, the aim of this study was to evaluate the effect of a maternal high-fat diet on the neurobehavioral, biochemical and inflammatory parameters of their adult female offspring. METHODS Wistar rats ± 90 days old were mated. The dams were allocated to consume a control (CTL) or high-fat (HFD) diet during pregnancy and lactation. After weaning, the female offspring from the CTL (N=10) and HFD (N=10) groups received standard chow. The offspring behavioral tests were started at 120 days old. Then, the somatic measures were evaluated followed by euthanasia, histological and biochemical analyses. RESULTS The HFD group had less ambulation and longer immobility time in the open field test compared to the CTL. The HFD group had lower HDL (48.4%) and a higher adiposity (71.8%) and LDL (62.2%) than the CTL. The CTL had a higher organic acid concentration in the intestine, mainly acetic and butyric acids, however the HFD had a higher citric and acetic acid concentration in the brain and ischemic lesion in the hippocampus with a higher NF-κB concentration. CONCLUSION The results demonstrate deleterious effects of a maternal HFD on the neurobehavioral and biochemical parameters of their offspring which may be associated with the role of organic acids and NF-κB in fetal programming.
Collapse
Affiliation(s)
- Anne Caroline Alves Vieira
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Rafael Oliveira Pinheiro
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Naís Lira Soares
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Maria Luiza Rolim Bezerra
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Davi Dos Santos Nascimento
- Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil; Laboratory of food microbiology and biochemistry, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil
| | - Adriano Francisco Alves
- Laboratory of General pathology, Department of Physiology and Pathology, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Maria Carolina de Paiva Sousa
- Laboratory of General pathology, Department of Physiology and Pathology, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Maria Letícia da Veiga Dutra
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Marcos Dos Santos Lima
- Department of Food Technology, Federal Institute of Sertão Pernambucano, Petrolina, Brazil; Post Graduate Program in Food Science and Technology, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Nilcimelly Rodrigues Donato
- Department of Nutrition, Center for Education and Health, Federal University of Campina Grande (UFCG), Cuité, Paraíba, Brazil
| | - Jailane de Souza Aquino
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil; Post Graduate Program in Food Science and Technology, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil.
| |
Collapse
|
9
|
Perinatal and post-weaning exposure to a high-fat diet causes histomorphometric, neuroplastic, and histopathological changes in the rat ileum. J Dev Orig Health Dis 2023; 14:231-241. [PMID: 36073012 DOI: 10.1017/s2040174422000514] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Exposure to a diet with a high saturated fat content can influence the characteristics of the gastrointestinal tract, causing losses in the absorption of nutrients and favoring the appearance of diseases. The objective was to assess the effects of a high-fat diet (HFD) in the perinatal (pregnancy and lactation) and post-weaning period on the histomorphometry, neuroplasticity, and histopathology of the ileum. Wistar rats were divided into four subgroups: Control/Control (CC, n = 10) rats fed a control diet (C) throughout the trial period; Control/HFD (CH, n = 9) rats fed diet C (perinatal) and HFD after weaning; HFD/Control (HC, n = 10) rats fed HFD (perinatal) and diet C (post-weaning); HFD/HFD (HH, n = 9) rats fed HFD throughout the experimental period. There was atrophy of the Ileum wall with a reduction in the muscular tunic, submucosa, and mucosa thickness in the HH group of 37%, 28%, and 46%, respectively (p < 0.0001). The depth of the crypts decreased by 29% (p < 0.0001) and height increased by 5% (p < 0.0013). Villus height decreased by 41% and 18% in HH and HC groups (p < 0.0001) and width decreased by 11% in the HH (p < 0.0001). The height of the enterocytes decreased by 18% in the HH (p < 0.0001). There was a decrease in the area of the myenteric and submucosal plexus ganglia in the HH and HC groups (p < 0.0001). The number, occupation, and granules of Paneth cells increased in the HH and HC groups (p < 0.0001). Intraepithelial lymphocytes (IELs) increased in all groups exposed to the HFD. Goblet cells decreased in groups CH and HH (p < 0.0001). The evidence from this study suggests that the HFD had altered the histomorphometry, neuroplasticity, and histopathology of the ileum of the rats.
Collapse
|
10
|
Radford-Smith DE, Anthony DC. Mechanisms of Maternal Diet-Induced Obesity Affecting the Offspring Brain and Development of Affective Disorders. Metabolites 2023; 13:455. [PMID: 36984895 PMCID: PMC10053489 DOI: 10.3390/metabo13030455] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/16/2023] [Accepted: 03/18/2023] [Indexed: 03/30/2023] Open
Abstract
Depression and metabolic disease are common disorders that share a bidirectional relationship and continue to increase in prevalence. Maternal diet and maternal behaviour both profoundly influence the developmental trajectory of offspring during the perinatal period. At an epidemiological level, both maternal depression and obesity during pregnancy have been shown to increase the risk of neuropsychiatric disease in the subsequent generation. Considerable progress has been made to understand the mechanisms by which maternal obesity disrupts the developing offspring gut-brain axis, priming offspring for the development of affective disorders. This review outlines such mechanisms in detail, including altered maternal care, the maternal microbiome, inflammation, breast milk composition, and maternal and placental metabolites. Subsequently, offspring may be prone to developing gut-brain interaction disorders with concomitant changes to brain energy metabolism, neurotransmission, and behaviour, alongside gut dysbiosis. The gut microbiome may act as a key modifiable, and therefore treatable, feature of the relationship between maternal obesity and the offspring brain function. Further studies examining the relationship between maternal nutrition, the maternal microbiome and metabolites, and offspring neurodevelopment are warranted to identify novel therapeutic targets.
Collapse
Affiliation(s)
- Daniel E. Radford-Smith
- Department of Psychiatry, University of Oxford, Warneford Hospital, Warneford Lane, Oxford OX37JX, UK
- Department of Chemistry, University of Oxford, Mansfield Road, Oxford OX13TA, UK
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX13QT, UK
| | - Daniel C. Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, Oxford OX13QT, UK
| |
Collapse
|
11
|
Guzzardi MA, La Rosa F, Iozzo P. Trust the gut: outcomes of gut microbiota transplant in metabolic and cognitive disorders. Neurosci Biobehav Rev 2023; 149:105143. [PMID: 36990372 DOI: 10.1016/j.neubiorev.2023.105143] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/21/2023] [Accepted: 03/24/2023] [Indexed: 03/29/2023]
Abstract
Type 2 diabetes mellitus (T2DM) is a main public health concern, with increasing prevalence and growingly premature onset in children, in spite of emerging and successful therapeutic options. T2DM promotes brain aging, and younger age at onset is associated with a higher risk of subsequent dementia. Preventive strategies should address predisposing conditions, like obesity and metabolic syndrome, and be started from very early and even prenatal life. Gut microbiota is an emerging target in obesity, diabetes and neurocognitive diseases, which could be safely modulated since pregnancy and infancy. Many correlative studies have supported its involvement in disease pathophysiology. Faecal material transplantation (FMT) studies have been conducted in clinical and preclinical settings to deliver cause-effect proof and mechanistic insights. This review provides a comprehensive overview of studies in which FMT was used to cure or cause obesity, metabolic syndrome, T2DM, cognitive decline and Alzheimer's disease, including the evidence available in early life. Findings were analysed to dissect consolidated from controversial results, highlighting gaps and possible future directions.
Collapse
Affiliation(s)
- Maria Angela Guzzardi
- Institute of Clinical Physiology (IFC), the National Research Council (CNR), via Moruzzi 1, 56124 Pisa, Italy.
| | - Federica La Rosa
- Institute of Clinical Physiology (IFC), the National Research Council (CNR), via Moruzzi 1, 56124 Pisa, Italy.
| | - Patricia Iozzo
- Institute of Clinical Physiology (IFC), the National Research Council (CNR), via Moruzzi 1, 56124 Pisa, Italy.
| |
Collapse
|
12
|
Kendig MD, Hasebe K, Tajaddini A, Kaakoush NO, Westbrook RF, Morris MJ. The Benefits of Switching to a Healthy Diet on Metabolic, Cognitive, and Gut Microbiome Parameters Are Preserved in Adult Rat Offspring of Mothers Fed a High-Fat, High-Sugar Diet. Mol Nutr Food Res 2023; 67:e2200318. [PMID: 36271770 PMCID: PMC10909468 DOI: 10.1002/mnfr.202200318] [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: 05/15/2022] [Revised: 09/05/2022] [Indexed: 01/19/2023]
Abstract
SCOPE Maternal obesity increases the risk of health complications in children, highlighting the need for effective interventions. A rat model of maternal obesity to examine whether a diet switch intervention could reverse the adverse effects of an unhealthy postweaning diet is used. METHODS AND RESULTS Male and female offspring born to dams fed standard chow or a high-fat, high-sugar "cafeteria" (Caf) diet are weaned onto chow or Caf diets until 22 weeks of age, when Caf-fed groups are switched to chow for 5 weeks. Adiposity, gut microbiota composition, and place recognition memory are assessed before and after the switch. Body weight and adiposity fall in switched groups but remain significantly higher than chow-fed controls. Nonetheless, the diet switch improves a deficit in place recognition memory observed in Caf-fed groups, increases gut microbiota species richness, and alters β diversity. Modeling indicate that adiposity most strongly predicts gut microbiota composition before and after the switch. CONCLUSION Maternal obesity does not alter the effects of switching diet on metabolic, microbial, or cognitive measures. Thus, a healthy diet intervention lead to major shifts in body weight, adiposity, place recognition memory, and gut microbiota composition, with beneficial effects preserved in offspring born to obese dams.
Collapse
Affiliation(s)
- Michael D. Kendig
- Department of PharmacologySchool of Medical SciencesUNSW SydneyNSW2052Australia
- School of Life SciencesUniversity of Technology SydneyNSW2007Australia
| | - Kyoko Hasebe
- Department of PharmacologySchool of Medical SciencesUNSW SydneyNSW2052Australia
| | - Aynaz Tajaddini
- Department of PharmacologySchool of Medical SciencesUNSW SydneyNSW2052Australia
| | | | | | - Margaret J. Morris
- Department of PharmacologySchool of Medical SciencesUNSW SydneyNSW2052Australia
| |
Collapse
|
13
|
Tang M, Marroquin E. The role of the gut microbiome in the intergenerational transmission of the obesity phenotype: A narrative review. Front Med (Lausanne) 2022; 9:1057424. [PMID: 36619646 PMCID: PMC9812955 DOI: 10.3389/fmed.2022.1057424] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Accepted: 12/07/2022] [Indexed: 12/24/2022] Open
Abstract
Obesity is considered an epidemic by the World Health Organization. In particular, maternal obesity can affect the development of obesity and other related metabolic disorders in infants. Recently, both animal and human studies have pointed to the importance of the gut microbiome in facilitating the transmission of the obesity phenotype from mother to offspring. The gut microbiome changes significantly during the progression of pregnancy, and the microbiota of the amniotic fluid and placenta have recently been shown to colonize the infant gut in utero. Microbial composition, diversity, and richness are significantly altered by maternal obesity, which in turn affects the infant's acquisition of the gut microbiome and their risk to develop metabolic disorders. C-section has also been shown to affect the colonization of the infant gut and offspring metabolic and immune health. This narrative review seeks to discuss the role of the gut microbiome in the transmission of the obesity phenotype from mother to child, as well as how birth delivery, breastfeeding, and probiotic interventions may modulate this relationship.
