1
|
Rosell-Cardona C, Collins MK, O'Riordan KJ, Goodson MS, Kelley-Loughnane N, Cryan JF, Clarke G. Acute stress enhances synaptic plasticity in male mice via a microbiota-dependent mechanism. Neuropharmacology 2025; 273:110434. [PMID: 40154944 DOI: 10.1016/j.neuropharm.2025.110434] [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: 01/07/2025] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/01/2025]
Abstract
Acute stress can enhance or impair synaptic plasticity depending on the nature, duration, and type of stress exposure as well as the brain region examined. The absence of a gut microbiome can also alter hippocampal plasticity. However, the possible interplay between synaptic plasticity, acute stress, and the gut microbiota remains unknown. Here, we examine this interaction and determine whether the gut microbiota impacts stress-induced alterations in hippocampal plasticity. Further, we explored whether exposure to the microbial metabolite butyrate is sufficient to counteract stress-induced alterations in synaptic plasticity. We used electrophysiological and molecular experiments in adult male C57/BL6 antibiotic-treated and acutely stressed mice. In electrophysiological experiments we treated hippocampal slices with 3 μM sodium butyrate to explore the effect of this microbial metabolite. We found the presence of the microbiota essential for the enhancement of both short- and long-term potentiation induced by 15 min of acute restraint stress. Furthermore, butyrate exposure effectively restored the stress-induced enhancement of potentiation in slices from microbiome-depleted animals while also enhancing long-term potentiation independent of stress. In addition, alterations of hippocampal synaptic plasticity markers were noted. Our findings highlight a critical new temporal role for gut-derived metabolites in defining the impact of acute stress on synaptic plasticity.
Collapse
Affiliation(s)
| | - Michael K Collins
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | | | - Michael S Goodson
- 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, OH, USA
| | - Nancy Kelley-Loughnane
- 711th Human Performance Wing, Air Force Research Laboratory, Wright-Patterson Air Force Base, Dayton, OH, USA
| | - John F Cryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Gerard Clarke
- APC Microbiome Ireland, University College Cork, Cork, Ireland; Department of Psychiatry and Neurobehavioural Science, University College Cork, Cork, Ireland.
| |
Collapse
|
2
|
Kiko H, Asaoka D, Nomura O, Nomoto Y, Sugano K, Matsuno K, Homma Y, Nishizaki Y, Yanagisawa N, Takeda T, Abe D, Oki S, Suzuki N, Akazawa Y, Ueda K, Ueyama H, Hojo M, Nagahara A, Isayama H, Miyauchi K. Association of Constipation and Geriatric Depressive Symptoms: Cross-Sectional Analysis Using Baseline Data from the JUSTICE-TOKYO Prospective Cohort Study. Diagnostics (Basel) 2025; 15:1537. [PMID: 40564857 PMCID: PMC12192452 DOI: 10.3390/diagnostics15121537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 06/05/2025] [Accepted: 06/13/2025] [Indexed: 06/28/2025] Open
Abstract
Objective: To clarify the relationship between constipation and depressive symptoms among the elderly. Methods: This single-center, cross-sectional study was performed using baseline data obtained at the time of enrollment in the prospective cohort of the JUSTICE-TOKYO study. Participants underwent assessments including patient profiling, drug use history, the Mini-Mental State Examination (MMSE), the Geriatric Depression Scale 15 (GDS-15), gastrointestinal-related quality of life (QOL), and the constipation scoring system (CSS). Geriatric depressive symptoms were evaluated based on GDS-15. We investigated correlations between GDS-15 scores and various abdominal symptoms and assessed risk factors for geriatric depressive symptoms using multiple regression analysis. Results: A total of 984 elderly participants (57% female, mean age 78.1 ± 6.1 year) were included. The GDS-15 scores were significantly correlated with body mass index (BMI) (r = -0.056) and MMSE (r = -0.092), reflex-related QOL (r = 0.253), pain-related QOL (r = 0.229), fullness-related QOL (r = 0.269), constipation-related QOL (r = 0.329), diarrhea-related QOL (r = 0.264), and CSS (r = 0.285) scores. Multiple regression analysis indicated that BMI (β = -0.069, p = 0.020) and MMSE (β = -0.074, p = 0.013), constipation-related QOL (β = 0.136, p = 0.002), reflex-related QOL (β = 0.126, p < 0.001), diarrhea-related QOL (β = 0.095, p = 0.006), and CSS (β = 0.098, p = 0.016) scores were significantly correlated with GDS-15 scores. Conclusions: Depressive symptoms among older individuals are associated with various abdominal symptoms, particularly constipation. However, the causality between depressive symptoms and constipation cannot be inferred due to the study's cross-sectional design.
Collapse
Affiliation(s)
- Hiroyuki Kiko
- Department of Gastroenterology, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan; (H.K.); (O.N.); (Y.N.)
| | - Daisuke Asaoka
- Department of Gastroenterology, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan; (H.K.); (O.N.); (Y.N.)
| | - Osamu Nomura
- Department of Gastroenterology, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan; (H.K.); (O.N.); (Y.N.)
| | - Yusuke Nomoto
- Department of Gastroenterology, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan; (H.K.); (O.N.); (Y.N.)
| | - Koji Sugano
- Department of Respiratory Medicine, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan; (K.S.); (K.M.)
| | - Kei Matsuno
- Department of Respiratory Medicine, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan; (K.S.); (K.M.)
| | - Yasuhiro Homma
- Department of Orthopedic Surgery, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan;
| | - Yuji Nishizaki
- Medical Technology Innovation Center, Juntendo University, Tokyo 113-8421, Japan; (Y.N.); (N.Y.)
| | - Naotake Yanagisawa
- Medical Technology Innovation Center, Juntendo University, Tokyo 113-8421, Japan; (Y.N.); (N.Y.)
| | - Tsutomu Takeda
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo 113-8421, Japan; (T.T.); (D.A.); (S.O.); (N.S.); (Y.A.); (K.U.); (H.U.); (M.H.); (A.N.); (H.I.)
| | - Daiki Abe
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo 113-8421, Japan; (T.T.); (D.A.); (S.O.); (N.S.); (Y.A.); (K.U.); (H.U.); (M.H.); (A.N.); (H.I.)
| | - Shotaro Oki
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo 113-8421, Japan; (T.T.); (D.A.); (S.O.); (N.S.); (Y.A.); (K.U.); (H.U.); (M.H.); (A.N.); (H.I.)
| | - Nobuyuki Suzuki
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo 113-8421, Japan; (T.T.); (D.A.); (S.O.); (N.S.); (Y.A.); (K.U.); (H.U.); (M.H.); (A.N.); (H.I.)
| | - Yoichi Akazawa
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo 113-8421, Japan; (T.T.); (D.A.); (S.O.); (N.S.); (Y.A.); (K.U.); (H.U.); (M.H.); (A.N.); (H.I.)
| | - Kumiko Ueda
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo 113-8421, Japan; (T.T.); (D.A.); (S.O.); (N.S.); (Y.A.); (K.U.); (H.U.); (M.H.); (A.N.); (H.I.)
| | - Hiroya Ueyama
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo 113-8421, Japan; (T.T.); (D.A.); (S.O.); (N.S.); (Y.A.); (K.U.); (H.U.); (M.H.); (A.N.); (H.I.)
| | - Mariko Hojo
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo 113-8421, Japan; (T.T.); (D.A.); (S.O.); (N.S.); (Y.A.); (K.U.); (H.U.); (M.H.); (A.N.); (H.I.)
| | - Akihito Nagahara
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo 113-8421, Japan; (T.T.); (D.A.); (S.O.); (N.S.); (Y.A.); (K.U.); (H.U.); (M.H.); (A.N.); (H.I.)
| | - Hiroyuki Isayama
- Department of Gastroenterology, Juntendo University School of Medicine, Tokyo 113-8421, Japan; (T.T.); (D.A.); (S.O.); (N.S.); (Y.A.); (K.U.); (H.U.); (M.H.); (A.N.); (H.I.)
| | - Katsumi Miyauchi
- Department of Cardiology, Juntendo Tokyo Koto Geriatric Medical Center, Tokyo 136-0075, Japan;
| |
Collapse
|
3
|
Qin N, Xie X, Deng R, Gao S, Zhu T. The role of the tryptophan metabolites in gut microbiota-brain axis and potential treatments: a focus on ischemic stroke. Front Pharmacol 2025; 16:1578018. [PMID: 40556758 PMCID: PMC12185428 DOI: 10.3389/fphar.2025.1578018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2025] [Accepted: 05/06/2025] [Indexed: 06/28/2025] Open
Abstract
Gut microbiota disturbances can elevate the risk of stroke by contributing to cerebrovascular events. Particularly, the gut tryptophan (TRP) metabolite is an essential mediator of the gut-brain axis. This review highlights the role of TRP metabolism in stroke, the influence of intestinal microbiomes on stroke pathology via TRP metabolism, and the gut-brain axis interactions. Recent studies indicate that various bioactive molecules produced via TRP metabolism can regulate various neurological functions and interrupt stroke pathophysiology. Moreover, the relationship between gut TRP metabolism and stroke development has been verified. TRP metabolism involves three pathways: kynurenine, 5-hydroxytryptamine, and indole, which potentially regulate post-stroke, may function as aryl hydrocarbon receptor agonists to modify neuronal excitotoxicity, and offer crucial targets for stroke treatment. This suggests that modulating TRP metabolite levels through various methods can enhance the prognosis of central nervous system diseases and restore microbiota-gut-brain axis functions.
Collapse
Affiliation(s)
- Na Qin
- Institute of Neuroregeneration & Neurorehabilitation, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Xiaodi Xie
- Institute of Neuroregeneration & Neurorehabilitation, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Rong Deng
- Institute of Neuroregeneration & Neurorehabilitation, Qingdao Medical College, Qingdao University, Qingdao, China
| | - Shiman Gao
- Women and Children’s Hospital, Qingdao University, Qingdao, China
| | - Ting Zhu
- Institute of Neuroregeneration & Neurorehabilitation, Qingdao Medical College, Qingdao University, Qingdao, China
| |
Collapse
|
4
|
Firoozi D, Masoumi SJ, Mohammad-Kazem Hosseini Asl S, Fararouei M, Jamshidi S. Effects of Short Chain Fatty Acid-Butyrate Supplementation on the Disease Severity, Inflammation, and Psychological Factors in Patients With Active Ulcerative Colitis: A Double-Blind Randomized Controlled Trial. J Nutr Metab 2025; 2025:3165876. [PMID: 40123849 PMCID: PMC11930386 DOI: 10.1155/jnme/3165876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 02/13/2025] [Indexed: 03/25/2025] Open
Abstract
Background: Depression and anxiety are common in UC patients due to gut microbiota dysbiosis and increased proinflammatory markers. Butyrate, a short-chain fatty acid, participates in the regulation of gut microbiota and inflammation and has neuroprotective effects in neurodegenerative disease. Therefore, we assessed the effects of sodium butyrate supplementation on the disease severity, inflammation, and psychological factors in active UC patients. Methods: This study was a randomized, parallel, double-blind controlled trial. Participants in the intervention (n = 18) and control (n = 18) groups received 600 mg/kg of sodium butyrate or rice starch as a placebo with their main meal, respectively, for 12 weeks. The partial Mayo score was used to evaluate disease severity, while the Westergren method was employed to assess the erythrocyte sedimentation rate (ESR). NLR and PLR were determined using an automated analyzer (XS-500i, Sysmex). Moreover, the psychological factors were assessed by the hospital anxiety depression scale (HADS) and the general health questionnaire (GHQ). Results: In comparison with placebo, sodium-butyrate supplementation significantly decreased the ESR level (-6.66 ± 1.56 vs. 3.00 ± 2.11, p=0.01), NLR (-0.24 ± 0.1 vs. 0.33 ± 0.23, p=0.02), Mayo score (-2.33 ± 0.41 vs. 0.22 ± 0.40, p < 0.001), HADS anxiety score (-2.77 ± 0.64 vs. 0.94 ± 0.63, p=0.001), HADS depression score (-2.38 ± 0.47 vs. 0.61 ± 0.33, p < 0.001), and GHQ total score (-12.11 ± 1.48 vs. 3.55 ± 1.39, p < 0.001). Conclusion: Butyrate could serve as an effective adjuvant treatment for reducing disease severity and alleviating psychological symptoms. This trial was registered on the Research Ethics Committee of Shiraz University of Medical Sciences, with the reference number IR.SUMS.SCHEANUT.REC.1400.037. Trial Registration: Iranian Registry of Clinical Trials: IRCT20211214053401N1.
