1
|
Sentürk NB, Kasapoglu B, Sahin E, Ozcan O, Ozansoy M, Ozansoy MB, Siyah P, Sezerman U, Sahin F. The Potential Role of Boron in the Modulation of Gut Microbiota Composition: An In Vivo Pilot Study. Pharmaceuticals (Basel) 2024; 17:1334. [PMID: 39458975 PMCID: PMC11510266 DOI: 10.3390/ph17101334] [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: 07/09/2024] [Revised: 09/13/2024] [Accepted: 09/24/2024] [Indexed: 10/28/2024] Open
Abstract
Background/Objectives: The role of the gut microbiome in the development and progression of many diseases has received increased attention in recent years. Boron, a trace mineral found in dietary sources, has attracted interest due to its unique electron depletion and coordination characteristics in chemistry, as well as its potential role in modulating the gut microbiota. This study investigates the effects of inorganic boron derivatives on the gut microbiota of mice. Methods: For three weeks, boric acid (BA), sodium pentaborate pentahydrate (NaB), and sodium perborate tetrahydrate (SPT) were dissolved (200 mg/kg each) in drinking water and administered to wild-type BALB/c mice. The composition of the gut microbiota was analyzed to determine the impact of these treatments. Results: The administration of BA significantly altered the composition of the gut microbiota, resulting in a rise in advantageous species such as Barnesiella and Alistipes. Additionally, there was a decrease in some taxa associated with inflammation and illness, such as Clostridium XIVb and Bilophila. Notable increases in genera like Treponema and Catellicoccus were observed, suggesting the potential of boron compounds to enrich microbial communities with unique metabolic functions. Conclusions: These findings indicate that boron compounds may have the potential to influence gut microbiota composition positively, offering potential prebiotic effects. Further research with additional analyses is necessary to fully understand the interaction between boron and microbiota and to explore the possibility of their use as prebiotic agents in clinical settings.
Collapse
Affiliation(s)
- Nermin Basak Sentürk
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, 34755 Istanbul, Turkey; (N.B.S.); (B.K.)
| | - Burcu Kasapoglu
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, 34755 Istanbul, Turkey; (N.B.S.); (B.K.)
- Abdi Ibrahim Pharmaceuticals, Biotechnological Products Production Facility (AbdiBio), 34538 Istanbul, Turkey
| | - Eray Sahin
- Biostatistics and Bioinformatics PhD Program, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
| | | | - Mehmet Ozansoy
- Department of Physiology, International School of Medicine, Istanbul Medipol University, 34810 Istanbul, Turkey;
| | - Muzaffer Beyza Ozansoy
- Department of Physical Therapy and Rehabilitation, Faculty of Health Sciences, Fenerbahçe University, 34758 Istanbul, Turkey;
| | - Pinar Siyah
- Department of Biochemistry, School of Pharmacy, Bahçeşehir University, 34353 Istanbul, Turkey;
| | - Ugur Sezerman
- Biostatistics and Bioinformatics PhD Program, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey;
- Department of Biostatistics and Medical Informatics, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, 34752 Istanbul, Turkey
| | - Fikrettin Sahin
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, 34755 Istanbul, Turkey; (N.B.S.); (B.K.)
| |
Collapse
|
2
|
Mabrok HB, Ramadan AA, Hamed IM, Mohamed DA. Obesity as Inducer of Cognitive Function Decline via Dysbiosis of Gut Microbiota in Rats. Brain Sci 2024; 14:807. [PMID: 39199499 PMCID: PMC11353248 DOI: 10.3390/brainsci14080807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/31/2024] [Accepted: 08/09/2024] [Indexed: 09/01/2024] Open
Abstract
Diet-induced obesity is a global phenomenon that affects the population worldwide with manifestations at both the phenotypic and genotypic levels. Cognitive function decline is a major global health challenge. The relation between obesity and cognitive function is a debatable issue. The main goal of the current research was to study the implications of obesity on cognitive function and gut microbiota diversity and its impact on plasma and brain metabolic parameters in rats. Obesity was induced in rats by feeding on a high-fat (HF) or a high-fat/high-sucrose (HFHS) diet. The results reveal that both the HF (0.683) and HFHS (0.688) diets were effective as obesity inducers, which was confirmed by a significant increase in the body mass index (BMI). Both diet groups showed dyslipidemia and elevation of oxidative stress, insulin resistance (IR), and inflammatory markers with alterations in liver and kidney functions. Obesity led to a reduction in cognitive function through a reduction in short-term memory by 23.8% and 30.7% in the rats fed HF and HFHS diets, respectively, and learning capacity and visuo-spatial memory reduced by 8.9 and 9.7 s in the rats fed an HF or HFHS diet, respectively. Bacteroidetes, Firmicutes, Proteobacteria, Fusobacteria, and Spirochaetes phyla were detected. The Firmicutes/Bacteroidetes ratio (F/B) significantly decreased in the HF group, while it increased in the HFHS group compared to the normal control. The two species, Bacteroides acidifaciens and Bacteroides ovatus, which are associated with IR, were drastically compromised by the high-fat/high-sucrose diet. Some species that have been linked to reduced inflammation showed a sharp decrease in the HFHS group, while Prevotella copri, which is linked to carbohydrate metabolism, was highly enriched. In conclusion: Obesity led to cognitive impairment through changes in short-term and visuo-spatial memory. A metagenomic analysis revealed alterations in the abundance of some microbial taxa associated with obesity, inflammation, and insulin resistance in the HF and HFHS groups.
Collapse
Grants
- a626035bfd925943, 4c6c6a0dc9645904, 175e6bf937114ef5, 18dca4e8f29e587c, aaf09103eb8bd6ee, 3740a1d4a23d772f, 1b07773fd3c8c954, 4f8fa1a570a3a4b7, 490e7e4e51713e71, 1e87a07edec11a96, 7642f29d62c1068b, c06bc3bf279a8491, c78b30a55528e880, e160d996ffb69ed4, 133 Discount Vouchers
Collapse
Affiliation(s)
| | | | | | - Doha A. Mohamed
- Nutrition and Food Science Department, Food Industries and Nutrition Institute, National Research Centre, Dokki, Cairo 12622, Egypt; (H.B.M.); (A.A.R.); (I.M.H.)
| |
Collapse
|
3
|
Ayten Ş, Bilici S. Modulation of Gut Microbiota Through Dietary Intervention in Neuroinflammation and Alzheimer's and Parkinson's Diseases. Curr Nutr Rep 2024; 13:82-96. [PMID: 38652236 PMCID: PMC11133127 DOI: 10.1007/s13668-024-00539-7] [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] [Accepted: 04/02/2024] [Indexed: 04/25/2024]
Abstract
PURPOSE OF REVIEW The gut microbiota plays a crucial role in the pathogenesis of neuroinflammation and Alzheimer's and Parkinson's diseases. One of the main modulators of the gut microbiota is the diet, which directly influences host homeostasis and biological processes. Some dietary patterns can affect neurodegenerative diseases' progression through gut microbiota composition, gut permeability, and the synthesis and secretion of microbial-derived neurotrophic factors and neurotransmitters. This comprehensive review critically assesses existing studies investigating the impact of dietary interventions on the modulation of the microbiota in relation to neurodegenerative diseases and neuroinflammation. RECENT FINDINGS There are limited studies on the effects of specific diets, such as the ketogenic diet, Mediterranean diet, vegetarian diet, and Western diet, on the progression of neuroinflammation and Alzheimer's and Parkinson's diseases through the gut-brain axis. The ketogenic diet displays promising potential in ameliorating the clinical trajectory of mild cognitive impairment and Alzheimer's disease. However, conflicting outcomes were observed among various studies, highlighting the need to consider diverse types of ketogenic diets and their respective effects on clinical outcomes and gut microbiota composition. Vegetarian and Mediterranean diets, known for their anti-inflammatory properties, can be effective against Parkinson's disease, which is related to inflammation in the gut environment. On the other hand, the westernization of dietary patterns was associated with reduced gut microbial diversity and metabolites, which ultimately contributed to the development of neuroinflammation and cognitive impairment. Various studies examining the impact of dietary interventions on the gut-brain axis with regard to neuroinflammation and Alzheimer's and Parkinson's diseases are thoroughly reviewed in this article. A strong mechanistic explanation is required to fully understand the complex interactions between various dietary patterns, gut microbiota, and microbial metabolites and the effects these interactions have on cognitive function and the progression of these diseases.