Collapse
Affiliation(s)
- Mabel Tang
- Department of BioSciences, Rice University, Houston, TX, United States
| | - Elisa Marroquin
- Department of Nutritional Sciences, Texas Christian University, Fort Worth, TX, United States,*Correspondence: Elisa Marroquin,
| |
Collapse
|
14
|
Ramírez V, González-Palacios P, Baca MA, González-Domenech PJ, Fernández-Cabezas M, Álvarez-Cubero MJ, Rodrigo L, Rivas A. Effect of exposure to endocrine disrupting chemicals in obesity and neurodevelopment: The genetic and microbiota link. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 852:158219. [PMID: 36007653 DOI: 10.1016/j.scitotenv.2022.158219] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/06/2022] [Accepted: 08/18/2022] [Indexed: 06/15/2023]
Abstract
Current evidence highlights the importance of the genetic component in obesity and neurodevelopmental disorders (attention-deficit hyperactivity disorder (ADHD), autism spectrum disorder (ASD) and intellectual disability (ID)), given that these diseases have reported an elevated heritability. Additionally, environmental stressors, such as endocrine disrupting chemicals (EDCs) have been classified as obesogens, neuroendocrine disruptors, and microbiota disrupting chemicals (MDCs). For this reason, the importance of this work lies in examining two possible biological mechanistic pathways linking obesity and neurodevelopmental/behavioural disorders: EDCs - gene and EDCs - microbiota interactions. First, we summarise the shared mechanisms of action of EDCs and the common genetic profile in the bidirectional link between obesity and neurodevelopment. In relation to interaction models, evidence from the reviewed studies reveals significant interactions between pesticides/heavy metals and gene polymorphisms of detoxifying and neurotransmission systems and metal homeostasis on cognitive development, ASD and ADHD symptomatology. Nonetheless, available literature about obesity is quite limited. Importantly, EDCs have been found to induce gut microbiota changes through gut-brain-microbiota axis conferring susceptibility to obesity and neurodevelopmental disorders. In view of the lack of studies assessing the impact of EDCs - gene interactions and EDCs - mediated dysbiosis jointly in obesity and neurodevelopment, we support considering genetics, EDCs exposure, and microbiota as interactive factors rather than individual contributors to the risk for developing obesity and neurodevelopmental disabilities at the same time.
Collapse
Affiliation(s)
- Viviana Ramírez
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain; GENYO. Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government PTS Granada - Avenida de la Ilustración, 114, 18016 Granada, Spain; "José Mataix Verdú" Institute of Nutrition and Food Technology (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18100 Granada, Spain
| | - Patricia González-Palacios
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain.
| | | | | | - María Fernández-Cabezas
- Department of Developmental and Educational Psychology, Faculty of Educational Sciences, University of Granada, 18011 Granada, Spain
| | - María Jesús Álvarez-Cubero
- GENYO. Centre for Genomics and Oncological Research: Pfizer / University of Granada / Andalusian Regional Government PTS Granada - Avenida de la Ilustración, 114, 18016 Granada, Spain; Department of Biochemistry and Molecular Biology III, Faculty of Medicine, University of Granada, 18016 Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, 18014 Granada, Spain
| | - Lourdes Rodrigo
- Department of Legal Medicine and Toxicology, Faculty of Medicine, University of Granada, 18016 Granada, Spain
| | - Ana Rivas
- Department of Nutrition and Food Science, Faculty of Pharmacy, University of Granada, 18071 Granada, Spain; "José Mataix Verdú" Institute of Nutrition and Food Technology (INYTA), Biomedical Research Centre (CIBM), University of Granada, 18100 Granada, Spain; Instituto de Investigación Biosanitaria ibs.GRANADA, 18014 Granada, Spain
| |
Collapse
|
15
|
Rubini E, Schenkelaars N, Rousian M, Sinclair KD, Wekema L, Faas MM, Steegers-Theunissen RPM, Schoenmakers S. Maternal obesity during pregnancy leads to derangements in one-carbon metabolism and the gut microbiota: implications for fetal development and offspring wellbeing. Am J Obstet Gynecol 2022; 227:392-400. [PMID: 35452650 DOI: 10.1016/j.ajog.2022.04.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/04/2022] [Accepted: 04/07/2022] [Indexed: 11/01/2022]
Abstract
A healthy diet before and during pregnancy is beneficial in acquiring essential B vitamins involved in 1-carbon metabolism, and in maintaining a healthy gut microbiota. Each play important roles in fetal development, immune-system remodeling, and pregnancy-nutrient acquisition. Evidence shows that there is a reciprocal interaction between the one-carbon metabolism and the gut microbiota given that dietary intake of B vitamins has been shown to influence the composition of the gut microbiota, and certain gut bacteria also synthesize B vitamins. This reciprocal interaction contributes to the individual's overall availability of B vitamins and, therefore, should be maintained in a healthy state during pregnancy. There is an emerging consensus that obese pregnant women often have derangements in 1-carbon metabolism and gut dysbiosis owing to high intake of nutritiously poor foods and a chronic systemic inflammatory state. For example, low folate and vitamin B12 in obese women coincide with the decreased presence of B vitamin-producing bacteria and increased presence of inflammatory-associated bacteria from approximately mid-pregnancy. These alterations are risk factors for adverse pregnancy outcomes, impaired fetal development, and disruption of fetal growth and microbiota formation, which may lead to potential long-term offspring metabolic and neurologic disorders. Therefore, preconceptional and pregnant obese women may benefit from dietary and lifestyle counseling to improve their dietary nutrient intake, and from monitoring their B vitamin levels and gut microbiome by blood tests and microbiota stool samples. In addition, there is evidence that some probiotic bacteria have folate biosynthetic capacity and could be used to treat gut dysbiosis. Thus, their use as an intervention strategy for obese women holds potential and should be further investigated. Currently, there are many knowledge gaps concerning the relationship between one-carbon metabolism and the gut microbiota, and future research should focus on intervention strategies to counteract B vitamin deficiencies and gut dysbiosis in obese pregnant women, commencing with the use of probiotic and prebiotic supplements.
Collapse
Affiliation(s)
- Eleonora Rubini
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Nicole Schenkelaars
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Melek Rousian
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Kevin D Sinclair
- School of Biosciences, University of Nottingham, Nottingham, United Kingdom
| | - Lieske Wekema
- Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Marijke M Faas
- Division of Medical Biology, Department of Pathology and Medical Biology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | | | - Sam Schoenmakers
- Department of Obstetrics and Gynaecology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
16
|
Lu X, Xue Z, Qian Y, Wei S, Qiao Y, Zhang W, Lu H. Changes in intestinal microflora and its metabolites underlie the cognitive impairment in preterm rats. Front Cell Infect Microbiol 2022; 12:945851. [PMID: 36061856 PMCID: PMC9437323 DOI: 10.3389/fcimb.2022.945851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/29/2022] [Indexed: 11/25/2022] Open
Abstract
Background The brain development of preterm infants is easily affected by various adverse extrauterine factors and complications, resulting in abnormal neurological and cognitive development. Recent studies have found that there is a significant correlation between intestinal microbial changes and cognitive behavior. Nevertheless, the correlation between the cognitive impairment and abnormal changes of intestinal microflora in the preterm newborn has been rarely elucidated. Aim To analyze the differences of fecal intestinal flora, short chain fatty acids (SCFAs) and microbiota-gut-brain axis (MGBA)-related serum factors between preterm birth with and without cognitive impairment. Methods Healthy female rats (body weight 410 ± 40 g) of 16-17 days of gestation were selected for the establishment of preterm cognitive impairment model and screened by Morris water maze navigation experiments. The pathological change of rat hippocampus was confirmed by HE staining. The abundance of fecal intestinal microflora was determined by 16sRNA sequencing, while the contents of fecal SCFAs were examined by gas chromatography. Results Compared with the control group, the cognitive impairment group had decreased abundance and diversity of intestinal microflora and increased abundance of Proteobacteria at the level of phylum. While the abundances of Alistipes, Bacteroides, Prevotella, and Lactobacillus decreased significantly at the level of order, family, and genus, the abundances of Staphylococcaceae, Enterococci, Psychrobacter, and Oligella increased significantly. Moreover, the levels of total SCFAs and acetic acid in the disease group were significantly lower. The fecal abundance of acetic acid was positively correlated with that of Lactobacillaceae or Peptostreptococcaceae, and negatively correlated with that of Aerococcaceae, and Alcaligenaceae in disease rats. Furthermore, cognitive impairment caused significantly decreased levels of 5-HT, GABA, and BDNF, and increased levels of GR, CRH, IL-6, and TNF-α in rat blood. Conclusion Alterations in intestinal microflora structure and the abundances of SCFAs contributed substantially to the cognitive impairment in preterm rats, which was associated with significant changes in MGBA-related soluble factors.
Collapse
Affiliation(s)
- Xiang Lu
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Zhengyang Xue
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yu Qian
- School of Medicine, Jiangsu University, Zhenjiang, China
| | - Shanjie Wei
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Yu Qiao
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Wen Zhang
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- School of Medicine, Jiangsu University, Zhenjiang, China
- *Correspondence: Wen Zhang, ; Hongyan Lu,
| | - Hongyan Lu
- Pediatrics, Affiliated Hospital of Jiangsu University, Zhenjiang, China
- *Correspondence: Wen Zhang, ; Hongyan Lu,
| |
Collapse
|
17
|
Abu Y, Tao J, Dutta R, Yan Y, Vitari N, Kolli U, Roy S. Brief Hydromorphone Exposure During Pregnancy Sufficient to Induce Maternal and Neonatal Microbial Dysbiosis. J Neuroimmune Pharmacol 2022; 17:367-375. [PMID: 34562195 PMCID: PMC10117152 DOI: 10.1007/s11481-021-10019-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 08/27/2021] [Indexed: 12/29/2022]
Abstract
Prenatal opioid exposure is associated with significantly adverse medical, developmental, and behavioral outcomes in offspring, though the underlying mechanisms driving these impairments are still unclear. Accumulating evidence implicates gut microbial dysbiosis as a potential modulator of these adverse effects. However, how opioid exposure during pregnancy alters the maternal and neonatal microbiome remain to be elucidated. Here, we utilize a murine model of brief hydromorphone exposure during pregnancy (gestation day 11-13; i.p.; 10 mg/kg) to examine its impact on the maternal and neonatal microbiome. Fecal samples were collected at various timepoints in dams (4 days post hydromorphone exposure, birth, and weaning) and offspring (2, 3, and 5 weeks) to interrogate longitudinal changes in the microbiome. Stomach contents at 2 weeks were also collected as a surrogate for breastmilk and microbial analysis was performed using 16S rRNA sequencing. Alongside alterations in the maternal gut microbial composition, offspring gut microbiota exhibited distinct communities at 2 and 3 weeks. Furthermore, functional profiling of microbial communities revealed significant differences in microbial community-level phenotypes gram-negative, gram-positive, and potentially pathogenic in maternal and/or neonatal hydromorphone exposed groups compared with controls. We also observed differences in stomach microbiota in opioid-exposed vs non-exposed offspring, which suggests breast milk may also play a role in shaping the development of the neonatal gut microbiota. Together, we provide evidence of maternal and neonatal microbial dysbiosis provoked even with brief hydromorphone exposure during pregnancy.