Collapse
Affiliation(s)
- Donya Firoozi
- Student Research Committee, School of Nutrition and Food Sciences, Shiraz University of Medical Sciences, Shiraz, Iran
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Science, Shiraz, Iran
| | - Seyed Jalil Masoumi
- Nutrition Research Center, School of Nutrition and Food Sciences, Shiraz University of Medical Science, Shiraz, Iran
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | - Mohammad Fararouei
- Department of Epidemiology, School of Public Health, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Sanaz Jamshidi
- Center for Cohort Study of Shiraz University of Medical Sciences Employees' Health, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
5
|
Li H, Meng H, Dang C, Liu P, Liu J, Yu X, Wang Z, Lilv, Sui X. Exploring potential causal relationships between gut microbiota, inflammatory factors, and postpartum depression: a Mendelian randomization analysis. BMC Pregnancy Childbirth 2025; 25:177. [PMID: 39962387 PMCID: PMC11834640 DOI: 10.1186/s12884-025-07304-w] [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: 05/03/2024] [Accepted: 02/07/2025] [Indexed: 02/20/2025] Open
Abstract
BACKGROUND Recent studies have suggested a potential correlation between ecological dysregulation of the gut microbiota (GM) and the onset and development of postpartum depression (PPD). In addition, inflammatory factors (IFs) have been reported to play an important role in the development of PPD. However, the causal connections among GM, IFs, and PPD remain to be understood. OBJECTIVE This study sought to determine if genetically predicted GM and IFs exert a causal effect on PPD and to study whether IFs mediate the causal effect of GM on PPD. METHODS Two-step and two-sample Mendelian randomization (MR) analyses, primarily employing the inverse variance weighted (IVW) method, were conducted to evaluate the causal relationship between GM, IFs, and PPD, and to assess potential mediating effects. Heterogeneity and horizontal pleiotropy tests were performed to evaluate the robustness of the findings and the strength of the causal associations. RESULTS Class Alphaproteobacteria, genus Family XIII AD3011 group exhibited a positive association with PPD risk; whereas, the family Clostridiales vadin BB60 group, family Veillonellaceae, genus Ruminococcaceae UCG011, and the inflammatory factors C-C motif chemokine ligand 5 (CCL5) and C-C motif chemokine ligand 3 (CCL3) demonstrated negative correlations with PPD risk. IFs did not exhibit a mediating role. No heterogeneity or horizontal pleiotropy was observed. CONCLUSIONS Our MR study offered genetic evidence that GM and IFs contribute to the pathogenesis of PPD, with no mediating effect of IFs. This enhances our understanding of PPD's pathological mechanisms and offers new perspectives for developing novel preventative and therapeutic strategies.
Collapse
Affiliation(s)
- Hui Li
- Shandong University of Traditional Chinese Medicine, Jingshi Road, Jinan, Shandong, 250014, China
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road, Jinan, Shandong, 250014, China
| | - Hongyan Meng
- Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jingshi Road, Jinan, Shandong, 250014, China
| | - Chunxiao Dang
- Shandong University of Traditional Chinese Medicine, Jingshi Road, Jinan, Shandong, 250014, China
| | - Pengfei Liu
- Shandong University of Traditional Chinese Medicine, Jingshi Road, Jinan, Shandong, 250014, China
| | - Jinxing Liu
- Shandong University of Traditional Chinese Medicine, Jingshi Road, Jinan, Shandong, 250014, China
| | - Xiao Yu
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road, Jinan, Shandong, 250014, China
| | - Zhonglin Wang
- Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jingshi Road, Jinan, Shandong, 250014, China.
| | - Lilv
- Qilu Hospital of Shandong University, Wenhua West Road, Jinan, Shandong, 250014, China.
| | - Xiaohui Sui
- Shandong University of Traditional Chinese Medicine, Jingshi Road, Jinan, Shandong, 250014, China
| |
Collapse
|
6
|
Zhang X, Hu H, Zhang Y, Hu S, Lu J, Peng W, Luo D. Dietary Capsaicin Exacerbates Gut Microbiota Dysbiosis and Mental Disorders in Type 1 Diabetes Mice. Nutrients 2025; 17:593. [PMID: 39940450 PMCID: PMC11821225 DOI: 10.3390/nu17030593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 01/27/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Background/Objectives: Diabetes mellitus is often accompanied by mental health complications, including anxiety, depression, and cognitive decline. Recent research suggested that capsaicin, the active component of chili peppers, may influence mental health. This study aimed to determine the effect of dietary capsaicin on mental disorders in a type 1 diabetes (T1D) mouse model, while also exploring the potential involvement of the microbiota-gut-brain axis. Methods: We induced T1D in mice using streptozotocin (STZ) and administered a diet supplemented with 0.005% capsaicin for five weeks. Behavioral assessments, including the open field test (OFT), tail suspension test (TST), forced swimming test (FST), elevated plus maze (EPM) test, and Morris water maze (MWM) test, were conducted to evaluate depressive and anxiety-like behaviors as well as cognitive function. Targeted and untargeted metabolomics analyses were performed to assess neurotransmitter levels in the hippocampus and serum metabolites, while 16S rRNA sequencing was utilized to analyze gut microbiota composition. Intestinal barriers were determined using western blot detection of the tight junction proteins ZO-1 and occludin. Results: Dietary capsaicin exacerbated anxiety and depressive-like behaviors along with cognitive declines in T1D mice. Capsaicin reduced gut microbiota diversity and levels of beneficial bacteria, while broad-spectrum antibiotic treatment further intensified anxiety and depression behaviors. Metabolomic analysis indicated that capsaicin disrupted metabolic pathways related to tryptophan and phenylalanine, leading to decreased neuroprotective metabolites, such as kynurenic acid, hippurate, and butyric acid. Additionally, capsaicin diminished the expression of ZO-1 and occludin, indicating increased intestinal permeability. Conclusions: Dietary capsaicin aggravates gut microbiota and metabolic disturbances in diabetic mice, thereby worsening anxiety, depression, and cognitive decline.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (X.Z.); (H.H.); (Y.Z.); (J.L.)
| | - Houjia Hu
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (X.Z.); (H.H.); (Y.Z.); (J.L.)
| | - Yue Zhang
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (X.Z.); (H.H.); (Y.Z.); (J.L.)
| | - Shuting Hu
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (S.H.); (W.P.)
| | - Jiaqin Lu
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (X.Z.); (H.H.); (Y.Z.); (J.L.)
| | - Weijie Peng
- School of Pharmaceutics, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (S.H.); (W.P.)
| | - Dan Luo
- Department of Physiology, School of Basic Medical Sciences, Jiangxi Medical College, Nanchang University, Nanchang 330006, China; (X.Z.); (H.H.); (Y.Z.); (J.L.)
| |
Collapse
|
7
|
Shukla S, Hsu CL. Alcohol Use Disorder and the Gut-Brain Axis: A Narrative Review of the Role of Gut Microbiota and Implications for Treatment. Microorganisms 2025; 13:67. [PMID: 39858835 PMCID: PMC11767426 DOI: 10.3390/microorganisms13010067] [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: 11/30/2024] [Revised: 12/21/2024] [Accepted: 12/28/2024] [Indexed: 01/27/2025] Open
Abstract
Alcohol use disorder (AUD) affects millions of people worldwide and can lead to deleterious physical and social consequences. Recent research has highlighted not only the effect of alcohol on the gut microbiome, but also the role of the gut microbiome and the gut-brain axis in the development and maintenance of alcohol use disorder. This review provides an overview of the reciprocal relationship between alcohol consumption and the gut microbiome, including the effects of alcohol on gut microbial composition, changes in gut microbial metabolites in response to alcohol consumption, and how gut microbial metabolites may modulate alcohol use behavior. We also discuss the gut-mediated mechanisms of neuroinflammation that contribute to and result from AUD, including disruption of the intestinal barrier, toll-like receptor signaling, and the activation of glial cells and immune cells. Finally, we review the current evidence on gut microbial-directed therapies for AUD and discuss the implications of this research for our understanding of the pathophysiology of AUD and future research directions.
Collapse
Affiliation(s)
- Shikha Shukla
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Cynthia L. Hsu
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
- Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA
| |
Collapse
|
8
|
Jeong S, K Davis C, Vemuganti R. Mechanisms of time-restricted feeding-induced neuroprotection and neuronal plasticity in ischemic stroke as a function of circadian rhythm. Exp Neurol 2025; 383:115045. [PMID: 39510297 DOI: 10.1016/j.expneurol.2024.115045] [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: 09/04/2024] [Revised: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 11/15/2024]
Abstract
Time-restricted feeding (TRF) is known to promote longevity and brain function, and potentially prevent neurological diseases. Animal studies show that TRF enhances brain-derived neurotrophic factor (BDNF) signaling and regulates autophagy and neuroinflammation, supporting synaptic plasticity, neurogenesis and neuroprotection. Feeding/fasting paradigms influence the circadian cycle, with TRF aligning circadian cycle-related gene expression, and thus altering physiological processes. Emerging evidence highlights the role of gut microbiota in neuronal plasticity, based on the observation that TRF significantly alters gut microbiota composition. Hence, the gut-brain axis may be crucial for maintaining cognitive functions and presents a potential therapeutic target for TRF-mediated neuroprotection. In the context of ischemic stroke where neuronal damage is extensive, TRF can be a preconditioning strategy to enhance synaptic plasticity and neuronal resilience, thus improving outcomes after stroke. This review discussed the link between TRF and circadian regulation in neuronal plasticity and its implications for recovery after stroke.
Collapse
Affiliation(s)
- Soomin Jeong
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Neuroscience Training Program, University of Wisconsin, Madison, WI, USA; William S. Middleton Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
9
|
Feng J, Cen Q, Cui Y, Hu X, Li M, Wang L, Wei J, Sun N, Wang J, Zhang A. Lactobacillus rhamnosus: An emerging probiotic with therapeutic potential for depression. Pharmacol Res 2025; 211:107541. [PMID: 39653301 DOI: 10.1016/j.phrs.2024.107541] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 11/18/2024] [Accepted: 12/06/2024] [Indexed: 12/16/2024]
Abstract
Depression, a complex psychological disorder, involves multiple biological pathways in its pathogenesis. In recent years, the gut-brain axis theory has provided novel insights into the pathogenesis of depression, particularly the crucial role of the gut microbiota in mood regulation. While there remains no universal consensus on the most efficacious strains for depression treatment, Lactobacillus rhamnosus has risen to prominence within the realm of probiotics for its potential to positively modulate depressive symptoms. This review preliminarily explores the clinical significance of Lactobacillus rhamnosus in the treatment of depression and summarizes the potential mechanisms by which Lactobacillus rhamnosus treats depression, including its regulation of gut microbiota, alterations in gene expression, improvement of intestinal barrier function, maintenance of neurotransmitter balance, suppression of inflammatory responses, modulation of the immune system, coping with oxidative stress, and optimization of metabolic processes. Future research needs to further explore these mechanisms and combine them with clinical research results to optimize treatment plans and provide more effective treatment options for patients with depression.