Collapse
Affiliation(s)
- Şerife Ayten
- Department of Nutrition and Dietetics, Gazi University, Ankara, Turkey.
| | - Saniye Bilici
- Department of Nutrition and Dietetics, Gazi University, Ankara, Turkey
| |
Collapse
|
4
|
Zhao H, Sun M, Zhang Y, Kong W, Fan L, Wang K, Xu Q, Chen B, Dong J, Shi Y, Wang Z, Wang S, Zhuang X, Li Q, Lin F, Yao X, Zhang W, Kong C, Zhang R, Feng D, Zhao X. Connecting the Dots: The Cerebral Lymphatic System as a Bridge Between the Central Nervous System and Peripheral System in Health and Disease. Aging Dis 2024; 15:115-152. [PMID: 37307828 PMCID: PMC10796102 DOI: 10.14336/ad.2023.0516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 05/16/2023] [Indexed: 06/14/2023] Open
Abstract
As a recently discovered waste removal system in the brain, cerebral lymphatic system is thought to play an important role in regulating the homeostasis of the central nervous system. Currently, more and more attention is being focused on the cerebral lymphatic system. Further understanding of the structural and functional characteristics of cerebral lymphatic system is essential to better understand the pathogenesis of diseases and to explore therapeutic approaches. In this review, we summarize the structural components and functional characteristics of cerebral lymphatic system. More importantly, it is closely associated with peripheral system diseases in the gastrointestinal tract, liver, and kidney. However, there is still a gap in the study of the cerebral lymphatic system. However, we believe that it is a critical mediator of the interactions between the central nervous system and the peripheral system.
Collapse
Affiliation(s)
- Hongxiang Zhao
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Meiyan Sun
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Yue Zhang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Wenwen Kong
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Lulu Fan
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Kaifang Wang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Qing Xu
- Department of Anesthesiology, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, China.
| | - Baiyan Chen
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Jianxin Dong
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Yanan Shi
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Zhengyan Wang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - ShiQi Wang
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Xiaoli Zhuang
- Department of Anesthesiology, Beijing Anzhen Hospital, Capital Medical University, Beijing, China.
| | - Qi Li
- Department of Anesthesiology, Ninth People's Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China.
| | - Feihong Lin
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| | - Xinyu Yao
- Department of Anesthesiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - WenBo Zhang
- Department of Neurosurgery, The Children’s Hospital of Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China.
| | - Chang Kong
- Department of Anesthesiology and Critical Care Medicine, Tianjin Nankai Hospital, Tianjin Medical University, Tianjin, China.
| | - Rui Zhang
- Department of Anesthesiology, Affiliated Hospital of Weifang Medical University, Weifang, China.
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| | - Dayun Feng
- Department of neurosurgery, Tangdu hospital, Fourth Military Medical University, Xi'an, China.
| | - Xiaoyong Zhao
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
- Department of Anesthesiology, Affiliated Hospital of Weifang Medical University, Weifang, China.
- Shandong Provincial Medicine and Health Key Laboratory of Clinical Anesthesia, School of Anesthesiology, Weifang Medical University, Weifang, China.
| |
Collapse
|
5
|
Stapleton S, Welch G, DiBerardo L, Freeman LR. Sex differences in a mouse model of diet-induced obesity: the role of the gut microbiome. Biol Sex Differ 2024; 15:5. [PMID: 38200579 PMCID: PMC10782710 DOI: 10.1186/s13293-023-00580-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Recent decades have seen an exponential rise in global obesity prevalence, with rates nearly doubling in a span of 40 years. A comprehensive knowledge base regarding the systemic effects of obesity is required to create new preventative and therapeutic agents effective at combating the current obesity epidemic. Previous studies of diet-induced obesity utilizing mouse models have demonstrated a difference in bodyweight gain by sex. In such studies, female mice gained significantly less weight than male mice when given the same high fat (HF) diet, indicating a resistance to diet-induced obesity. Research has also shown sex differences in gut microbiome composition between males and females, indicated to be in part a result of sex hormones. Understanding metabolic differences between sexes could assist in the development of new measures for obesity prevention and treatment. This study aimed to characterize sex differences in weight gain, plasma lipid profiles, fecal microbiota composition, and fecal short chain fatty acid levels. We hypothesized a role for the gut microbiome in these sex differences that would be normalized following microbiome depletion. METHODS A mouse model was used to study these effects. Mice were divided into treatment groups by sex, diet, and presence/absence of an antibiotic cocktail to deplete genera in the gut microbiome. We hypothesized that sex differences would be present both in bodyweight gain and systemic measures of obesity, including hormone and circulating free fatty acid levels. RESULTS We determined statistically significant differences for sex and/or treatment for the outcome measures. We confirm previous findings in which male mice gained significantly more weight than female mice fed the same high fat diet. However, sex differences persisted following antibiotic administration for microbiome depletion. CONCLUSIONS We conclude that sex differences in the gut microbiome may contribute to sex differences in obesity, but they do not explain all of the differences.
Collapse
Affiliation(s)
| | - Grace Welch
- Department of Biology, Furman University, Greenville, SC, USA
| | | | - Linnea R Freeman
- Department of Biology, Furman University, Greenville, SC, USA.
- Neurosciences, Furman University, Greenville, SC, USA.
| |
Collapse
|
6
|
Stapleton S, Welch G, DiBerardo L, Freeman LR. Sex differences in a mouse model of diet-induced obesity: the role of the gut microbiome. RESEARCH SQUARE 2023:rs.3.rs-3496738. [PMID: 37961721 PMCID: PMC10635401 DOI: 10.21203/rs.3.rs-3496738/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Recent decades have seen an exponential rise in global obesity prevalence, with rates nearly doubling in a span of forty years. A comprehensive knowledge base regarding the systemic effects of obesity is required to create new preventative and therapeutic agents effective at combating the current obesity epidemic. Previous studies of diet-induced obesity utilizing mouse models have demonstrated a difference in bodyweight gain by sex. In such studies, female mice gained significantly less weight than male mice when given the same high fat (HF) diet, indicating a resistance to diet-induced obesity. Research has also shown sex differences in gut microbiome composition between males and females, indicated to be in part a result of sex hormones. Understanding metabolic differences between sexes could assist in the development of new measures for obesity prevention and treatment. This study aimed to characterize sex differences in weight gain, plasma lipid profiles, fecal microbiota composition, and fecal short chain fatty acid levels. We hypothesized a role for the gut microbiome in these sex differences that would be normalized following microbiome depletion. Methods A mouse model was used to study these effects. Mice were divided into treatment groups by sex, diet, and presence/absence of an antibiotic cocktail to deplete genera in the gut microbiome. We hypothesized that sex differences would be present both in bodyweight gain and systemic measures of obesity, including hormone and circulating free fatty acid levels. Results We determined statistically significant differences for sex and/or treatment for the outcome measures. We confirm previous findings in which male mice gained significantly more weight than female mice fed the same high fat diet. However, sex differences persisted following antibiotic administration for microbiome depletion. Conclusions We conclude that sex differences in the gut microbiome may contribute to sex differences in obesity, but they do not explain all of the differences.