Collapse
Affiliation(s)
- Yaa Abu
- Medical Scientist Training Program, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Junyi Tao
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Rajib Dutta
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Yan Yan
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Nicolas Vitari
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Udhghatri Kolli
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA
| | - Sabita Roy
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
18
|
Cusick JA, Wellman CL, Demas GE. Maternal stress and the maternal microbiome have sex-specific effects on offspring development and aggressive behavior in Siberian hamsters (Phodopus sungorus). Horm Behav 2022; 141:105146. [PMID: 35276524 DOI: 10.1016/j.yhbeh.2022.105146] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Revised: 02/18/2022] [Accepted: 02/21/2022] [Indexed: 11/29/2022]
Abstract
The gut microbiome, a community of commensal, symbiotic and pathogenic bacteria, fungi, and viruses, interacts with many physiological systems to affect behavior. Prenatal experiences, including exposure to maternal stress and different maternal microbiomes, are important sources of organismal variation that can affect offspring development. These physiological systems do not act in isolation and can have long-term effects on offspring development and behavior. Here we investigated the interactive effects of maternal stress and manipulations of the maternal microbiome on offspring development and social behavior using Siberian hamsters, Phodopus sungorus. We exposed pregnant females to either a social stressor, antibiotics, both the social stressor and antibiotics, or no treatment (i.e., control) over the duration of their pregnancy and quantified male and female offspring growth, gut microbiome composition and diversity, stress-induced cortisol concentrations, and social behavior. Maternal antibiotic exposure altered the gut microbial communities of male and female offspring. Maternal treatment also had sex-specific effects on aspects of offspring development and aggressive behavior. Female offspring produced by stressed mothers were more aggressive than other female offspring. Female, but not male, offspring produced by mothers exposed to the combined treatment displayed low levels of aggression, suggesting that alteration of the maternal microbiome attenuated the effects of prenatal stress in a sex-specific manner. Maternal treatment did not affect non-aggressive behavior in offspring. Collectively, our study offers insight into how maternal systems can interact to affect offspring in sex-specific ways and highlights the important role of the maternal microbiome in mediating offspring development and behavior.
Collapse
Affiliation(s)
- Jessica A Cusick
- Department of Biology, Utah Valley University, United States of America; Department of Biology, Indiana University, United States of America; Animal Behavior Program, Indiana University, United States of America.
| | - Cara L Wellman
- Animal Behavior Program, Indiana University, United States of America; Department of Psychological and Brain Sciences, Indiana University, United States of America; Program in Neuroscience, Indiana University, United States of America
| | - Gregory E Demas
- Department of Biology, Indiana University, United States of America; Animal Behavior Program, Indiana University, United States of America; Program in Neuroscience, Indiana University, United States of America
| |
Collapse
|
19
|
Rosa JM, Formolo DA, Yu J, Lee TH, Yau SY. The Role of MicroRNA and Microbiota in Depression and Anxiety. Front Behav Neurosci 2022; 16:828258. [PMID: 35299696 PMCID: PMC8921933 DOI: 10.3389/fnbeh.2022.828258] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/07/2022] [Indexed: 12/12/2022] Open
Abstract
Depression and anxiety are devastating disorders. Understanding the mechanisms that underlie the development of depression and anxiety can provide new hints on novel treatments and preventive strategies. Here, we summarize the latest findings reporting the novel roles of gut microbiota and microRNAs (miRNAs) in the pathophysiology of depression and anxiety. The crosstalk between gut microbiota and the brain has been reported to contribute to these pathologies. It is currently known that some miRNAs can regulate bacterial growth and gene transcription while also modulate the gut microbiota composition, suggesting the importance of miRNAs in gut and brain health. Treatment and prevention strategies for neuropsychiatric diseases, such as physical exercise, diet, and probiotics, can modulate the gut microbiota composition and miRNAs expressions. Nonetheless, there are critical questions to be addressed to understand further the mechanisms involved in the interaction between the gut microbiota and miRNAs in the brain. This review summarizes the recent findings of the potential roles of microbiota and miRNA on the neuropathology of depression and anxiety, and its potential as treatment strategies.
Collapse
Affiliation(s)
- Julia M. Rosa
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Mental Health Research Center (MHRC), Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Research Institute for Smart Aging (RISA), Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Douglas A. Formolo
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Mental Health Research Center (MHRC), Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Research Institute for Smart Aging (RISA), Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Jiasui Yu
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Mental Health Research Center (MHRC), Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Research Institute for Smart Aging (RISA), Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| | - Thomas H. Lee
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Neurocentre Magendie, INSERM U1215, University of Bordeaux, Bordeaux, France
| | - Suk-yu Yau
- Department of Rehabilitation Sciences, Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Mental Health Research Center (MHRC), Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
- Research Institute for Smart Aging (RISA), Hong Kong Polytechnic University, Hung Hom, Hong Kong SAR, China
| |
Collapse
|
20
|
Guzzardi MA, Ederveen THA, Rizzo F, Weisz A, Collado MC, Muratori F, Gross G, Alkema W, Iozzo P. Maternal pre-pregnancy overweight and neonatal gut bacterial colonization are associated with cognitive development and gut microbiota composition in pre-school-age offspring. Brain Behav Immun 2022; 100:311-320. [PMID: 34920092 DOI: 10.1016/j.bbi.2021.12.009] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 11/03/2021] [Accepted: 12/11/2021] [Indexed: 12/17/2022] Open
Abstract
Maternal gestational obesity is a risk factor for offspring's neurodevelopment and later neuro-cognitive disorders. Altered gut microbiota composition has been found in patients with neurocognitive disorders, and in relation to maternal metabolic health. We explored the associations between gut microbiota and cognitive development during infancy, and their link with maternal obesity. In groups of children from the Pisa birth Cohort (PISAC), we analysed faecal microbiota composition by 16S rRNA marker gene sequencing of first-pass meconium samples and of faecal samples collected at age 3, 6, 12, 24, 36 months, and its relationship with maternal gestational obesity or diabetes, and with cognitive development, as measured from 6 to 60 months of age by the Griffith's Mental Development Scales. Gut microbiota composition in the first phases of life is dominated by Bifidobacteria (Actinobacteria phylum), with contribution of Escherichia/Shigella and Klebsiella genera (Proteobacteria phylum), whereas Firmicutes become more dominant at 36 months of age. Maternal overweight leads to lower abundance of Bifidobacterium, Blautia and Ruminococcus, and lower practical reasoning scores in the offspring at the age of 36 months. In the whole population, microbiota in the first-pass meconium samples shows much higher alpha diversity compared to later samples, and its composition, particularly Bifidobacterium and Veillonella abundances, correlates with practical reasoning scores at 60 months of age. Maternal overweight correlates with bacterial colonization and with the development of reasoning skills at pre-school age. Associations between neonatal gut colonization and later cognitive function provide new perspectives of primary (antenatal) prevention of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Maria Angela Guzzardi
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy.
| | - Thomas H A Ederveen
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (Radboudumc), Nijmegen, the Netherlands.
| | - Francesca Rizzo
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana, University of Salerno, Baronissi, SA, Italy; Genome Research Center for Health (CRGS), Baronissi, SA, Italy.
| | - Alessandro Weisz
- Laboratory of Molecular Medicine and Genomics, Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana, University of Salerno, Baronissi, SA, Italy; Genome Research Center for Health (CRGS), Baronissi, SA, Italy.
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain.
| | | | - Gabriele Gross
- Medical and Scientific Affairs, Nutrition, RB Mead Johnson Nutrition Institute, Nijmegen, the Netherlands.
| | - Wynand Alkema
- Center for Molecular and Biomolecular Informatics (CMBI), Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Center (Radboudumc), Nijmegen, the Netherlands.
| | - Patricia Iozzo
- Institute of Clinical Physiology (IFC), National Research Council (CNR), Pisa, Italy.
| |
Collapse
|
21
|
Jantsch J, Tassinari ID, Giovenardi M, Bambini-Junior V, Guedes RP, de Fraga LS. Mood Disorders Induced by Maternal Overnutrition: The Role of the Gut-Brain Axis on the Development of Depression and Anxiety. Front Cell Dev Biol 2022; 10:795384. [PMID: 35155424 PMCID: PMC8826230 DOI: 10.3389/fcell.2022.795384] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/10/2022] [Indexed: 11/13/2022] Open
Abstract
Since the first evidence suggesting that maternal nutrition can impact the development of diseases in the offspring, much has been elucidated about its effects on the offspring’s nervous system. Animal studies demonstrated that maternal obesity can predispose the offspring to greater chances of metabolic and neurodevelopmental diseases. However, the mechanisms underlying these responses are not well established. In recent years, the role of the gut-brain axis in the development of anxiety and depression in people with obesity has emerged. Studies investigating changes in the maternal microbiota during pregnancy and also in the offspring demonstrate that conditions such as maternal obesity can modulate the microbiota, leading to long-term outcomes in the offspring. Considering that maternal obesity has also been linked to the development of psychiatric conditions (anxiety and depression), the gut-brain axis is a promising target to be further explored in these neuropsychiatric contexts. In the present study, we review the relationship between maternal obesity and anxious and depressive features, exploring the gut-brain axis as a potential mechanism underlying this relationship.
Collapse
Affiliation(s)
- Jeferson Jantsch
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Isadora D’Ávila Tassinari
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Márcia Giovenardi
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Victorio Bambini-Junior
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire (UCLan), Preston, United Kingdom
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster, United Kingdom
| | - Renata Padilha Guedes
- Programa de Pós-Graduação em Biociências, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
| | - Luciano Stürmer de Fraga
- Programa de Pós-Graduação em Ciências Biológicas: Fisiologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- *Correspondence: Luciano Stürmer de Fraga,
| |
Collapse
|
22
|
Dreisbach C, Morgan H, Cochran C, Gyamfi A, Henderson WA, Prescott S. Metabolic and Microbial Changes Associated With Diet and Obesity During Pregnancy: What Can We Learn From Animal Studies? Front Cell Infect Microbiol 2022; 11:795924. [PMID: 35118010 PMCID: PMC8804207 DOI: 10.3389/fcimb.2021.795924] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 12/13/2021] [Indexed: 12/22/2022] Open
Abstract
The intestinal microbiota changes throughout pregnancy and influences maternal metabolic adaptations to support fetal growth. Obesity induces alterations to the microbiota that include decreased microbial diversity and shifts in microbial composition, though specific species changes are inconsistent between published studies. In animal models, probiotics and exercise moderate maternal weight gain and partially correct the maternal microbiota. Supplemental Escherichia coli, however, exacerbate maternal obesity during the perinatal period, lending weight to the theory that inflammation-induced gut epithelial barrier leak influences metabolic dysregulation. Although birth weight is not always altered when offspring are exposed to an obesogenic diet during gestation, insulin resistance and lipid metabolism are impacted through adulthood in association with this exposure and can lead to increased body weight in adulthood. Postnatal offspring growth is accelerated in response to maternal overnutrition during lactation. Offspring microbiota, metabolism, and behavior are altered in response to early exposure to high fat and high sucrose diets. Consequences to this exposure include impaired glucose and insulin homeostasis, fatty liver, and neurobehavioral deficits that can be ameliorated by improving the microbial environment. In this mini review, we provide an overview of the use of translational animal models to understand the mechanisms associated with changes to the gastrointestinal microbiota due to maternal obesity and the microbial impact on the metabolic changes of pregnancy.