Collapse
Affiliation(s)
- Jing Feng
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Qiuyu Cen
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Yanru Cui
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Xiaomin Hu
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Min Li
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Linjie Wang
- Department of Rehabilitation Medicine, General Hospital of Western Theater Command, Chengdu, China
| | - Juanfang Wei
- College of Physical Education and Health, Geely University of China, Chengdu, China
| | - Nianyi Sun
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Junyu Wang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| | - Anren Zhang
- Department of Rehabilitation Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai, China.
| |
Collapse
|
10
|
Varghese SM, Patel S, Nandan A, Jose A, Ghosh S, Sah RK, Menon B, K V A, Chakravarty S. Unraveling the Role of the Blood-Brain Barrier in the Pathophysiology of Depression: Recent Advances and Future Perspectives. Mol Neurobiol 2024; 61:10398-10447. [PMID: 38730081 DOI: 10.1007/s12035-024-04205-5] [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: 08/03/2023] [Accepted: 04/19/2024] [Indexed: 05/12/2024]
Abstract
Depression is a highly prevalent psychological disorder characterized by persistent dysphoria, psychomotor retardation, insomnia, anhedonia, suicidal ideation, and a remarkable decrease in overall well-being. Despite the prevalence of accessible antidepressant therapies, many individuals do not achieve substantial improvement. Understanding the multifactorial pathophysiology and the heterogeneous nature of the disorder could lead the way toward better outcomes. Recent findings have elucidated the substantial impact of compromised blood-brain barrier (BBB) integrity on the manifestation of depression. BBB functions as an indispensable defense mechanism, tightly overseeing the transport of molecules from the periphery to preserve the integrity of the brain parenchyma. The dysfunction of the BBB has been implicated in a multitude of neurological disorders, and its disruption and consequent brain alterations could potentially serve as important factors in the pathogenesis and progression of depression. In this review, we extensively examine the pathophysiological relevance of the BBB and delve into the specific modifications of its components that underlie the complexities of depression. A particular focus has been placed on examining the effects of peripheral inflammation on the BBB in depression and elucidating the intricate interactions between the gut, BBB, and brain. Furthermore, this review encompasses significant updates on the assessment of BBB integrity and permeability, providing a comprehensive overview of the topic. Finally, we outline the therapeutic relevance and strategies based on BBB in depression, including COVID-19-associated BBB disruption and neuropsychiatric implications. Understanding the comprehensive pathogenic cascade of depression is crucial for shaping the trajectory of future research endeavors.
Collapse
Affiliation(s)
- Shamili Mariya Varghese
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Shashikant Patel
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad, Telangana, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Amritasree Nandan
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Anju Jose
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Soumya Ghosh
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad, Telangana, 500007, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Ranjay Kumar Sah
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Bindu Menon
- Department of Psychiatry, Amrita School of Medicine, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India
| | - Athira K V
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, AIMS Health Sciences Campus, Kochi, Kerala, 682 041, India.
| | - Sumana Chakravarty
- Applied Biology Division, CSIR-Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad, Telangana, 500007, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India.
| |
Collapse
|
11
|
Sun D, Luo J, Ye W, Wang C, Deng Q, Fang Z, Sun L, Gooneratne R. Ziziphus Jujube Polysaccharides inhibit over-abundance of fecal butyric acid in mildly stressed growing mice to ameliorate depression-like behavior. FOOD BIOSCI 2024; 62:104875. [DOI: 10.1016/j.fbio.2024.104875] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
12
|
Wang L, Qin N, Shi L, Liu R, Zhu T. Gut Microbiota and Tryptophan Metabolism in Pathogenesis of Ischemic Stroke: A Potential Role for Food Homologous Plants. Mol Nutr Food Res 2024; 68:e2400639. [PMID: 39551995 DOI: 10.1002/mnfr.202400639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/15/2024] [Indexed: 11/19/2024]
Abstract
SCOPE The intestinal flora is involved in the maintenance of human health and the development of diseases, and is closely related to the brain. As an essential amino acid, tryptophan (TRP) participates in a variety of physiological functions in the body and affects the growth and health of the human body. TRP catabolites produced by the gut microbiota are important signaling molecules for microbial communities and host-microbe interactions, and play an important role in maintaining health and disease pathogenesis. METHODS AND RESULTS The review first demonstrates the evidence of TRP metabolism in stroke and the relationship between gut microbiota and TRP metabolism. Furthermore, the review reveals that food homologous plants (FHP) bioactive compounds have been shown to regulate various metabolic pathways of the gut microbiota, including the biosynthesis of valine, leucine, isoleucine, and vitamin B6 metabolism. The most notable metabolic alteration is in TRP metabolism. CONCLUSION The interaction between gut microbiota and TRP metabolism offers a plausible explanation for the notable bioactivities of FHP in the treatment of ischemic stroke (IS). This review enhances the comprehension of the underlying mechanisms associated with the bioactivity of FHP on IS.
Collapse
Affiliation(s)
- Lei Wang
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Na Qin
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
| | - Liuliu Shi
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
- Department of Neurosurgery, The Affiliated Hospital of Qingdao University, Qingdao, 266000, PR China
| | - Rujuan Liu
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
- Department of Rehabilitation Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, PR China
| | - Ting Zhu
- Institute of Neuroregeneration and Neurorehabilitation, Department of Pathophysiology, School of Basic Medicine, Qingdao University, Qingdao, 266071, PR China
| |
Collapse
|
13
|
Sun Y, Fan C, Lei D. Association between gut microbiota and postpartum depression: A bidirectional Mendelian randomization study. J Affect Disord 2024; 362:615-622. [PMID: 39029663 DOI: 10.1016/j.jad.2024.07.057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 07/04/2024] [Accepted: 07/14/2024] [Indexed: 07/21/2024]
Abstract
BACKGROUNDS Empirical investigations have shown an association between gut microbiota and postpartum depression (PPD); nevertheless, the precise cause-and-effect relationship between these two variables remains ambiguous. This research aimed to examine the possible reciprocal causal relationship between the gut microbiota and PPD. METHODS In this work, we used Mendelian randomization (MR) to analyze the relationship between the gut microbiota (n = 18,340) and PPD (n = 67,205). We obtained the relevant SNPs from publicly accessible genome-wide association studies (GWAS). The SNP estimations were combined by the inverse-variance weighted (IVW) method, including sensitivity analyses such as weighted median, MR Egger, and MR Pleiotropy Residual Sum and Outlier (PRESSO). RESULTS We have identified strong correlations between six bacterial characteristics and the likelihood of developing PPD. Our research revealed that the genus Ruminococcaceae UCG010, the family Veillonellaceae, and the class Clostridia had a beneficial effect on preventing PPD. The class Alphaproteobacteria, genus Slackia, and order NB1n were found to have a significant negative impact on PPD. The sensitivity studies conducted on these bacterial features consistently confirmed these finding. LIMITATIONS It is crucial to acknowledge that our study was conducted just within a European society, which may restrict its applicability to other groups. CONCLUSIONS The findings from our MR investigation indicate a potential causal relationship between certain kinds of gut bacteria and PPD. Additional investigation is required to elucidate the influence of gut microbiota on the advancement of PPD.
Collapse
Affiliation(s)
- Yonghao Sun
- Fujian Provincial Key Laboratory of Reproductive Health Research, School of Medicine, Xiamen University, Xiamen, China
| | - Cuifang Fan
- Department of Obstetrics, Renmin Hospital of Wuhan University, Wuhan 430000, China.
| | - Di Lei
- Department of Obstetrics, Renmin Hospital of Wuhan University, Wuhan 430000, China.
| |
Collapse
|
14
|
Lockwood MB, Sung C, Alvernaz SA, Lee JR, Chin JL, Nayebpour M, Bernabé BP, Tussing-Humphreys LM, Li H, Spaggiari M, Martinino A, Park CG, Chlipala GE, Doorenbos AZ, Green SJ. The Gut Microbiome and Symptom Burden After Kidney Transplantation: An Overview and Research Opportunities. Biol Res Nurs 2024; 26:636-656. [PMID: 38836469 DOI: 10.1177/10998004241256031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2024]
Abstract
Many kidney transplant recipients continue to experience high symptom burden despite restoration of kidney function. High symptom burden is a significant driver of quality of life. In the post-transplant setting, high symptom burden has been linked to negative outcomes including medication non-adherence, allograft rejection, graft loss, and even mortality. Symbiotic bacteria (microbiota) in the human gastrointestinal tract critically interact with the immune, endocrine, and neurological systems to maintain homeostasis of the host. The gut microbiome has been proposed as an underlying mechanism mediating symptoms in several chronic medical conditions including irritable bowel syndrome, chronic fatigue syndrome, fibromyalgia, and psychoneurological disorders via the gut-brain-microbiota axis, a bidirectional signaling pathway between the enteric and central nervous system. Post-transplant exposure to antibiotics, antivirals, and immunosuppressant medications results in significant alterations in gut microbiota community composition and function, which in turn alter these commensal microorganisms' protective effects. This overview will discuss the current state of the science on the effects of the gut microbiome on symptom burden in kidney transplantation and future directions to guide this field of study.