Collapse
|
7
|
Effects of High-Fat and High-Fat High-Sugar Diets in the Anxiety, Learning and Memory, and in the Hippocampus Neurogenesis and Neuroinflammation of Aged Rats. Nutrients 2023; 15:nu15061370. [PMID: 36986100 PMCID: PMC10053405 DOI: 10.3390/nu15061370] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Revised: 03/09/2023] [Accepted: 03/09/2023] [Indexed: 03/14/2023] Open
Abstract
High-caloric diets induce several deleterious alterations in the human body, including the brain. However, information on the effects of these diets on the elderly brain is scarce. Therefore, we studied the effects of 2 months of treatment with high-fat (HF) and high-fat-high-sugar (HFHS) diets on aged male Wistar rats at 18 months. Anxiety levels were analyzed using the open-field and plus-maze tests, while learning and memory processes were analyzed using the Morris water maze test. We also analyzed neurogenesis using doublecortin (DCX) and neuroinflammation using glial fibrillary acidic protein (GFAP). In aged rats, the HFHS diet impaired spatial learning, memory, and working memory and increased anxiety levels, associated with a reduction in the number of DCX cells and an increase in GFAP cells in the hippocampus. In contrast, the effects of the HF diet were lighter, impairing spatial memory and working memory, and associated with a reduction in DCX cells in the hippocampus. Thus, our results suggest that aged rats are highly susceptible to high-caloric diets, even if they only started in the elderly, with an impact on cognition and emotions. Furthermore, diets rich in saturated fats and sugar are more detrimental to aged rats than high-fat diets are.
Collapse
|
8
|
Asbjornsdottir B, Miranda-Ribera A, Fiorentino M, Konno T, Cetinbas M, Lan J, Sadreyev RI, Gudmundsson LS, Gottfredsson M, Lauth B, Birgisdottir BE, Fasano A. Prophylactic Effect of Bovine Colostrum on Intestinal Microbiota and Behavior in Wild-Type and Zonulin Transgenic Mice. Biomedicines 2022; 11:biomedicines11010091. [PMID: 36672598 PMCID: PMC9855927 DOI: 10.3390/biomedicines11010091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/22/2022] [Accepted: 12/26/2022] [Indexed: 12/31/2022] Open
Abstract
The microbiota-gut-brain axis (MGBA) involves bidirectional communication between intestinal microbiota and the gastrointestinal (GI) tract, central nervous system (CNS), neuroendocrine/neuroimmune systems, hypothalamic-pituitary-adrenal (HPA) axis, and enteric nervous system (ENS). The intestinal microbiota can influence host physiology and pathology. Dysbiosis involves the loss of beneficial microbial input or signal, diversity, and expansion of pathobionts, which can lead to loss of barrier function and increased intestinal permeability (IP). Colostrum, the first milk from mammals after birth, is a natural source of nutrients and is rich in oligosaccharides, immunoglobulins, growth factors, and anti-microbial components. The aim of this study was to investigate if bovine colostrum (BC) administration might modulate intestinal microbiota and, in turn, behavior in two mouse models, wild-type (WT) and Zonulin transgenic (Ztm)-the latter of which is characterized by dysbiotic microbiota, increased intestinal permeability, and mild hyperactivity-and to compare with control mice. Bioinformatics analysis of the microbiome showed that consumption of BC was associated with increased taxonomy abundance (p = 0.001) and diversity (p = 0.004) of potentially beneficial species in WT mice and shifted dysbiotic microbial community towards eubiosis in Ztm mice (p = 0.001). BC induced an anxiolytic effect in WT female mice compared with WT female control mice (p = 0.0003), and it reduced anxiogenic behavior in Ztm female mice compared with WT female control mice (p = 0.001), as well as in Ztm male mice compared with WT BC male mice (p = 0.03). As evidenced in MGBA interactions, BC supplementation may well be applied for prophylactic approaches in the future. Further research is needed to explore human interdependencies between intestinal microbiota, including eubiosis and pathobionts, and neuroinflammation, and the potential value of BC for human use. The MGH Institutional Animal Care and Use Committee authorized the animal study (2013N000013).
Collapse
Affiliation(s)
- Birna Asbjornsdottir
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
- School of Health Sciences, Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
- Unit for Nutrition Research, Landspitali University Hospital and Faculty of Food Science and Nutrition, University of Iceland, 101 Reykjavik, Iceland
- Correspondence:
| | - Alba Miranda-Ribera
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
| | - Maria Fiorentino
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
| | - Takumi Konno
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
| | - Murat Cetinbas
- Department of Molecular Biology and Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jinggang Lan
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
| | - Ruslan I. Sadreyev
- Department of Molecular Biology and Pathology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Larus S. Gudmundsson
- School of Health Sciences, Faculty of Pharmaceutical Sciences, University of Iceland, 101 Reykjavik, Iceland
| | - Magnus Gottfredsson
- School of Health Sciences, Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
- Department of Scientific Affairs, Landspitali University Hospital, 101 Reykjavik, Iceland
- Department of Infectious Diseases, Landspitali University Hospital, 101 Reykjavik, Iceland
| | - Bertrand Lauth
- School of Health Sciences, Faculty of Medicine, University of Iceland, 101 Reykjavik, Iceland
- Department of Child and Adolescent Psychiatry, Landspitali University Hospital, 105 Reykjavik, Iceland
| | - Bryndis Eva Birgisdottir
- Unit for Nutrition Research, Landspitali University Hospital and Faculty of Food Science and Nutrition, University of Iceland, 101 Reykjavik, Iceland
| | - Alessio Fasano
- Department of Pediatric Gastroenterology and Nutrition, Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02152, USA
- Department of Pediatrics, Harvard Medical School, Harvard University, Boston, MA 02114, USA
| |
Collapse
|
9
|
Kumar S, Raj VS, Ahmad A, Saini V. Amoxicillin modulates gut microbiota to improve short-term high-fat diet induced pathophysiology in mice. Gut Pathog 2022; 14:40. [PMID: 36229889 PMCID: PMC9563906 DOI: 10.1186/s13099-022-00513-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/02/2022] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND A high-fat diet (HFD) induced perturbation of gut microbiota is a major contributory factor to promote the pathophysiology of HFD-associated metabolic syndrome. The HFD could also increase the susceptibility to the microbial infections warranting the use of antibiotics which are independently capable of impacting both gut microbiota and metabolic syndrome. Further, the usage of antibiotics in individuals consuming HFD can impact mitochondrial function that can be associated with an elevated risk of chronic conditions like inflammatory bowel disease (IBD). Despite this high propensity to infections in individuals on HFD, the link between duration of HFD and antibiotic treatment, and its impact on diversity of the gut microbiome and features of metabolic syndrome is not well established. In this study, we have addressed these knowledge gaps by examining how the gut microbiota profile changes in HFD-fed mice receiving antibiotic intervention in the form of amoxicillin. We also determine whether antibiotic treatment in HFD-fed mice may adversely impact the ability of immune cells to clear microbial infections. METHODS AND RESULTS We have subjected mice to HFD and chow diet (CD) for 3 weeks, and a subset of these mice on both diets received antibiotic intervention in the form of amoxicillin in the 3rd week. Body weight and food intake were recorded for 3 weeks. After 21 days, all animals were weighted and sacrificed. Subsequently, these animals were evaluated for basic haemato-biochemical and histopathological attributes. We used 16S rRNA sequencing followed by bioinformatics analysis to determine changes in gut microbiota in these mice. We observed that a HFD, even for a short-duration, could successfully induce the partial pathophysiology typical of a metabolic syndrome, and substantially modulated the gut microbiota in mice. The short course of amoxicillin treatment to HFD-fed mice resulted in beneficial effects by significantly reducing fasting blood glucose and skewing the number of thrombocytes towards a normal range. Remarkably, we observed a significant remodelling of gut microbiota in amoxicillin-treated HFD-fed mice. Importantly, some gut microbes associated with improved insulin sensitivity and recovery from metabolic syndrome only appeared in amoxicillin-treated HFD-fed mice reinforcing the beneficial effects of antibiotic treatment in the HFD-associated metabolic syndrome. Moreover, we also observed the presence of gut-microbiota unique to amoxicillin-treated HFD-fed mice that are also known to improve the pathophysiology associated with metabolic syndrome. However, both CD-fed as well as HFD-fed mice receiving antibiotics showed an increase in intestinal pathogens as is typically observed for antibiotic treatment. Importantly though, infection studies with S. aureus and A. baumannii, revealed that macrophages isolated from amoxicillin-treated HFD-fed mice are comparable to those isolated from mice receiving only HFD or CD in terms of susceptibility, and progression of microbial infection. This finding clearly indicated that amoxicillin treatment does not introduce any additional deficits in the ability of macrophages to combat microbial infections. CONCLUSIONS Our results showed that amoxicillin treatment in HFD-fed mice exert a beneficial influence on the pathophysiological attributes of metabolic syndrome which correlates with a significant remodelling of gut microbiota. A novel observation was the increase in microbes known to improve insulin sensitivity following amoxicillin treatment during short-term intake of HFD. Even though there is a minor increase in gut-resistant intestinal pathogens in amoxicillin-treated groups, there is no adverse impact on macrophages with respect to their susceptibility and ability to control infections. Taken together, this study provides a proof of principle for the exploration of amoxicillin treatment as a potential therapy in the people affected with metabolic syndrome.
