1
|
Oikonomou I, Papageorgiou A, de Lastic AL, Moulias A, Georgopoulou GA, Mouzaki A, Koufou EE, Tsigkas G, Gogos C, Davlouros P, Assimakopoulos SF. Gut barrier dysfunction, endotoxemia and inflammatory response in STEMI patients and effect of primary PCI. Am J Med Sci 2024; 368:485-493. [PMID: 38969287 DOI: 10.1016/j.amjms.2024.07.004] [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: 12/22/2023] [Revised: 06/05/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
BACKGROUND Gut-derived bacterial and endotoxin translocation induce systemic inflammation, which exerts a pivotal pathogenetic role in all phases of atherosclerosis. OBJECTIVES To investigate prospectively the gut barrier function, endotoxin translocation and inflammatory response in ST-elevation myocardial infarction (STEMI) patients undergoing primary percutaneous coronary artery intervention (PPCI). METHODS Twenty-seven patients with STEMI that underwent successful PPCI were subjected to peripheral blood sampling at 3-time points; before PPCI (day0), 24 h (day1) and 96 h (day4) after PPCI and were compared with 20 chronic coronary syndrome (CCS) patients and 11 healthy controls. Serum ZO-1, I-FABP and endotoxin concentrations were determined by ELISA. Concentrations of cytokines IL-1β, -6, -8, -10 and TNF-α were determined by flow cytometry. RESULTS Patients with STEMI before PPCI (day0) had increased serum ZO-1 and endotoxin, both at significantly higher levels compared to CCS patients. STEMI induced also significant increases of the cytokines IL-6, -8 and -10. After PPCI, a significant improvement of gut barrier integrity (ZO-1) and endotoxemia was observed from the first day. At day4 post PPCI, systemic endotoxin and cytokines IL-6, -8 and -10 levels were reduced to control levels. Serum ZO-1 levels were positively correlated with systemic IL-10 concentrations (r = 0.471). CONCLUSION STEMI is associated with gut barrier dysfunction, systemic endotoxemia and inflammatory response, which improve rapidly following successful PPCI.
Collapse
Affiliation(s)
- Ioanna Oikonomou
- Department of Internal Medicine and Division of Infectious Diseases, University of Patras Medical School, Patras, Greece
| | - Angeliki Papageorgiou
- Division of Cardiology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Anne-Lise de Lastic
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Athanasios Moulias
- Division of Cardiology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | | | - Athanasia Mouzaki
- Laboratory of Immunohematology, Division of Hematology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Eleni-Evangelia Koufou
- Division of Cardiology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Grigorios Tsigkas
- Division of Cardiology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Charalambos Gogos
- Department of Internal Medicine and Division of Infectious Diseases, University of Patras Medical School, Patras, Greece
| | - Periklis Davlouros
- Division of Cardiology, Department of Internal Medicine, University of Patras Medical School, Patras, Greece
| | - Stelios F Assimakopoulos
- Department of Internal Medicine and Division of Infectious Diseases, University of Patras Medical School, Patras, Greece.
| |
Collapse
|
2
|
Baenas I, Camacho-Barcia L, Miranda-Olivos R, Solé-Morata N, Misiolek A, Jiménez-Murcia S, Fernández-Aranda F. Probiotic and prebiotic interventions in eating disorders: A narrative review. EUROPEAN EATING DISORDERS REVIEW 2024; 32:1085-1104. [PMID: 38297469 DOI: 10.1002/erv.3069] [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: 05/29/2023] [Revised: 11/17/2023] [Accepted: 12/29/2023] [Indexed: 02/02/2024]
Abstract
AIMS The review aimed to summarise and discuss findings focused on therapeutic probiotic and prebiotic interventions in eating disorders (ED). METHODS Using PubMed/MEDLINE, Cochrane Library, and Web of Science all published studies were retrieved until February 2023, following PRISMA guidelines. From the 111 initial studies, 5 met the inclusion criteria for this review. RESULTS All studies included in this narrative review were focused on anorexia nervosa (AN). Three longitudinal, randomised, controlled trials aimed to evaluate interventions with probiotics (Lactobacillus reuteri, yoghurt with Lactobacillus, and Streptococcus) in children and adolescents. These studies primarily emphasised medical outcomes and anthropometric measures following the administration of probiotics. However, the findings yielded mixed results in terms of short-term weight gain or alterations in specific immunological parameters. With a lower level of evidence, supplementation with synbiotics (probiotic + prebiotic) has been associated with improvements in microbiota diversity and attenuation of inflammatory responses. CONCLUSIONS Research on probiotics and prebiotics in ED is limited, primarily focussing on anorexia nervosa (AN). Their use in AN regarding medical and anthropometric outcomes needs further confirmation and future research should be warranted to assess their impact on psychological and ED symptomatology, where there is a notable gap in the existing literature.
Collapse
Affiliation(s)
- Isabel Baenas
- Department of Clinical Psychology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviours, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Instituto Salud Carlos III, Barcelona, Spain
- Doctoral Program in Medicine and Translational Research, University of Barcelona (UB), Barcelona, Spain
| | - Lucía Camacho-Barcia
- Department of Clinical Psychology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviours, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Instituto Salud Carlos III, Barcelona, Spain
| | - Romina Miranda-Olivos
- Department of Clinical Psychology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviours, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Instituto Salud Carlos III, Barcelona, Spain
- Doctoral Program in Medicine and Translational Research, University of Barcelona (UB), Barcelona, Spain
| | - Neus Solé-Morata
- Department of Clinical Psychology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Alejandra Misiolek
- Proyecto Autoestima Relaciones y Trastornos Alimenticios (ART), Barcelona, Spain
| | - Susana Jiménez-Murcia
- Department of Clinical Psychology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviours, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Instituto Salud Carlos III, Barcelona, Spain
- Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona (UB), L'Hospitalet de Llobregat, Barcelona, Spain
- Psychology Services, University of Barcelona, Barcelona, Spain
| | - Fernando Fernández-Aranda
- Department of Clinical Psychology, Bellvitge University Hospital, L'Hospitalet de Llobregat, Barcelona, Spain
- Psychoneurobiology of Eating and Addictive Behaviours, Neuroscience Program, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, Barcelona, Spain
- CIBER Physiopathology of Obesity and Nutrition (CIBERobn), Instituto Salud Carlos III, Barcelona, Spain
- Department of Clinical Sciences, School of Medicine and Health Sciences, University of Barcelona (UB), L'Hospitalet de Llobregat, Barcelona, Spain
| |
Collapse
|
3
|
Bu LK, Jia PP, Huo WB, Pei DS. Assessment of Probiotics' Impact on Neurodevelopmental and Behavioral Responses in Zebrafish Models: Implications for Autism Spectrum Disorder Therapy. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10335-y. [PMID: 39090455 DOI: 10.1007/s12602-024-10335-y] [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] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
Autism spectrum disorder (ASD) is a neurodevelopmental disorder; the prevalence of which has been on the rise with unknown causes. Alterations in the gut-brain axis have been widely recognized in ASD patients, and probiotics are considered to potentially benefit the rescuing of autism-like behaviors. However, the effectiveness and mechanisms of multiple probiotics on zebrafish models are still not clearly revealed. This study aims to use the germ-free (GF) and conventionally raised (CR) AB wild-type zebrafish and the mutant Tbr1b-/- and Katnal2-/- lines as human-linked ASD animal models to evaluate the effects of multiple probiotics on mitigating developmental and behavioral defects. Results showed that the addition of probiotics increased the basic important developmental indexes, such as body length, weight, and survival rate of treated zebrafish. Moreover, the Lactobacillus plantarum and Lactobacillus rhamnosus affected the behavior of CR zebrafish by increasing their mobility, lowering the GF zebrafish manic, and mitigating transgenic zebrafish abnormal behavior. Moreover, the expression levels of key genes related to gamma-aminobutyric acid (GABA), dopamine (DA), and serotonin (5-HT) as important neuropathways to influence the appearance and development of autism-related disorders, including gad1b, tph1a, htr3a, th, and slc6a3, were significantly activated by some of the probiotics' treatment at some extent. Taken together, this study indicates the beneficial effects of different probiotics, which may provide a novel understanding of probiotic function in related diseases' therapy.
Collapse
Affiliation(s)
- Ling-Kang Bu
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Pan-Pan Jia
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - Wen-Bo Huo
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China
| | - De-Sheng Pei
- School of Public Health, Chongqing Medical University, Chongqing, 400016, China.
- Chongqing Miankai Biotechnology Research Institute Co., Ltd., Chongqing, 400025, China.
| |
Collapse
|
4
|
Ferrari S, Mulè S, Rosso G, Parini F, Galla R, Molinari C, Uberti F. An Innovative Probiotic-Based Supplement to Mitigate Molecular Factors Connected to Depression and Anxiety: An In Vitro Study. Int J Mol Sci 2024; 25:4774. [PMID: 38731995 PMCID: PMC11083558 DOI: 10.3390/ijms25094774] [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: 02/27/2024] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 05/13/2024] Open
Abstract
The gut-brain axis is a bidirectional relationship between the microbiota and the brain; genes related to the brain and gut synaptic formation are similar. Research on the causal effects of gut microbiota on human behavior, brain development, and function, as well as the underlying molecular processes, has emerged in recent decades. Probiotics have been shown in several trials to help reduce anxiety and depressive symptoms. Because of this, probiotic combinations have been tested in in vitro models to see whether they might modulate the gut and alleviate depression and anxiety. Therefore, we sought to determine whether a novel formulation might affect the pathways controlling anxiety and depression states and alter gut barrier activities in a 3D model without having harmful side effects. Our findings indicate that B. bifidum novaBBF7 10 mg/mL, B. longum novaBLG2 5 mg/mL, and L. paracasei TJB8 10 mg/mL may influence the intestinal barrier and enhance the synthesis of short-chain fatty acids. Additionally, the probiotics studied did not cause neuronal damage and, in combination, exert a protective effect against the condition of anxiety and depression triggered by L-Glutamate. All these findings show that probiotics can affect gut function to alter the pathways underlying anxiety and depression.
Collapse
Affiliation(s)
- Sara Ferrari
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, 13100 Vercelli, Italy (F.P.); (C.M.)
| | - Simone Mulè
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, 13100 Vercelli, Italy (F.P.); (C.M.)
| | - Giorgia Rosso
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, 13100 Vercelli, Italy (F.P.); (C.M.)
| | - Francesca Parini
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, 13100 Vercelli, Italy (F.P.); (C.M.)
| | - Rebecca Galla
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, 13100 Vercelli, Italy (F.P.); (C.M.)
- Noivita Srls, Spin Off of the University of Piemonte Orientale, Via Solaroli 17, 28100 Novara, Italy
| | - Claudio Molinari
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, 13100 Vercelli, Italy (F.P.); (C.M.)
| | - Francesca Uberti
- Laboratory of Physiology, Department for Sustainable Development and Ecological Transition, 13100 Vercelli, Italy (F.P.); (C.M.)
| |
Collapse
|
5
|
Dziedzic A, Maciak K, Bliźniewska-Kowalska K, Gałecka M, Kobierecka W, Saluk J. The Power of Psychobiotics in Depression: A Modern Approach through the Microbiota-Gut-Brain Axis: A Literature Review. Nutrients 2024; 16:1054. [PMID: 38613087 PMCID: PMC11013390 DOI: 10.3390/nu16071054] [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: 02/13/2024] [Revised: 03/28/2024] [Accepted: 04/02/2024] [Indexed: 04/14/2024] Open
Abstract
The microbiota-gut-brain (MGB) axis is a complex communication network linking the gut, microbiota, and brain, influencing various aspects of health and disease. Dysbiosis, a disturbance in the gut microbiome equilibrium, can significantly impact the MGB axis, leading to alterations in microbial composition and function. Emerging evidence highlights the connection between microbiota alterations and neurological and psychiatric disorders, including depression. This review explores the potential of psychobiotics in managing depressive disorders, emphasizing their role in restoring microbial balance and influencing the MGB axis. Psychobiotics exhibit positive effects on the intestinal barrier, immune response, cortisol levels, and the hypothalamic-pituitary-adrenal (HPA) axis. Studies suggest that probiotics may serve as an adjunct therapy for depression, especially in treatment-resistant cases. This review discusses key findings from studies on psychobiotics interventions, emphasizing their impact on the gut-brain axis and mental health. The increasing acceptance of the expanded concept of the MGB axis underscores the importance of microorganisms in mental well-being. As our understanding of the microbiome's role in health and disease grows, probiotics emerge as promising agents for addressing mental health issues, providing new avenues for therapeutic interventions in depressive disorders.