Collapse
Affiliation(s)
- Mark B Lockwood
- Department of Biobehavioral Nursing Science, University of Illinois Chicago College of Nursing, Chicago, IL, USA
| | - Choa Sung
- Post-Doctoral Fellow, Department of Biobehavioral Nursing Science, University of Illinois Chicago College of Nursing, Chicago, IL, USA
| | - Suzanne A Alvernaz
- Graduate Student, Department of Biomedical Engineering, University of Illinois ChicagoColleges of Engineering and Medicine, Chicago, IL, USA
| | - John R Lee
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medicine, New York, NY, USA
| | - Jennifer L Chin
- Medical Student, Touro College of Osteopathic Medicine, Middletown, NY, USA
| | - Mehdi Nayebpour
- Virginia BioAnalytics LLC, Washington, District of Columbia, USA
| | - Beatriz Peñalver Bernabé
- Graduate Student, Department of Biomedical Engineering, University of Illinois ChicagoColleges of Engineering and Medicine, Chicago, IL, USA
| | - Lisa M Tussing-Humphreys
- Department of Kinesiology and Nutrition, College of Applied Health Sciences, University of Illinois Chicago, Chicago, IL, USA
| | - Hongjin Li
- Department of Biobehavioral Nursing Science, University of Illinois Chicago College of Nursing, Chicago, IL, USA
| | - Mario Spaggiari
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago, Chicago, IL, USA
| | - Alessandro Martinino
- Division of Transplantation, Department of Surgery, University of Illinois at Chicago, Chicago, IL, USA
| | - Chang G Park
- Department of Population Health Nursing Science, Office of Research Facilitation, University of Illinois Chicago, Chicago, IL, USA
| | - George E Chlipala
- Research Core Facility, Research Resources Center, University of Illinois Chicago, Chicago, IL, USA
| | - Ardith Z Doorenbos
- Department of Biobehavioral Nursing Science, University of Illinois ChicagoCollege of Nursing, Chicago, IL, USA
| | - Stefan J Green
- Department of Internal Medicine, Division of Infectious Diseases, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
15
|
Alatan H, Liang S, Shimodaira Y, Wu X, Hu X, Wang T, Luo J, Iijima K, Jin F. Supplementation with Lactobacillus helveticus NS8 alleviated behavioral, neural, endocrine, and microbiota abnormalities in an endogenous rat model of depression. Front Immunol 2024; 15:1407620. [PMID: 39346901 PMCID: PMC11428200 DOI: 10.3389/fimmu.2024.1407620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 08/13/2024] [Indexed: 10/01/2024] Open
Abstract
Introduction Major depressive disorder is a condition involving microbiota-gut-brain axis dysfunction. Increasing research aims to improve depression through gut microbiota regulation, including interventions such as probiotics, prebiotics, and fecal microbiota transplants. However, most research focuses on exogenous depression induced by chronic stress or drugs, with less attention given to endogenous depression. Additionally, research on gut mycobiota in depression is significantly less than that on gut bacteria. Methods In the present study, Wistar-Kyoto rats were used as an endogenous depression and treatment-resistant depression model, while Wistar rats served as controls. Differences between the two rat strains in behavior, gut bacteria, gut mycobiota, nervous system, endocrine system, immune system, and gut barrier were evaluated. Additionally, the effects of Lactobacillus helveticus NS8 supplementation were investigated. Results Wistar-Kyoto rats demonstrated increased depressive-like behaviors in the forced swimming test, reduced sucrose preference in the sucrose preference test, and decreased locomotor activity in the open field test. They also exhibited abnormal gut bacteria and mycobiota, characterized by higher bacterial α-diversity but lower fungal α-diversity, along with increased butyrate, L-tyrosine, and L-phenylalanine biosynthesis from bacteria. Furthermore, these rats showed dysfunction in the microbiota-gut-brain axis, evidenced by a hypo-serotonergic system, hyper-noradrenergic system, defective hypothalamic-pituitary-adrenal axis, compromised gut barrier integrity, heightened serum inflammation, and diminished gut immunity. A 1-month L. helveticus NS8 intervention increased the fecal abundance of L. helveticus; reduced the abundance of Bilophila and Debaryomycetaceae; decreased immobility time but increased climbing time in the forced swimming test; reduced hippocampal corticotropin-releasing hormone levels; decreased hypothalamic norepinephrine levels; increased hippocampal glucocorticoid receptor, brain-derived neurotrophic factor dopamine, and 5-hydroxyindoleacetic acid content; and improved the gut microbiota, serotonergic, and noradrenergic system. Conclusion The depressive phenotype of Wistar-Kyoto rats is not only attributed to their genetic context but also closely related to their gut microbiota. Abnormal gut microbiota and a dysfunctional microbiota-gut-brain axis play important roles in endogenous depression, just as they do in exogenous depression. Supplementing with probiotics such as L. helveticus NS8 is likely a promising approach to improve endogenous depression and treatment-resistant depression.
Collapse
Affiliation(s)
- Husile Alatan
- Department of Gastroenterology and Neurology, Akita University Graduate School of Medicine, Akita, Japan
| | - Shan Liang
- Mirai Food Academic Institute of Japan, Akita, Japan
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Yosuke Shimodaira
- Department of Gastroenterology and Neurology, Akita University Graduate School of Medicine, Akita, Japan
| | - Xiaoli Wu
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Xu Hu
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Tao Wang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Jia Luo
- Psychology College, Sichuan Normal University, Chengdu, China
| | - Katsunori Iijima
- Department of Gastroenterology and Neurology, Akita University Graduate School of Medicine, Akita, Japan
| | - Feng Jin
- Mirai Food Academic Institute of Japan, Akita, Japan
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
16
|
Shi M, Li Z, Tang Z, Zhou H, Huang X, Wei Y, Li X, Li X, Shi H, Qin D. Exploring the pathogenesis and treatment of PSD from the perspective of gut microbiota. Brain Res Bull 2024; 215:111022. [PMID: 38936669 DOI: 10.1016/j.brainresbull.2024.111022] [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: 04/07/2024] [Revised: 05/30/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Post-stroke depression (PSD) is a psychological disease that can occur following a stroke and is associated with serious consequences. Research on the pathogenesis and treatment of PSD is still in the infancy stage. Patients with PSD often exhibit gastrointestinal symptoms; therefore the role of gut microbiota in the pathophysiology and potential treatment effects of PSD has become a hot topic of research. In this review, describe the research on the pathogenesis and therapy of PSD. We also describe how the gut microbiota influences neurotransmitters, the endocrine system, energy metabolism, and the immune system. It was proposed that the gut microbiota is involved in the pathogenesis and treatment of PSD through the regulation of neurotransmitter levels, vagal signaling, hypothalamic-pituitary-adrenal axis activation and inhibition, hormone secretion and release, in addition to immunity and inflammation.
Collapse
Affiliation(s)
- Mingqin Shi
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Zhenmin Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Zhengxiu Tang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Haimei Zhou
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Xiaoyi Huang
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Yuanyuan Wei
- First Clinical Medical College, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Xinyao Li
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| | - Xiahuang Li
- The People's Hospital of Mengzi, The Affiliated Hospital of Yunnan University of Chinese Medicine, Mengzi Honghe, China.
| | - Hongling Shi
- Department of Rehabilitation Medicine, The Third People's Hospital of Yunnan Province, Kunming Yunnan, China.
| | - Dongdong Qin
- School of Basic Medical Sciences, Yunnan University of Chinese Medicine, Kunming Yunnan, China.
| |
Collapse
|
17
|
Jing JQ, Jia SJ, Yang CJ. Physical activity promotes brain development through serotonin during early childhood. Neuroscience 2024; 554:34-42. [PMID: 39004411 DOI: 10.1016/j.neuroscience.2024.07.015] [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: 11/21/2023] [Revised: 05/22/2024] [Accepted: 07/09/2024] [Indexed: 07/16/2024]
Abstract
Early childhood serves as a critical period for neural development and skill acquisition when children are extremely susceptible to the external environment and experience. As a crucial experiential stimulus, physical activity is believed to produce a series of positive effects on brain development, such as cognitive function, social-emotional abilities, and psychological well-being. The World Health Organization recommends that children engage in sufficient daily physical activity, which has already been strongly advocated in the practice of preschool education. However, the mechanisms by which physical activity promotes brain development are still unclear. The role of neurotransmitters, especially serotonin, in promoting brain development through physical activity has received increasing attention. Physical activity has been shown to stimulate the secretion of serotonin by increasing the bioavailability of free tryptophan and enriching the diversity of gut microbiota. Due to its important role in modulating neuronal proliferation, differentiation, synaptic morphogenesis, and synaptic transmission, serotonin can regulate children's explicit cognitive and social interaction behavior in the early stages of life. Therefore, we hypothesized that serotonin emerges as a pivotal transmitter that mediates the relationship between physical activity and brain development during early childhood. Further systematic reviews and meta-analyses are needed to specifically explore whether the type, intensity, dosage, duration, and degree of voluntariness of PA may affect the role of serotonin in the relationship between physical activity and brain function. This review not only helps us understand the impact of exercise on development but also provides a solid theoretical basis for increasing physical activity during early childhood.
Collapse
Affiliation(s)
- Jia-Qi Jing
- Faculty of Education, East China Normal University, Shanghai, China
| | - Si-Jia Jia
- Faculty of Education, East China Normal University, Shanghai, China
| | - Chang-Jiang Yang
- Faculty of Education, East China Normal University, Shanghai, China.
| |
Collapse
|
18
|
Chen CW, Chen HC, She SC, Lai CT, Chen WJ, Kuo TBJ, Yang CCH. Levilactobacillus brevis SG031 modulates mood-related behaviors and attenuates stress-related sleep disturbance and autonomic dysfunction via gut microbiota modulation in Wistar-Kyoto rats. Life Sci 2024; 351:122804. [PMID: 38852801 DOI: 10.1016/j.lfs.2024.122804] [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: 03/20/2024] [Revised: 05/22/2024] [Accepted: 06/04/2024] [Indexed: 06/11/2024]
Abstract
AIMS The probiotic bacterium Levilactobacillus brevis (L. brevis) has been proposed as a potential solution to manage mood disorders and alleviate stress-related sleep disturbances. However, the underlying mechanisms of its effects have not been fully elucidated. The aim of this study was to explore the impact and potential mechanisms of L. brevis SG031 supplementation on anxiety/depression-like behaviors and stress-induced changes in sleep patterns and sleep-related autonomic function. MAIN METHODS Male Wistar-Kyoto rats were administered low, medium, or high doses of L. brevis SG031 or a vehicle for 4 weeks, followed by behavioral tests to evaluate anxiety and depression. After an additional 2 weeks of SG031 or vehicle administration, a cage-exchange paradigm was performed with 24-hour physiological signal measurements under different stress conditions. Fecal samples were collected to construct a 16S rRNA library and assess fecal short-chain fatty acids (SCFAs). KEY FINDINGS High-dose SG031 administration yielded reduced depression-like responses and enhanced social interaction in behavioral tests. It also exhibited a protective effect against stress-induced sleep disturbance characterized by decreased sleep time, increased awake time, and autonomic dysfunction during sleep. Fecal examination indicated that high-dose SG031 administration exerted beneficial effects on gut health by maintaining the gut microbial abundance, preserving stability of the microbial composition, and enriching the gut with SCFAs, which were associated with improvements in sleep and autonomic function. SIGNIFICANCE These findings collectively underscore the multifaceted potential of SG031 in addressing mental health and stress-related sleep challenges through the modulation of the gut microbiota.
Collapse
Affiliation(s)
- Chieh-Wen Chen
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Sleep Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Health and Leisure Management, Yuanpei University of Medical Technology, Hsinchu, Taiwan
| | - Hung-Chang Chen
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Sleep Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Medical Education, Taichung Veterans General Hospital, Taichung, Taiwan
| | - Sheng-Chieh She
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Sleep Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Ting Lai
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Sleep Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Wei-Jen Chen
- College of Management, Chang Jung Christian University, Tainan, Taiwan
| | - Terry B J Kuo
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Sleep Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Education and Research, Taipei City Hospital, Taipei, Taiwan; Center for Mind and Brain Medicine, Tsaotun Psychiatric Center, Ministry of Health and Welfare, Nantou, Taiwan
| | - Cheryl C H Yang
- Institute of Brain Science, National Yang Ming Chiao Tung University, Taipei, Taiwan; Sleep Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Brain Research Center, National Yang Ming Chiao Tung University, Taipei, Taiwan; Department of Education and Research, Taipei City Hospital, Taipei, Taiwan.
| |
Collapse
|
19
|
Hu A, Zaongo SD, Harypursat V, Wang X, Ouyang J, Chen Y. HIV-associated neurocognitive disorder: key implications of the microbiota-gut-brain axis. Front Microbiol 2024; 15:1428239. [PMID: 39155987 PMCID: PMC11327151 DOI: 10.3389/fmicb.2024.1428239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 07/23/2024] [Indexed: 08/20/2024] Open
Abstract
HIV-associated neurocognitive disorder (HAND) is now recognized to be relatively common in people living with HIV (PLWH), and remains a common cause of cognitive impairment. Unfortunately, the fundamental pathogenic processes underlying this specific outcome of HIV infection have not as yet been fully elucidated. With increased interest in research related to the microbiota-gut-brain axis, the gut-brain axis has been shown to play critical roles in regulating central nervous system disorders such as Alzheimer's disease and Parkinson's disease. PLWH are characterized by a particular affliction, referred to as gut-associated dysbiosis syndrome, which provokes an alteration in microbial composition and diversity, and of their associated metabolite composition within the gut. Interestingly, the gut microbiota has also been recognized as a key element, which both positively and negatively influences human brain health, including the functioning and development of the central nervous system (CNS). In this review, based on published evidence, we critically discuss the relevant interactions between the microbiota-gut-brain axis and the pathogenesis of HAND in the context of HIV infection. It is likely that HAND manifestation in PLWH mainly results from (i) gut-associated dysbiosis syndrome and a leaky gut on the one hand and (ii) inflammation on the other hand. In other words, the preceding features of HIV infection negatively alter the composition of the gut microbiota (microbes and their associated metabolites) and promote proinflammatory immune responses which singularly or in tandem damage neurons and/or induce inadequate neuronal signaling. Thus, HAND is fairly prevalent in PLWH. This work aims to demonstrate that in the quest to prevent and possibly treat HAND, the gut microbiota may ultimately represent a therapeutically targetable "host factor."