Collapse
Affiliation(s)
- Suresh Kumar
- National Institute of Biologicals, Ministry of Health & Family Welfare, Govt. of India, Noida, 201309, India.
| | - V Samuel Raj
- Center for Drug, Design, Discovery and Development (C4D), SRM University, Delhi-NCR, 131029, Sonepat, Haryana, India
| | - Ayaan Ahmad
- Laboratory of Infection Biology and Translational Research, Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India
| | - Vikram Saini
- Laboratory of Infection Biology and Translational Research, Department of Biotechnology, All India Institute of Medical Sciences, New Delhi, India.
- Biosafety Laboratory-3, Centralized Core Research Facility (CCRF), All India Institute of Medical Sciences (AIIMS), New Delhi, India.
| |
Collapse
|
10
|
Koblinsky ND, Power KA, Middleton L, Ferland G, Anderson ND. The Role of the Gut Microbiome in Diet and Exercise Effects on Cognition: A Review of the Intervention Literature. J Gerontol A Biol Sci Med Sci 2022; 78:195-205. [PMID: 35977540 PMCID: PMC9951060 DOI: 10.1093/gerona/glac166] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Indexed: 11/13/2022] Open
Abstract
Interest in the gut-brain axis and its implications for neurodegenerative diseases, such as Alzheimer's disease and related dementias, is growing. Microbial imbalances in the gastrointestinal tract, which are associated with impaired cognition, may represent a therapeutic target for lowering dementia risk. Multicomponent lifestyle interventions are a promising dementia risk reduction strategy and most often include diet and exercise, behaviors that are also known to modulate the gut microbiome. A better understanding of the role of the gut microbiome in diet and exercise effects on cognition may help to optimize these lifestyle interventions. The purpose of this review is to summarize findings from diet and exercise interventions that have investigated cognitive changes via effects on the microbiome. We aim to discuss the underlying mechanisms, highlight current gaps in the field, and provide new research directions. There is evidence mainly from rodent studies supporting the notion that microbiota changes mediate the effects of diet and exercise on cognition, with potential mechanisms including end-product metabolites and regulation of local and systemic inflammation. The field lacks whole diet and exercise interventions, especially those involving human participants. It is further limited by heterogeneous rodent models, outcome assessments, and the absence of proper mediation analyses. Trials including older adults with dementia risk factors, factorial designs of diet and exercise, and pre and post measures of microbiota, end-product metabolites, and inflammation would help to elucidate and potentially leverage the role of the microbiome in lowering dementia risk through lifestyle modification.
Collapse
Affiliation(s)
- Noah D Koblinsky
- Rotman Research Institute, Baycrest Health Sciences, Toronto, Ontario, Canada
| | - Krista A Power
- School of Nutrition Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Laura Middleton
- Department of Kinesiology and Health Sciences, University of Waterloo, Waterloo, Ontario, Canada
| | - Guylaine Ferland
- Montreal Heart Institute Research Centre, Montreal, Quebec, Canada,Departments of Psychology and Psychiatry, University of Toronto, Toronto, Ontario, Canada
| | - Nicole D Anderson
- Address correspondence to: Nicole D. Anderson, PhD, CPsych, Rotman Research Institute, Baycrest Health Sciences, 3560 Bathurst St., M6A 2E1 Toronto, ON, Canada. E-mail:
| |
Collapse
|
11
|
Zhang T, Wu X, Yuan H, Huang S, Park S. Mitigation of Memory Impairment with Fermented Fucoidan and λ-Carrageenan Supplementation through Modulating the Gut Microbiota and Their Metagenome Function in Hippocampal Amyloid-β Infused Rats. Cells 2022; 11:cells11152301. [PMID: 35892598 PMCID: PMC9367263 DOI: 10.3390/cells11152301] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/13/2022] [Accepted: 07/23/2022] [Indexed: 11/17/2022] Open
Abstract
Attenuating acetylcholinesterase and insulin/insulin-like growth factor-1 signaling in the hippocampus is associated with Alzheimer’s disease (AD) development. Fucoidan and carrageenan are brown and red algae, respectively, with potent antibacterial, anti-inflammatory, antioxidant and antiviral activities. This study examined how low-molecular-weight (MW) and high-MW fucoidan and λ-carrageenan would improve memory impairment in Alzheimer’s disease-induced rats caused by an infusion of toxic amyloid-β(Aβ). Fucoidan and λ-carrageenan were dissected into low-MW by Luteolibacter algae and Pseudoalteromonas carrageenovora. Rats receiving an Aβ(25–35) infusion in the CA1 region of the hippocampus were fed dextrin (AD-Con), 1% high-MW fucoidan (AD-F-H), 1% low-MW fucoidan (AD-F-L), 1% high-MW λ-carrageenan (AD-C-H), and 1% low-MW λ-carrageenan (AD-C-L) for six weeks. Rats to receive saline infusion (Normal-Con) had an AD-Con diet. The AD-F-L group showed an improved memory function, which manifested as an enhanced Y-maze spontaneous alternation test, water maze, and passive avoidance tests, similar to the Normal-Con group. AD-F-L also potentiated hippocampal insulin signaling and increased the expression of ciliary neurotrophic factor (CNTF) and brain-derived neurotrophic factor (BDNF) in the hippocampus. AD-C-L improved the memory function mainly by increasing the BDNF content. AD-F-H and AD-C-H did not improve the memory function. Compared to AD-Con, the ascending order of AD-C-H, AD-F-H, AD-C-L, and AD-F-L increased insulin signaling by enhancing the pSTAT3→pAkt→pGSK-3β pathway. AD-F-L improved glucose tolerance the most. Compared to AD-CON, the AD-F-L treatment increased the serum acetate concentrations and compensated for the defect of cerebral glucose metabolism. AD-Con increased Clostridium, Terrisporobacter and Sporofaciens compared to Normal-Con, and AD-F-L and AD-C-L increased Akkermentia. In conclusion, AD-F-L and AD-C-L alleviated the memory function in the rats with induced AD symptoms by modulating.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Bioconvergence System, Hoseo University, Asan 31499, Korea; (T.Z.); (X.W.); (H.Y.); (S.H.)
| | - Xuangao Wu
- Department of Bioconvergence System, Hoseo University, Asan 31499, Korea; (T.Z.); (X.W.); (H.Y.); (S.H.)
| | - Heng Yuan
- Department of Bioconvergence System, Hoseo University, Asan 31499, Korea; (T.Z.); (X.W.); (H.Y.); (S.H.)
| | - Shaokai Huang
- Department of Bioconvergence System, Hoseo University, Asan 31499, Korea; (T.Z.); (X.W.); (H.Y.); (S.H.)
| | - Sunmin Park
- Department of Bioconvergence System, Hoseo University, Asan 31499, Korea; (T.Z.); (X.W.); (H.Y.); (S.H.)