Collapse
Affiliation(s)
- Angela Dziedzic
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biochemistry, Pomorska 141/143, 90-236 Lodz, Poland; (K.M.); (W.K.); (J.S.)
| | - Karina Maciak
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biochemistry, Pomorska 141/143, 90-236 Lodz, Poland; (K.M.); (W.K.); (J.S.)
| | | | - Małgorzata Gałecka
- Department of Psychotherapy, Medical University of Lodz, Aleksandrowska 159, 91-229 Lodz, Poland;
| | - Weronika Kobierecka
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biochemistry, Pomorska 141/143, 90-236 Lodz, Poland; (K.M.); (W.K.); (J.S.)
| | - Joanna Saluk
- University of Lodz, Faculty of Biology and Environmental Protection, Department of General Biochemistry, Pomorska 141/143, 90-236 Lodz, Poland; (K.M.); (W.K.); (J.S.)
| |
Collapse
|
6
|
Tanelian A, Nankova B, Miari M, Sabban EL. Microbial composition, functionality, and stress resilience or susceptibility: unraveling sex-specific patterns. Biol Sex Differ 2024; 15:20. [PMID: 38409102 PMCID: PMC10898170 DOI: 10.1186/s13293-024-00590-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/31/2024] [Indexed: 02/28/2024] Open
Abstract
BACKGROUND Following exposure to traumatic stress, women are twice as likely as men to develop mood disorders. Yet, individual responses to such stress vary, with some people developing stress-induced psychopathologies while others exhibit resilience. The factors influencing sex-related disparities in affective disorders as well as variations in resilience remain unclear; however, emerging evidence suggests differences in the gut microbiota play a role. In this study, using the single prolonged stress (SPS) model of post-traumatic stress disorder, we investigated pre- and post-existing differences in microbial composition, functionality, and metabolites that affect stress susceptibility or resilience in each sex. METHODS Male and female Sprague-Dawley rats were randomly assigned to control or SPS groups. Two weeks following SPS, the animals were exposed to a battery of behavioral tests and decapitated a day later. Based on their anxiety index, they were further categorized as SPS-resilient (SPS-R) or SPS-susceptible (SPS-S). On the day of dissection, cecum, and selected brain tissues were isolated. Stool samples were collected before and after SPS, whereas urine samples were taken before and 30 min into the SPS. RESULTS Before SPS exposure, the sympathoadrenal axis exhibited alterations within male subgroups only. Expression of tight junction protein claudin-5 was lower in brain of SPS-S males, but higher in SPS-R females following SPS. Across the study, alpha diversity remained consistently lower in males compared to females. Beta diversity revealed distinct separations between male and female susceptible groups before SPS, with this separation becoming evident in the resilient groups following SPS. At the genus level, Lactobacillus, Lachnospiraceae_Incertae_Sedis, and Barnesiella exhibited sex-specific alterations, displaying opposing abundances in each sex. Additionally, sex-specific changes were observed in microbial predictive functionality and targeted functional modules both before and after SPS. Alterations in the microbial short-chain fatty acids (SCFAs), were also observed, with major and minor SCFAs being lower in SPS-susceptible males whereas branched-chain SCFAs being higher in SPS-susceptible females. CONCLUSION This study highlights distinct pre- and post-trauma differences in microbial composition, functionality, and metabolites, associated with stress resilience in male and female rats. The findings underscore the importance of developing sex-specific therapeutic strategies to effectively address stress-related disorders. Highlights SPS model induces divergent anxiety and social behavioral responses to traumatic stress in both male and female rodents. SPS-resilient females displayed less anxiety-like behavior and initiated more interactions towards a juvenile rat than SPS-resilient males. Sex-specific pre-existing and SPS-induced differences in the gut microbial composition and predictive functionality were observed in susceptible and resilient rats. SPS-resilient males displayed elevated cecal acetate levels, whereas SPS-susceptible females exhibited heightened branched-chain SCFAs.
Collapse
Affiliation(s)
- Arax Tanelian
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, 10595, USA
| | - Bistra Nankova
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, 10595, USA
- Division of Newborn Medicine, Departments of Pediatrics, New York Medical College, Valhalla, NY, 10595, USA
| | - Mariam Miari
- Department of Clinical Sciences in Malmo, Lund University Diabetes Center, Malmo, Sweden
| | - Esther L Sabban
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, NY, 10595, USA.
- Department of Psychiatry and Behavioral Science, New York Medical College, Valhalla, NY, 10595, USA.
| |
Collapse
|
7
|
Wlaź P, Wiater A, Majewska M, Wyska E, Grąz M, Śliwa-Dominiak J, Gapińska N, Socała K. Effect of dietary supplementation with Lactobacillus helveticus R0052 on seizure thresholds and antiseizure potency of sodium valproate in mice. Psychopharmacology (Berl) 2024; 241:327-340. [PMID: 37966492 PMCID: PMC10805985 DOI: 10.1007/s00213-023-06489-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023]
Abstract
OBJECTIVE Both animal and human studies, though limited, showed that multi-strain probiotic supplementation may reduce the number of seizures and/or seizure severity. Here, we evaluated the effect of a single strain probiotic supplementation on seizure susceptibility, antiseizure efficacy of sodium valproate, and several behavioral parameters in mice. METHODS Lactobacillus helveticus R0052 was given orally for 28 days. Its influence on seizure thresholds was evaluated in the ivPTZ- and electrically-induced seizure tests. The effect on the antiseizure potency of valproate was assessed in the scPTZ test. We also investigated the effects of probiotic supplementation on anxiety-related behavior (in the elevated plus maze and light/dark box tests), motor coordination (in the accelerating rotarod test), neuromuscular strength (in the grip-strength test), and spontaneous locomotor activity. Serum and brain concentrations of valproate as well as cecal contents of SCFAs and lactate were determined using HPLC method. RESULTS L. helveticus R0052 significantly increased the threshold for the 6 Hz-induced psychomotor seizure. There was also a slight increase in the threshold for myoclonic and clonic seizure in the ivPTZ test. L. helveticus R0052 did not affect the threshold for tonic seizures both in the maximal electroshock- and ivPTZ-induced seizure tests. No changes in the antiseizure potency of valproate against the PTZ-induced seizures were reported. Interestingly, L. helveticus R0052 increased valproate concentration in serum, but not in the brain. Moreover, L. helveticus R0052 did not produce any significant effects on anxiety-related behavior, motor coordination, neuromuscular strength, and locomotor activity. L. helveticus R0052 supplementation resulted in increased concentrations of total SCFAs, acetate, and butyrate. CONCLUSIONS Altogether, this study shows that a single-strain probiotic - L. helveticus R0052 may decrease seizure susceptibility and this effect can be mediated, at least in part, by increased production of SCFAs. In addition, L. helveticus R0052 may affect bioavailability of valproate, which warrants further investigations.
Collapse
Affiliation(s)
- Piotr Wlaź
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033, Lublin, Poland
| | - Adrian Wiater
- Department of Industrial and Environmental Microbiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033, Lublin, Poland
| | - Małgorzata Majewska
- Department of Industrial and Environmental Microbiology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033, Lublin, Poland
| | - Elżbieta Wyska
- Department of Pharmacokinetics and Physical Pharmacy, Jagiellonian University Medical College, Medyczna 9, PL 30-688, Kraków, Poland
| | - Marcin Grąz
- Department of Biochemistry and Biotechnology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033, Lublin, Poland
| | - Joanna Śliwa-Dominiak
- R&D and Scientific Department, Sanprobi Sp. z o.o Sp.k., Quality Control and Microbiology Laboratory, Kurza Stopka 5/C, PL 70-535, Szczecin, Poland
| | - Nikola Gapińska
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033, Lublin, Poland
| | - Katarzyna Socała
- Department of Animal Physiology and Pharmacology, Institute of Biological Sciences, Faculty of Biology and Biotechnology, Maria Curie-Skłodowska University, Akademicka 19, PL 20-033, Lublin, Poland.
| |
Collapse
|
8
|
Beurel E. Stress in the microbiome-immune crosstalk. Gut Microbes 2024; 16:2327409. [PMID: 38488630 PMCID: PMC10950285 DOI: 10.1080/19490976.2024.2327409] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 03/04/2024] [Indexed: 03/19/2024] Open
Abstract
The gut microbiota exerts a mutualistic interaction with the host in a fragile ecosystem and the host intestinal, neural, and immune cells. Perturbations of the gastrointestinal track composition after stress have profound consequences on the central nervous system and the immune system. Reciprocally, brain signals after stress affect the gut microbiota highlighting the bidirectional communication between the brain and the gut. Here, we focus on the potential role of inflammation in mediating stress-induced gut-brain changes and discuss the impact of several immune cells and inflammatory molecules of the gut-brain dialogue after stress. Understanding the impact of microbial changes on the immune system after stress might provide new avenues for therapy.
Collapse
Affiliation(s)
- Eléonore Beurel
- Department of Psychiatry and Behavioral Sciences, Miller School of Medicine, University of Miami, Miami, FL, USA
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
9
|
Tang J, Wei Y, Pi C, Zheng W, Zuo Y, Shi P, Chen J, Xiong L, Chen T, Liu H, Zhao Q, Yin S, Ren W, Cao P, Zeng N, Zhao L. The therapeutic value of bifidobacteria in cardiovascular disease. NPJ Biofilms Microbiomes 2023; 9:82. [PMID: 37903770 PMCID: PMC10616273 DOI: 10.1038/s41522-023-00448-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 10/03/2023] [Indexed: 11/01/2023] Open
Abstract
There has been an increase in cardiovascular morbidity and mortality over the past few decades, making cardiovascular disease (CVD) the leading cause of death worldwide. However, the pathogenesis of CVD is multi-factorial, complex, and not fully understood. The gut microbiome has long been recognized to play a critical role in maintaining the physiological and metabolic health of the host. Recent scientific advances have provided evidence that alterations in the gut microbiome and its metabolites have a profound influence on the development and progression of CVD. Among the trillions of microorganisms in the gut, bifidobacteria, which, interestingly, were found through the literature to play a key role not only in regulating gut microbiota function and metabolism, but also in reducing classical risk factors for CVD (e.g., obesity, hyperlipidemia, diabetes) by suppressing oxidative stress, improving immunomodulation, and correcting lipid, glucose, and cholesterol metabolism. This review explores the direct and indirect effects of bifidobacteria on the development of CVD and highlights its potential therapeutic value in hypertension, atherosclerosis, myocardial infarction, and heart failure. By describing the key role of Bifidobacterium in the link between gut microbiology and CVD, we aim to provide a theoretical basis for improving the subsequent clinical applications of Bifidobacterium and for the development of Bifidobacterium nutritional products.
Collapse
Affiliation(s)
- Jia Tang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Yumeng Wei
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Chao Pi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Central Nervous System Drug Key Laboratory of Sichuan Province, School of Pharmacy of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wenwu Zheng
- Department of Cardiology, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Ying Zuo
- Department of Comprehensive Medicine, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Shi
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Jinglin Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Linjin Xiong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Tao Chen
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Huiyang Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Qianjiao Zhao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Suyu Yin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy of Southwest Medical University, Luzhou, 646000, P.R. China
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Wei Ren
- National Traditional Chinese Medicine Clinical Research Base and Drug Research Center of the Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China
| | - Peng Cao
- The Affiliated Hospital of Traditional Chinese and Western Medicine Nanjing University of Chinese Medicine, Nanjing, Jiangsu, 210028, P.R. China.
| | - Nan Zeng
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
| | - Ling Zhao
- Luzhou Key Laboratory of Traditional Chinese Medicine for Chronic Diseases Jointly Built by Sichuan and Chongqing, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
- Chengdu University of Traditional Chinese Medicine State Key Laboratory of Southwestern Chinese Medicine Resources, 1166 Liutai Avenue, Wenjiang District, Chengdu, Sichuan, 611137, P.R. China.
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Development Planning Department of Southwest Medical University, Luzhou, Sichuan, 646000, P.R. China.
| |
Collapse
|
10
|
Ross K. Psychobiotics: Are they the future intervention for managing depression and anxiety? A literature review. Explore (NY) 2023; 19:669-680. [PMID: 36868988 PMCID: PMC9940471 DOI: 10.1016/j.explore.2023.02.007] [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: 08/29/2022] [Revised: 02/12/2023] [Accepted: 02/18/2023] [Indexed: 02/22/2023]
Abstract
Mental health is a public health concern among professional organizations, clinicians, and consumers alike, especially in light of the COVID-19 pandemic. Indeed, the World Health Organization has identified mental health as an epidemic of the 21st century contributing to the global health burden, which highlights the urgency to develop economical, accessible, minimally invasive interventions to effectively manage depression, anxiety, and stress. Nutritional approaches, including the use of probiotics and psychobiotics to manage depression and anxiety, have elicited interest in recent years. This review aimed to summarize evidence from studies including animal models, cell cultures, and human subjects. Overall, the current evidence suggests that 1) Specific strains of probiotics can reduce depressive symptoms and anxiety; 2) Symptoms may be reduced through one or more possible mechanisms of action, including impact on the synthesis of neurotransmitters such as serotonin and GABA, modulation of inflammatory cytokines, or enhancing stress responses through effects on stress hormones and the HPA axis; and 3) While psychobiotics may offer therapeutic benefits to manage depression and anxiety, further research, particularly human studies, is needed to better characterize their mode of action and understand optimal dosing in the context of nutritional interventions.