Collapse
Affiliation(s)
- Aizhen Hu
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Silvere D. Zaongo
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Vijay Harypursat
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| | - Xin Wang
- Phase I Clinical Trial Center, Chonggang General Hospital, Chongqing, China
| | - Jing Ouyang
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
| | - Yaokai Chen
- Clinical Research Center, Chongqing Public Health Medical Center, Chongqing, China
- Department of Infectious Diseases, Chongqing Public Health Medical Center, Chongqing, China
| |
Collapse
|
20
|
Di Majo D, Ricciardi N, Di Liberto V, Allegra M, Frinchi M, Urone G, Scordino M, Massaro A, Mudò G, Ferraro G, Sardo P, Giglia G, Gambino G. The remarkable impact of Opuntia Ficus Indica fruit administration on metabolic syndrome: Correlations between cognitive functions, oxidative stress and lipid dysmetabolism in the high-fat, diet-fed rat model. Biomed Pharmacother 2024; 177:117028. [PMID: 38959603 DOI: 10.1016/j.biopha.2024.117028] [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: 04/15/2024] [Revised: 06/21/2024] [Accepted: 06/21/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND A wealth of evidence underscores the bioactive properties of nutraceuticals and functional foods in addressing oxyinflammatory-based diseases with implications at both peripheral and central levels. Opuntia ficus-indica (OFI) is well-documented for its health-promoting attributes, though its fruit (OFIF) remains relatively understudied. Not only poses Metabolic Syndrome (MetS) cardiometabolic risks but also contributes significantly to cognitive impairment, especially in crucial brain areas such as hippocampus and hypothalamus. METHODS Following 8 weeks of HFD to induce MetS, rats received OFIF oral supplementation for 4 weeks to evaluate cognitive and affective modifications using behavioural paradigms, i.e. open field, burrowing, white-dark box, novelty-suppressed feeding, and object recognition tests. Our investigation extended to biochemical evaluations of lipid homeostasis, central and peripheral oxidative stress and neurotrophic pathways, correlating these measures together with circulating leptin levels. RESULTS Our data revealed that OFIF modulation of leptin positively correlates with systemic and brain oxidative stress, with markers of increased anxiety-like behaviour and impaired lipid homeostasis. On the other hand, leptin levels reduced by OFIF are associated with improved antioxidant barriers, declarative memory and neurotrophic signalling. DISCUSSION This study underscores OFIF neuroactive potential in the context of MetS-associated cognitive impairment, offering insights into its mechanisms and implications for future therapeutic strategies.
Collapse
Affiliation(s)
- Danila Di Majo
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Section of Human Physiology, School of Medicine, University of Palermo, Palermo 90127, Italy; Post-Graduate School of Nutrition and Food Science, School of Medicine, University of Palermo, Palermo 90127, Italy
| | - Nicolò Ricciardi
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Section of Human Physiology, School of Medicine, University of Palermo, Palermo 90127, Italy
| | - Valentina Di Liberto
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Section of Human Physiology, School of Medicine, University of Palermo, Palermo 90127, Italy
| | - Mario Allegra
- Post-Graduate School of Nutrition and Food Science, School of Medicine, University of Palermo, Palermo 90127, Italy; Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Palermo 90128, Italy
| | - Monica Frinchi
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Section of Human Physiology, School of Medicine, University of Palermo, Palermo 90127, Italy
| | - Giulia Urone
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Section of Human Physiology, School of Medicine, University of Palermo, Palermo 90127, Italy
| | - Miriana Scordino
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Section of Human Physiology, School of Medicine, University of Palermo, Palermo 90127, Italy
| | - Alessandro Massaro
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Palermo 90128, Italy
| | - Giuseppa Mudò
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Section of Human Physiology, School of Medicine, University of Palermo, Palermo 90127, Italy
| | - Giuseppe Ferraro
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Section of Human Physiology, School of Medicine, University of Palermo, Palermo 90127, Italy; Post-Graduate School of Nutrition and Food Science, School of Medicine, University of Palermo, Palermo 90127, Italy
| | - Pierangelo Sardo
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Section of Human Physiology, School of Medicine, University of Palermo, Palermo 90127, Italy; Post-Graduate School of Nutrition and Food Science, School of Medicine, University of Palermo, Palermo 90127, Italy
| | - Giuseppe Giglia
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Section of Human Physiology, School of Medicine, University of Palermo, Palermo 90127, Italy; Post-Graduate School of Nutrition and Food Science, School of Medicine, University of Palermo, Palermo 90127, Italy.
| | - Giuditta Gambino
- Department of Biomedicine Neuroscience and Advanced Diagnostics, Section of Human Physiology, School of Medicine, University of Palermo, Palermo 90127, Italy; Post-Graduate School of Nutrition and Food Science, School of Medicine, University of Palermo, Palermo 90127, Italy
| |
Collapse
|
21
|
Bokoliya SC, Russell J, Dorsett Y, Panier HA, Singh V, Daddi L, Yuan H, Dedon LR, Liu Z, Zhou Y, Min Z, Barson JR, Covault J, Bubier JA, Zhou Y. Short-chain fatty acid valerate reduces voluntary alcohol intake in male mice. MICROBIOME 2024; 12:108. [PMID: 38886761 PMCID: PMC11181657 DOI: 10.1186/s40168-024-01829-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 05/04/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND Despite serious health and social consequences, effective intervention strategies for habitual alcohol binge drinking are lacking. The development of novel therapeutic and preventative approaches is highly desirable. Accumulating evidence in the past several years has established associations between the gut microbiome and microbial metabolites with drinking behavior, but druggable targets and their underlying mechanism of action are understudied. RESULTS Here, using a drink-in-the-dark mouse model, we identified a microbiome metabolite-based novel treatment (sodium valerate) that can reduce excessive alcohol drinking. Sodium valerate is a sodium salt of valeric acid short-chain fatty acid with a similar structure as γ-aminobutyric acid (GABA). Ten days of oral sodium valerate supplementation attenuates excessive alcohol drinking by 40%, reduces blood ethanol concentration by 53%, and improves anxiety-like or approach-avoidance behavior in male mice, without affecting overall food and water intake. Mechanistically, sodium valerate supplementation increases GABA levels across stool, blood, and amygdala. It also significantly increases H4 acetylation in the amygdala of mice. Transcriptomics analysis of the amygdala revealed that sodium valerate supplementation led to changes in gene expression associated with functional pathways including potassium voltage-gated channels, inflammation, glutamate degradation, L-DOPA degradation, and psychological behaviors. 16S microbiome profiling showed that sodium valerate supplementation shifts the gut microbiome composition and decreases microbiome-derived neuroactive compounds through GABA degradation in the gut microbiome. CONCLUSION Our findings suggest that sodium valerate holds promise as an innovative therapeutic avenue for the reduction of habitual binge drinking, potentially through multifaceted mechanisms. Video Abstract.
Collapse
Affiliation(s)
- Suresh C Bokoliya
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Jordan Russell
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Yair Dorsett
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Hunter A Panier
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Vijender Singh
- Computational Biology Core, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Lauren Daddi
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Hanshu Yuan
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Liv R Dedon
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
- Calhoun Cardiology Center, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Zhongmao Liu
- Department of Statistics, University of Connecticut, Storrs, CT, 06269, USA
| | - Yuqi Zhou
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | - Zefang Min
- Department of Statistics, University of Connecticut, Storrs, CT, 06269, USA
| | - Jessica R Barson
- Department of Neurobiology & Anatomy, Drexel University College of Medicine, Philadelphia, PA, 19129, USA
| | - Jonathan Covault
- Department of Psychiatry, University of Connecticut Health Center, Farmington, CT, 06030, USA
| | | | - Yanjiao Zhou
- Department of Medicine, University of Connecticut Health Center, Farmington, CT, 06030, USA.
| |
Collapse
|
22
|
Meckel KR, Simpson SS, Godino A, Peck EG, Sens JP, Leonard MZ, George O, Calipari ES, Hofford RS, Kiraly DD. Microbial short-chain fatty acids regulate drug seeking and transcriptional control in a model of cocaine seeking. Neuropsychopharmacology 2024; 49:386-395. [PMID: 37528220 PMCID: PMC10724273 DOI: 10.1038/s41386-023-01661-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 08/03/2023]
Abstract
Cocaine use disorder represents a public health crisis with no FDA-approved medications for its treatment. A growing body of research has detailed the important connections between the brain and the resident population of bacteria in the gut, the gut microbiome, in psychiatric disease models. Acute depletion of gut bacteria results in enhanced reward in a mouse cocaine place preference model, and repletion of bacterially-derived short-chain fatty acid (SCFA) metabolites reverses this effect. However, the role of the gut microbiome and its metabolites in modulating cocaine-seeking behavior after prolonged abstinence is unknown. Given that relapse prevention is the most clinically challenging issue in treating substance use disorders, studies examining the effects of microbiome manipulations in relapse-relevant models are critical. Here, male Sprague-Dawley rats received either untreated water or antibiotics to deplete the gut microbiome and its metabolites. Rats were trained to self-administer cocaine and subjected to either within-session threshold testing to evaluate motivation for cocaine or 21 days of abstinence followed by a cue-induced cocaine-seeking task to model relapse behavior. Microbiome depletion did not affect cocaine acquisition on an fixed-ratio 1 schedule. However, microbiome-depleted rats exhibited significantly enhanced motivation for low dose cocaine on a within-session threshold task. Similarly, microbiome depletion increased cue-induced cocaine-seeking following prolonged abstinence and altered transcriptional regulation in the nucleus accumbens. In the absence of a normal microbiome, repletion of bacterially-derived SCFA metabolites reversed the behavioral and transcriptional changes associated with microbiome depletion. These findings suggest that gut bacteria, via their metabolites, are key regulators of drug-seeking behaviors, positioning the microbiome as a potential translational research target.
Collapse
Affiliation(s)
- Katherine R Meckel
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Biology, Swarthmore College, Swarthmore, PA, 19081, USA
| | - Sierra S Simpson
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Arthur Godino
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Emily G Peck
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Physiology & Pharmacology, Wake Forest University School of Medicine, Atrium Wake Forest Baptist Health, Winston-Salem, NC, 27101, USA
| | - Jonathon P Sens
- Department of Physiology & Pharmacology, Wake Forest University School of Medicine, Atrium Wake Forest Baptist Health, Winston-Salem, NC, 27101, USA
| | - Michael Z Leonard
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
| | - Olivier George
- Department of Psychiatry, University of California San Diego, La Jolla, CA, USA
| | - Erin S Calipari
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Pharmacology, Vanderbilt University, Nashville, TN, USA
- Vanderbilt Brain Institute, Vanderbilt University, 865F Light Hall, 2215 Garland Avenue, Nashville, TN, 37232, USA
- Vanderbilt Center for Addiction Research, Vanderbilt University, Nashville, TN, USA
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, USA
- Department of Psychiatry and Behavioral Sciences, Vanderbilt University, Nashville, TN, USA
| | - Rebecca S Hofford
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Physiology & Pharmacology, Wake Forest University School of Medicine, Atrium Wake Forest Baptist Health, Winston-Salem, NC, 27101, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | - Drew D Kiraly
- Nash Family Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Physiology & Pharmacology, Wake Forest University School of Medicine, Atrium Wake Forest Baptist Health, Winston-Salem, NC, 27101, USA.