- Department of Food and Nutrition, Obesity/Diabetes Research Center, Hoseo University, Asan 31499, Korea
- Correspondence: ; Tel.: +82-41-540-5633; Fax: +82-41-540-5638
| |
Collapse
|
12
|
Saxena A, Moran RRM, Bullard MR, Bondy EO, Smith MF, Morris L, Fogle N, Singh J, Jarvis B, Ray T, Saxena J, Freeman LR. Sex differences in the fecal microbiome and hippocampal glial morphology following diet and antibiotic treatment. PLoS One 2022; 17:e0265850. [PMID: 35385494 PMCID: PMC8985946 DOI: 10.1371/journal.pone.0265850] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 03/08/2022] [Indexed: 12/31/2022] Open
Abstract
Rising obesity rates have become a major public health concern within the United States. Understanding the systemic and neural effects of obesity is crucial in designing preventive and therapeutic measures. In previous studies, administration of a high fat diet has induced significant weight gain for mouse models of obesity. Interestingly, sex differences in high-fat diet-induced weight gain have been observed, with female mice gaining significantly less weight compared to male mice on the same high-fat diet. It has also been observed that consumption of a high-fat diet can increase neurogliosis, but the mechanism by which this occurs is still not fully understood. Recent research has suggested that the gut microbiome may mediate diet-induced glial activation. The current study aimed to (1) analyze changes to the gut microbiome following consumption of a high fat (HF) diet as well as antibiotic treatment, (2) evaluate hippocampal microgliosis and astrogliosis, and (3) identify sex differences within these responses. We administered a low fat (Research Diets D12450 K) or high fat diet (Research Diets D12451) to male and female C57Bl/6 mice for sixteen weeks. Mice received an antibiotic cocktail containing 0.5g/L of vancomycin, 1.0 g/L ampicillin, 1.0 g/L neomycin, and 1.0 g/L metronidazole in their drinking water during the last six weeks of the study and were compared to control mice receiving normal drinking water throughout the study. We observed a significant reduction in gut microbiome diversity for groups that received the antibiotic cocktail, as determined by Illumina next-generation sequencing. Male mice fed the HF diet (± antibiotics) had significantly greater body weights compared to all other groups. And, female mice fed the low fat (LF) diet and administered antibiotics revealed significantly decreased microgliosis and astrogliosis in the hippocampus compared to LF-fed females without antibiotics. Interestingly, male mice fed the LF diet and administered antibiotics revealed significantly increased microgliosis, but decreased astrogliosis, compared to LF-fed males without antibiotics. The observed sex differences in LF-fed mice given antibiotics brings forward questions about sex differences in nutrient metabolism, gut microbiome composition, and response to antibiotics.
Collapse
Affiliation(s)
- Anju Saxena
- Neurosciences, Furman University, Greenville, South Carolina, United States of America
| | - Roberta R. M. Moran
- Neurosciences, Furman University, Greenville, South Carolina, United States of America
| | - Meghan R. Bullard
- Department of Biology, Furman University, Greenville, South Carolina, United States of America
| | - Emma O. Bondy
- Neurosciences, Furman University, Greenville, South Carolina, United States of America
| | - Matthew Foster Smith
- Neurosciences, Furman University, Greenville, South Carolina, United States of America
| | - Lainie Morris
- Department of Biology, Furman University, Greenville, South Carolina, United States of America
| | - Nicaella Fogle
- Neurosciences, Furman University, Greenville, South Carolina, United States of America
| | - Jagroop Singh
- Department of Biology, Furman University, Greenville, South Carolina, United States of America
| | - Brendan Jarvis
- Neurosciences, Furman University, Greenville, South Carolina, United States of America
| | - Tammy Ray
- Neurosciences, Furman University, Greenville, South Carolina, United States of America
| | - Juhi Saxena
- Neurosciences, Furman University, Greenville, South Carolina, United States of America
| | - Linnea Ruth Freeman
- Neurosciences, Furman University, Greenville, South Carolina, United States of America
- Department of Biology, Furman University, Greenville, South Carolina, United States of America
| |
Collapse
|
13
|
Alemohammad SMA, Noori SMR, Samarbafzadeh E, Noori SMA. The role of the gut microbiota and nutrition on spatial learning and spatial memory: a mini review based on animal studies. Mol Biol Rep 2022; 49:1551-1563. [PMID: 35028854 DOI: 10.1007/s11033-021-07078-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 12/09/2021] [Indexed: 02/07/2023]
Abstract
The gut-brain axis is believed to constitute a bidirectional communication mechanism that affects both mental and digestive processes. Recently, the role of the gut microbiota in cognitive performance has been the focus of much research. In this paper, we discuss the effects of gut microbiota and nutrition on spatial memory and learning. Studies have shown the influence of diet on cognitive capabilities such as spatial learning and memory. It has been reported that a high-fat diet can alter gut microbiota which subsequently leads to changes in spatial learning and memory. Some microorganisms in the gut that can significantly affect spatial learning and memory are Akkermansia muciniphila, Bifidobacterium, Lactobacillus, Firmicutes, Bacteroidetes, and Helicobacter pylori. For example, a reduction in the amount of A. muciniphila in the gut leads to increased intestinal permeability and induces immune response in the brain which then negatively affects cognitive performances. We suggest that more studies should be carried out regarding the indirect effects of nutrition on cognitive activities via alteration in gut microbiota.
Collapse
Affiliation(s)
| | - Seyed Mohammad Reza Noori
- Department of Medical Physics and Biomedical Engineering, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Samarbafzadeh
- Department of Psychiatry and Behavioral Medicine, Carilion Clinic, Roanoke, VA, USA
| | - Seyyed Mohammad Ali Noori
- Toxicology Research Center, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran. .,Department of Nutrition, School of Allied Medical Sciences, Medical Basic Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| |
Collapse
|
14
|
Borah S, Sarkar P, Sharma HK. Zederone Improves the Fecal Microbial Profile in Dementia Induced Rat Model: A First Report. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2021; 21:335-342. [PMID: 34455974 DOI: 10.2174/1871527320666210827114227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 09/27/2020] [Accepted: 04/11/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Dementia correlates with Alzheimer's disease, Parkinson's disease, frontotemporal and cerebrovascular diseases. There are supporting shreds of evidence on the pharmacological activity of Curcuma caesia (Zingiberaceae family) for its antioxidant, antidepressant, analgesic, anticonvulsant, and anti-acetylcholinesterase effect. OBJECTIVE This study aims to analyze the fecal microbial profile in Zederone treated demented rat model. METHOD In our study, isolation and characterization of Zederone were carried out from Curcuma caesia rhizomes, followed by estimation of its memory-enhancing effect on Aluminium-induced demented rat, which was evaluated by behavioural study on radial 8 arm maze. Moreover the detection of amyloid plaque formation was carried out using fluorescent microscopy of the congo red-stained rat brain tissues of the cerebral neocortex region. This study included eighteen female Wistar Albino rats that were divided into three groups that consisted of six rats in each group. The study of fecal microbial profile by metagenomic DNA extraction followed by next-generation sequencing was carried out to establish the correlation between gut microbes and amyloid plaques in dementia. RESULTS Zederone could be characterized as pale yellow colored, needle-shaped crystals with 96.57% purity. This compound at 10 mg/kg body weight showed cognition improving capacity (p ≤ 0.0001) with a reduction of accumulated amyloid plaques that were detected in the demented group in fluorescence microscope and fecal microbiome study divulged an increased shift towards Lactobacillus genera in the treated group from Bacteroides in the demented group. CONCLUSION This sesquiterpenoid compound would assist in the modulation of gut bacterial dysbiosis and act as a better therapeutic drug for dementia and other neurological disorders.