Collapse
Affiliation(s)
- Kim Ross
- Sonoran University of Health Sciences, 2140 E. Broadway Rd. Tempe, AZ 85282, United States.
| |
Collapse
|
11
|
Liu Q, Zhu Y, Li G, Guo T, Jin M, Xi D, Wang S, Liu X, Guo S, Liu H, Fan J, Liu R. Irisin ameliorates myocardial ischemia-reperfusion injury by modulating gut microbiota and intestinal permeability in rats. PLoS One 2023; 18:e0291022. [PMID: 37656700 PMCID: PMC10473488 DOI: 10.1371/journal.pone.0291022] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Accepted: 08/18/2023] [Indexed: 09/03/2023] Open
Abstract
Recently, myocardial ischemia-reperfusion (I/R) injury was suggested associated with intestinal flora. However, irisin has demonstrated beneficial effects on myocardial I/R injury, thus increasing interest in exploring its mechanism. Therefore, whether irisin interferes in gut microbiota and gut mucosal barrier during myocardial I/R injury was investigated in the present study. Irisin was found to reduce the infiltration of inflammatory cells and fracture in myocardial tissue, myocardial enzyme levels, and the myocardial infarction (MI) area. In addition, the data showed that irisin reverses I/R-induced gut dysbiosis as indicated by the decreased abundance of Actinobacteriota and the increased abundance of Firmicutes, and maintains intestinal barrier integrity, reduces metabolic endotoxemia, and inhibits the production of proinflammatory cytokines interleukin 1β (IL-1β), interleukin 6 (IL-6), and tumor necrosis factor α (TNF-α). Based on the results, irisin could be a good candidate for ameliorating myocardial I/R injury and associated diseases by alleviating gut dysbiosis, endothelial dysfunction and anti-inflammatory properties.
Collapse
Affiliation(s)
- Qingqing Liu
- LinFen Central Hospital, LinFen, China
- Linfen Key Laboratory of Basic and Clinical Research on Coronary Heart Disease, Linfen Clinical Medical Research Center, LinFen, China
| | - Yu Zhu
- LinFen Central Hospital, LinFen, China
- Linfen Key Laboratory of Basic and Clinical Research on Coronary Heart Disease, Linfen Clinical Medical Research Center, LinFen, China
- School of Sport Medicine and Rehabilitation, Beijing Sport University, Beijing, China
| | - Guangyao Li
- LinFen Central Hospital, LinFen, China
- Linfen Key Laboratory of Basic and Clinical Research on Coronary Heart Disease, Linfen Clinical Medical Research Center, LinFen, China
| | - Tiantian Guo
- LinFen Central Hospital, LinFen, China
- Linfen Key Laboratory of Basic and Clinical Research on Coronary Heart Disease, Linfen Clinical Medical Research Center, LinFen, China
| | - Mengtong Jin
- LinFen Central Hospital, LinFen, China
- Linfen Key Laboratory of Basic and Clinical Research on Coronary Heart Disease, Linfen Clinical Medical Research Center, LinFen, China
| | - Duan Xi
- LinFen Central Hospital, LinFen, China
| | | | - Xuezhi Liu
- LinFen Central Hospital, LinFen, China
- Linfen Key Laboratory of Basic and Clinical Research on Coronary Heart Disease, Linfen Clinical Medical Research Center, LinFen, China
- Department of Cardiovascular Surgery, Linfen Central Hospital, Linfen, China
| | - Shuming Guo
- LinFen Central Hospital, LinFen, China
- Linfen Key Laboratory of Basic and Clinical Research on Coronary Heart Disease, Linfen Clinical Medical Research Center, LinFen, China
| | - Hui Liu
- LinFen Central Hospital, LinFen, China
- Linfen Key Laboratory of Basic and Clinical Research on Coronary Heart Disease, Linfen Clinical Medical Research Center, LinFen, China
- Department of Cardiovascular Surgery, Linfen Central Hospital, Linfen, China
| | - Jiamao Fan
- LinFen Central Hospital, LinFen, China
- Linfen Key Laboratory of Basic and Clinical Research on Coronary Heart Disease, Linfen Clinical Medical Research Center, LinFen, China
- Department of Cardiology, Linfen Central Hospital, Linfen, China
| | - Ronghua Liu
- LinFen Central Hospital, LinFen, China
- Linfen Key Laboratory of Basic and Clinical Research on Coronary Heart Disease, Linfen Clinical Medical Research Center, LinFen, China
| |
Collapse
|
12
|
Torraville SE, Flynn CM, Kendall TL, Yuan Q. Life Experience Matters: Enrichment and Stress Can Influence the Likelihood of Developing Alzheimer's Disease via Gut Microbiome. Biomedicines 2023; 11:1884. [PMID: 37509523 PMCID: PMC10377385 DOI: 10.3390/biomedicines11071884] [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: 05/22/2023] [Revised: 06/21/2023] [Accepted: 06/30/2023] [Indexed: 07/30/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease, characterized by the presence of β-amyloid (Aβ) plaques and neurofibrillary tangles (NFTs) formed from abnormally phosphorylated tau proteins (ptau). To date, there is no cure for AD. Earlier therapeutic efforts have focused on the clinical stages of AD. Despite paramount efforts and costs, pharmaceutical interventions including antibody therapies targeting Aβ have largely failed. This highlights the need to alternate treatment strategies and a shift of focus to early pre-clinical stages. Approximately 25-40% of AD cases can be attributed to environmental factors including chronic stress. Gut dysbiosis has been associated with stress and the pathogenesis of AD and can increase both Aβ and NFTs in animal models of the disease. Both stress and enrichment have been shown to alter AD progression and gut health. Targeting stress-induced gut dysbiosis through probiotic supplementation could provide a promising intervention to delay disease progression. In this review, we discuss the effects of stress, enrichment, and gut dysbiosis in AD models and the promising evidence from probiotic intervention studies.
Collapse
Affiliation(s)
- Sarah E Torraville
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Cassandra M Flynn
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Tori L Kendall
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| | - Qi Yuan
- Biomedical Sciences, Faculty of Medicine, Memorial University of Newfoundland, St. John's, NL A1B 3V6, Canada
| |
Collapse
|
13
|
Shukla AK, Kumari A, Kumar A. Gut brain regulation using psychobiotics for improved neuropsychological illness. Dev Psychobiol 2023; 65:e22404. [PMID: 37338246 DOI: 10.1002/dev.22404] [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: 10/06/2022] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 06/21/2023]
Abstract
"Psychobiotics" are a novel class of probiotics that are beneficial to the health and functional efficiency of our brain and psychology. The main hold on command in ill conditions of the brain and psychology is overtaken by these psychobiotic bacteria (a dietary supplement) via the action/determined role of bacterial neurochemicals or neuroactive substances that are released by them in the intestinal epithelium after their ingestion. Although these psychobiotics flourish in the gut of the host consuming them, the effect is widely spread to the brain due to the communication between the gut and the brain via the bidirectional gut-brain axis. The nervous system involved in this directional process includes both the enteric nervous system and the central nervous system. With time, several corroborations have proved the effectiveness of psychobiotics in terms of mental illnesses and brain disorders. In the prevailing situation of the coronavirus pandemic, psychobiotics may serve as an aid because a majority of the population worldwide is already suffering from psychological issues due to changes in lifestyle and dietary habits, and in need of an immediate solution to cope with it. Moreover, the in silico approach is also vital for the development of biological relevance to neurosubstances.
Collapse
Affiliation(s)
- Adarsh Kumar Shukla
- Department of Nutrition Biology, Central University of Haryana, Mahendragarh, India
| | - Anita Kumari
- Department of Nutrition Biology, Central University of Haryana, Mahendragarh, India
| | - Ashwani Kumar
- Department of Nutrition Biology, Central University of Haryana, Mahendragarh, India
| |
Collapse
|
14
|
Li J, Wang J, Wang M, Zheng L, Cen Q, Wang F, Zhu L, Pang R, Zhang A. Bifidobacterium: a probiotic for the prevention and treatment of depression. Front Microbiol 2023; 14:1174800. [PMID: 37234527 PMCID: PMC10205982 DOI: 10.3389/fmicb.2023.1174800] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Depression is a common psychological disease, which has become one of the main factors affecting human health. It has a serious impact on individuals, families, and society. With the prevalence of COVID-19, the incidence of depression has further increased worldwide. It has been confirmed that probiotics play a role in preventing and treating depression. Especially, Bifidobacterium is the most widely used probiotic and has positive effects on the treatment of depression. The mechanisms underlying its antidepressant effects might include anti-inflammation and regulation of tryptophan metabolism, 5-hydroxytryptamine synthesis, and the hypothalamus-pituitary-adrenal axis. In this mini-review, the relationship between Bifidobacterium and depression was summarized. It is hoped that Bifidobacterium-related preparations would play a positive role in the prevention and treatment of depression in the future.
Collapse
Affiliation(s)
- Jiayu Li
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Junyu Wang
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Meiyu Wang
- Rehabilitation and Wellness Care Centre, Tianfu College of Swufe, Chengdu, China
| | - Li Zheng
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Qiuyu Cen
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Fangfang Wang
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Li Zhu
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Rizhao Pang
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People’s Hospital Affiliated to Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
15
|
Liao J, Zhang Y, Ma C, Wu G, Zhang W. Microbiome-metabolome reveals that the Suxiao Jiuxin pill attenuates acute myocardial infarction associated with fatty acid metabolism. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116529. [PMID: 37086873 DOI: 10.1016/j.jep.2023.116529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE The Suxiao Jiuxin pill (SJP) is a Chinese medical patent drug on the national essential drug list of China, with well-established cardiovascular protective effects in the clinic. However, the mechanisms underlying the protective effects of SJP on cardiovascular disease have not yet been elucidated clearly, especially its relationship with the gut microbiota. AIM OF THE STUDY This study aimed to investigate the cardioprotective effect of SJP against isoproterenol-induced acute myocardial infarction (AMI) by integrating the gut microbiome and metabolome. METHODS A rat model of AMI was generated using isoproterenol. Firstly, the effect of antibiotic (ABX) treatment on the blood absorption and excretion of the main components of SJP were studied. Secondly, 16S rRNA sequencing and untargeted metabolomics were used to discover the improvement of SJP treatment on gut microbiota and host metabolism in AMI rats. Finally, targeted metabolomics was used to verify the effects of SJP treatment on host metabolism in AMI rats. RESULT The results showed that ABX treatment could affect the blood absorption and fecal excretion of the main active components of SJP. At the same time, SJP can restore the richness and diversity of gut microbiota, and multiple gut microbiota (including Jeotgalicoccus, Lachnospiraceae, and Blautia) are significantly associated with fatty acids. Untargeted metabolomics also found that SJP could restore the levels of various fatty acid metabolites in serum and cecal contents (p < 0.01, FC > 1.5 and VIP >1). Targeted metabolomics further confirmed that 41, 21, and 39 fatty acids were significantly altered in serum, cecal contents, and heart samples, respectively. Interestingly, these fatty acids belong to the class of eicosanoids, and SJP can significantly downregulate these eicosanoids in AMI rats. CONCLUSION The results of this study suggest that SJP may exert its cardioprotective effects by remodeling the gut microbiota and host fatty acid metabolism.
Collapse
Affiliation(s)
- Jingyu Liao
- School of Pharmacy, Guangdong Pharmaceutical University, Guangdong, 510006, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yuhao Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Chi Ma
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Gaosong Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| | - Weidong Zhang
- School of Pharmacy, Guangdong Pharmaceutical University, Guangdong, 510006, China; Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China; School of Pharmacy, Second Military Medical University, Shanghai, 200433, China.
| |
Collapse
|
16
|
Gawlik-Kotelnicka O, Margulska A, Skowrońska A, Strzelecki D. PRO-DEMET Randomized Controlled Trial on Probiotics in Depression—Pilot Study Results. Nutrients 2023; 15:nu15061400. [PMID: 36986132 PMCID: PMC10058314 DOI: 10.3390/nu15061400] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/11/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023] Open
Abstract
There is a pressing need to identify new treatment options for depression and its comorbidities. Depression often coexists with metabolic complications, and the two may share a pathophysiological overlap, including inflammation and microbiota changes. Microbiota interventions (e.g., probiotics) may represent a safe and easy-to-use treatment option as an adjunctive therapy in patients only partially responsive to pharmacologic treatment. (1) Objective: The paper presents the results of a feasibility and pilot study. The study is an internal part of a randomized controlled trail (RCT) of the effect of probiotic supplementation on psychometric, anthropometric, metabolic, and inflammatory parameters in adult patients with depressive disorders depending on the presence of metabolic syndrome. (2) Methods: The trial has a four-arm, parallel-group, prospective, randomized, double-blind, controlled design. Sixty participants received a probiotic preparation containing Lactobacillus helveticus Rosell®-52 and Bifidobacterium longum Rosell®-175 over 60 days. The feasibility of the study design was assessed, as well as the rates of recruitment, eligibility, consent, and study completion. The following were assessed: depressive, anxiety and stress symptoms, quality of life, blood pressure, body mass index and waist circumference, complete blood count with differential, serum levels of C-reactive protein, high-density lipoprotein cholesterol, triglycerides, fasting glucose, some secondary markers of inflammation and metabolic health, as well as noninvasive biomarkers of liver fibrosis (APRI and FIB-4). (3) Results: The study was found to be generally feasible. The eligibility rate was 52% of recruited participants with 80% completing the study protocol. No differences in sociodemographic or anthropometric factors or basic laboratory findings were found between the placebo and probiotic group at the start of the intervention period. Importantly, the proportion of recruited participants fulfilling the criteria of metabolic syndrome was too low. (4) Conclusions: Whilst the whole study protocol was feasible, some different timepoint procedures require modification. The major weakness of the recruitment methods was that the percentage of metabolic arms participants was insufficient. Overall, the full RCT design on probiotics in depression with vs. without metabolic syndrome was shown to be feasible with little modification.