Collapse
Affiliation(s)
- Caitlin Dreisbach
- Data Science Institute, Columbia University, New York, NY, United States
| | - Hailey Morgan
- College of Nursing, University of South Florida, Tampa, FL, United States
| | - Caroline Cochran
- School of Nursing, Columbia University, New York, NY, United States
| | - Adwoa Gyamfi
- School of Medicine, University of Connecticut, Farmington, CT, United States
| | - Wendy Ann Henderson
- School of Medicine, University of Connecticut, Farmington, CT, United States
- School of Nursing, University of Connecticut, Storrs, CT, United States
| | - Stephanie Prescott
- College of Nursing, University of South Florida, Tampa, FL, United States
- *Correspondence: Stephanie Prescott,
| |
Collapse
|
23
|
Solano-Aguilar GI, Lakshman S, Jang S, Gupta R, Molokin A, Schroeder SG, Gillevet PM, Urban JF. The Effects of Consuming White Button Mushroom Agaricus bisporus on the Brain and Liver Metabolome Using a Targeted Metabolomic Analysis. Metabolites 2021; 11:metabo11110779. [PMID: 34822437 PMCID: PMC8625434 DOI: 10.3390/metabo11110779] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/29/2021] [Accepted: 11/11/2021] [Indexed: 11/21/2022] Open
Abstract
A targeted metabolomic analysis was performed on tissues derived from pigs fed diets supplemented with white button mushrooms (WBM) to determine the effect on the liver and brain metabolome. Thirty-one pigs were fed a grower diet alone or supplemented with either three or six servings of freeze-dried WBM for six weeks. Tissue metabolomes were analyzed using targeted liquid chromatography-mass spectrometry (LC-MS) combined with chemical similarity enrichment analysis (ChemRICH) and correlated to WBM-induced changes in fecal microbiome composition. Results indicated that WBM can differentially modulate metabolites in liver, brain cortex and hippocampus of healthy pigs. Within the glycero-phospholipids, there was an increase in alkyl-acyl-phosphatidyl-cholines (PC-O 40:3) in the hippocampus of pigs fed six servings of WBM. A broader change in glycerophospholipids and sphingolipids was detected in the liver with a reduction in several lipid species in pigs fed both WBM diets but with an increase in amino acids known as precursors of neurotransmitters in the cortex of pigs fed six servings of WBM. Metabolomic changes were positively correlated with increased abundance of Cryomorphaceae, Lachnospiraceae, Flammeovirgaceae and Ruminococcaceae in the microbiome suggesting that WBM can also positively impact tissue metabolite composition.
Collapse
Affiliation(s)
- Gloria I. Solano-Aguilar
- Diet Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture Northeast Area, Beltsville, MD 20705, USA; (S.L.); (S.J.); (A.M.); (J.F.U.J.)
- Correspondence: ; Tel.: +1-301-504-8068
| | - Sukla Lakshman
- Diet Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture Northeast Area, Beltsville, MD 20705, USA; (S.L.); (S.J.); (A.M.); (J.F.U.J.)
| | - Saebyeol Jang
- Diet Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture Northeast Area, Beltsville, MD 20705, USA; (S.L.); (S.J.); (A.M.); (J.F.U.J.)
| | - Richi Gupta
- Microbiome Analysis Center, George Mason University, Science & Technology Campus, Manassas, VA 20108, USA; (R.G.); (P.M.G.)
| | - Aleksey Molokin
- Diet Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture Northeast Area, Beltsville, MD 20705, USA; (S.L.); (S.J.); (A.M.); (J.F.U.J.)
| | - Steven G. Schroeder
- Animal Genomics and Improvement Laboratory, Beltsville Agricultural Research Center, Agricultural Research Service, U.S. Department of Agriculture Northeast Area, Beltsville, MD 20705, USA;
| | - Patrick M. Gillevet
- Microbiome Analysis Center, George Mason University, Science & Technology Campus, Manassas, VA 20108, USA; (R.G.); (P.M.G.)
| | - Joseph F. Urban
- Diet Genomics and Immunology Laboratory, Beltsville Human Nutrition Research Center, Agricultural Research Service, U.S. Department of Agriculture Northeast Area, Beltsville, MD 20705, USA; (S.L.); (S.J.); (A.M.); (J.F.U.J.)
| |
Collapse
|
24
|
Cusick JA, Wellman CL, Demas GE. The call of the wild: using non-model systems to investigate microbiome-behaviour relationships. J Exp Biol 2021; 224:jeb224485. [PMID: 33988717 PMCID: PMC8180253 DOI: 10.1242/jeb.224485] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
On and within most sites across an animal's body live complex communities of microorganisms. These microorganisms perform a variety of important functions for their hosts, including communicating with the brain, immune system and endocrine axes to mediate physiological processes and affect individual behaviour. Microbiome research has primarily focused on the functions of the microbiome within the gastrointestinal tract (gut microbiome) using biomedically relevant laboratory species (i.e. model organisms). These studies have identified important connections between the gut microbiome and host immune, neuroendocrine and nervous systems, as well as how these connections, in turn, influence host behaviour and health. Recently, the field has expanded beyond traditional model systems as it has become apparent that the microbiome can drive differences in behaviour and diet, play a fundamental role in host fitness and influence community-scale dynamics in wild populations. In this Review, we highlight the value of conducting hypothesis-driven research in non-model organisms and the benefits of a comparative approach that assesses patterns across different species or taxa. Using social behaviour as an intellectual framework, we review the bidirectional relationship between the gut microbiome and host behaviour, and identify understudied mechanisms by which these effects may be mediated.
Collapse
Affiliation(s)
- Jessica A. Cusick
- Department of Biology, Indiana University, Biology Building 142, 1001 East Third Street, Bloomington, IN 47405, USA
- Animal Behavior Program, Indiana University, 409 N. Park Avenue, Bloomington, IN 47405, USA
| | - Cara L. Wellman
- Animal Behavior Program, Indiana University, 409 N. Park Avenue, Bloomington, IN 47405, USA
- Department of Psychological and Brain Sciences, Indiana University, 1101 E. 10th Street, Bloomington, IN 47405-7007, USA
- Program in Neuroscience, Indiana University, Psychology Building, 1101 E 10th Street Bloomington, IN 47405-2204, USA
| | - Gregory E. Demas
- Department of Biology, Indiana University, Biology Building 142, 1001 East Third Street, Bloomington, IN 47405, USA
- Animal Behavior Program, Indiana University, 409 N. Park Avenue, Bloomington, IN 47405, USA
- Program in Neuroscience, Indiana University, Psychology Building, 1101 E 10th Street Bloomington, IN 47405-2204, USA
| |
Collapse
|
25
|
Valeri F, Endres K. How biological sex of the host shapes its gut microbiota. Front Neuroendocrinol 2021; 61:100912. [PMID: 33713673 DOI: 10.1016/j.yfrne.2021.100912] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 02/10/2021] [Accepted: 03/03/2021] [Indexed: 02/06/2023]
Abstract
The gut microbiota is a complex system, consisting of a dynamic population of microorganisms, involved in the regulation of the host's homeostasis. A vast number of factors are driving the gut microbiota composition including diet, antibiotics, environment, and lifestyle. However, in the past decade, a growing number of studies also focused on the role of sex in relationship to changes in the gut microbiota composition in animal experiments as well as in human beings. Despite the progress in investigation techniques, still little is known about the mechanism behind the observed sex-related differences. In this review, we summarized current knowledge on the sex-dependent differences of the intestinal commensals and discuss the probable direct impact of sex hormones and more indirect effects such as dietary habits or antibiotics. While we have to conclude limited data on specific developmental stages, a clear role for sexual hormones and most probably for testosterone emerges.
Collapse
Affiliation(s)
- Francesco Valeri
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, Mainz 55131, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center, Johannes Gutenberg-University Mainz, Mainz 55131, Germany.
| |
Collapse
|
26
|
Moradi K, Ashraf-Ganjouei A, Tavolinejad H, Bagheri S, Akhondzadeh S. The interplay between gut microbiota and autism spectrum disorders: A focus on immunological pathways. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110091. [PMID: 32891667 DOI: 10.1016/j.pnpbp.2020.110091] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 08/21/2020] [Accepted: 08/30/2020] [Indexed: 12/23/2022]
Abstract
Autism spectrum disorders (ASD) are a group of neurodevelopmental disorders characterized by impairments in social and cognitive activities, stereotypical and repetitive behaviors and restricted areas of interest. A remarkable proportion of ASD patients represent immune dysregulation as well as gastrointestinal complications. Hence, a novel concept has recently emerged, addressing the possible intercommunication between the brain, the immune system, the gut and its commensals. Here, we provide an overview of how gut microbes and their metabolites are associated with neurobehavioral features of ASD through various immunologic mechanisms. Moreover, we discuss the potential therapeutic options that could modify these features.
Collapse
Affiliation(s)
- Kamyar Moradi
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ashraf-Ganjouei
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Tavolinejad
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayna Bagheri
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahin Akhondzadeh
- Psychiatric Research Center, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
Abstract
Gastrointestinal disorders are one of the most common medical conditions that are comorbid with autism spectrum disorders. These comorbidities can cause greater severity in autism spectrum disorder symptoms, other associated clinical manifestations, and lower quality of life if left untreated. Clinicians need to understand how these gastrointestinal issues present and apply effective therapies. Effective treatment of gastrointestinal problems in autism spectrum disorder may result in marked improvements in autism spectrum disorder behavioral outcomes. This article discusses the gastrointestinal disorders commonly associated with autism spectrum disorders, how they present, and studied risk factors.
Collapse
Affiliation(s)
- Moneek Madra
- Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Irving Medical Center, 622 West 168th Street, New York, NY 10025, USA,Institute of Human Nutrition, Columbia University Irving Medical Center, 630 West 168th Street, PH1512E, New York, NY 10032, USA
| | - Roey Ringel
- Department of Pediatrics, Morgan Stanley Children’s Hospital, Columbia University Irving Medical Center, 622 West 168th Street, New York, NY 10025, USA,Columbia College, Columbia University, New York, NY, USA
| | - Kara Gross Margolis
- Department of Pediatrics, Morgan Stanley Children's Hospital, Columbia University Irving Medical Center, 622 West 168th Street, New York, NY 10025, USA; Institute of Human Nutrition, Columbia University Irving Medical Center, 630 West 168th Street, PH1512E, New York, NY 10032, USA.
| |
Collapse
|
28
|
Troyer EA, Kohn JN, Ecklu-Mensah G, Aleti G, Rosenberg DR, Hong S. Searching for host immune-microbiome mechanisms in obsessive-compulsive disorder: A narrative literature review and future directions. Neurosci Biobehav Rev 2021; 125:517-534. [PMID: 33639178 DOI: 10.1016/j.neubiorev.2021.02.034] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Revised: 02/09/2021] [Accepted: 02/22/2021] [Indexed: 12/20/2022]
Abstract
Obsessive-compulsive disorder (OCD) is disabling and often treatment-refractory. Host immunity and gut microbiota have bidirectional communication with each other and with the brain. Perturbations to this axis have been implicated in neuropsychiatric disorders, but immune-microbiome signaling in OCD is relatively underexplored. We review support for further pursuing such investigations in OCD, including: 1) gut microbiota has been associated with OCD, but causal pathogenic mechanisms remain unclear; 2) early environmental risk factors for OCD overlap with critical periods of immune-microbiome development; 3) OCD is associated with increased risk of immune-mediated disorders and changes in immune parameters, which are separately associated with the microbiome; and 4) gut microbiome manipulations in animal models are associated with changes in immunity and some obsessive-compulsive symptoms. Theoretical pathogenic mechanisms could include microbiota programming of cytokine production, promotion of expansion and trafficking of peripheral immune cells to the CNS, and regulation of microglial function. Immune-microbiome signaling in OCD requires further exploration, and may offer novel insights into pathogenic mechanisms and potential treatment targets for this disabling disorder.