Collapse
Affiliation(s)
- Sudarshana Borah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, Assam-786004. India
| | - Priyanka Sarkar
- Wellcome trust- DBT (India Alliance) Lab, Asian Healthcare Foundation, Asian Institute of Gastroenterology, Hyderabad-500032. India
| | - Hemanta Kumar Sharma
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh, Assam-786004. India
| |
Collapse
|
15
|
de Figueiredo CS, Sandre PC, Portugal LCL, Mázala-de-Oliveira T, da Silva Chagas L, Raony Í, Ferreira ES, Giestal-de-Araujo E, Dos Santos AA, Bomfim POS. COVID-19 pandemic impact on children and adolescents' mental health: Biological, environmental, and social factors. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110171. [PMID: 33186638 PMCID: PMC7657035 DOI: 10.1016/j.pnpbp.2020.110171] [Citation(s) in RCA: 281] [Impact Index Per Article: 93.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 11/02/2020] [Accepted: 11/06/2020] [Indexed: 12/11/2022]
Abstract
Since the Coronavirus disease 2019 (COVID-19) pandemic, caused by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) was announced, we had an unprecedented change in the way we organize ourselves socially and in our daily routine. Children and adolescents were also greatly impacted by the abrupt withdrawal from school, social life and outdoor activities. Some of them also experienced domestic violence growing. The stress they are subjected to directly impacts their mental health on account of increased anxiety, changes in their diets and in school dynamics, fear or even failing to scale the problem. Our aim is to bring up a discussion under different aspects and to alert public health and government agents about the need for surveillance and care of these individuals. We hope that the damage to their mental health as a result of the side effect of this pandemic can be mitigated by adequate and timely intervention.
Collapse
Affiliation(s)
- Camila Saggioro de Figueiredo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, 24020-141, Brazil
| | - Poliana Capucho Sandre
- NuPEDEN, Nucleus for Research, Education, Dissemination and Neurosciences Popularization, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; Thymus Research Laboratory, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Liana Catarina Lima Portugal
- NuPEDEN, Nucleus for Research, Education, Dissemination and Neurosciences Popularization, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; Department of Physiology and Pharmacology, Laboratory of Neurophysiology of Behavior, Biomedical Institute, Federal Fluminense University, Brazil
| | - Thalita Mázala-de-Oliveira
- NuPEDEN, Nucleus for Research, Education, Dissemination and Neurosciences Popularization, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil
| | - Luana da Silva Chagas
- NuPEDEN, Nucleus for Research, Education, Dissemination and Neurosciences Popularization, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, 24020-141, Brazil
| | - Ícaro Raony
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, 24020-141, Brazil; School of Medicine, Federal Fluminense University, Niterói 24033-900, Brazil
| | - Elenn Soares Ferreira
- NuPEDEN, Nucleus for Research, Education, Dissemination and Neurosciences Popularization, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil
| | - Elizabeth Giestal-de-Araujo
- Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, 24020-141, Brazil; National Institute of Science and Technology on Neuroimmunomodulation - INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil
| | - Aline Araujo Dos Santos
- Department of Physiology and Pharmacology, Laboratory of Neurochemical Interactions, Biomedical Institute, Federal Fluminense University, Brazil
| | - Priscilla Oliveira-Silva Bomfim
- NuPEDEN, Nucleus for Research, Education, Dissemination and Neurosciences Popularization, Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói 24020-141, Brazil; Department of Neurobiology and Program of Neurosciences, Institute of Biology, Federal Fluminense University, Niterói, 24020-141, Brazil; National Institute of Science and Technology on Neuroimmunomodulation - INCT-NIM, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil; Rio de Janeiro Neuroinflammation Research Network, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21040-360, Brazil.
| |
Collapse
|
16
|
Moridi H, Sarihi A, Habibitabar E, Shateri H, Salehi I, Komaki A, Karimi J, Karimi SA. Effects of post-training administration of LY341495, as an mGluR2/3 antagonist on spatial memory deficit in rats fed with high-fat diet. IBRO Rep 2020; 9:241-246. [PMID: 33024878 PMCID: PMC7527618 DOI: 10.1016/j.ibror.2020.09.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Accepted: 09/07/2020] [Indexed: 12/12/2022] Open
Abstract
High-fat diets (HFDs) adversely influence glutamate metabolism and neurotransmission. The precise role of the group II metabotropic glutamate receptors (mGluR2/3) antagonist on spatial memory deficit following consumption of HFD has not yet been clarified. Therefore, in this study, we examined the effects of post-training administration of mGluR2/3 antagonism; LY341495 on spatial memory in rats fed with HFD (for 10 weeks) by using Morris Water Maze (MWM) task. The training session for testing memory acquisition in MWM consisted of 4 trials per day for 4 consecutive days. Twenty-four hours after the last training session the spatial probe test (retention) was given. Intraperitoneal injection (i.p) injection of LY341495 was done 30 min before probe test. Our results showed that 10 weeks consumption of HFD had no significant effect on escape latency and swimming distance in memory acquisition. Our finding showed that consumption of a HFD leads to reference memory impairment in the probe test. HFD animals spent less time in the target zone in compare with control animals. Also, LY341495 improved HFD-induced reference memory (retention) impairment. HFD animals treated with LY341495 spent more time in the target zone in compare with HFD animals. Escape latencies to find the visible platform during visual task were same in all experimental groups, indicating no visual impairment in the animals. We propose that a HFD may act through mGluR2/3 within the brain to reduce synaptic plasticity, which impairs memory retrieval, and post-training administration of LY341495 can reduce HFD-induced reference memory impairment.
Collapse
Affiliation(s)
- Heresh Moridi
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran.,Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Abdolrahman Sarihi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Elahe Habibitabar
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran.,Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Hossein Shateri
- Student Research Committee, Hamadan University of Medical Sciences, Hamadan, Iran.,Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Iraj Salehi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Jamshid Karimi
- Department of Biochemistry, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Asaad Karimi
- Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran.,Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
17
|
Luca M, Chattipakorn SC, Sriwichaiin S, Luca A. Cognitive-Behavioural Correlates of Dysbiosis: A Review. Int J Mol Sci 2020; 21:4834. [PMID: 32650553 PMCID: PMC7402132 DOI: 10.3390/ijms21144834] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 07/06/2020] [Accepted: 07/07/2020] [Indexed: 02/07/2023] Open
Abstract
Evidence suggests an association between an altered gut microbiota (dysbiosis), cognitive performance and behaviour. This paper provides an overview of the current literature regarding the cognitive-behavioural correlates of dysbiosis, with special attention on the clinical and biochemical mechanisms underlying the association between dysbiosis, cognition (mild cognitive impairment and dementia) and behaviour (depression, schizophrenia, addiction). After providing an overview of the evidence, the review discusses the molecular aspects that could account for the cognitive-behavioural correlates of dysbiosis. Shedding light on this topic could provide insights regarding the pathogenesis of these burdening neuropsychiatric disorders and even suggest future therapeutic strategies.