Collapse
Affiliation(s)
- Oliwia Gawlik-Kotelnicka
- Department of Affective and Psychotic Disorders, Medical University of Lodz, 92-216 Lodz, Poland
- Correspondence:
| | - Aleksandra Margulska
- Department of Adolescent Psychiatry, Medical University of Lodz, 92-216 Lodz, Poland
| | - Anna Skowrońska
- Department of Affective and Psychotic Disorders, Medical University of Lodz, 92-216 Lodz, Poland
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, 92-216 Lodz, Poland
| |
Collapse
|
17
|
Johnson D, Letchumanan V, Thum CC, Thurairajasingam S, Lee LH. A Microbial-Based Approach to Mental Health: The Potential of Probiotics in the Treatment of Depression. Nutrients 2023; 15:nu15061382. [PMID: 36986112 PMCID: PMC10053794 DOI: 10.3390/nu15061382] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/06/2023] [Accepted: 03/10/2023] [Indexed: 03/16/2023] Open
Abstract
Probiotics are currently the subject of intensive research pursuits and also represent a multi-billion-dollar global industry given their vast potential to improve human health. In addition, mental health represents a key domain of healthcare, which currently has limited, adverse-effect prone treatment options, and probiotics may hold the potential to be a novel, customizable treatment for depression. Clinical depression is a common, potentially debilitating condition that may be amenable to a precision psychiatry-based approach utilizing probiotics. Although our understanding has not yet reached a sufficient level, this could be a therapeutic approach that can be tailored for specific individuals with their own unique set of characteristics and health issues. Scientifically, the use of probiotics as a treatment for depression has a valid basis rooted in the microbiota-gut-brain axis (MGBA) mechanisms, which play a role in the pathophysiology of depression. In theory, probiotics appear to be ideal as adjunct therapeutics for major depressive disorder (MDD) and as stand-alone therapeutics for mild MDD and may potentially revolutionize the treatment of depressive disorders. Although there is a wide range of probiotics and an almost limitless range of therapeutic combinations, this review aims to narrow the focus to the most widely commercialized and studied strains, namely Lactobacillus and Bifidobacterium, and to bring together the arguments for their usage in patients with major depressive disorder (MDD). Clinicians, scientists, and industrialists are critical stakeholders in exploring this groundbreaking concept.
Collapse
Affiliation(s)
- Dinyadarshini Johnson
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Vengadesh Letchumanan
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Pathogen Resistome Virulome and Diagnostic Research Group (PathRiD), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
| | - Chern Choong Thum
- Department of Psychiatry, Hospital Sultan Abdul Aziz Shah, Persiaran Mardi-UPM, Serdang 43400, Malaysia
| | - Sivakumar Thurairajasingam
- Clinical School Johor Bahru, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Johor Bahru 80100, Malaysia
- Correspondence: (S.T.); or (L.-H.L.)
| | - Learn-Han Lee
- Novel Bacteria and Drug Discovery Research Group (NBDD), Microbiome and Bioresource Research Strength (MBRS), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Pathogen Resistome Virulome and Diagnostic Research Group (PathRiD), Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway 47500, Malaysia
- Correspondence: (S.T.); or (L.-H.L.)
| |
Collapse
|
18
|
Marques LS, Jung JT, Zborowski VA, Pinheiro RC, Nogueira CW, Zeni G. Emotional-Single Prolonged Stress: A promising model to illustrate the gut-brain interaction. Physiol Behav 2023; 260:114070. [PMID: 36574940 DOI: 10.1016/j.physbeh.2022.114070] [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: 06/17/2022] [Revised: 12/08/2022] [Accepted: 12/23/2022] [Indexed: 12/25/2022]
Abstract
Excessive stress can precipitate depression and anxiety diseases, and damage gastrointestinal functionality and microbiota changes, favoring the development of functional gastrointestinal disorders (FGIDs) - defined by dysregulation in the brain-gut interaction. Therefore, the present study investigated if Emotional-Single Prolonged Stress (E-SPS) induces depressive/anxiety-like phenotype and gut dysfunction in adult Swiss male mice. For this, mice of the E-SPS group were subjected to three stressors sequential exposure: immobilization, swimming, and odor of the predator for 7 days (incubation period). Next, animals performed behavior tests and 24 h later, samples of feces, blood, and colon tissue were collected. E-SPS increased the plasma corticosterone levels, immobility time in the tail suspension and forced swim test, decreased the grooming time in the splash test, OAT%, and OAE% in the elevated plus-maze test, as well as increased anxiety index. Mice of E-SPS had increased % of intestinal transit rate, % of fecal moisture content, and fecal pellets number, and decreased Claudin1 content in the colon. E-SPS decreased the relative abundance of Bacteroidetes phylum, Bacteroidia class, Bacteroidales order, Muribaculaceae and Porphyromonadaceae family, Muribaculum, and Duncaniella genus. However, E-SPS increased Firmicutes and Actinobacteria phylum, Coriobacteriales order, and the ratio of Firmicutes/Bacteroidetes, and demonstrated Mucispirillum genus presence. The present study showed that E-SPS induced depressive/anxiety-like phenotype, predominant diarrhea gut dysfunction, and modulated the gut bacterial microbiota profile in male adult Swiss mice. E-SPS might be a promising model for future studies on the brain-gut interaction and the development of FGIDs with psychological comorbidities.
Collapse
Affiliation(s)
- Luiza S Marques
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Juliano Tk Jung
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Vanessa A Zborowski
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Roberto C Pinheiro
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Cristina W Nogueira
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil
| | - Gilson Zeni
- Laboratory of Synthesis, Reactivity, Pharmacological and Toxicological Evaluation of Organochalcogen Compounds, Department of Biochemistry and Molecular Biology, Center of Natural and Exact Sciences, Federal University of Santa Maria, Santa Maria, RS 97105-900, Brazil.
| |
Collapse
|
19
|
Tanelian A, Nankova B, Cheriyan A, Arens C, Hu F, Sabban EL. Differences in gut microbiota associated with stress resilience and susceptibility to single prolonged stress in female rodents. Neurobiol Stress 2023; 24:100533. [PMID: 36970450 PMCID: PMC10034505 DOI: 10.1016/j.ynstr.2023.100533] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Exposure to traumatic stress is a major risk factor for the development of neuropsychiatric disorders in a subpopulation of individuals, whereas others remain resilient. The determinants of resilience and susceptibility remain unclear. Here, we aimed to characterize the microbial, immunological, and molecular differences between stress-susceptible and stress-resilient female rats before and after exposure to a traumatic experience. Animals were randomly divided into unstressed controls (n = 10) and experimental groups (n = 16) exposed to Single Prolonged Stress (SPS), an animal model of PTSD. Fourteen days later, all rats underwent a battery of behavioral tests and were sacrificed the following day to collect different organs. Stool samples were collected before and after SPS. Behavioral analyses revealed divergent responses to SPS. The SPS treated animals were further subdivided into SPS-resilient (SPS-R) and SPS-susceptible (SPS-S) subgroups. Comparative analysis of fecal 16S sequencing before and after SPS exposure indicated significant differences in the gut microbial composition, functionality, and metabolites of the SPS-R and SPS-S subgroups. In line with the observed distinct behavioral phenotypes, the SPS-S subgroup displayed higher blood-brain barrier permeability and neuroinflammation relative to the SPS-R and/or controls. These results indicate, for the first time, pre-existing and trauma-induced differences in the gut microbial composition and functionality of female rats that relate to their ability to cope with traumatic stress. Further characterization of these factors will be crucial for understanding susceptibility and fostering resilience, especially in females, who are more likely than males to develop mood disorders.
Collapse
|
20
|
The Influence of Probiotic Supplementation on the Severity of Anxiety and Depressive Symptoms; Function and Composition of Gut Microbiota; and Metabolic, Inflammation, and Oxidative Stress Markers in Patients with Depression-A Study Protocol. Metabolites 2023; 13:metabo13020182. [PMID: 36837799 PMCID: PMC9966580 DOI: 10.3390/metabo13020182] [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: 12/10/2022] [Revised: 01/11/2023] [Accepted: 01/20/2023] [Indexed: 01/27/2023] Open
Abstract
This article aims to present the theoretical basis, methodology, and design of a clinical trial we will conduct. The study will be prospective, randomized, placebo-controlled, and double-blind. Each intervention period will last 8 weeks and the trial will be conducted on 100 patients in total, who will be randomly divided into two groups consisting of 50 patients each. We plan to investigate the impact of Lactobacillus helveticus Rosell and Bifidobacterium longum Rosell on the depressive, anxiety, and stress levels in patients with depressive disorders with possible comorbid anxiety. In addition to assessing the influence of probiotics on the clinical condition, we also plan to study the clinical and biochemical parameters of metabolic syndrome, which often coexists with depression. Both depressive and metabolic issues may have part of their etiopathology in common, e.g., inflammation, oxidative stress, and dysbiosis. This is why we will additionally investigate the parameters related to gut microbiota, inflammatory, and oxidative statuses. Thus, the primary endpoint of the study will be the change in depression score measured with the Montgomery-Åsberg Depression Rating Scale. The secondary endpoints will include changes in anxiety and stress levels, as well as metabolic, inflammation, and oxidative stress parameters.
Collapse
|
21
|
QI S, LIU D, MA B, YANG L, YU H. Screening of the most efficacious lactic acid bacteria strain for myocardial infarction recovery and verification and exploration of its functions and mechanisms. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2023; 42:13-23. [PMID: 36660592 PMCID: PMC9816043 DOI: 10.12938/bmfh.2021-044] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 04/12/2022] [Indexed: 02/01/2023]
Abstract
Screening efficient strains by cell platform is cost-effective, but to date, no screening experiments have been performed for targeted lactic acid bacteria with hypoxic/reoxygenation (H/R)-treated cardiomyocytes, and their effects on the phosphoinositide 3-kinase (PI3K)/protein kinase b (Akt)/endothelial nitric oxide synthase (eNOS) pathway in myocardial infarction (MI) are unclear. Here we activated 102 strains of lactic acid bacteria and inoculated them into MRS medium for fermentation. The fermentation supernatants of the lactic acid bacteria were incubated with an H/R model of H9C2 cells. We found that Bifidobacterium longum ZL0210 had the greatest potential for inhibiting the apoptosis of H/R-induced H9C2 cells. Furthermore, it significantly increased the expression of heme oxygenase-1 (HO-1) and quinone oxidoreductase 1 (NQO1) in H9C2 cardiomyocytes, as well as the Bcl-2/Bax protein ratio, protecting damaged myocardial cells via an anti-apoptotic pathway. Intragastric administration of B. longum ZL0210 to mice for one week before and after establishment of an MI model drastically attenuated the myocardial cell hypertrophy and fibrosis of the MI mice. Meanwhile, B. longum ZL0210 significantly reduced the secretion of myocardial enzymes, increased the activity of antioxidant enzymes, and inhibited lipid-oxidative malondialdehyde (MDA) levels. Moreover, it upregulated the expression of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) protein and the phosphorylation levels of PI3K, Akt, and eNOS, resulting in increased NO contents. In summary, we screened 102 strains of lactic acid bacteria with a cell platform and determined that B. longum ZL0210 was a favorable candidate for protecting the myocardium. We are the first to reveal the protective effects of B. longum ZL0210 for MI via activation of the PI3K/Akt/eNOS pathway through TRAIL.