Collapse
Affiliation(s)
- Emily A Troyer
- Department of Psychiatry, University of California San Diego, La Jolla, California, United States.
| | - Jordan N Kohn
- Department of Psychiatry, University of California San Diego, La Jolla, California, United States
| | - Gertrude Ecklu-Mensah
- Department of Medicine and Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, United States
| | - Gajender Aleti
- Department of Psychiatry, University of California San Diego, La Jolla, California, United States
| | - David R Rosenberg
- Department of Psychiatry and Behavioral Neurosciences, Wayne State University, Detroit, Michigan, United States
| | - Suzi Hong
- Department of Psychiatry, University of California San Diego, La Jolla, California, United States; Herbert Wertheim School of Public Health and Human Longevity Science, University of California San Diego, La Jolla, California, United States
| |
Collapse
|
29
|
Carranza-Naval MJ, Vargas-Soria M, Hierro-Bujalance C, Baena-Nieto G, Garcia-Alloza M, Infante-Garcia C, del Marco A. Alzheimer's Disease and Diabetes: Role of Diet, Microbiota and Inflammation in Preclinical Models. Biomolecules 2021; 11:biom11020262. [PMID: 33578998 PMCID: PMC7916805 DOI: 10.3390/biom11020262] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 02/05/2021] [Accepted: 02/05/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Epidemiological studies show the association between AD and type 2 diabetes (T2DM), although the mechanisms are not fully understood. Dietary habits and lifestyle, that are risk factors in both diseases, strongly modulate gut microbiota composition. Also, the brain-gut axis plays a relevant role in AD, diabetes and inflammation, through products of bacterial metabolism, like short-chain fatty acids. We provide a comprehensive review of current literature on the relation between dysbiosis, altered inflammatory cytokines profile and microglia in preclinical models of AD, T2DM and models that reproduce both diseases as commonly observed in the clinic. Increased proinflammatory cytokines, such as IL-1β and TNF-α, are widely detected. Microbiome analysis shows alterations in Actinobacteria, Bacteroidetes or Firmicutes phyla, among others. Altered α- and β-diversity is observed in mice depending on genotype, gender and age; therefore, alterations in bacteria taxa highly depend on the models and approaches. We also review the use of pre- and probiotic supplements, that by favoring a healthy microbiome ameliorate AD and T2DM pathologies. Whereas extensive studies have been carried out, further research would be necessary to fully understand the relation between diet, microbiome and inflammation in AD and T2DM.
Collapse
Affiliation(s)
- Maria Jose Carranza-Naval
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
- Salus Infirmorum, Universidad de Cadiz, 11005 Cadiz, Spain
| | - Maria Vargas-Soria
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
| | - Carmen Hierro-Bujalance
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
| | - Gloria Baena-Nieto
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
- Department of Endocrinology, Jerez Hospital, Jerez de la Frontera, 11407 Cadiz, Spain
| | - Monica Garcia-Alloza
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
| | - Carmen Infante-Garcia
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
- Correspondence: (C.I.-G.); (A.d.M.)
| | - Angel del Marco
- Division of Physiology, School of Medicine, Universidad de Cadiz, 11003 Cadiz, Spain; (M.J.C.-N.); (M.V.-S.); (C.H.-B.); (M.G.-A.)
- Instituto de Investigacion e Innovacion en Ciencias Biomedicas de la Provincia de Cadiz (INIBICA), 11009 Cadiz, Spain;
- Correspondence: (C.I.-G.); (A.d.M.)
| |
Collapse
|
30
|
O'Connor R, Moloney GM, Fulling C, O'Riordan KJ, Fitzgerald P, Bastiaanssen TFS, Schellekens H, Dinan TG, Cryan JF. Maternal antibiotic administration during a critical developmental window has enduring neurobehavioural effects in offspring mice. Behav Brain Res 2021; 404:113156. [PMID: 33571573 DOI: 10.1016/j.bbr.2021.113156] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 01/05/2021] [Accepted: 01/29/2021] [Indexed: 02/08/2023]
Abstract
Rates of perinatal maternal antibiotic use have increased in recent years linked to prophylactic antibiotic use following Caesarean section delivery. This antibiotic use is necessary and beneficial in the short-term; however, long-term consequences on brain and behaviour have not been studied in detail. Here, we endeavoured to determine whether maternal administration of antibiotics during a critical window of development in early life has lasting effects on brain and behaviour in offspring mice. To this end we studied two different antibiotic preparations (single administration of Phenoxymethylpenicillin at 31 mg/kg/day; and a cocktail consisting of, ampicillin 1 mg/mL; vancomycin 0.5 mg/mL; metronidazole 1 mg/mL; ciprofloxacin 0.2 mg/mL and imipenem 0.25 mg/mL). It was observed that early life exposure to maternal antibiotics led to persistent alterations in anxiety, sociability and cognitive behaviours. These effects in general were greater in animals treated with the broad-spectrum antibiotic cocktail compared to a single antibiotic with the exception of deficits in social recognition which were more robustly observed in Penicillin V exposed animals. Given the prevalence of maternal antibiotic use, our findings have potentially significant translational relevance, particularly considering the implications on infant health during this critical period and into later life.
Collapse
Affiliation(s)
- Rory O'Connor
- APC Microbiome Ireland, University College Cork, Ireland
| | - Gerard M Moloney
- Department of Anatomy & Neuroscience, University College Cork, Ireland
| | | | | | - Pat Fitzgerald
- APC Microbiome Ireland, University College Cork, Ireland
| | - Thomaz F S Bastiaanssen
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - Harriët Schellekens
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland
| | - Timothy G Dinan
- APC Microbiome Ireland, University College Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Ireland
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Ireland; Department of Anatomy & Neuroscience, University College Cork, Ireland.
| |
Collapse
|
31
|
Marzullo P, Di Renzo L, Pugliese G, De Siena M, Barrea L, Muscogiuri G, Colao A, Savastano S. From obesity through gut microbiota to cardiovascular diseases: a dangerous journey. INTERNATIONAL JOURNAL OF OBESITY SUPPLEMENTS 2020; 10:35-49. [PMID: 32714511 PMCID: PMC7371682 DOI: 10.1038/s41367-020-0017-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The co-existence of humans and gut microbiota started millions of years ago. Until now, a balance gradually developed between gut bacteria and their hosts. It is now recognized that gut microbiota are key to form adequate immune and metabolic functions and, more in general, for the maintenance of good health. Gut microbiota are established before birth under the influence of maternal nutrition and metabolic status, which can impact the future metabolic risk of the offspring in terms of obesity, diabetes, and cardiometabolic disorders during the lifespan. Obesity and diabetes are prone to disrupt the gut microbiota and alter the gut barrier permeability, leading to metabolic endotoxaemia with its detrimental consequences on health. Specific bacterial sequences are now viewed as peculiar signatures of the metabolic syndrome across life stages in each individual, and are linked to pathogenesis of cardiovascular diseases (CVDs) via metabolic products (metabolites) and immune modulation. These mechanisms have been linked, in association with abnormalities in microbial richness and diversity, to an increased risk of developing arterial hypertension, systemic inflammation, nonalcoholic fatty liver disease, coronary artery disease, chronic kidney disease, and heart failure. Emerging strategies for the manipulation of intestinal microbiota represent a promising therapeutic option for the prevention and treatment of CVD especially in individuals prone to CV events.
Collapse
Affiliation(s)
- Paolo Marzullo
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Division of General Medicine, IRCCS Istituto Auxologico Italiano, 28923 Piancavallo, Verbania Italy
| | - Laura Di Renzo
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00136 Rome, Italy
| | - Gabriella Pugliese
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Martina De Siena
- Division of Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
| | - Luigi Barrea
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Giovanna Muscogiuri
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Annamaria Colao
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - Silvia Savastano
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
| | - on behalf of Obesity Programs of nutrition, Education, Research and Assessment (OPERA) Group
- Department of Translational Medicine, Università del Piemonte Orientale, 28100 Novara, Italy
- Division of General Medicine, IRCCS Istituto Auxologico Italiano, 28923 Piancavallo, Verbania Italy
- Section of Clinical Nutrition and Nutrigenomic, Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00136 Rome, Italy
- Unit of Endocrinology, Dipartimento di Medicina Clinica e Chirurgia, Federico II University, 80131 Naples, Italy
- Division of Gastroenterology, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
- Digestive Endoscopy Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS—Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
32
|
Abstract
Gastrointestinal disorders are one of the most common medical conditions that are comorbid with autism spectrum disorders. These comorbidities can cause greater severity in autism spectrum disorder symptoms, other associated clinical manifestations, and lower quality of life if left untreated. Clinicians need to understand how these gastrointestinal issues present and apply effective therapies. Effective treatment of gastrointestinal problems in autism spectrum disorder may result in marked improvements in autism spectrum disorder behavioral outcomes. This article discusses the gastrointestinal disorders commonly associated with autism spectrum disorders, how they present, and studied risk factors.
Collapse
Affiliation(s)
- Moneek Madra
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York
| | - Roey Ringel
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,Columbia College, Columbia University, New York, New York
| | - Kara G. Margolis
- Morgan Stanley Children’s Hospital, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York,Institute of Human Nutrition, Columbia University Irving Medical Center, New York, New York
| |
Collapse
|
33
|
Shook LL, Kislal S, Edlow AG. Fetal brain and placental programming in maternal obesity: A review of human and animal model studies. Prenat Diagn 2020; 40:1126-1137. [PMID: 32362000 DOI: 10.1002/pd.5724] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Revised: 03/26/2020] [Accepted: 04/21/2020] [Indexed: 12/13/2022]
Abstract
Both human epidemiologic and animal model studies demonstrate that prenatal and lactational exposure to maternal obesity and high-fat diet are associated with adverse neurodevelopmental outcomes in offspring. Neurodevelopmental outcomes described in offspring of obese women include cognitive impairment, autism spectrum disorder (ASD), attention deficit hyperactivity disorder, anxiety and depression, disordered eating, and propensity for reward-driven behavior, among others. This review synthesizes human and animal data linking maternal obesity and high-fat diet consumption to abnormal fetal brain development, and neurodevelopmental and psychiatric morbidity in offspring. It highlights key mechanisms by which maternal obesity and maternal diet impact fetal and offspring development, and sex differences in offspring programming. In addition, we review placental effects of maternal obesity, and the role the placenta might play as an indicator vs mediator of fetal programming.
Collapse
Affiliation(s)
- Lydia L Shook
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA
| | - Sezen Kislal
- Massachusetts General Hospital Research Institute, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| | - Andrea G Edlow
- Division of Maternal-Fetal Medicine, Department of Obstetrics & Gynecology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, USA.,Massachusetts General Hospital Research Institute, Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, Massachusetts, USA
| |
Collapse
|
34
|
Abstract
While there are numerous medical comorbidities associated with ASD, gastrointestinal (GI) issues have a significant impact on quality of life for these individuals. Recent findings continue to support the relationship between the gut microbiome and both GI symptoms and behavior, but the heterogeneity within the autism spectrum requires in-depth clinical characterization of these clinical cohorts. Large, diverse, well-controlled studies in this area of research are still needed. Although there is still much to discover about the brain-gut-microbiome axis in ASD, microbially mediated therapies, specifically probiotics and fecal microbiota transplantation have shown promise in the treatment of GI symptoms in ASD, with potential benefit to the core behavioral symptoms of ASD as well. Future research and clinical trials must increasingly consider complex phenotypes in ASD in stratification of large datasets as well as in design of inclusion criteria for individual therapeutic interventions.
Collapse
Affiliation(s)
- Virginia Saurman
- Department of Pediatrics, Columbia University Medical Center, 620 West 168th Street, New York, NY 10032, USA
| | - Kara G. Margolis
- Department of Pediatrics, Columbia University Medical Center, 620 West 168th Street, New York, NY 10032, USA
| | - Ruth Ann Luna
- Department of Pathology and Immunology, Texas Children’s Microbiome Center, Baylor College of Medicine, Texas Children’s Hospital, Feigin Tower, 1102 Bates Avenue, Suite 955, Houston, TX 77030, USA
| |
Collapse
|
35
|
Kamimura I, Kaneko R, Morita H, Mogi K, Kikusui T. Microbial colonization history modulates anxiety-like and complex social behavior in mice. Neurosci Res 2020; 168:64-75. [PMID: 32017965 DOI: 10.1016/j.neures.2020.01.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 12/27/2019] [Accepted: 01/06/2020] [Indexed: 12/17/2022]
Abstract
Microbiome composition has a pivotal role in neurobehavioral development. However, there is limited information about the role of the microbiome in sociability of mice in complex social contexts. Germ-free (GF) mice were reared in a microbiota-free environment until postnatal day 21 and then transferred to a room containing specific pathogen free (SPF) mice. At 9 weeks old, group social behaviors were measured for three GF mice and three SPF mice unfamiliar to each other. GF mice spent less time in the center area of the arena and there were longer inter-individual distances compared with SPF mice. GF mice also had decreased brain-derived neurotrophic factor (BDNF) and increased ΔFosB mRNA in the prefrontal cortex compared to SPF mice. There were differences in the gut microbiome composition between GF and SPF mice; however, if cohabitating after weaning, then their microbiome composition became equivalent and group differences in behavior and BDNF and ΔFosB mRNA expression disappeared. These results demonstrate that the bacterial community can modulate neural systems that are involved in sociability and anxiety during the developmental period and suggest that sociability and anxiety can be shaped depending on the microbiome environment through interaction with conspecifics.