Collapse
Affiliation(s)
- Maria Luca
- Department of Medical and Surgical Sciences and Advanced Technologies, “GF Ingrassia”, University of Catania, 95123 Catania, Italy;
| | - Siriporn C. Chattipakorn
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (S.C.C.); (S.S.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Department of Oral Biology and Diagnostic Sciences, Faculty of Dentistry, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sirawit Sriwichaiin
- Neurophysiology Unit, Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; (S.C.C.); (S.S.)
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Antonina Luca
- Department of Medical and Surgical Sciences and Advanced Technologies, “GF Ingrassia”, University of Catania, 95123 Catania, Italy;
| |
Collapse
|
18
|
Arivazhagan L, Ruiz HH, Wilson R, Manigrasso M, Gugger PF, Fisher EA, Moore KJ, Ramasamy R, Schmidt AM. An Eclectic Cast of Cellular Actors Orchestrates Innate Immune Responses in the Mechanisms Driving Obesity and Metabolic Perturbation. Circ Res 2020; 126:1565-1589. [PMID: 32437306 PMCID: PMC7250004 DOI: 10.1161/circresaha.120.315900] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The escalating problem of obesity and its multiple metabolic and cardiovascular complications threatens the health and longevity of humans throughout the world. The cause of obesity and one of its chief complications, insulin resistance, involves the participation of multiple distinct organs and cell types. From the brain to the periphery, cell-intrinsic and intercellular networks converge to stimulate and propagate increases in body mass and adiposity, as well as disturbances of insulin sensitivity. This review focuses on the roles of the cadre of innate immune cells, both those that are resident in metabolic organs and those that are recruited into these organs in response to cues elicited by stressors such as overnutrition and reduced physical activity. Beyond the typical cast of innate immune characters invoked in the mechanisms of metabolic perturbation in these settings, such as neutrophils and monocytes/macrophages, these actors are joined by bone marrow-derived cells, such as eosinophils and mast cells and the intriguing innate lymphoid cells, which are present in the circulation and in metabolic organ depots. Upon high-fat feeding or reduced physical activity, phenotypic modulation of the cast of plastic innate immune cells ensues, leading to the production of mediators that affect inflammation, lipid handling, and metabolic signaling. Furthermore, their consequent interactions with adaptive immune cells, including myriad T-cell and B-cell subsets, compound these complexities. Notably, many of these innate immune cell-elicited signals in overnutrition may be modulated by weight loss, such as that induced by bariatric surgery. Recently, exciting insights into the biology and pathobiology of these cell type-specific niches are being uncovered by state-of-the-art techniques such as single-cell RNA-sequencing. This review considers the evolution of this field of research on innate immunity in obesity and metabolic perturbation, as well as future directions.
Collapse
Affiliation(s)
- Lakshmi Arivazhagan
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Henry H. Ruiz
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Robin Wilson
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Michaele Manigrasso
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Paul F. Gugger
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Edward A. Fisher
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, NYU Langone Medical Center, New York 10016
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, New York 10016
| | - Kathryn J. Moore
- The Leon H. Charney Division of Cardiology, Department of Medicine, The Marc and Ruti Bell Program in Vascular Biology, NYU Langone Medical Center, New York 10016
- NYU Cardiovascular Research Center, NYU Grossman School of Medicine, New York, New York 10016
| | - Ravichandran Ramasamy
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| | - Ann Marie Schmidt
- Diabetes Research Program, Division of Endocrinology, Diabetes and Metabolism, Department of Medicine, NYU Grossman School of Medicine, New York, New York 10016
| |
Collapse
|
19
|
Daly CM, Saxena J, Singh J, Bullard MR, Bondy EO, Saxena A, Buffalino RE, Melville MF, Freeman LR. Sex differences in response to a high fat, high sucrose diet in both the gut microbiome and hypothalamic astrocytes and microglia. Nutr Neurosci 2020; 25:321-335. [PMID: 32297553 DOI: 10.1080/1028415x.2020.1752996] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Objectives: Obesity is a major epidemic in our population and has emerged as a primary health concern. Consumption of a high fat, high sugar (HFHS) diet can specifically lead to gut dysbiosis, increased inflammation, and neuroinflammation. Interestingly, sex differences in the response to a HFHS diet are emerging. In this study, we investigated the effects of a HFHS diet compared to a low fat, low sugar (LFLS) diet in 8 week old male and female C57Bl/6 mice.Methods: The diet was administered for 14 weeks; body weights and food consumption were evaluated weekly.Results: Male and female mice fed the HFHS diet gained significantly more weight than LFLS-fed mice. However, in agreement with previous studies, males gained significantly more weight on the HFHS diet compared to females fed the same diet. Importantly, we determined significant sex and diet-induced differences to gut microbiome composition using next generation Illumina sequencing. We also observed significantly less astrocyte densitometry and no significant change to microglial morphology in the hypothalamus of Female HFHS compared to Female LFLS. On the other hand, Male HFHS revealed no change to hypothalamic astrogliosis, but increased microgliosis compared to Male LFLS.Discussion: In this study, we determined sex and diet-induced differences in both the gut and the brain, however, future studies will need to be performed in order to test the direct role of the gut microbiome to weight gain and neuroinflammation in male and female mice.
Collapse
Affiliation(s)
| | - Juhi Saxena
- Neuroscience, Furman University, Greenville, SC, USA
| | - Jagroop Singh
- Department of Biology, Furman University, Greenville, SC, USA
| | | | - Emma O Bondy
- Neuroscience, Furman University, Greenville, SC, USA
| | - Anju Saxena
- Neuroscience, Furman University, Greenville, SC, USA
| | | | | | - Linnea R Freeman
- Neuroscience, Furman University, Greenville, SC, USA.,Department of Biology, Furman University, Greenville, SC, USA
| |
Collapse
|
20
|
Leigh SJ, Morris MJ. Diet, inflammation and the gut microbiome: Mechanisms for obesity-associated cognitive impairment. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165767. [PMID: 32171891 DOI: 10.1016/j.bbadis.2020.165767] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/18/2020] [Accepted: 03/09/2020] [Indexed: 12/11/2022]
Abstract
Poor diet and obesity are associated with cognitive impairment throughout adulthood, and increased dementia risk in aging. Here we review the current literature interrogating the mechanisms by which diets high in fat, or fat and sugar lead to cognitive impairment, focusing on changes to gut microbiome composition, inflammatory signalling and blood-brain barrier integrity. Preclinical studies indicate weight gain is not necessary for diet-induced cognitive impairment. Rather, gut microbiome composition, and systemic and central inflammatory processes appear to contribute to diet-induced cognitive impairment. While both obese humans and rodents exhibit reduced blood-brain barrier integrity, cognitive impairments precede these changes, suggesting other mechanisms may underly diet-induced cognitive changes. Other potential candidates include hormone, glucoregulatory and cardiovascular changes. Poor diet and obesity act through multiple mechanisms to affect cognitive health and the challenge for future research is to identify key processes that can be reversed to improve cognition and quality of life.