Collapse
Affiliation(s)
- Shanshan QI
- The Third Affiliated Hospital of Qiqihar Medical University,
No. 27 Taishun Street, Tiefeng District, Qiqihar, Heilongjiang, 161000, P.R. China
| | - Donghua LIU
- Qiqihar Jianhua Hospital, The Second Community Health Service
Center of Cultural Street, Qiqihar, Heilongjiang, 161000, P.R. China
| | - Bo MA
- The Third Affiliated Hospital of Qiqihar Medical University,
No. 27 Taishun Street, Tiefeng District, Qiqihar, Heilongjiang, 161000, P.R. China
| | - Lei YANG
- The Third Affiliated Hospital of Qiqihar Medical University,
No. 27 Taishun Street, Tiefeng District, Qiqihar, Heilongjiang, 161000, P.R. China
| | - Haitao YU
- The Third Affiliated Hospital of Qiqihar Medical University,
No. 27 Taishun Street, Tiefeng District, Qiqihar, Heilongjiang, 161000, P.R. China,*Corresponding author. Haitao Yu (E-mail: )
| |
Collapse
|
22
|
Dysbiotic microbiota contributes to the extent of acute myocardial infarction in rats. Sci Rep 2022; 12:16517. [PMID: 36192578 PMCID: PMC9530207 DOI: 10.1038/s41598-022-20826-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 09/19/2022] [Indexed: 11/26/2022] Open
Abstract
Increasing evidence suggests that the intestinal microbiota composition could play a role in specific pathologies such as hypertension, obesity and diabetes. This study aims to demonstrate that the intestinal microbiota modulated by a diet creating dysbiosis increased the size of the myocardial infarction and that probiotics could attenuate this effect. To do this, microbiota transplants from rats fed a dysbiotic or non-dysbiotic diet in the presence or absence of probiotics were performed for 10 days on rats whose microbiota had been previously suppressed by antibiotic therapy. Then, the anterior coronary artery of the transplanted rats was occluded for 30 min. Infarct size was measured after 24 h of reperfusion, while signaling pathways were evaluated after 15 min of reperfusion. Intestinal resistance, plasma concentration of LPS (lipopolysaccharides), activation of NF-κB and Akt and composition of the microbiota were also measured. Our results demonstrate a larger infarct size in animals transplanted with the dysbiotic microbiota without probiotics compared to the other groups, including those that received the dysbiotic microbiota with probiotics. This increase in infarct size correlates with a higher firmicutes/bacteroidetes ratio, NF-kB phosphorylation and plasma LPS concentration, and a decrease in intestinal barrier resistance and Akt. These results indicate that dysbiotic microbiota promotes an increase in infarct size, an effect that probiotics can attenuate.
Collapse
|
23
|
Probiotics in the Management of Mental and Gastrointestinal Post-COVID Symptomes. J Clin Med 2022; 11:jcm11175155. [PMID: 36079082 PMCID: PMC9457065 DOI: 10.3390/jcm11175155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 01/30/2023] Open
Abstract
Patients with “post-COVID” syndrome manifest with a variety of signs and symptoms that continue/develop after acute COVID-19. Among the most common are gastrointestinal (GI) and mental symptoms. The reason for symptom occurrence lies in the SARS-CoV-2 capability of binding to exact receptors, among other angiotensin converting enzyme 2 (ACE2) receptors in gastrointestinal lining and neuropilin-1 (NRP-1) in the nervous system, which leads to loss of gastrointestinal and blood-brain barriers integrity and function. The data are mounting that SARS-CoV-2 can trigger systemic inflammation and lead to disruption of gut-brain axis (GBA) and the development of disorders of gut brain interaction (DGBIs). Functional dyspepsia (FD) and irritable bowel syndrome (IBS) are the most common DGBIs syndromes. On the other hand, emotional disorders have also been demonstrated as DGBIs. Currently, there are no official recommendations or recommended procedures for the use of probiotics in patients with COVID-19. However, it can be assumed that many doctors, pharmacists, and patients will want to use a probiotic in the treatment of this disease. In such cases, strains with documented activity should be used. There is a constant need to plan and conduct new trials on the role of probiotics and verify their clinical efficacy for counteracting the negative consequences of COVID-19 pandemic. Quality control is another important but often neglected aspect in trials utilizing probiotics in various clinical entities. It determines the safety and efficacy of probiotics, which is of utmost importance in patients with post-acute COVID-19 syndrome.
Collapse
|
24
|
Attention-Deficit/Hyperactivity Disorder and the Gut Microbiota–Gut–Brain Axis: Closing Research Gaps through Female Inclusion in Study Design. WOMEN 2022. [DOI: 10.3390/women2030023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The gastrointestinal tract harbors a densely populated community of microbes that exhibits sexual dimorphism. Dysbiosis of this community has been associated with chronic human disease states ranging from metabolic diseases to neuropsychiatric disorders (NPDs). The gut microbiota–gut–brain axis (GMGBA) is a bi-directional pathway that facilitates the interaction of the gut microflora with host physiological functions. Recently, research surrounding the potential roles of the GMGBA in the development of NPDs (e.g., depression, anxiety, and autism spectrum disorders (ASDs)) has increased. However, the role of the GMGBA in attention-deficit/hyperactivity disorder (ADHD), an NPD that affects an estimated 8.4% of children (5.1% of female and 11.5% of male children) and 4% of adults (with a male–female odds ratio of 1.6) in the United States, remains understudied. Herein, we synthesize the current literature regarding the GMGBA, ADHD, and the potentially relevant intersections between the GMGBA and ADHD. Recommendations are presented for pathways of future research into the role(s) of the GMGBA in ADHD etiology and symptomatology. Particular focus is given to the potential for the variable of host sex to act as an outcome modifier of the relationship between the GMGBA and ADHD.
Collapse
|
25
|
Hashikawa-Hobara N, Otsuka A, Okujima C, Hashikawa N. Lactobacillus paragasseri OLL2809 Improves Depression-Like Behavior and Increases Beneficial Gut Microbes in Mice. Front Neurosci 2022; 16:918953. [PMID: 35837127 PMCID: PMC9274989 DOI: 10.3389/fnins.2022.918953] [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: 04/13/2022] [Accepted: 06/07/2022] [Indexed: 11/21/2022] Open
Abstract
Lactobacillus paragasseri OLL2809 is a probiotic bacterial strain isolated from healthy human feces. While OLL2809 has been studied for its immunomodulatory activities, its effect on depressive-like behaviors remains unclear. In this study, we used a mouse model of social defeat stress (SDS) to investigate whether oral administration of OLL2809 ameliorates depressive-like behavior. C57BL6 male mice were administered OLL2809 for 2 weeks following a 4-week period of SDS. Although OLL2809 did not affect serum corticosterone levels, it ameliorated depression-like behaviors, and it induced neurite outgrowth in the hippocampal dentate gyrus. The 16S rRNA amplicon sequence analyses revealed that family level gut microbiota composition was affected by stress and OLL2809 administration. Additionally, Akkermansia muciniphila, Bifidobacterium, and Lactobacillus were significantly increased by OLL2809 treatment. LEfSe analysis suggested that the antidepressive effect of OLL2809 may be mediated by increases in other microorganisms, such as Erysipelotrichaceae uncultured. Our findings suggest that L. paragasseri OLL2809 may have potential in microbiome therapeutics.
Collapse
|
26
|
Li Y, Kong D, Bi K, Luo H. Related Effects of Methamphetamine on the Intestinal Barrier via Cytokines, and Potential Mechanisms by Which Methamphetamine May Occur on the Brain-Gut Axis. Front Med (Lausanne) 2022; 9:783121. [PMID: 35620725 PMCID: PMC9128015 DOI: 10.3389/fmed.2022.783121] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 03/23/2022] [Indexed: 11/28/2022] Open
Abstract
Methamphetamine (METH) is an illegal drug widely abused in many countries. Methamphetamine abuse is a major health and social problem all over the world. However, the effects of METH on the digestive system have rarely been reported. Previous studies and clinical cases have shown that METH use can lead to the impaired intestinal barrier function and severe digestive diseases. METH can cause multiple organ dysfunction, especially in the central nervous system (CNS). The gut microbiota are involved in the development of various CNS-related diseases via the gut-brain axis (GBA). Here, we describe the related effects of METH on the intestinal barrier via cytokines and the underlying mechanisms by which METH may occur in the brain-gut axis.
Collapse
Affiliation(s)
- Yuansen Li
- Department of Intestine and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Deshenyue Kong
- Department of Intestine and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Ke Bi
- Department of Intestine and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China
| | - Huayou Luo
- Department of Intestine and Hernia Surgery, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,NHC Key Laboratory of Drug Addiction Medicine, Kunming Medical University, Kunming, China.,Yunnan Institute of Digestive Disease, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
27
|
Sasi M, Kumar S, Hasan M, S R A, Garcia-Gutierrez E, Kumari S, Prakash O, Nain L, Sachdev A, Dahuja A. Current trends in the development of soy-based foods containing probiotics and paving the path for soy-synbiotics. Crit Rev Food Sci Nutr 2022; 63:9995-10013. [PMID: 35611888 DOI: 10.1080/10408398.2022.2078272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the world of highly processed foods, special attention is drawn to the nutrient composition and safety of consumed food products. Foods fortified with probiotic bacteria confer beneficial effects on human health and are categorized as functional foods. The salubrious activities of probiotics include the synthesis of vital bioactives, prevention of inflammatory diseases, anticancerous, hypocholesterolemic, and antidiarrheal effects. Soy foods are exemplary delivery vehicles for probiotics and prebiotics and there are diverse strategies to enhance their functionality like employing mixed culture fermentation, engineering probiotics, and incorporating prebiotics in fermented soy foods. High potential is ascribed to the concurrent use of probiotics and prebiotics in one product, termed as "synbiotics," which implicates synergy, in which a prebiotic ingredient particularly favors the growth and activity of a probiotic micro-organism. The insights on emended bioactive profile, metabolic role, and potential health benefits of advanced soy-based probiotic and synbiotic hold a promise which can be profitably implemented to meet consumer needs. This article reviews the available knowledge about strategies to enhance the nutraceutical potential, mechanisms, and health-promoting effects of advanced soy-based probiotics. Traditional fermentation merged with diverse strategies to improve the efficiency and health benefits of probiotics considered vital, are also discussed.
Collapse
Affiliation(s)
- Minnu Sasi
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Sandeep Kumar
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
- Quality and Productivity Improvement Division, ICAR-Indian Institute of Natural Resins and Gums, Ranchi, India
| | - Muzaffar Hasan
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
- Agro Produce Processing Division, ICAR-Central Institute of Agricultural Engineering, Bhopal, India
| | - Arpitha S R
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | | | - Sweta Kumari
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Om Prakash
- National Centre for Microbial Resource (NCMR), National Centre for Cell Science, Pune, India
| | - Lata Nain
- Division of Microbiology, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Archana Sachdev
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Anil Dahuja
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| |
Collapse
|
28
|
Shor EK, Brown SP, Freeman DA. Bacteria and Bellicosity: Photoperiodic Shifts in Gut Microbiota Drive Seasonal Aggressive Behavior in Male Siberian Hamsters. J Biol Rhythms 2022; 37:296-309. [PMID: 35502701 DOI: 10.1177/07487304221092105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The existence of a microbiome-gut-brain axis has been established wherein gut microbiota significantly impacts host behavior and physiology, with increasing evidence suggesting a role for the gut microbiota in maintaining host homeostasis. Communication between the gut microbiota and the host is bidirectional, and shifts in the composition of the gut microbiota are dependent on both internal and external cues (host-derived signals, such as stress and immunity, and endocrine and environmental signals, such as photoperiod). Although there is host-driven seasonal variation in the composition of the microbiota, the mechanisms linking photoperiod, gut microbiota, and host behavior have not been characterized. The results of the present study suggest that seasonal changes in the gut microbiota drive seasonal changes in aggression. Implanting short-day Siberian hamsters (Phodopus sungorus) with fecal microbiota from long-day hamsters resulted in a reversal of seasonal aggression, whereby short-day hamsters displayed aggression levels typical of long-day hamsters. In addition, there are correlations between aggressive behavior and several bacterial taxa. These results implicate the gut microbiota as part of the photoperiodic mechanism regulating seasonal host behavior and contribute toward a more comprehensive understanding of the relationships between the microbiota, host, and environment.