Collapse
Affiliation(s)
- Itsuka Kamimura
- Department of Animal Science and Biotechnology, Azabu University, Japan
| | - Ryou Kaneko
- Graduate School of Environmental and Life Science, Okayama University, Japan
| | - Hidetoshi Morita
- Graduate School of Environmental and Life Science, Okayama University, Japan
| | - Kazutaka Mogi
- Department of Animal Science and Biotechnology, Azabu University, Japan
| | - Takefumi Kikusui
- Department of Animal Science and Biotechnology, Azabu University, Japan.
| |
Collapse
|
36
|
Ward-Flanagan R, Scavuzzo C, Mandhane PJ, Bolduc FV, Dickson CT. Prenatal fruit juice exposure enhances memory consolidation in male post-weanling Sprague-Dawley rats. PLoS One 2020; 15:e0227938. [PMID: 31990931 PMCID: PMC6986755 DOI: 10.1371/journal.pone.0227938] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 12/28/2019] [Indexed: 11/18/2022] Open
Abstract
Objectives Nutritional intake during gestation is known to impact health outcomes for progeny. Correlational evidence in humans suggests that increased fruit consumption of pregnant mothers enhances infant cognitive development. Moreover, wild-type Drosophila supplemented with a combination of orange and tomato juice showed robust enhancements in performance on an associative olfactory memory task. The current study aimed to experimentally test the effects of prenatal fruit juice exposure in a non-human, mammalian model of learning and memory. Methods Across three separate birth cohorts, pregnant rats were given access to diluted tomato and orange juice (N = 2 per cohort), with control rats (N = 2 per cohort) receiving only water, in addition to standard rodent chow, throughout the duration of gestation, ending at parturition. Following weaning, male offspring were tested for learning and memory in a spatial version of the circular water maze and an auditory-cued fear-conditioning task. Results All pregnant rats increased fluid and food intake over the gestational period. Fruit juice-fed pregnant rats had increased fluid intake compared to control pregnant rats. When testing progeny, there were no effects of prenatal fruit juice on spatial learning, while it appeared to impair learning in fear conditioning relative to controls. However, we measured significant enhancements in both spatial memory and conditioned fear memory in the prenatal fruit-juice group compared to controls. Measures of vigilance, in response to the conditioned cue, were increased in prenatal fruit rats compared to controls, suggesting less generalized, and more adaptive, anxiety behaviours. Discussion Our results corroborate the human and Drosophila findings of prenatal fruit effects on behaviour, specifically that prenatal fruit juice exposure may be beneficial for early-life memory consolidation in rats.
Collapse
Affiliation(s)
- Rachel Ward-Flanagan
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Claire Scavuzzo
- Department of Psychology, University of Alberta, Edmonton, AB, Canada
| | - Piush J. Mandhane
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Francois V. Bolduc
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
- Department of Medical Genetics, University of Alberta, Edmonton, AB, Canada
| | - Clayton T. Dickson
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
- Department of Psychology, University of Alberta, Edmonton, AB, Canada
- Department of Physiology, University of Alberta, Edmonton, AB, Canada
- * E-mail:
| |
Collapse
|
37
|
Bruce-Keller AJ, Richard AJ, Fernandez-Kim SO, Ribnicky DM, Salbaum JM, Newman S, Carmouche R, Stephens JM. Fenugreek Counters the Effects of High Fat Diet on Gut Microbiota in Mice: Links to Metabolic Benefit. Sci Rep 2020; 10:1245. [PMID: 31988303 PMCID: PMC6985225 DOI: 10.1038/s41598-020-58005-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 01/07/2020] [Indexed: 02/06/2023] Open
Abstract
Fenugreek (Trigonella foenum-graecum) is an annual herbaceous plant and a staple of traditional health remedies for metabolic conditions including high cholesterol and diabetes. While the mechanisms of the beneficial actions of fenugreek remain unknown, a role for intestinal microbiota in metabolic homeostasis is likely. To determine if fenugreek utilizes intestinal bacteria to offset the adverse effects of high fat diets, C57BL/6J mice were fed control/low fat (CD) or high fat (HFD) diets each supplemented with or without 2% (w/w) fenugreek for 16 weeks. The effects of fenugreek and HFD on gut microbiota were comprehensively mapped and then statistically assessed in relation to effects on metrics of body weight, hyperlipidemia, and glucose tolerance. 16S metagenomic analyses revealed robust and significant effects of fenugreek on gut microbiota, with alterations in both alpha and beta diversity as well as taxonomic redistribution under both CD and HFD conditions. As previously reported, fenugreek attenuated HFD-induced hyperlipidemia and stabilized glucose tolerance without affecting body weight. Finally, fenugreek specifically reversed the dysbiotic effects of HFD on numerous taxa in a manner tightly correlated with overall metabolic function. Collectively, these data reinforce the essential link between gut microbiota and metabolic syndrome and suggest that the preservation of healthy populations of gut microbiota participates in the beneficial properties of fenugreek in the context of modern Western-style diets.
Collapse
Affiliation(s)
- Annadora J Bruce-Keller
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, 70808, USA.
| | - Allison J Richard
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, 70808, USA
| | - Sun-Ok Fernandez-Kim
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, 70808, USA
| | - David M Ribnicky
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, 08901, USA
| | - J Michael Salbaum
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, 70808, USA
| | - Susan Newman
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, 70808, USA
| | - Richard Carmouche
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, 70808, USA
| | - Jacqueline M Stephens
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, 70808, USA
| |
Collapse
|
38
|
Huang YC, Huang LT, Sheen JM, Hou CY, Yeh YT, Chiang CP, Lin IC, Tiao MM, Tsai CC, Lin YJ, Chen CC, Tain YL, Yu HR. Resveratrol treatment improves the altered metabolism and related dysbiosis of gut programed by prenatal high-fat diet and postnatal high-fat diet exposure. J Nutr Biochem 2019; 75:108260. [PMID: 31707285 DOI: 10.1016/j.jnutbio.2019.108260] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2018] [Revised: 05/21/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023]
Abstract
A maternal high-fat (HF) diet sensitizes offspring to the adverse effects of postnatal HF intake and can lead to metabolic dysregulation. Resveratrol, a natural polyphenolic compound found in grapes and red wine, could help to relieve metabolic syndrome dysregulation. Since the gut microbiota is known to be closely related to metabolic homeostasis, this study aimed to investigate the impact of a combination of maternal and postweaning HF diets on the gut microbiota and whether resveratrol could relieve the gut dysbiosis associated with metabolic dysregulation. Sprague-Dawley dams were sustained on either a chow or HF diet before mating, during pregnancy and during lactation. Their offspring were randomly fed chow or a HF diet after weaning. Four experimental groups were generated: CC (maternal/postnatal chow diet), HC (maternal HF/postnatal chow diet), CH (maternal chow/postnatal high-fat diet) and HH (maternal/postnatal HF diet). A fifth group consisted of HH with resveratrol treatment. We found that both maternal and postnatal HF exposure has a distinct effect on the gut microbiota metagenome of offspring. Maternal HF diet exposure decreased plasma acetate, propionate and butyrate level, while postnatal HF diet exposure decreased plasma acetate level in adult life. The metabolic dysregulation programed by the maternal and postnatal HF diets was related to the relevant gut microbiota. Resveratrol treatment ameliorated the altered plasma propionate level related to maternal HF and postnatal HF diet treatment. Resveratrol treatment also improved most of the altered metabolic dysregulation and related dysbiosis programmed by maternal and postnatal HF diet exposure.
Collapse
Affiliation(s)
- Yi-Chuan Huang
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Li-Tung Huang
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Jiunn-Ming Sheen
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Chih-Yao Hou
- Department of Seafood Science, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan
| | - Yao-Tsung Yeh
- Aging and Disease Prevention Research Center, Fooyin University, Kaohsiung, Taiwan; Department of Education and Research, Fooyin University Hospital, Pingtung, Taiwan; Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, Taiwan
| | - Chih-Po Chiang
- Aging and Disease Prevention Research Center, Fooyin University, Kaohsiung, Taiwan; Department of Education and Research, Fooyin University Hospital, Pingtung, Taiwan; Department of Medical Laboratory Sciences and Biotechnology, Fooyin University, Kaohsiung, Taiwan
| | - I-Chun Lin
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Mao-Meng Tiao
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Ching-Chou Tsai
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital
| | - Yu-Ju Lin
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital
| | - Chih-Cheng Chen
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - You-Lin Tain
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan
| | - Hong-Ren Yu
- Department of Pediatrics, Chang Gung Memorial Hospital-Kaohsiung Medical Center, Graduate Institute of Clinical Medical Science, Chang Gung University College of Medicine, Kaohsiung, Taiwan.
| |
Collapse
|
39
|
Rivell A, Mattson MP. Intergenerational Metabolic Syndrome and Neuronal Network Hyperexcitability in Autism. Trends Neurosci 2019; 42:709-726. [PMID: 31495451 PMCID: PMC6779523 DOI: 10.1016/j.tins.2019.08.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 07/17/2019] [Accepted: 08/06/2019] [Indexed: 12/13/2022]
Abstract
We review evidence that suggests a role for excessive consumption of energy-dense foods, particularly fructose, and consequent obesity and insulin resistance (metabolic syndrome) in the recent increase in prevalence of autism spectrum disorders (ASD). Maternal insulin resistance, obesity, and diabetes may predispose offspring to ASD by mechanisms involving chronic activation of anabolic cellular pathways and a lack of metabolic switching to ketosis resulting in a deficit in GABAergic signaling and neuronal network hyperexcitability. Metabolic reprogramming by epigenetic DNA and chromatin modifications may contribute to alterations in gene expression that result in ASD. These mechanistic insights suggest that interventions that improve metabolic health such as intermittent fasting and exercise may ameliorate developmental neuronal network abnormalities and consequent behavioral manifestations in ASD.
Collapse
Affiliation(s)
- Aileen Rivell
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA
| | - Mark P Mattson
- Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD, USA; Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
40
|
Lu A, Petrullo L, Carrera S, Feder J, Schneider-Crease I, Snyder-Mackler N. Developmental responses to early-life adversity: Evolutionary and mechanistic perspectives. Evol Anthropol 2019; 28:249-266. [PMID: 31498945 DOI: 10.1002/evan.21791] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 03/28/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023]
Abstract
Adverse ecological and social conditions during early life are known to influence development, with rippling effects that may explain variation in adult health and fitness. The adaptive function of such developmental plasticity, however, remains relatively untested in long-lived animals, resulting in much debate over which evolutionary models are most applicable. Furthermore, despite the promise of clinical interventions that might alleviate the health consequences of early-life adversity, research on the proximate mechanisms governing phenotypic responses to adversity have been largely limited to studies on glucocorticoids. Here, we synthesize the current state of research on developmental plasticity, discussing both ultimate and proximate mechanisms. First, we evaluate the utility of adaptive models proposed to explain developmental responses to early-life adversity, particularly for long-lived mammals such as humans. In doing so, we highlight how parent-offspring conflict complicates our understanding of whether mothers or offspring benefit from these responses. Second, we discuss the role of glucocorticoids and a second physiological system-the gut microbiome-that has emerged as an additional, clinically relevant mechanism by which early-life adversity can influence development. Finally, we suggest ways in which nonhuman primates can serve as models to study the effects of early-life adversity, both from evolutionary and clinical perspectives.