Collapse
|
21
|
Jaiswal AK, Makhija S, Stahr N, Sandey M, Suryawanshi A, Saxena A, Dagur PK, McCoy JP, Levine SJ, Mishra A. Dendritic Cell-Restricted Progenitors Contribute to Obesity-Associated Airway Inflammation via Adam17-p38 MAPK-Dependent Pathway. Front Immunol 2020; 11:363. [PMID: 32184787 PMCID: PMC7058657 DOI: 10.3389/fimmu.2020.00363] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Accepted: 02/14/2020] [Indexed: 01/08/2023] Open
Abstract
Proliferation of dendritic cell (DC)—restricted progenitor cells in bone marrow compartment is tightly regulated at steady state and responds to multiple tissue-specific triggers during disturbed homeostasis such as obesity. DCs in the lung stem from a rapidly dividing DC-restricted progenitor cells and are effective at generating adaptive immune responses in allergic airway inflammation. Precisely, how DC-restricted progenitor expansion and differentiation are influenced by airway inflammation to maintain constant supply of myeloid DCs is poorly understood. Here we show that a high fat diet (HFD) induces oxidative stress and accelerates the expansion of DC- restricted progenitor cells in bone marrow and correlates with persistent induction of p38 mitogen activated protein kinase (MAPK), which is blocked with a selective p38α/β MAPK inhibitor. Mice fed a HFD and sensitized to inhaled allergen house dust mite (HDM) led to alterations of DC- restricted progenitor cells that were characterized by increased expansion and seeding of lung DCs in airway inflammation. Mechanistically, we establish that the expansion induced by HFD dysregulates the expression of a disintegrin and metallopeptidase domain 17 (Adam17) and is required for p38 MAPK activation in DC-restricted progenitors. These results demonstrates that obesity produces persistent changes in DC precursors and that elevation of Adam17 expression is tightly coupled to p38 MAPK and is a key driver of proliferation. Altogether, these data provide phenotypic and mechanistic insight into dendritic cell supply chain in obesity-associated airway inflammation.
Collapse
Affiliation(s)
- Anil Kumar Jaiswal
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Sangeet Makhija
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Natalie Stahr
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Maninder Sandey
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Amol Suryawanshi
- Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| | - Ankit Saxena
- Flow Cytometry Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Pradeep K Dagur
- Flow Cytometry Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - J Philip McCoy
- Flow Cytometry Core Facility, Division of Intramural Research, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Stewart J Levine
- Laboratory of Asthma and Lung Inflammation, Division of Intramural Research, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Amarjit Mishra
- Laboratory of Lung Inflammation, College of Veterinary Medicine, Auburn University, Auburn, AL, United States.,Department of Pathobiology, College of Veterinary Medicine, Auburn University, Auburn, AL, United States
| |
Collapse
|
22
|
Illouz T, Madar R, Okun E. A modified Barnes maze for an accurate assessment of spatial learning in mice. J Neurosci Methods 2020; 334:108579. [PMID: 31926999 DOI: 10.1016/j.jneumeth.2020.108579] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2019] [Revised: 12/17/2019] [Accepted: 01/05/2020] [Indexed: 11/19/2022]
Abstract
BACKGROUND The Morris water maze (MWM) and the Barnes maze (BM) are among the most widely-used paradigms for assessing spatial learning in rodents, with specific advantages and disadvantages for each apparatus. Compared with the intense water-related stress exerted during the MWM, the BM exhibits a milder light-induced stress, while suffering from biasing animals towards non-spatial strategies such as serial search, a heuristic non-spatial search strategy. To overcome this problem, we have developed a modified Barnes maze (MBM) apparatus that recapitulates natural environments more accurately without inducing undesirable exploration strategy bias. NEW METHOD Apparatus. A circular 122 cm-wide table with 40 randomly placed holes. One target hole is leading to an escape chamber. Task. Three target locations were examined, varying in their distance from the center. C57BL6/j male mice were given three trials per day to find the target. Following acquisition, a probe test was performed by removing the escape chamber. RESULTS Spatial-encoding-depended reduction in latency to reach the target was observed, along with improvement in path efficiency with test progress. Mice tested with peripheral and distal targets outperformed mice tested with a central target. A robust exploration pattern was identified in the probe test. COMPARISON WITH EXISTING METHOD The MBM mimics natural environment to a higher degree of accuracy than the BM, without eliciting bias towards non-spatial searching strategies. CONCLUSIONS Spatial learning in the MBM is a target-location sensitive process, providing flexibility in task difficulty. Along with overcoming biases towards non-spatial strategies, the MBM represents an improvement over the well-validated BM.
Collapse
Affiliation(s)
- Tomer Illouz
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan, 5290002, Israel.
| | - Ravit Madar
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan, 5290002, Israel.
| | - Eitan Okun
- The Leslie and Susan Gonda Multidisciplinary Brain Research Center, Bar-Ilan University, Ramat Gan, 5290002, Israel; The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, 5290002, Israel; The Paul Feder Laboratory on Alzheimer's Disease Research, Bar-Ilan University, Ramat Gan, 5290002, Israel.
| |
Collapse
|
23
|
Higarza SG, Arboleya S, Gueimonde M, Gómez-Lázaro E, Arias JL, Arias N. Neurobehavioral dysfunction in non-alcoholic steatohepatitis is associated with hyperammonemia, gut dysbiosis, and metabolic and functional brain regional deficits. PLoS One 2019; 14:e0223019. [PMID: 31539420 PMCID: PMC6754158 DOI: 10.1371/journal.pone.0223019] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 09/11/2019] [Indexed: 12/11/2022] Open
Abstract
Non-alcoholic steatohepatitis (NASH) is one of the most prevalent diseases worldwide. While it has been suggested to cause nervous impairment, its neurophysiological basis remains unknown. Therefore, the aim of this study is to unravel the effects of NASH, through the interrelationship of liver, gut microbiota, and nervous system, on the brain and human behavior. To this end, 40 Sprague-Dawley rats were divided into a control group that received normal chow and a NASH group that received a high-fat, high-cholesterol diet. Our results show that 14 weeks of the high-fat, high-cholesterol diet induced clinical conditions such as NASH, including steatosis and increased levels of ammonia. Rats in the NASH group also demonstrated evidence of gut dysbiosis and decreased levels of short-chain fatty acids in the gut. This may explain the deficits in cognitive ability observed in the NASH group, including their depressive-like behavior and short-term memory impairment characterized in part by deficits in social recognition and prefrontal cortex-dependent spatial working memory. We also reported the impact of this NASH-like condition on metabolic and functional processes. Brain tissue demonstrated lower levels of metabolic brain activity in the prefrontal cortex, thalamus, hippocampus, amygdala, and mammillary bodies, accompanied by a decrease in dopamine levels in the prefrontal cortex and cerebellum and a decrease in noradrenalin in the striatum. In this article, we emphasize the important role of ammonia and gut-derived bacterial toxins in liver-gut-brain neurodegeneration and discuss the metabolic and functional brain regional deficits and behavioral impairments in NASH.
Collapse
Affiliation(s)
- Sara G. Higarza
- Institute of Neurosciences of the Principality of Asturias (INEUROPA), Asturias, Spain
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Asturias, Spain
| | - Silvia Arboleya
- Department of Microbiology and Biochemistry of Dairy Products, Institute of Dairy Products of the Principality of Asturias (IPLA-CSIC), Asturias, Spain
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Institute of Dairy Products of the Principality of Asturias (IPLA-CSIC), Asturias, Spain
| | - Eneritz Gómez-Lázaro
- Department of Basic Psychological Processes and their Development, Basque Country University, San Sebastián, Basque Country, Spain
| | - Jorge L. Arias
- Institute of Neurosciences of the Principality of Asturias (INEUROPA), Asturias, Spain
- Laboratory of Neuroscience, Department of Psychology, University of Oviedo, Oviedo, Asturias, Spain
| | - Natalia Arias
- Institute of Neurosciences of the Principality of Asturias (INEUROPA), Asturias, Spain
- Department of Basic and Clinical Neuroscience, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, England, United Kingdom
| |
Collapse
|