Collapse
Affiliation(s)
- Elyan K Shor
- Department of Biological Sciences, Center for Biodiversity Research, The University of Memphis, Memphis, Tennessee, USA
| | - Shawn P Brown
- Department of Biological Sciences, Center for Biodiversity Research, The University of Memphis, Memphis, Tennessee, USA
| | - David A Freeman
- Department of Biological Sciences, Center for Biodiversity Research, The University of Memphis, Memphis, Tennessee, USA
| |
Collapse
|
29
|
Zhao K, Yao M, Zhang X, Xu F, Shao X, Wei Y, Wang H. Flavonoids and intestinal microbes interact to alleviate depression. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:1311-1318. [PMID: 34625972 DOI: 10.1002/jsfa.11578] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 09/03/2021] [Accepted: 10/08/2021] [Indexed: 06/13/2023]
Abstract
Flavonoids have a variety of biological activities that are beneficial to human health. However, owing to low bioavailability, most flavonoids exert beneficial effects in the intestine through metabolism by the flora into a variety of structurally different derivatives. Also, flavonoids can modulate the type and structure of intestinal microorganisms to improve human health. It has been reported that the development of depression is accompanied by changes in the type and number of intestinal microorganisms, and gut microbes can significantly improve depressive symptoms through the gut-brain axis. Therefore, the interaction between flavonoids and intestinal microbes to alleviate depression is discussed. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ke Zhao
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Mei Yao
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Xin Zhang
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Feng Xu
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Xingfeng Shao
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Yingying Wei
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| | - Hongfei Wang
- Department of Food Science and Engineering, Ningbo University, Ningbo, P.R. China
| |
Collapse
|
30
|
One Giant Leap from Mouse to Man: The Microbiota-Gut-Brain Axis in Mood Disorders and Translational Challenges Moving towards Human Clinical Trials. Nutrients 2022; 14:nu14030568. [PMID: 35276927 PMCID: PMC8840472 DOI: 10.3390/nu14030568] [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: 12/24/2021] [Revised: 01/24/2022] [Accepted: 01/26/2022] [Indexed: 12/13/2022] Open
Abstract
The microbiota–gut–brain axis is a bidirectional communication pathway that enables the gut microbiota to communicate with the brain through direct and indirect signaling pathways to influence brain physiology, function, and even behavior. Research has shown that probiotics can improve several aspects of health by changing the environment within the gut, and several lines of evidence now indicate a beneficial effect of probiotics on mental and brain health. Such evidence has prompted the arrival of a new term to the world of biotics research: psychobiotics, defined as any exogenous influence whose effect on mental health is bacterially mediated. Several taxonomic changes in the gut microbiota have been reported in neurodevelopmental disorders, mood disorders such as anxiety and depression, and neurodegenerative disorders such as Alzheimer’s disease. While clinical evidence supporting the role of the gut microbiota in mental and brain health, and indeed demonstrating the beneficial effects of probiotics is rapidly accumulating, most of the evidence to date has emerged from preclinical studies employing different animal models. The purpose of this review is to focus on the role of probiotics and the microbiota–gut–brain axis in relation to mood disorders and to review the current translational challenges from preclinical to clinical research.
Collapse
|
31
|
Chen J, Chen X, Ho CL. Recent Development of Probiotic Bifidobacteria for Treating Human Diseases. Front Bioeng Biotechnol 2022; 9:770248. [PMID: 35004640 PMCID: PMC8727868 DOI: 10.3389/fbioe.2021.770248] [Citation(s) in RCA: 65] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Accepted: 12/08/2021] [Indexed: 12/12/2022] Open
Abstract
Bifidobacterium is a non-spore-forming, Gram-positive, anaerobic probiotic actinobacterium and commonly found in the gut of infants and the uterine region of pregnant mothers. Like all probiotics, Bifidobacteria confer health benefits on the host when administered in adequate amounts, showing multifaceted probiotic effects. Examples include B. bifidum, B. breve, and B. longum, common Bifidobacterium strains employed to prevent and treat gastrointestinal disorders, including intestinal infections and cancers. Herein, we review the latest development in probiotic Bifidobacteria research, including studies on the therapeutic impact of Bifidobacterial species on human health and recent efforts in engineering Bifidobacterium. This review article would provide readers with a wholesome understanding of Bifidobacteria and its potentials to improve human health.
Collapse
Affiliation(s)
- Jun Chen
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China
| | - Xinyi Chen
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China
| | - Chun Loong Ho
- Department of Biomedical Engineering, Southern University of Science and Technology (SUSTech), Shenzhen, China
| |
Collapse
|
32
|
What do experimental animal models of mood disorders tell clinicians about influence of probiotics on the gut-brain axis? POSTEP HIG MED DOSW 2022. [DOI: 10.2478/ahem-2022-0042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
It is commonly pointed out that enteric microbiota have a significant impact on the behavioral and neurophysiological parameters relevant to brain-gut axis disorders. Accordingly, many data have demonstrated that probiotics can alter the central nervous system function via this gut-brain axis and commensal bacteria consumption can ameliorate stress-related neuropsychiatric disorders. Thus, modulating the enteric microbiota is increasingly considered a new therapeutic approach for these disorders, although so far there is a lack of reliable pre-clinical and clinical data confirming the usefulness of probiotics in the treatment of affective disorders. In this review, we discuss various mechanisms linking specific probiotic bacteria with behaviors related to anhedonia and the exact mechanisms of their action, including data provided by using animal models and tests. Finally, we point to potential clinical impact resulting from future studies investigating the gut-brain axis activity with respect to the efficacy of probiotic treatment of mental disorders.
Collapse
|
33
|
Zheng Y, Bek MK, Prince NZ, Peralta Marzal LN, Garssen J, Perez Pardo P, Kraneveld AD. The Role of Bacterial-Derived Aromatic Amino Acids Metabolites Relevant in Autism Spectrum Disorders: A Comprehensive Review. Front Neurosci 2021; 15:738220. [PMID: 34744609 PMCID: PMC8568365 DOI: 10.3389/fnins.2021.738220] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Accepted: 09/30/2021] [Indexed: 12/27/2022] Open
Abstract
In recent years, the idea of the gut microbiota being involved in the pathogenesis of autism spectrum disorders (ASD) has attracted attention through numerous studies. Many of these studies report microbial dysregulation in the gut and feces of autistic patients and in ASD animal models. The host microbiota plays a large role in metabolism of ingested foods, and through the production of a range of metabolites it may be involved in neurodevelopmental disorders such as ASD. Two specific microbiota-derived host metabolites, p-cresol sulfate and 4-ethylphenyl sulfate, have been associated with ASD in both patients and animal models. These metabolites originate from bacterially produced p-cresol and 4-ethylphenol, respectively. p-Cresol and 4-ethylphenol are produced through aromatic amino acid fermentation by a range of commensal bacteria, most notably bacteria from the Clostridioides genus, which are among the dysregulated bacteria frequently detected in ASD patients. Once produced, these metabolites are suggested to enter the bloodstream, pass the blood–brain-barrier and affect microglial cells in the central nervous system, possibly affecting processes like neuroinflammation and microglial phagocytosis. This review describes the current knowledge of microbial dysbiosis in ASD and elaborates on the relevance and synthesis pathways of two specific ASD-associated metabolites that may form a link between the microbiota and the brain in autism. While the two discussed metabolites are promising candidates for biomarkers and (nutritional) intervention targets, more research into the role of these metabolites in ASD is required to causally connect these metabolites to ASD pathophysiology.
Collapse
Affiliation(s)
- Yuanpeng Zheng
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Marie K Bek
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Naika Z Prince
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Lucia N Peralta Marzal
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands.,Global Centre of Excellence Immunology, Danone Nutricia Research, Utrecht, Netherlands
| | - Paula Perez Pardo
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| | - Aletta D Kraneveld
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
34
|
Lim EY, Song EJ, Kim JG, Jung SY, Lee SY, Shin HS, Nam YD, Kim YT. Lactobacillus intestinalis YT2 restores the gut microbiota and improves menopausal symptoms in ovariectomized rats. Benef Microbes 2021; 12:503-516. [PMID: 34463192 DOI: 10.3920/bm2020.0217] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
There are many studies focusing on the alleviation of menopausal symptoms; however, little is known about the role of gut microorganisms in menopausal symptoms. Ovariectomized (OVX) rats were administered a novel strain (YT2) of Lactobacillus intestinalis (a species with significantly reduced abundance in OVX rats) and the potential probiotic effect on the improvement of menopausal symptoms was evaluated. Of note, the gut microbial composition completely shifted after ovariectomy in rats. Treatment with L. intestinalis YT2 significantly alleviated menopausal symptoms, such as increased fat mass, decreased bone mineral density, increased pain sensitivity, depression-like behaviour, and cognitive impairment. Additionally, the administration of L. intestinalis YT2 restored the intestinal microbial composition, including an increased Firmicutes/Bacteroides ratio. L. intestinalis YT2 also promoted gut barrier integrity by increasing the mRNA levels of tight junction-related markers. In conclusion, L. intestinalis YT2 treatment alleviated menopausal symptoms via the modulation of the gut microbiota. Importantly, these results suggest that L. intestinalis YT2 should be considered as a therapeutic probiotic agent for menopausal women.
Collapse
Affiliation(s)
- E Y Lim
- Division of Food Functionality, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea.,Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - E-J Song
- Division of Food Functionality, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - J G Kim
- Food Functional Evaluation Support Team, Technical Assistance Center, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - S Y Jung
- Division of Food Functionality, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea.,Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - S-Y Lee
- Division of Food Functionality, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea.,Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - H S Shin
- Division of Food Functionality, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea.,Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Y-D Nam
- Division of Food Functionality, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea
| | - Y T Kim
- Division of Food Functionality, Korea Food Research Institute, Wanju-gun 55365, Republic of Korea.,Department of Food Biotechnology, Korea University of Science and Technology, Daejeon 34113, Republic of Korea
| |
Collapse
|
35
|
Ge T, Yao X, Zhao H, Yang W, Zou X, Peng F, Li B, Cui R. Gut microbiota and neuropsychiatric disorders: Implications for neuroendocrine-immune regulation. Pharmacol Res 2021; 173:105909. [PMID: 34543739 DOI: 10.1016/j.phrs.2021.105909] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/08/2021] [Accepted: 09/15/2021] [Indexed: 12/17/2022]
Abstract
Recently, increasing evidence has shown gut microbiota dysbiosis might be implicated in the physiological mechanisms of neuropsychiatric disorders. Altered microbial community composition, diversity and distribution traits have been reported in neuropsychiatric disorders. However, the exact pathways by which the intestinal microbiota contribute to neuropsychiatric disorders remain largely unknown. Given that the onset and progression of neuropsychiatric disorders are characterized with complicated alterations of neuroendocrine and immunology, both of which can be continually affected by gut microbiota via "microbiome-gut-brain axis". Thus, we assess the complicated crosstalk between neuroendocrine and immunological regulation might underlie the mechanisms of gut microbiota associated with neuropsychiatric disorders. In this review, we summarized clinical and preclinical evidence on the role of the gut microbiota in neuropsychiatry disorders, especially in mood disorders and neurodevelopmental disorders. This review may elaborate the potential mechanisms of gut microbiota implicating in neuroendocrine-immune regulation and provide a comprehensive understanding of physiological mechanisms for neuropsychiatric disorders.
Collapse
Affiliation(s)
- Tongtong Ge
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Xiaoxiao Yao
- Department of Hepatopancreatobiliary Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Haisheng Zhao
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Wei Yang
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Xiaohan Zou
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Fanzhen Peng
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China
| | - Ranji Cui
- Jilin Provincial Key Laboratory on Molecular and Che mical Genetic, Second Hospital of Jilin University, Changchun, China.
| |
Collapse
|
36
|
Forouzan S, McGrew K, Kosten TA. Drugs and bugs: Negative affect, psychostimulant use and withdrawal, and the microbiome. Am J Addict 2021; 30:525-538. [PMID: 34414622 DOI: 10.1111/ajad.13210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND OBJECTIVES A growing body of literature demonstrates that the human microbiota plays a crucial role in health and disease states, as well as in the body's response to stress. In addition, the microbiome plays a role in psychological well-being and regulating negative affect. Regulation of negative affect is a factor in psychostimulant abuse disorders. We propose a risk chain in which stress leads to negative affect that places an individual at risk to develop or relapse to psychostimulant abuse disorder. Stress, negative affect, and psychostimulant use all alter the gut microbiome. METHODS This review brings together the literature on affective disorders, stress, and psychostimulant abuse disorders to assess possible modulatory actions of the gut-brain axis to regulate these conditions. RESULTS Studies reviewed across the various disciplines suggest that the dysbiosis resulting from drug use, drug withdrawal, or stress may cause an individual to be more susceptible to addiction and relapse. Probiotics and prebiotics reduce stress and negative affect. SCIENTIFIC SIGNIFICANCE Treatment during the withdrawal phase of psychostimulant abuse disorder, when the microbiome is altered, may ameliorate the symptoms of stress and negative affect leading to a reduced risk of relapse to psychostimulant use.
Collapse
Affiliation(s)
- Shadab Forouzan
- Department of Psychology, Texas Institute for Measurement, Evaluation and Statistics (TIMES), University of Houston, Houston, Texas, USA
| | - Keely McGrew
- Department of Psychology, Texas Institute for Measurement, Evaluation and Statistics (TIMES), University of Houston, Houston, Texas, USA
| | - Therese A Kosten
- Department of Psychology, Texas Institute for Measurement, Evaluation and Statistics (TIMES), University of Houston, Houston, Texas, USA
| |
Collapse
|
37
|
Wu L, Shen F, Wang W, Qi C, Wang C, Shang A, Xuan S. The effect of multispecies probiotics on cognitive reactivity to sad mood in patients with Crohn’s disease. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
38
|
Yang Y, Li X, Chen S, Xiao M, Liu Z, Li J, Cheng Y. Mechanism and therapeutic strategies of depression after myocardial infarction. Psychopharmacology (Berl) 2021; 238:1401-1415. [PMID: 33594503 DOI: 10.1007/s00213-021-05784-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 02/04/2021] [Indexed: 01/08/2023]
Abstract
Depression resulted as an important factor associated with the myocardial infarction (MI) prognosis. Patients with MI also have a higher risk for developing depression. Although the issue of depression after MI has become a matter of clinical concern, the molecular mechanism underlying depression after MI remains unclear, whereby several strategies suggested have not got ideal effects, such as selective serotonin reuptake inhibitors. In this review, we summarized and discussed the occurrence mechanism of depression after MI, such as 5-hydroxytryptamine (5-HT) dysfunction, altered hypothalamus-pituitary-adrenal (HPA) axis function, gut microbiota imbalance, exosomal signal transduction, and inflammation. In addition, we offered a succinct overview of treatment, as well as some promising molecules especially from natural products for the treatment of depression after MI.