Collapse
Affiliation(s)
- Amy Lu
- Department of Anthropology, Stony Brook University, Stony Brook, New York
| | - Lauren Petrullo
- Interdepartmental Doctoral Program in Anthropological Sciences, Stony Brook University, Stony Brook, New York
| | - Sofia Carrera
- Department of Psychology, University of Michigan, Ann Arbor, Michigan
| | - Jacob Feder
- Interdepartmental Doctoral Program in Anthropological Sciences, Stony Brook University, Stony Brook, New York
| | - India Schneider-Crease
- Department of Anthropology, Stony Brook University, Stony Brook, New York.,Department of Psychology, University of Washington, Seattle, Washington
| | - Noah Snyder-Mackler
- Department of Psychology, University of Washington, Seattle, Washington.,Center for Studies in Demography and Ecology, University of Washington, Seattle, Washington
| |
Collapse
|
41
|
Daniel S, Doron M, Fishman B, Koren G, Lunenfeld E, Levy A. The safety of amoxicillin and clavulanic acid use during the first trimester of pregnancy. Br J Clin Pharmacol 2019; 85:2856-2863. [PMID: 31486528 DOI: 10.1111/bcp.14118] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Revised: 08/13/2019] [Accepted: 08/23/2019] [Indexed: 11/29/2022] Open
Abstract
AIMS The goal of the current study was to assess the risk for major congenital malformations following first-trimester exposure to amoxicillin, or amoxicillin and clavulanic acid (ACA). METHODS A population-based retrospective cohort study was conducted, by linking 4 computerized databases: maternal and infant hospitalization records, drug dispensing database of Clalit Health Services in Israel and data concerning pregnancy terminations. Multivariate negative-binomial regression was used to assess the risk for major malformations following first-trimester exposure, adjusted for mother's age, ethnicity (Bedouin vs Jewish), parity, diabetes mellitus, lack of perinatal care, and the year of birth. RESULTS The study included 101 615 pregnancies, of which 6919 (6.8%) were exposed to amoxicillin: 1045 (1.0%) to amoxicillin only and 6041 (5.9%) to ACA. No significant association was found, in the univariate and multivariate analyses, between first-trimester exposure to amoxicillin or ACA and major malformations in general (crude relative risk, 1.05 95% confidence interval 0.95-1.16; adjusted relative risk 1.09, 95% confidence interval 0.98-1.20), or for major malformations according to organ systems. No dose-response relationship was found between exposure in terms of the defined daily dose and major malformations. CONCLUSION Exposure to amoxicillin and ACA during the first trimester of pregnancy was not associated with an increased risk of major congenital malformations.
Collapse
Affiliation(s)
- Sharon Daniel
- Department of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Clalit Health Services (Southern District), Beer-Sheva, Israel
| | - Maya Doron
- Department of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Boris Fishman
- Department of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | | | - Eitan Lunenfeld
- Department of Obstetrics and Gynecology, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Soroka Medical Center, Beer-Sheva, Israel
| | - Amalia Levy
- Department of Public Health, Ben-Gurion University of the Negev, Beer-Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
42
|
Martin CL, Jima D, Sharp GC, McCullough LE, Park SS, Gowdy KM, Skaar D, Cowley M, Maguire RL, Fuemmeler B, Collier D, Relton CL, Murphy SK, Hoyo C. Maternal pre-pregnancy obesity, offspring cord blood DNA methylation, and offspring cardiometabolic health in early childhood: an epigenome-wide association study. Epigenetics 2019; 14:325-340. [PMID: 30773972 PMCID: PMC6557549 DOI: 10.1080/15592294.2019.1581594] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 01/25/2019] [Accepted: 01/28/2019] [Indexed: 01/03/2023] Open
Abstract
Pre-pregnancy obesity is an established risk factor for adverse sex-specific cardiometabolic health in offspring. Epigenetic alterations, such as in DNA methylation (DNAm), are a hypothesized link; however, sex-specific epigenomic targets remain unclear. Leveraging data from the Newborn Epigenetics Study (NEST) cohort, linear regression models were used to identify CpG sites in cord blood leukocytes associated with pre-pregnancy obesity in 187 mother-female and 173 mother-male offsprings. DNAm in cord blood was measured using the Illumina HumanMethylation450k BeadChip. Replication analysis was conducted among the Avon Longitudinal Study of Parents and Children (ALSPAC) cohort. Associations between pre-pregnancy obesity-associated CpG sites and offspring BMI z-score (BMIz) and blood pressure (BP) percentiles at 4-5-years of age were also examined. Maternal pre-pregnacy obesity was associated with 876 CpGs in female and 293 CpGs in male offspring (false discovery rate <5%). Among female offspring, 57 CpG sites, including the top 18, mapped to the TAPBP gene (range of effect estimates: -0.83% decrease to 4.02% increase in methylation). CpG methylation differences in the TAPBP gene were also observed among males (range of effect estimates: -0.30% decrease to 2.59% increase in methylation). While technically validated, none of the TAPBP CpG sites were replicated in ALSPAC. In NEST, methylation differences at CpG sites of the TAPBP gene were associated with BMI z-score (cg23922433 and cg17621507) and systolic BP percentile (cg06230948) in female and systolic (cg06230948) and diastolic (cg03780271) BP percentile in male offspring. Together, these findings suggest sex-specific effects, which, if causal, may explain observed sex-specific effects of maternal obesity.
Collapse
Affiliation(s)
- Chantel L Martin
- a Department of Epidemiology , Gillings School of Global Public Health, University of North Carolina , Chapel Hill , NC , USA
| | - Dereje Jima
- b Center of Human Health and the Environment , North Carolina State University , Raleigh , USA
- c Bioinformatics Research Center , North Carolina State University , Raleigh , NC , USA
| | - Gemma C Sharp
- d Medical Research Integrative Epidemiology Unit , Bristol Medical School, Population Health Sciences, University of Bristol , Bristol , UK
| | - Lauren E McCullough
- e Department of Epidemiology , Rollins School of Public Health, Emory University , Atlanta , GA , USA
| | - Sarah S Park
- f Department of Biological Sciences , North Carolina State University , Raleigh , NC , USA
| | - Kymberly M Gowdy
- g Department of Pharmacology and Toxicology , Brody School of Medicine, East Carolina University , Greenville , NC , USA
| | - David Skaar
- f Department of Biological Sciences , North Carolina State University , Raleigh , NC , USA
| | - Michael Cowley
- f Department of Biological Sciences , North Carolina State University , Raleigh , NC , USA
| | - Rachel L Maguire
- f Department of Biological Sciences , North Carolina State University , Raleigh , NC , USA
| | - Bernard Fuemmeler
- h Department of Health Behavior and Policy , Virginia Commonwealth University , Richmond , VA , USA
| | - David Collier
- i Department of Pediatrics , Brody School of Medicine, East Carolina University , Greenville , NC , USA
| | - Caroline L Relton
- d Medical Research Integrative Epidemiology Unit , Bristol Medical School, Population Health Sciences, University of Bristol , Bristol , UK
| | - Susan K Murphy
- j Division of Reproductive Sciences, Department of Obstetrics and Gynecology , Duke University School of Medicine , Durham , NC , USA
| | - Cathrine Hoyo
- b Center of Human Health and the Environment , North Carolina State University , Raleigh , USA
- f Department of Biological Sciences , North Carolina State University , Raleigh , NC , USA
| |
Collapse
|
43
|
Long-term behavioural effects of maternal obesity in C57BL/6J mice. Physiol Behav 2019; 199:306-313. [DOI: 10.1016/j.physbeh.2018.11.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 11/04/2018] [Accepted: 11/04/2018] [Indexed: 12/22/2022]
|
44
|
Mattson MP. An Evolutionary Perspective on Why Food Overconsumption Impairs Cognition. Trends Cogn Sci 2019; 23:200-212. [PMID: 30670325 DOI: 10.1016/j.tics.2019.01.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 11/02/2018] [Accepted: 01/07/2019] [Indexed: 01/01/2023]
Abstract
Brain structures and neuronal networks that mediate spatial navigation, decision-making, sociality, and creativity evolved, in part, to enable success in food acquisition. Here, I discuss evidence suggesting that the reason that overconsumption of energy-rich foods negatively impacts cognition is that signaling pathways that evolved to respond adaptively to food scarcity are relatively disengaged in the setting of continuous food availability. Obesity impairs cognition and increases the risk for some psychiatric disorders and dementias. Moreover, maternal and paternal obesity predispose offspring to poor cognitive outcomes by epigenetic molecular mechanisms. Neural signaling pathways that evolved to bolster cognition in settings of food insecurity can be stimulated by intermittent fasting and exercise to support the cognitive health of current and future generations.
Collapse
Affiliation(s)
- Mark P Mattson
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Laboratory of Neurosciences, National Institute on Aging Intramural Research Program, Baltimore, MD 21224, USA.
| |
Collapse
|
45
|
Zhou L, Kang L, Xiao X, Jia L, Zhang Q, Deng M. "Gut Microbiota-Circadian Clock Axis" in Deciphering the Mechanism Linking Early-Life Nutritional Environment and Abnormal Glucose Metabolism. Int J Endocrinol 2019; 2019:5893028. [PMID: 31534453 PMCID: PMC6732598 DOI: 10.1155/2019/5893028] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/11/2019] [Accepted: 07/16/2019] [Indexed: 02/07/2023] Open
Abstract
The prevalence of diabetes mellitus (DM) has been increasing dramatically worldwide, but the pathogenesis is still unknown. A growing amount of evidence suggests that an abnormal developmental environment in early life increases the risk of developing metabolic diseases in adult life, which is referred to as the "metabolic memory" and the Developmental Origins of Health and Disease (DOHaD) hypothesis. The mechanism of "metabolic memory" has become a hot topic in the field of DM worldwide and could be a key to understanding the pathogenesis of DM. In recent years, several large cohort studies have shown that shift workers have a higher risk of developing type 2 diabetes mellitus (T2DM) and worse control of blood glucose levels. Furthermore, a maternal high-fat diet could lead to metabolic disorders and abnormal expression of clock genes and clock-controlled genes in offspring. Thus, disorders of circadian rhythm might play a pivotal role in glucose metabolic disturbances, especially in terms of early adverse nutritional environments and the development of metabolic diseases in later life. In addition, as a peripheral clock, the gut microbiota has its own circadian rhythm that fluctuates with periodic feeding and has been widely recognized for its significant role in metabolism. In light of the important roles of the gut microbiota and circadian clock in metabolic health and their interconnected regulatory relationship, we propose that the "gut microbiota-circadian clock axis" might be a novel and crucial mechanism to decipher "metabolic memory." The "gut microbiota-circadian clock axis" is expected to facilitate the future development of a novel target for the prevention and intervention of diabetes during the early stage of life.