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA.
- Department of Psychiatry, Wake Forest University School of Medicine, Atrium Wake Forest Baptist Health, Winston-Salem, NC, 27101, USA.
| |
Collapse
|
23
|
Zhou HB, Lu SZ, Yu ZS, Zhang JL, Mei ZN. Mechanisms for the biological activity of Gastrodia elata Blume and its constituents: A comprehensive review on sedative-hypnotic, and antidepressant properties. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 123:155251. [PMID: 38056151 DOI: 10.1016/j.phymed.2023.155251] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 11/15/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
BACKGROUND Insomnia and depressive disorder are two common symptoms with a reciprocal causal relationship in clinical practice, which are usually manifested in comorbid form. Several medications have been widely used in the treatment of insomnia and depression, but most of these drugs show non-negligible side effects. Currently, many treatments are indicated for insomnia and depressive symptom, including Chinese herbal medicine such as Gastrodia elata Blume (G. elata), which has excellent sedative-hypnotic and antidepressant effects in clinical and animal studies. PURPOSE To summarize the mechanisms of insomnia and depression and the structure-activity mechanism for G. elata to alleviate these symptoms, particularly by hypothalamic-pituitary-adrenal (HPA) axis and intestinal flora, aiming to discover new approaches for the treatment of insomnia and depression. METHODS The following electronic databases were searched from the beginning to November 2023: PubMed, Web of Science, Google Scholar, Wanfang Database, and CNKI. The following keywords of G. elata were used truncated with other relevant topic terms, such as depression, insomnia, antidepressant, sedative-hypnotic, neuroprotection, application, safety, and toxicity. RESULTS Natural compounds derived from G. elata could alleviate insomnia and depressive disorder, which is involved in monoamine neurotransmitters, inflammatory response, oxidative stress, and gut microbes, etc. Several clinical trials showed that G. elata-derived natural compounds that treat depression and insomnia have significant and safe therapeutic effects, but further well-designed clinical and toxicological studies are needed. CONCLUSION G. elata exerts a critical role in treating depression and insomnia due to its multi-targeting properties and fewer side effects. However, more clinical and toxicological studies should be performed to further explore the sedative-hypnotic and antidepressant mechanisms of G. elata and provide more evidence and recommendations for its clinical application. Our review provides an overview of G. elata treating insomnia with depression for future research direction.
Collapse
Affiliation(s)
- Hai-Bo Zhou
- College of Food Science and Technology, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, Hubei Province 430070, China
| | - Sheng-Ze Lu
- College of Food Science and Technology, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, Hubei Province 430070, China
| | - Zhong-Shun Yu
- College of Food Science and Technology, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, Hubei Province 430070, China
| | - Jiu-Liang Zhang
- College of Food Science and Technology, Huazhong Agricultural University, No.1, Shizishan Street, Hongshan District, Wuhan, Hubei Province 430070, China; Key Laboratory of Environment Correlative Dietology, Ministry of Education, Wuhan, 430070, China.
| | - Zhi-Nan Mei
- College of Plant Science & Technology, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
24
|
Nohesara S, Abdolmaleky HM, Zhou JR, Thiagalingam S. Microbiota-Induced Epigenetic Alterations in Depressive Disorders Are Targets for Nutritional and Probiotic Therapies. Genes (Basel) 2023; 14:2217. [PMID: 38137038 PMCID: PMC10742434 DOI: 10.3390/genes14122217] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/08/2023] [Accepted: 12/11/2023] [Indexed: 12/24/2023] Open
Abstract
Major depressive disorder (MDD) is a complex disorder and a leading cause of disability in 280 million people worldwide. Many environmental factors, such as microbes, drugs, and diet, are involved in the pathogenesis of depressive disorders. However, the underlying mechanisms of depression are complex and include the interaction of genetics with epigenetics and the host immune system. Modifications of the gut microbiome and its metabolites influence stress-related responses and social behavior in patients with depressive disorders by modulating the maturation of immune cells and neurogenesis in the brain mediated by epigenetic modifications. Here, we discuss the potential roles of a leaky gut in the development of depressive disorders via changes in gut microbiota-derived metabolites with epigenetic effects. Next, we will deliberate how altering the gut microbiome composition contributes to the development of depressive disorders via epigenetic alterations. In particular, we focus on how microbiota-derived metabolites such as butyrate as an epigenetic modifier, probiotics, maternal diet, polyphenols, drugs (e.g., antipsychotics, antidepressants, and antibiotics), and fecal microbiota transplantation could positively alleviate depressive-like behaviors by modulating the epigenetic landscape. Finally, we will discuss challenges associated with recent therapeutic approaches for depressive disorders via microbiome-related epigenetic shifts, as well as opportunities to tackle such problems.
Collapse
Affiliation(s)
- Shabnam Nohesara
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Hamid Mostafavi Abdolmaleky
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boson, MA 02215, USA;
| | - Jin-Rong Zhou
- Nutrition/Metabolism Laboratory, Department of Surgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boson, MA 02215, USA;
| | - Sam Thiagalingam
- Department of Medicine (Biomedical Genetics), Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
- Department of Pathology & Laboratory Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
25
|
Tsugiyama LE, Macedo Moraes RC, Cavalcante Moraes YA, Francis-Oliveira J. Promising new pharmacological targets for depression: The search for efficacy. Drug Discov Today 2023; 28:103804. [PMID: 37865307 DOI: 10.1016/j.drudis.2023.103804] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 08/31/2023] [Accepted: 10/16/2023] [Indexed: 10/23/2023]
Abstract
Pharmacological treatment of major depressive disorder (MDD) still relies on the use of serotonergic drugs, despite their limited efficacy. A few mechanistically new drugs have been developed in recent years, but many fail in clinical trials. Several hypotheses have been proposed to explain MDD pathophysiology, indicating that physiological processes such as neuroplasticity, circadian rhythms, and metabolism are potential targets. Here, we review the current state of pharmacological treatments for MDD, as well as the preclinical and clinical evidence for an antidepressant effect of molecules that target non-serotonergic systems. We offer some insights into the challenges facing the development of new antidepressant drugs, and the prospect of finding more effectiveness for each target discussed.
Collapse
Affiliation(s)
- Lucila Emiko Tsugiyama
- Kansai Medical University, Graduate School of Medicine, iPS Cell Applied Medicine, Hirakata, Osaka, Japan
| | - Ruan Carlos Macedo Moraes
- University of Alabama at Birmingham, Department of Psychiatry and Behavioral Neurobiology, Birmingham, AL, USA; Biomedical Sciences Institute, Department of Human Physiology, Sao Paulo University, Sao Paulo, Brazil
| | | | - Jose Francis-Oliveira
- University of Alabama at Birmingham, Department of Psychiatry and Behavioral Neurobiology, Birmingham, AL, USA; Biomedical Sciences Institute, Department of Human Physiology, Sao Paulo University, Sao Paulo, Brazil.
| |
Collapse
|
26
|
Brushett S, Gacesa R, Vich Vila A, Brandao Gois M, Andreu-Sánchez S, Swarte J, Klaassen M, Collij V, Sinha T, Bolte L, Wu J, Swertz M, de Kroon M, Reijneveld S, Wijmenga C, Weersma R, Fu J, van Loo H, Kurilshikov A, Zhernakova A. Gut feelings: the relations between depression, anxiety, psychotropic drugs and the gut microbiome. Gut Microbes 2023; 15:2281360. [PMID: 38017662 PMCID: PMC10730195 DOI: 10.1080/19490976.2023.2281360] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Accepted: 11/06/2023] [Indexed: 11/30/2023] Open
Abstract
The gut microbiome is involved in the bi-directional relationship of the gut - brain axis. As most studies of this relationship are small and do not account for use of psychotropic drugs (PTDs), we explored the relations of the gut microbiome with several internalizing disorders, while adjusting for PTDs and other relevant medications, in 7,656 Lifelines participants from the Northern Netherlands (5,522 controls and 491 participants with at least one internalizing disorder). Disorders included dysthymia, major depressive disorder (MDD), any depressive disorder (AnyDep: dysthymia or MDD), generalized anxiety disorder (GAD) and any anxiety disorder (AnyAnx: GAD, social phobia and panic disorder). Compared to controls, 17 species were associated with depressive disorders and 3 were associated with anxiety disorders. Around 90% of these associations remained significant (FDR <0.05) after adjustment for PTD use, suggesting that the disorders, not PTD use, drove these associations. Negative associations were observed for the butyrate-producing bacteria Ruminococcus bromii in participants with AnyDep and for Bifidobacterium bifidum in AnyAnx participants, along with many others. Tryptophan and glutamate synthesis modules and the 3,4-Dihydroxyphenylacetic acid synthesis module (related to dopamine metabolism) were negatively associated with MDD and/or dysthymia. After additional adjustment for functional gastrointestinal disorders and irritable bowel syndrome, these relations remained either statistically (FDR <0.05) or nominally (P < 0.05) significant. Overall, multiple bacterial species and functional modules were associated with internalizing disorders, including gut - brain relevant components, while associations to PTD use were moderate. These findings suggest that internalizing disorders rather than PTDs are associated with gut microbiome differences relative to controls.
Collapse
Affiliation(s)
- S. Brushett
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Health Sciences, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - R. Gacesa
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - A. Vich Vila
- Department of Microbiology and Immunology, Rega Institute for Medical Research, Leuven, Belgium
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| | - M.F. Brandao Gois
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - S. Andreu-Sánchez
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - J.C. Swarte
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - M.A.Y. Klaassen
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - V. Collij
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - T. Sinha
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - L.A. Bolte
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - J. Wu
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - M. Swertz
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Genomics Coordination Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - M.L.A. de Kroon
- Department of Health Sciences, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - S.A. Reijneveld
- Department of Health Sciences, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - C. Wijmenga
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - R.K. Weersma
- Department of Gastroenterology and Hepatology, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - J. Fu
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
- Department of Pediatrics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - H.M. van Loo
- Department of Psychiatry, Interdisciplinary Center Psychopathology and Emotion regulation, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - A. Kurilshikov
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| | - A. Zhernakova
- Department of Genetics, University of Groningen and University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
27
|
Bokoliya SC, Russell J, Dorsett Y, Panier H, Singh V, Daddi L, Yuan H, Dedon LR, Liu Z, Barson JR, Covault J, Bubier JA, Zhou Y. Short-chain-fatty acid valerate reduces voluntary alcohol intake in male mice. RESEARCH SQUARE 2023:rs.3.rs-3496323. [PMID: 37961441 PMCID: PMC10635392 DOI: 10.21203/rs.3.rs-3496323/v1] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Background Despite serious health and social consequences, effective intervention strategies for habitual alcohol binge drinking are lacking. Development of novel therapeutic and preventative approaches is highly desirable. Accumulating evidence in the past several years has established associations between the gut microbiome and microbial metabolites with drinking behavior, but druggable targets and their underlying mechanism of action are understudied. Results Here, using a drink-in-the-dark mouse model, we identified a microbiome metabolite-based novel treatment (sodium valerate) that can reduce excessive alcohol drinking. Sodium valerate is a sodium salt of valeric acidshort-chain-fatty-acid with similar structure as γ-aminobutyric acid (GABA). Ten days of oral sodium valerate supplementation attenuates excessive alcohol drinking by 40%, reduces blood ethanol concentration by 53%, and improves anxiety-like or approach-avoidance behavior in male mice, without affecting overall food and water intake. Mechanistically, sodium valerate supplementation increases GABA levels across stool, blood, and amygdala. It also significantly increases H4 acetylation in the amygdala of mice. Transcriptomics analysis of the amygdala revealed that sodium valerate supplementation led to changes in gene expression associated with functional pathways including potassium voltage-gated channels, inflammation, glutamate degradation, L-DOPA degradation, and psychological behaviors. 16S microbiome profiling showed that sodium valerate supplementation shifts the gut microbiome composition and decreases microbiome-derived neuroactive compounds through GABA degradation in the gut microbiome. Conclusion Our findings suggest that the sodium valerate holds promise as an innovative therapeutic avenue for the reduction of habitual binge drinking, potentially through multifaceted mechanisms.