Collapse
Affiliation(s)
- Ying Yang
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Xuping Li
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Sixuan Chen
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Mingzhu Xiao
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Zhongqiu Liu
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China
| | - Jingyan Li
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China.
| | - Yuanyuan Cheng
- Guangdong Key Laboratory for Translational Cancer Research of Chinese Medicine, Joint Laboratory for Translational Cancer Research of Chinese Medicine of the Ministry of Education of the People's Republic of China, International Institute for Translational Chinese Medicine, School of Pharmaceutical Science, Guangzhou University of Chinese Medicine, Guangzhou, 510006, Guangdong, China.
| |
Collapse
|
39
|
Methiwala HN, Vaidya B, Addanki VK, Bishnoi M, Sharma SS, Kondepudi KK. Gut microbiota in mental health and depression: role of pre/pro/synbiotics in their modulation. Food Funct 2021; 12:4284-4314. [PMID: 33955443 DOI: 10.1039/d0fo02855j] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The microbiome residing in the human gut performs a wide range of biological functions. Recently, it has been elucidated that a change in dietary habits is associated with alteration in the gut microflora which results in increased health risks and vulnerability towards various diseases. Falling in line with the same concept, depression has also been shown to increase its prevalence around the globe, especially in the western world. Various research studies have suggested that changes in the gut microbiome profile further result in decreased tolerance of stress. Although currently available medications help in relieving the symptoms of depressive disorders briefly, these drugs are not able to completely reverse the multifactorial pathology of depression. The discovery of the communication pathway between gut microbes and the brain, i.e. the Gut-Brain Axis, has led to new areas of research to find more effective and safer alternatives to current antidepressants. The use of probiotics and prebiotics has been suggested as being effective in various preclinical studies and clinical trials for depression. Therefore, in the present review, we address the new antidepressant mechanisms via gut microbe alterations and provide insight into how these can provide an alternative to antidepressant therapy without the side effects and risk of adverse drug reactions.
Collapse
Affiliation(s)
- Hasnain N Methiwala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India.
| | | | | | | | | | | |
Collapse
|
40
|
Gawlik-Kotelnicka O, Strzelecki D. Probiotics as a Treatment for "Metabolic Depression"? A Rationale for Future Studies. Pharmaceuticals (Basel) 2021; 14:ph14040384. [PMID: 33924064 PMCID: PMC8074252 DOI: 10.3390/ph14040384] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/16/2021] [Accepted: 04/18/2021] [Indexed: 02/06/2023] Open
Abstract
Depression and metabolic diseases often coexist, having several features in common, e.g., chronic low-grade inflammation and intestinal dysbiosis. Different microbiota interventions have been proposed to be used as a treatment for these disorders. In the paper, we review the efficacy of probiotics in depressive disorders, obesity, metabolic syndrome and its liver equivalent based on the published experimental studies, clinical trials and meta-analyses. Probiotics seem to be effective in reducing depressive symptoms when administered in addition to antidepressants. Additionally, probiotics intake may ameliorate some of the clinical components of metabolic diseases. However, standardized methodology regarding probiotics use in clinical trials has not been established yet. In this narrative review, we discuss current knowledge on the recently used methodology with its strengths and limitations and propose criteria that may be implemented to create a new study of the effectiveness of probiotics in depressive disorders comorbid with metabolic abnormalities. We put across our choice on type of study population, probiotics genus, strains, dosages and formulations, intervention period, as well as primary and secondary outcome measures.
Collapse
|
41
|
Gawlik-Kotelnicka O, Skowrońska A, Margulska A, Czarnecka-Chrebelska KH, Łoniewski I, Skonieczna-Żydecka K, Strzelecki D. The Influence of Probiotic Supplementation on Depressive Symptoms, Inflammation, and Oxidative Stress Parameters and Fecal Microbiota in Patients with Depression Depending on Metabolic Syndrome Comorbidity-PRO-DEMET Randomized Study Protocol. J Clin Med 2021; 10:jcm10071342. [PMID: 33804999 PMCID: PMC8036404 DOI: 10.3390/jcm10071342] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 03/17/2021] [Accepted: 03/20/2021] [Indexed: 12/14/2022] Open
Abstract
There is a huge need to search for new treatment options and potential biomarkers of therapeutic response to antidepressant treatment. Depression and metabolic syndrome often coexist, while a pathophysiological overlap, including microbiota changes, may play a role. The paper presents a study protocol that aims to assess the effect of probiotic supplementation on symptoms of depression, anxiety and stress, metabolic parameters, inflammatory and oxidative stress markers, as well as fecal microbiota in adult patients with depressive disorders depending on the co-occurrence of metabolic syndrome. The trial will be a four-arm, parallel-group, prospective, randomized, double-blind, controlled design that will include 200 participants and will last 20 weeks (ClinicalTrials.gov identifier: NCT04756544). The probiotic preparation will contain Lactobacillus helveticus Rosell®-52, Bifidobacterium longum Rosell®-175. We will assess the level of depression, anxiety and stress, quality of life, blood pressure, body mass index and waist circumference, white blood cells count, serum levels of C-reactive protein, high-density lipoprotein (HDL) cholesterol, triglycerides, fasting glucose, fecal microbiota composition and the level of some fecal microbiota metabolites, as well as serum inflammatory markers and oxidative stress parameters. The proposed trial may establish a safe and easy-to-use adjunctive treatment option in a subpopulation of depressive patients only partially responsive to pharmacologic therapy.
Collapse
Affiliation(s)
- Oliwia Gawlik-Kotelnicka
- Department of Affective and Psychotic Disorders, Medical University of Lodz, 92-216 Lodz, Poland; (A.S.); (D.S.)
- Correspondence:
| | - Anna Skowrońska
- Department of Affective and Psychotic Disorders, Medical University of Lodz, 92-216 Lodz, Poland; (A.S.); (D.S.)
| | - Aleksandra Margulska
- Admission Department, Central Teaching Hospital of Medical University of Lodz, 92-216 Lodz, Poland;
| | | | - Igor Łoniewski
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (I.Ł.); (K.S.-Ż.)
| | - Karolina Skonieczna-Żydecka
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, 71-460 Szczecin, Poland; (I.Ł.); (K.S.-Ż.)
| | - Dominik Strzelecki
- Department of Affective and Psychotic Disorders, Medical University of Lodz, 92-216 Lodz, Poland; (A.S.); (D.S.)
| |
Collapse
|
42
|
Trzeciak P, Herbet M. Role of the Intestinal Microbiome, Intestinal Barrier and Psychobiotics in Depression. Nutrients 2021; 13:927. [PMID: 33809367 PMCID: PMC8000572 DOI: 10.3390/nu13030927] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/07/2021] [Accepted: 03/08/2021] [Indexed: 12/18/2022] Open
Abstract
The intestinal microbiota plays an important role in the pathophysiology of depression. As determined, the microbiota influences the shaping and modulation of the functioning of the gut-brain axis. The intestinal microbiota has a significant impact on processes related to neurotransmitter synthesis, the myelination of neurons in the prefrontal cortex, and is also involved in the development of the amygdala and hippocampus. Intestinal bacteria are also a source of vitamins, the deficiency of which is believed to be related to the response to antidepressant therapy and may lead to exacerbation of depressive symptoms. Additionally, it is known that, in periods of excessive activation of stress reactions, the immune system also plays an important role, negatively affecting the tightness of the intestinal barrier and intestinal microflora. In this review, we have summarized the role of the gut microbiota, its metabolites, and diet in susceptibility to depression. We also describe abnormalities in the functioning of the intestinal barrier caused by increased activity of the immune system in response to stressors. Moreover, the presented study discusses the role of psychobiotics in the prevention and treatment of depression through their influence on the intestinal barrier, immune processes, and functioning of the nervous system.
Collapse
Affiliation(s)
| | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8b Street, 20-090 Lublin, Poland;
| |
Collapse
|
43
|
Tremblay A, Lingrand L, Maillard M, Feuz B, Tompkins TA. The effects of psychobiotics on the microbiota-gut-brain axis in early-life stress and neuropsychiatric disorders. Prog Neuropsychopharmacol Biol Psychiatry 2021; 105:110142. [PMID: 33069817 DOI: 10.1016/j.pnpbp.2020.110142] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 09/28/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Psychobiotics are considered among potential avenues for modulating the bidirectional communication between the gastrointestinal tract and central nervous system, defined as the microbiota-gut-brain axis (MGBA). Even though causality has not yet been established, intestinal dysbiosis has emerged as a hallmark of several diseases, including neuropsychiatric disorders (NPDs). The fact that the microbiota and central nervous system are co-developing during the first years of life has provided a paradigm suggesting a potential role of psychobiotics for earlier interventions. Studies in animal models of early-life stress (ELS) have shown that they can counteract the pervasive effects of stress during this crucial developmental period, and rescue behavioral symptoms related to anxiety and depression later in life. In humans, evidence from clinical studies on the efficacy of psychobiotics at improving mental outcomes in most NPDs remain limited, except for major depressive disorder for which more studies are available. Consequently, the beneficial effect of psychobiotics on depression-related outcomes in adults are becoming clearer. While the specific mechanisms at play remain elusive, the effect of psychobiotics are generally considered to involve the hypothalamic-pituitary-adrenal axis, intestinal permeability, and inflammation. It is anticipated that future clinical studies will explore the potential role of psychobiotics at mitigating the risk developing NPDs in vulnerable individuals or in the context of childhood adversity. However, such studies remain challenging at present in terms of design and target populations; the profound impact of stress on the proper development of the MGBA during the first year of life is becoming increasingly recognized, but the trajectories post-ELS in humans and the mechanisms by which stress affects the susceptibility to various NPDs are still ill-defined. As psychobiotics are likely to exert both shared and specific mechanisms, a better definition of target subpopulations would allow to tailor psychobiotics selection by aligning mechanistic properties with known pathophysiological mechanisms or risk factors. Here we review the available evidence from clinical and preclinical studies supporting a role for psychobiotics at ameliorating depression-related outcomes, highlighting the knowledge gaps and challenges associated with conducting longitudinal studies to address outstanding key questions in the field.
Collapse
Affiliation(s)
- Annie Tremblay
- Rosell® Institute for Microbiome and Probiotics, 6100 Royalmount Avenue, Montreal, Quebec H4P 2R2, Canada
| | - Lucie Lingrand
- Lallemand Health Solutions, 19 Rue des Briquetiers, 31702 Blagnac, France
| | - Morgane Maillard
- Lallemand Health Solutions, 19 Rue des Briquetiers, 31702 Blagnac, France
| | - Berengere Feuz
- Lallemand Health Solutions, 19 Rue des Briquetiers, 31702 Blagnac, France
| | - Thomas A Tompkins
- Rosell® Institute for Microbiome and Probiotics, 6100 Royalmount Avenue, Montreal, Quebec H4P 2R2, Canada.
| |
Collapse
|
44
|
Qiu X, Wu G, Wang L, Tan Y, Song Z. Lactobacillus delbrueckii alleviates depression-like behavior through inhibiting toll-like receptor 4 (TLR4) signaling in mice. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:366. [PMID: 33842587 PMCID: PMC8033381 DOI: 10.21037/atm-20-4411] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background The intestinal flora can influence behavior through the microbiota-gut-brain axis and is closely related to the occurrence and development of nervous system diseases such as depression. Probiotics like Lactobacillus may regulate the balance of the intestinal flora and play an active role in preventing and treating depression. Methods Eight-week-old C57BL/6J mice (n=32) were randomly and equally divided into a normal control group, a control + Lac group, a model group, and a model + Lac group. The model and model + Lac groups were intraperitoneally injected with 1.2 mg/kg lipopolysaccharide for 7 days, and the behavior of the mice was assessed 24 hours later. The normal and model groups received intragastric administration of saline daily, while the control + Lac and model + Lac groups were given 109 cfu Lac intragastrically daily for 7 days. The inhibitory effect of Lac and its fermentation products on depression-related bacteria were examined in vitro. Results Lac effectively inhibited the production of depression-like behaviors in mice. The expression levels of zonula occludens-1 (ZO-1) and E-cadherin in the small intestine in the model group were significantly decreased, but Lac abrogated this effect. Overactivation of microglia and decreased expression of dopamine transporter (DAT) in brain tissues, which are closely related to depression, were also abrogated by Lac treatment. Furthermore, the expression of toll-like receptor 4 (TLR4) and nod-like receptor protein-3 (NLRP3), as well as the level of interleukin-1 beta (IL-1β) in the intestine and brain, were all significantly increased; however, these effects were subsequently abrogated by Lac. Moreover, Lac inhibited dysbiosis through its metabolites. Conclusions Lac has a remarkable antidepressant function, which it performs through the inhibition of dysbiosis (via its metabolites) and pattern recognition receptor TLR4 signaling.