Collapse
Affiliation(s)
- Liyuan Zhou
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lin Kang
- Department of Endocrinology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Xinhua Xiao
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Lijing Jia
- Department of Endocrinology, The Second Clinical Medical College of Jinan University, Shenzhen People's Hospital, Shenzhen 518020, China
| | - Qian Zhang
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Mingqun Deng
- Key Laboratory of Endocrinology, Translational Medicine Center, Ministry of Health, Department of Endocrinology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
46
|
Wankhade UD, Zhong Y, Kang P, Alfaro M, Chintapalli SV, Piccolo BD, Mercer KE, Andres A, Thakali KM, Shankar K. Maternal High-Fat Diet Programs Offspring Liver Steatosis in a Sexually Dimorphic Manner in Association with Changes in Gut Microbial Ecology in Mice. Sci Rep 2018; 8:16502. [PMID: 30405201 PMCID: PMC6220325 DOI: 10.1038/s41598-018-34453-0] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022] Open
Abstract
The contributions of maternal diet and obesity in shaping offspring microbiome remain unclear. Here we employed a mouse model of maternal diet-induced obesity via high-fat diet feeding (HFD, 45% fat calories) for 12 wk prior to conception on offspring gut microbial ecology. Male and female offspring were provided access to control or HFD from weaning until 17 wk of age. Maternal HFD-associated programming was sexually dimorphic, with male offspring from HFD dams showing hyper-responsive weight gain to postnatal HFD. Likewise, microbiome analysis of offspring cecal contents showed differences in α-diversity, β-diversity and higher Firmicutes in male compared to female mice. Weight gain in offspring was significantly associated with abundance of Lachnospiraceae and Clostridiaceae families and Adlercreutzia, Coprococcus and Lactococcus genera. Sex differences in metagenomic pathways relating to lipid metabolism, bile acid biosynthesis and immune response were also observed. HFD-fed male offspring from HFD dams also showed worse hepatic pathology, increased pro-inflammatory cytokines, altered expression of bile acid regulators (Cyp7a1, Cyp8b1 and Cyp39a1) and serum bile acid concentrations. These findings suggest that maternal HFD alters gut microbiota composition and weight gain of offspring in a sexually dimorphic manner, coincident with fatty liver and a pro-inflammatory state in male offspring.
Collapse
Affiliation(s)
- Umesh D Wankhade
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ying Zhong
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Ping Kang
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Maria Alfaro
- Molecular Genetic Pathology Laboratory, Arkansas Children's Hospital, Little Rock, Arkansas, USA
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Brian D Piccolo
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kelly E Mercer
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Aline Andres
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Keshari M Thakali
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA. .,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA.
| |
Collapse
|
47
|
House JS, Mendez M, Maguire RL, Gonzalez-Nahm S, Huang Z, Daniels J, Murphy SK, Fuemmeler BF, Wright FA, Hoyo C. Periconceptional Maternal Mediterranean Diet Is Associated With Favorable Offspring Behaviors and Altered CpG Methylation of Imprinted Genes. Front Cell Dev Biol 2018; 6:107. [PMID: 30246009 PMCID: PMC6137242 DOI: 10.3389/fcell.2018.00107] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 08/20/2018] [Indexed: 12/28/2022] Open
Abstract
Background: Maternal diet during pregnancy has been shown to influence the child neuro-developmental outcomes. Studies examining effects of dietary patterns on offspring behavior are sparse. Objective: Determine if maternal adherence to a Mediterranean diet is associated with child behavioral outcomes assessed early in life, and to evaluate the role of differentially methylated regions (DMRs) regulating genomically imprinted genes in these associations. Methods: Among 325 mother/infant pairs, we used regression models to evaluate the association between tertiles of maternal periconceptional Mediterranean diet adherence (MDA) scores derived from a Food Frequency Questionnaire, and social and emotional scores derived from the Infant Toddler Social and Emotional Assessment (ITSEA) questionnaire in the second year of life. Methylation of nine genomically imprinted genes was measured to determine if MDA was associated with CpG methylation. Results: Child depression was inversely associated with maternal MDA (Bonferroni-corrected p = 0.041). While controlling for false-discovery, compared to offspring of women with the lowest MDA tertile, those with MDA scores in middle and high MDA tertiles had decreased odds for atypical behaviors [OR (95% CI) = 0.40 (0.20, 0.78) for middle and 0.40 (0.17, 0.92) for highest tertile], for maladaptive behaviors [0.37 (0.18, 0.72) for middle tertile and 0.42 (0.18, 0.95) for highest tertile] and for an index of autism spectrum disorder behaviors [0.46 (0.23, 0.90) for middle and 0.35 (0.15, 0.80) for highest tertile]. Offspring of women with the highest MDA tertile were less likely to exhibit depressive [OR = 0.28 (0.12, 0.64)] and anxiety [0.42 (0.18, 0.97)] behaviors and increased odds of social relatedness [2.31 (1.04, 5.19)] behaviors when compared to low MDA mothers. Some associations varied by sex. Perinatal MDA score was associated with methylation differences for imprinted control regions of PEG10/SGCE [females: Beta (95% CI) = 1.66 (0.52, 2.80) - Bonferroni-corrected p = 0.048; males: -0.56 (-1.13, -0.00)], as well as both MEG3 and IGF2 in males [0.97 (0.00, 1.94)] and -0.92 (-1.65, -0.19) respectively. Conclusion: In this ethnically diverse cohort, maternal adherence to a Mediterranean diet in early pregnancy was associated with favorable neurobehavioral outcomes in early childhood and with sex-dependent methylation differences of MEG3, IGF2, and SGCE/PEG10 DMRs.
Collapse
Affiliation(s)
- John S House
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States.,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States
| | - Michelle Mendez
- Department of Environmental and Occupational Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Rachel L Maguire
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| | - Sarah Gonzalez-Nahm
- Department of Health, Behavior and Society, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Zhiqing Huang
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, United States
| | - Julie Daniels
- Department of Epidemiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Susan K Murphy
- Department of Obstetrics and Gynecology, Duke University Medical Center, Durham, NC, United States
| | - Bernard F Fuemmeler
- Department of Health Behavior and Policy, Virginia Commonwealth University, Richmond, VA, United States
| | - Fred A Wright
- Bioinformatics Research Center, North Carolina State University, Raleigh, NC, United States.,Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States.,Department of Statistics, North Carolina State University, Raleigh, NC, United States
| | - Cathrine Hoyo
- Center for Human Health and the Environment, North Carolina State University, Raleigh, NC, United States.,Department of Biological Sciences, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
48
|
Rackers HS, Thomas S, Williamson K, Posey R, Kimmel MC. Emerging literature in the Microbiota-Brain Axis and Perinatal Mood and Anxiety Disorders. Psychoneuroendocrinology 2018; 95:86-96. [PMID: 29807325 PMCID: PMC6348074 DOI: 10.1016/j.psyneuen.2018.05.020] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 05/15/2018] [Accepted: 05/15/2018] [Indexed: 12/15/2022]
Abstract
Perinatal Mood and Anxiety Disorders (PMAD) are common and can cause significant morbidity and mortality for mother and child. A healthy perinatal period requires significant adaptations; however, systems can become imbalanced resulting in depressive and anxiety symptoms. The interface between the microbiome, the immune system, and the stress system may be a model for understanding mechanisms underlying PMAD. Emerging literature from general populations regarding immune, hormone, and HPA axis changes in relation to the microbiome combined with literature on immune, gonadotropin, and stress systems in the perinatal period provides a background. We systematically investigated literature in the developing field of the microbiome in relation to PMAD. Our inclusion criteria were 1) reporting measure of maternal mood, stress, or anxious or depressed behavior; 2) in the perinatal period, defined as pregnancy through one year postpartum; and 3) reporting measure of maternal microbiome including manipulations of the microbiome through prebiotics, probiotics, or interventions with microbial byproducts. The review identified research studying associations between stress and maternal microbiome; dietary impacts on microbial composition, mood, and stress; and the relationship between the microbiome and the immune system through immunoregulatory mechanisms. Important themes identified include: the importance of studying the maternal microbiome and measures of stress, anxiety, and depression and that multi-hit models will be needed as research strives to determine the effects of multiple mechanisms working in concert.
Collapse
Affiliation(s)
- Hannah S Rackers
- Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, 304 MacNider Hall, CB #7160, Chapel Hill, NC, 27599-7160, United States.
| | - Stephanie Thomas
- Department of Nutrition, Gillings School of Global Public Health, University of North Carolina at Chapel Hill, 245 Rosenau Hall, CB # 7461, Chapel Hill, NC, 27599-7461, United States.
| | - Kelsey Williamson
- University of North Carolina at Chapel Hill School School of Medicine, Chapel Hill, NC, 27599, United States.
| | - Rachael Posey
- Health Sciences Library, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, United States.
| | - Mary C Kimmel
- Department of Psychiatry, University of North Carolina at Chapel Hill School of Medicine, 304 MacNider Hall, CB #7160, Chapel Hill, NC, 27599-7160, United States.
| |
Collapse
|
49
|
The role of gut microbiota in the effects of maternal obesity during pregnancy on offspring metabolism. Biosci Rep 2018; 38:BSR20171234. [PMID: 29208770 PMCID: PMC5897743 DOI: 10.1042/bsr20171234] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 11/28/2017] [Accepted: 12/05/2017] [Indexed: 02/07/2023] Open
Abstract
Obesity is considered a global epidemic. Specifically, obesity during pregnancy programs an increased risk of the offspring developing metabolic disorders in addition to the adverse effects on the mother per se Large numbers of human and animal studies have demonstrated that the gut microbiota plays a pivotal role in obesity and metabolic diseases. Similarly, maternal obesity during pregnancy is associated with alterations in the composition and diversity of the intestine microbial community. Recently, the microbiota in the placenta, amniotic fluid, and meconium in healthy gestations has been investigated, and the results supported the "in utero colonization hypothesis" and challenged the traditional "sterile womb" that has been acknowledged worldwide for more than a century. Thus, the offspring microbiota, which is crucial for the immune and metabolic function and further health in the offspring, might be established prior to birth. As a detrimental intrauterine environment, maternal obesity influences the microbial colonization and increases the risk of metabolic diseases in offspring. This review discusses the role of the microbiota in the impact of maternal obesity during pregnancy on offspring metabolism and further analyzes related probiotic or prebiotic interventions to prevent and treat obesity and metabolic diseases.
Collapse
|
50
|
Stiemsma LT, Michels KB. The Role of the Microbiome in the Developmental Origins of Health and Disease. Pediatrics 2018; 141:e20172437. [PMID: 29519955 PMCID: PMC5869344 DOI: 10.1542/peds.2017-2437] [Citation(s) in RCA: 214] [Impact Index Per Article: 35.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/29/2017] [Indexed: 12/15/2022] Open
Abstract
Although the prominent role of the microbiome in human health has been established, the early-life microbiome is now being recognized as a major influence on long-term human health and development. Variations in the composition and functional potential of the early-life microbiome are the result of lifestyle factors, such as mode of birth, breastfeeding, diet, and antibiotic usage. In addition, variations in the composition of the early-life microbiome have been associated with specific disease outcomes, such as asthma, obesity, and neurodevelopmental disorders. This points toward this bacterial consortium as a mediator between early lifestyle factors and health and disease. In addition, variations in the microbial intrauterine environment may predispose neonates to specific health outcomes later in life. A role of the microbiome in the Developmental Origins of Health and Disease is supported in this collective research. Highlighting the early-life critical window of susceptibility associated with microbiome development, we discuss infant microbial colonization, beginning with the maternal-to-fetal exchange of microbes in utero and up through the influence of breastfeeding in the first year of life. In addition, we review the available disease-specific evidence pointing toward the microbiome as a mechanistic mediator in the Developmental Origins of Health and Disease.
Collapse
Affiliation(s)
- Leah T Stiemsma
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California
| | - Karin B Michels
- Department of Epidemiology, Fielding School of Public Health, University of California, Los Angeles, Los Angeles, California
| |
Collapse
|