Collapse
|
28
|
Tung TH, Lai WD, Lee HC, Su KP, Panunggal B, Huang SY. Attenuation of Chronic Stress-Induced Depressive-like Symptoms by Fish Oil via Alleviating Neuroinflammation and Impaired Tryptophan Metabolism in Aging Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:14550-14561. [PMID: 37769277 PMCID: PMC10915802 DOI: 10.1021/acs.jafc.3c01784] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/30/2023]
Abstract
The prevalence of depression is increasing, and geriatric depression, in particular, is difficult to recognize and treat. Depression in older adults is often accompanied by neuroinflammation in the central nervous system (CNS). Neuroinflammation affects the brain's physiological and immune functions through several pathways and induces depressive symptoms. This study investigated the relationship among depression, neuroinflammation, and fish oil supplementation. Thirty-six male Sprague-Dawley rats were used in an aging-related depression animal model to simulate geriatric depression. Cognitive function, depressive-like symptoms, peripheral nervous system and CNS inflammation status, and the tryptophan-related metabolic pathway were analyzed. The geriatric depression animal model was associated with depressive-like behaviors and cognitive impairment. The integrity of the blood-brain barrier was compromised, resulting in increased expression of ionized calcium-binding adapter molecule 1 and the glial fibrillary acidic protein in the brain, indicating increased neuroinflammation. Tryptophan metabolism was also negatively affected. The geriatric-depressive-like rats had high levels of neurotoxic 5-hydroxyindoleacetic acid and kynurenine in their hippocampus. Fish oil intake improved depressive-like symptoms and cognitive impairment, reduced proinflammatory cytokine expression, activated the brain's glial cells, and increased the interleukin-10 level in the prefrontal cortex. Thus, fish oil intervention could ameliorate abnormal neurobehaviors and neuroinflammation and elevate the serotonin level in the hippocampus.
Collapse
Affiliation(s)
- Te-Hsuan Tung
- School
of Nutrition and Health Sciences, Taipei
Medical University, Taipei 110301, Taiwan
| | - Wen-De Lai
- School
of Nutrition and Health Sciences, Taipei
Medical University, Taipei 110301, Taiwan
| | - Hsiu-Chuan Lee
- School
of Nutrition and Health Sciences, Taipei
Medical University, Taipei 110301, Taiwan
| | - Kuan-Pin Su
- Department
of Psychiatry & Mind-Body Interface Laboratory (MBI-Lab), China Medical University Hospital, Taichung 404018, Taiwan
- College of
Medicine, China Medical University, Taichung 404018, Taiwan
| | - Binar Panunggal
- School
of Nutrition and Health Sciences, Taipei
Medical University, Taipei 110301, Taiwan
- Department
of Nutrition Science, Faculty of Medicine, Diponegoro University, Semarang 50275, Indonesia
- Center
of Nutrition Research, Diponegoro University, Semarang 50275, Indonesia
| | - Shih-Yi Huang
- School
of Nutrition and Health Sciences, Taipei
Medical University, Taipei 110301, Taiwan
- Graduate
Institute of Metabolism and Obesity Sciences, Taipei Medical University, Taipei 110301, Taiwan
- Nutrition
Research Centre, Taipei Medical University
Hospital, Taipei 110301, Taiwan
| |
Collapse
|
29
|
Moțățăianu A, Șerban G, Andone S. The Role of Short-Chain Fatty Acids in Microbiota-Gut-Brain Cross-Talk with a Focus on Amyotrophic Lateral Sclerosis: A Systematic Review. Int J Mol Sci 2023; 24:15094. [PMID: 37894774 PMCID: PMC10606032 DOI: 10.3390/ijms242015094] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 09/27/2023] [Accepted: 10/06/2023] [Indexed: 10/29/2023] Open
Abstract
Amyotrophic lateral sclerosis is a devastating neurodegenerative disease characterized by the gradual loss of motor neurons in the brain and spinal cord, leading to progressive motor function decline. Unfortunately, there is no effective treatment, and its increasing prevalence is linked to an aging population, improved diagnostics, heightened awareness, and changing lifestyles. In the gastrointestinal system, the gut microbiota plays a vital role in producing metabolites, neurotransmitters, and immune molecules. Short-chain fatty acids, of interest for their potential health benefits, are influenced by a fiber- and plant-based diet, promoting a diverse and balanced gut microbiome. These fatty acids impact the body by binding to receptors on enteroendocrine cells, influencing hormones like glucagon-like peptide-1 and peptide YY, which regulate appetite and insulin sensitivity. Furthermore, these fatty acids impact the blood-brain barrier, neurotransmitter levels, and neurotrophic factors, and directly stimulate vagal afferent nerves, affecting gut-brain communication. The vagus nerve is a crucial link between the gut and the brain, transmitting signals related to appetite, inflammation, and various processes. Dysregulation of this pathway can contribute to conditions like obesity and irritable bowel syndrome. Emerging evidence suggests the complex interplay among these fatty acids, the gut microbiota, and environmental factors influences neurodegenerative processes via interconnected pathways, including immune function, anti-inflammation, gut barrier, and energy metabolism. Embracing a balanced, fiber-rich diet may foster a diverse gut microbiome, potentially impacting neurodegenerative disease risk. Comprehensive understanding requires further research into interventions targeting the gut microbiome and fatty acid production and their potential therapeutic role in neurodegeneration.
Collapse
Affiliation(s)
- Anca Moțățăianu
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Târgu Mures, Romania
- Department of Neurology, University of Medicine, Pharmacy, Science and Technology of Târgu Mures ‘George Emil Palade’, 540142 Târgu Mures, Romania
| | - Georgiana Șerban
- Doctoral School, University of Medicine, Pharmacy, Science and Technology of Târgu Mures ‘George Emil Palade’, 540142 Târgu Mures, Romania
| | - Sebastian Andone
- 1st Neurology Clinic, Mures County Clinical Emergency Hospital, 540136 Târgu Mures, Romania
- Department of Neurology, University of Medicine, Pharmacy, Science and Technology of Târgu Mures ‘George Emil Palade’, 540142 Târgu Mures, Romania
| |
Collapse
|
30
|
Jeong S, Chokkalla AK, Davis CK, Vemuganti R. Post-stroke depression: epigenetic and epitranscriptomic modifications and their interplay with gut microbiota. Mol Psychiatry 2023; 28:4044-4055. [PMID: 37188778 PMCID: PMC10646155 DOI: 10.1038/s41380-023-02099-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 04/21/2023] [Accepted: 05/02/2023] [Indexed: 05/17/2023]
Abstract
Epigenetic and epitranscriptomic modifications that regulate physiological processes of an organism at the DNA and RNA levels, respectively, are novel therapeutic candidates for various neurological diseases. Gut microbiota and its metabolites are known to modulate DNA methylation and histone modifications (epigenetics), as well as RNA methylation especially N6-methyladenosine (epitranscriptomics). As gut microbiota as well as these modifications are highly dynamic across the lifespan of an organism, they are implicated in the pathogenesis of stroke and depression. The lack of specific therapeutic interventions for managing post-stroke depression emphasizes the need to identify novel molecular targets. This review highlights the interaction between the gut microbiota and epigenetic/epitranscriptomic pathways and their interplay in modulating candidate genes that are involved in post-stroke depression. This review further focuses on the three candidates, including brain-derived neurotrophic factor, ten-eleven translocation family proteins, and fat mass and obesity-associated protein based on their prevalence and pathoetiologic role in post-stroke depression.
Collapse
Affiliation(s)
- Soomin Jeong
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA
| | - Anil K Chokkalla
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Charles K Davis
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.
- Neuroscience Training Program, University of Wisconsin, Madison, WI, USA.
- William S. Middleton Veterans Hospital, Madison, WI, USA.
| |
Collapse
|
31
|
Zhang S, Lu B, Wang G. The role of gut microbiota in the pathogenesis and treatment of postpartum depression. Ann Gen Psychiatry 2023; 22:36. [PMID: 37759312 PMCID: PMC10523734 DOI: 10.1186/s12991-023-00469-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/11/2023] [Accepted: 09/22/2023] [Indexed: 09/29/2023] Open
Abstract
Postpartum depression (PPD) is a common complication of pregnancy in women, and its pathogenesis mainly involves disturbances of the neuroendocrine regulation, immune system, neurotransmitters, hormone secretion, and the gut microbiome. Gut microbes play essential physiological and pathological roles in the gut-brain axis' pathways which are involved in various central nervous system (CNS) and psychiatric disorders, including PPD. Numerous studies have identified the fundamental role of the gut-brain axis in the pathogenesis and treatment of PPD patients and also correlates with other pathogenic mechanisms of PPD. Disturbances in gut microbes are associated with the disruption of multiple signaling pathways and systems that ultimately lead to PPD development. This review aimed to elucidate the potential connections between gut microbes and the established PPD network, and this might serve as a guide for the development of new efficient diagnostic, therapeutic, and prognostic strategies in the management of PPD.
Collapse
Affiliation(s)
| | - Baili Lu
- Wuhan Mental Health Center, Wuhan, China
| | - Gang Wang
- Wuhan Mental Health Center, Wuhan, China.
| |
Collapse
|
32
|
Magzal F, Turroni S, Fabbrini M, Barone M, Vitman Schorr A, Ofran A, Tamir S. A personalized diet intervention improves depression symptoms and changes microbiota and metabolite profiles among community-dwelling older adults. Front Nutr 2023; 10:1234549. [PMID: 37794974 PMCID: PMC10547149 DOI: 10.3389/fnut.2023.1234549] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 08/21/2023] [Indexed: 10/06/2023] Open
Abstract
Introduction The impact of diet on mental well-being and gut microorganisms in humans is well recognized. However, research on the connections between food nutrients, gut microbiota, and mental health remains limited. To address this, the present study aimed to assess the effects of a personalized diet, based on individual needs and aligned with the Mediterranean diet principles, on depression symptoms, quality of life, nutritional intake, and gut microbiota changes among older adults living in the community. Methods The intervention involved regular visits from a registered dietitian, who provided tailored dietary recommendations. During the 6-month study, participants completed questionnaires to evaluate their depression levels, quality of life, and dietary habits. Additionally, they provided stool samples for analysis of gut microbiota and metabolites. Results The results demonstrated that the personalized dietary intervention reduced depression symptoms and improved the quality of life among older adults. Furthermore, significant changes in the intake of certain nutrients, such as folate, lutein, zeaxanthin, EPA, and DHA, were observed following the intervention. Moreover, the intervention was associated with increased diversity in the gut microbiome and reduced total short-chain fatty acids, the main metabolites produced by gut microorganisms. The study also revealed correlations between food nutrients, gut microbiota, and mental health parameters. Discussion In conclusion, this research highlights the potential advantages of personalized dietary interventions in managing depression and enhancing overall well-being among older populations. It also sheds light on the role of gut microbiota and its metabolites in these effects. The findings offer valuable insights into the significance of nutrition and gut health for mental well-being in older adults.