Collapse
Affiliation(s)
- Xiangjie Qiu
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Guojun Wu
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Lili Wang
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Yurong Tan
- Department of Medical Microbiology, Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhi Song
- Department of General Surgery, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
45
|
Partrick KA, Rosenhauer AM, Auger J, Arnold AR, Ronczkowski NM, Jackson LM, Lord MN, Abdulla SM, Chassaing B, Huhman KL. Ingestion of probiotic (Lactobacillus helveticus and Bifidobacterium longum) alters intestinal microbial structure and behavioral expression following social defeat stress. Sci Rep 2021; 11:3763. [PMID: 33580118 PMCID: PMC7881201 DOI: 10.1038/s41598-021-83284-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 01/28/2021] [Indexed: 02/08/2023] Open
Abstract
Social stress exacerbates anxious and depressive behaviors in humans. Similarly, anxiety- and depressive-like behaviors are triggered by social stress in a variety of non-human animals. Here, we tested whether oral administration of the putative anxiolytic probiotic strains Lactobacillus helveticus R0052 and Bifidobacterium longum R0175 reduces the striking increase in anxiety-like behavior and changes in gut microbiota observed following social defeat stress in Syrian hamsters. We administered the probiotic at two different doses for 21 days, and 16S rRNA gene amplicon sequencing revealed a shift in microbial structure following probiotic administration at both doses, independently of stress. Probiotic administration at either dose increased anti-inflammatory cytokines IL-4, IL-5, and IL-10 compared to placebo. Surprisingly, probiotic administration at the low dose, equivalent to the one used in humans, significantly increased social avoidance and decreased social interaction. This behavioral change was associated with a reduction in microbial richness in this group. Together, these results demonstrate that probiotic administration alters gut microbial composition and may promote an anti-inflammatory profile but that these changes may not promote reductions in behavioral responses to social stress.
Collapse
Affiliation(s)
- Katherine A Partrick
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Anna M Rosenhauer
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Jérémie Auger
- Rosell Institute for Microbiome and Probiotics, Montreal, QC, Canada
| | - Amanda R Arnold
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Nicole M Ronczkowski
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Lanaya M Jackson
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Magen N Lord
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Sara M Abdulla
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA
| | - Benoit Chassaing
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA.,INSERM U1016, Team "Mucosal Microbiota in Chronic Inflammatory Diseases", CNRS UMR 8104, Université de Paris, Paris, France.,Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, USA
| | - Kim L Huhman
- Neuroscience Institute, Center for Behavioral Neuroscience, Georgia State University, PO Box 5030, Atlanta, GA, 30303-5030, USA.
| |
Collapse
|
46
|
Abildgaard A, Kern T, Pedersen O, Hansen T, Lund S, Wegener G. A diet-induced gut microbiota component and related plasma metabolites are associated with depressive-like behaviour in rats. Eur Neuropsychopharmacol 2021; 43:10-21. [PMID: 32933808 DOI: 10.1016/j.euroneuro.2020.09.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 08/30/2020] [Accepted: 09/04/2020] [Indexed: 02/07/2023]
Abstract
It is well-established in preclinical studies that various probiotics may improve behaviours related to psychiatric disease. We have previously shown that probiotics protected against high-fat diet (HFD)-induced depressive-like behaviour in Flinders Sensitive Line (FSL) rats, whereas FSL rats on control (CON) diet were unaffected. Therefore, we hypothesised that a dysmetabolic component of depression may exist that involves the gut microbiota and that such component may be reflected in the plasma metabolome. The aims of the present study post hoc analyses were 1) to study the effect of probiotics on gut microbiota composition and its association with depressive-like behaviour in FSL rats, and 2) to identify plasma metabolites associated with gut microbiota and depressive-like behaviour. Forty-six FSL rats were fed CON or HFD and treated with multi-species probiotics (nine Bifidobacterium, Lactococcus and Lactobacillus species) for 12 weeks. Faecal samples were collected for 16S rRNA (VR4) gene amplicon sequencing (Illumina MiSeq), and an untargeted plasma metabolomics was performed. We found that probiotics increased the relative faecal abundance of the Bifidobacterium, Lactococcus and Lactobacillus genera in HFD-fed rats only. Also, a HFD-induced microbiota component associated with depressive-like behaviour was identified, and probiotics improved the component score. Finally, the plasma levels of 44 metabolites correlated with the depression-related microbiota component, and three such metabolites had good predictive ability for depressive-like behaviour. Potentially, our findings imply that a subtype of depression characterised by a diet-induced, pro-depressant gut microbiota may exist and that analysis of related plasma metabolites may reveal aberrant microbiota functioning related to depression.
Collapse
Affiliation(s)
- Anders Abildgaard
- Translational Neuropsychiatry Unit, Aarhus University, Skovagervej 2, Risskov, Denmark; Department of Clinical Biochemistry, Aarhus University Hospital, Palle Juul-Jensens Boulevard 99, Aarhus N, Denmark.
| | - Timo Kern
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, Blegdamsvej 3B, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, Blegdamsvej 3B, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, Blegdamsvej 3B, University of Copenhagen, 2200 Copenhagen, Denmark
| | - Sten Lund
- Steno Diabetes Centre, Aarhus University Hospital, Hedeager 3, Aarhus N, Denmark
| | - Gregers Wegener
- Translational Neuropsychiatry Unit, Aarhus University, Skovagervej 2, Risskov, Denmark
| |
Collapse
|
47
|
Microbiota, a New Playground for the Omega-3 Polyunsaturated Fatty Acids in Cardiovascular Diseases. Mar Drugs 2021; 19:md19020054. [PMID: 33498729 PMCID: PMC7931107 DOI: 10.3390/md19020054] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/19/2021] [Accepted: 01/20/2021] [Indexed: 02/06/2023] Open
Abstract
Several cardioprotective mechanisms attributed to Omega-3 polyunsaturated fatty acids (PUFAs) have been studied and widely documented. However, in recent years, studies have supported the concept that the intestinal microbiota can play a much larger role than we had anticipated. Microbiota could contribute to several pathologies, including cardiovascular diseases. Indeed, an imbalance in the microbiota has often been reported in patients with cardiovascular disease and produces low-level inflammation. This inflammation contributes to, more or less, long-term development of cardiovascular diseases. It can also worsen the symptoms and the consequences of these pathologies. According to some studies, omega-3 PUFAs in the diet could restore this imbalance and mitigate its harmful effects on cardiovascular diseases. Many mechanisms are involved and included: (1) a reduction of bacteria producing trimethylamine (TMA); (2) an increase in bacteria producing butyrate, which has anti-inflammatory properties; and (3) a decrease in the production of pro-inflammatory cytokines. Additionally, omega-3 PUFAs would help maintain better integrity in the intestinal barrier, thereby preventing the translocation of intestinal contents into circulation. This review will summarize the effects of omega-3 PUFAs on gut micro-biota and the potential impact on cardiac health.
Collapse
|
48
|
Gut-brain axis: A matter of concern in neuropsychiatric disorders…! Prog Neuropsychopharmacol Biol Psychiatry 2021; 104:110051. [PMID: 32758517 DOI: 10.1016/j.pnpbp.2020.110051] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 07/25/2020] [Accepted: 07/26/2020] [Indexed: 01/09/2023]
Abstract
The gut microbiota is composed of a large number of microbes, usually regarded as commensal bacteria. It has become gradually clear that gastrointestinal microbiota affects gut pathophysiology and the central nervous system (CNS) function by modulating the signaling pathways of the microbiota-gut-brain (MGB) axis. This bidirectional MGB axis communication primarily acts through neuroendocrine, neuroimmune, and autonomic nervous systems (ANS) mechanisms. Accumulating evidence reveals that gut microbiota interacts with the host brain, and its modulation may play a critical role in the pathology of neuropsychiatric disorders. Recently, neuroscience research has established the significance of gut microbiota in the development of brain systems that are essential to stress-related behaviors, including depression and anxiety. Application of modulators of the MGB, such as psychobiotics (e.g., probiotics), prebiotics, and specific diets, may be a promising therapeutic approach for neuropsychiatric disorders. The present review article primarily focuses on the relevant features of the disturbances of the MGB axis in the pathophysiology of neuropsychiatric disorders and its potential mechanisms.
Collapse
|
49
|
Forouzan S, Hoffman KL, Kosten TA. Methamphetamine exposure and its cessation alter gut microbiota and induce depressive-like behavioral effects on rats. Psychopharmacology (Berl) 2021; 238:281-292. [PMID: 33097978 DOI: 10.1007/s00213-020-05681-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/07/2020] [Indexed: 02/06/2023]
Abstract
RATIONALE Methamphetamine is a highly abused psychostimulant drug and its use remains a major public health concern worldwide with limited effective treatment options. Accumulative evidence reveals the influence of gut microbiota on the brain, behavior, and health as a part of the gut-brain axis but its involvement in modulating this substance use disorder remains poorly understood. OBJECTIVE We sought to determine whether methamphetamine exposure and cessation or withdrawal alter the intestinal gut microbiota as well as characterize cessation-induced behavioral changes. METHODS Male, Sprague-Dawley rats were administered methamphetamine (2 mg/kg; s.c.) or vehicle (n = 8 per group) twice per day for 14 consecutive days. On various days before, during, and after administration, fecal samples were collected and tests of anxiety- and depressive-like behaviors were conducted. RESULTS Methamphetamine administration and cessation did not alter the relative abundance of bacteria but significantly changed the composition of gut bacteria through 16S rRNA sequencing. These changes were normalized after 7 days of methamphetamine cessation. Moreover, acute methamphetamine cessation induced depressive-like behavior, with an increase in immobility in the forced swim test but did not alter anxiety-like behaviors in tests of open field test or elevated plus maze. CONCLUSIONS These findings provide direct evidence that methamphetamine and its cessation cause gut dysbiosis and that the latter associates with depressive-like behavior in rodents. Our observation will contribute to a better understanding of the function of gut microbiota in the process of substance use disorders and guide the choice of target therapeutics.
Collapse
Affiliation(s)
- Shadab Forouzan
- Department of Psychology, Texas Institute for Measurement, Evaluation and Statistics (TIMES), University of Houston, Health and Biomedical Sciences Building 1, 4849 Calhoun Road, Houston, TX, 77204-6022, USA
| | - Kristi L Hoffman
- Molecular VIrology and Microbiology Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Therese A Kosten
- Department of Psychology, Texas Institute for Measurement, Evaluation and Statistics (TIMES), University of Houston, Health and Biomedical Sciences Building 1, 4849 Calhoun Road, Houston, TX, 77204-6022, USA.
| |
Collapse
|
50
|
Lewis CV, Taylor WR. Intestinal barrier dysfunction as a therapeutic target for cardiovascular disease. Am J Physiol Heart Circ Physiol 2020; 319:H1227-H1233. [PMID: 32986965 PMCID: PMC7792706 DOI: 10.1152/ajpheart.00612.2020] [Citation(s) in RCA: 68] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 09/16/2020] [Accepted: 09/27/2020] [Indexed: 02/06/2023]
Abstract
The gut microbiome and intestinal dysfunction have emerged as potential contributors to the development of cardiovascular disease (CVD). Alterations in gut microbiome are well documented in hypertension, atherosclerosis, and heart failure and have been investigated as a therapeutic target. However, a perhaps underappreciated but related role for intestinal barrier function has become evident. Increased intestinal permeability is observed in patients and mouse models of CVD. This increased intestinal permeability can enhance systemic inflammation, alter gut immune function, and has been demonstrated as predictive of adverse cardiovascular outcomes. The goal of this review is to examine the evidence supporting a role for intestinal barrier function in cardiovascular disease and its prospect as a novel therapeutic target. We outline key studies that have investigated intestinal permeability in hypertension, coronary artery disease, atherosclerosis, heart failure, and myocardial infarction. We highlight the central mechanisms involved in the breakdown of barrier function and look at emerging evidence for restored barrier function as a contributor to promising treatment strategies such as short chain fatty acid, probiotic, and renin angiotensin system-targeted therapeutics. Recent studies of more selective targeting of the intestinal barrier to improve disease outcomes are also examined. We suggest that although current data supporting a contribution of intestinal permeability to CVD pathogenesis are largely associative, it appears to be a promising avenue for further investigation. Additional studies of the mechanisms of barrier restoration in CVD and testing of intestinal barrier-targeted compounds will be required to confirm their potential as a new class of CVD therapeutic.
Collapse
Affiliation(s)
- Caitlin V Lewis
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - W Robert Taylor
- Division of Cardiology, Department of Medicine, Emory University School of Medicine, Atlanta, Georgia
- Cardiology Division, Atlanta Veterans Affairs Medical Center, Atlanta, Georgia
- Department of Biomedical Engineering, Emory University School of Medicine and Georgia Institute of Technology, Atlanta, Georgia
| |
Collapse
|