Collapse
Affiliation(s)
- Faiga Magzal
- Laboratory of Human Health and Nutrition Sciences, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Department of Nutrition, Tel Hai College, Upper Galilee, Israel
| | - Silvia Turroni
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
| | - Marco Fabbrini
- Unit of Microbiome Science and Biotechnology, Department of Pharmacy and Biotechnology, University of Bologna, Bologna, Italy
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | - Monica Barone
- Microbiomics Unit, Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | | | - Snait Tamir
- Laboratory of Human Health and Nutrition Sciences, MIGAL-Galilee Research Institute, Kiryat Shmona, Israel
- Department of Nutrition, Tel Hai College, Upper Galilee, Israel
| |
Collapse
|
33
|
Zhang Y, Chen X, Mo X, Xiao R, Cheng Q, Wang H, Liu L, Xie P. Enterogenic metabolomics signatures of depression: what are the possibilities for the future. Expert Rev Proteomics 2023; 20:397-418. [PMID: 37934939 DOI: 10.1080/14789450.2023.2279984] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Accepted: 10/24/2023] [Indexed: 11/09/2023]
Abstract
INTRODUCTION An increasing number of studies indicate that the microbiota-gut-brain axis is an important pathway involved in the onset and progression of depression. The responses of the organism (or its microorganisms) to external cues cannot be separated from a key intermediate element: their metabolites. AREAS COVERED In recent years, with the rapid development of metabolomics, an increasing amount of metabolites has been detected and studied, especially the gut metabolites. Nevertheless, the increasing amount of metabolites described has not been reflected in a better understanding of their functions and metabolic pathways. Moreover, our knowledge of the biological interactions among metabolites is also incomplete, which limits further studies on the connections between the microbial-entero-brain axis and depression. EXPERT OPINION This paper summarizes the current knowledge on depression-related metabolites and their involvement in the onset and progression of this disease. More importantly, this paper summarized metabolites from the intestine, and defined them as enterogenic metabolites, to further clarify the function of intestinal metabolites and their biochemical cross-talk, providing theoretical support and new research directions for the prevention and treatment of depression.
Collapse
Affiliation(s)
- Yangdong Zhang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xueyi Chen
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Faculty of Basic Medicine, Department of Pathology, Chongqing Medical University, Chongqing, China
| | - Xiaolong Mo
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Xiao
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Faculty of Basic Medicine, Department of Pathology, Chongqing Medical University, Chongqing, China
| | - Qisheng Cheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Haiyang Wang
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lanxiang Liu
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Diseases, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
34
|
Scriven M, McSweeney A, O'Carroll T, Morkl S, Butler MI. The Muscle-Gut-Brain Axis and Psychiatric Illness. Adv Biol (Weinh) 2023; 7:e2200214. [PMID: 37080945 DOI: 10.1002/adbi.202200214] [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: 08/03/2022] [Revised: 02/07/2023] [Indexed: 04/22/2023]
Abstract
The microbiota-gut-brain axis (MGBA) has been the subject of much research over the past decade, offering an exciting new paradigm for the treatment of psychiatric disorders. In this review, the MGBA is extended to include skeletal muscle and the potential role of an expanded "muscle-gut-brain axis" (MuGBA) in conditions such as anxiety and depression is discussed. There is evidence, from both preclinical and human studies, of bidirectional links between the gut microbiome and skeletal muscle function and structure. The therapeutic role of exercise in reducing depressive and anxiety symptoms is widely recognised, and the potential role of the gut microbiota-skeletal muscle link is discussed within this context. Potential pathways of communication involved in the MuGBA including the tryptophan-kynurenine pathway, intestinal permeability, immune modulation, and bacterial metabolites such as short-chain-fatty-acids are explored.
Collapse
Affiliation(s)
- Mary Scriven
- St Loman's Psychiatric Hospital, Delvin Road, Mullingar, County Westmeath, N91T3PR, Ireland
| | - Angela McSweeney
- Department of Psychiatry, Cork University Hospital, Wilton Road, Cork, T12DC4A, Ireland
| | | | - Sabrina Morkl
- Medical University of Graz, Department of Psychiatry and Psychotherapeutic Medicine, Graz, 8010, Austria
| | - Mary I Butler
- Department of Psychiatry, Cork University Hospital, Wilton Road, Cork, T12DC4A, Ireland
| |
Collapse
|
35
|
Song Y, Wang X, Ma W, Yang Y, Yan S, Sun J, Zhu X, Tang Y. Graves' disease as a driver of depression: a mechanistic insight. Front Endocrinol (Lausanne) 2023; 14:1162445. [PMID: 37152963 PMCID: PMC10157224 DOI: 10.3389/fendo.2023.1162445] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/05/2023] [Indexed: 05/09/2023] Open
Abstract
Graves' disease (GD) is characterized by diffuse enlargement and overactivity of the thyroid gland, which may be accompanied by other physical symptoms. Among them, depression can dramatically damage patients' quality of life, yet its prevalence in GD has not received adequate attention. Some studies have established a strong correlation between GD and increased risk of depression, though the data from current study remains limited. The summary of mechanistic insights regarding GD and depression has underpinned possible pathways by which GD contributes to depression. In this review, we first summarized the clinical evidence that supported the increased prevalence of depression by GD. We then concentrated on the mechanistic findings related to the acceleration of depression in the context of GD, as mounting evidence has indicated that GD promotes the development of depression through various mechanisms, including triggering autoimmune responses, inducing hormonal disorders, and influencing the thyroid-gut-microbiome-brain axis. Finally, we briefly presented potential therapeutic approaches to decreasing the risk of depression among patients with GD.
Collapse
Affiliation(s)
- Yifei Song
- Beijing University of Chinese Medicine, Beijing, China
| | - Xinying Wang
- Beijing University of Chinese Medicine, Beijing, China
| | - Wenxin Ma
- Centre for Evidence-Based Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| | - Yan Yang
- Tongling Municipal hospital, Anhui, China
| | - Shuxin Yan
- Beijing University of Chinese Medicine, Beijing, China
| | - Jiapan Sun
- Department of Geriatrics, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, Guangdong, China
| | - Xiaoyun Zhu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yang Tang
- Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
36
|
Vieira ACA, Pinheiro RO, Soares NL, Bezerra MLR, Nascimento DDS, Alves AF, Sousa MCDP, Dutra MLDV, Lima MDS, Donato NR, Aquino JDS. Maternal high-fat diet alters the neurobehavioral, biochemical and inflammatory parameters of their adult female rat offspring. Physiol Behav 2023; 266:114180. [PMID: 37037382 DOI: 10.1016/j.physbeh.2023.114180] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 03/24/2023] [Accepted: 03/31/2023] [Indexed: 04/12/2023]
Abstract
BACKGROUND Lipid metabolism dysregulations have been associated with depressive and anxious behaviors which can affect pregnant and lactating individuals, with indications that such changes extend to the offspring. Therefore, the aim of this study was to evaluate the effect of a maternal high-fat diet on the neurobehavioral, biochemical and inflammatory parameters of their adult female offspring. METHODS Wistar rats ± 90 days old were mated. The dams were allocated to consume a control (CTL) or high-fat (HFD) diet during pregnancy and lactation. After weaning, the female offspring from the CTL (N=10) and HFD (N=10) groups received standard chow. The offspring behavioral tests were started at 120 days old. Then, the somatic measures were evaluated followed by euthanasia, histological and biochemical analyses. RESULTS The HFD group had less ambulation and longer immobility time in the open field test compared to the CTL. The HFD group had lower HDL (48.4%) and a higher adiposity (71.8%) and LDL (62.2%) than the CTL. The CTL had a higher organic acid concentration in the intestine, mainly acetic and butyric acids, however the HFD had a higher citric and acetic acid concentration in the brain and ischemic lesion in the hippocampus with a higher NF-κB concentration. CONCLUSION The results demonstrate deleterious effects of a maternal HFD on the neurobehavioral and biochemical parameters of their offspring which may be associated with the role of organic acids and NF-κB in fetal programming.
Collapse
Affiliation(s)
- Anne Caroline Alves Vieira
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Rafael Oliveira Pinheiro
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Naís Lira Soares
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Maria Luiza Rolim Bezerra
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Davi Dos Santos Nascimento
- Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil; Laboratory of food microbiology and biochemistry, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil
| | - Adriano Francisco Alves
- Laboratory of General pathology, Department of Physiology and Pathology, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Maria Carolina de Paiva Sousa
- Laboratory of General pathology, Department of Physiology and Pathology, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Maria Letícia da Veiga Dutra
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Marcos Dos Santos Lima
- Department of Food Technology, Federal Institute of Sertão Pernambucano, Petrolina, Brazil; Post Graduate Program in Food Science and Technology, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil
| | - Nilcimelly Rodrigues Donato
- Department of Nutrition, Center for Education and Health, Federal University of Campina Grande (UFCG), Cuité, Paraíba, Brazil
| | - Jailane de Souza Aquino
- Laboratory of Experimental Nutrition, Department of Nutrition, Federal University of Paraíba, UFPB, João Pessoa PB, Brazil; Post Graduate Program in Nutrition Sciences, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil; Post Graduate Program in Food Science and Technology, Federal University of Paraíba (UFPB), João Pessoa, Paraíba, Brazil.
| |
Collapse
|
37
|
Cho H, Jo M, Oh H, Lee Y, Park Y. Synergistic antidepressant-like effect of n-3 polyunsaturated fatty acids and probiotics through the brain-gut axis in rats exposed to chronic mild stress. J Nutr Biochem 2023; 116:109326. [PMID: 36963732 DOI: 10.1016/j.jnutbio.2023.109326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 03/17/2023] [Accepted: 03/17/2023] [Indexed: 03/26/2023]
Abstract
N-3 polyunsaturated fatty acids (PUFA) and probiotics have antidepressant-like effects, but the underlying mechanisms are unclear. We hypothesized that n-3 PUFA combined with live and dead probiotics synergistically improves depression by modulating the hypothalamic-pituitary-adrenal (HPA) axis and serotonergic pathways through the brain-gut axis. Rats were randomly divided into seven groups (n = 8/group): non-chronic mild stress (CMS) with n-6 PUFA, CMS with n-3 PUFA, n-6 PUFA, live probiotics, dead probiotics, n-3 PUFA and live probiotics, and n-3 PUFA and dead probiotics. Diets of n-6 and n-3 PUFA and oral supplementation of live and dead probiotics were provided for 12 weeks, and CMS was performed for the last 5 weeks. N-3 PUFA and probiotics improved depressive behaviors and modulated the brain and gut HPA axis by synergistically increasing glucocorticoid receptor expression and decreasing corticotropin-releasing factor expression and blood levels of adrenocorticotropic hormone and corticosterone. N-3 PUFA and probiotics upregulated the brain serotonergic pathway through serotonin levels and expression of brain-derived neurotrophic factor, phosphorylated cAMP response binding protein, and 5-hydroxytryptamine 1A receptor while downregulating the gut serotonergic pathway. Furthermore, n-3 PUFA and probiotics increased the abundance of Ruminococcaceae, brain and gut short chain fatty acid levels, and occludin expression while decreasing the expression of tumor necrosis factor-α, interleukin-1β, and prostaglandin E2 and blood lipopolysaccharides levels. There was no significant difference between the live and dead probiotics. In conclusion, n-3 PUFA and probiotics had synergistic antidepressant-like effects on the HPA axis and serotonergic pathways of the brain and gut through the brain-gut axis.
Collapse
Affiliation(s)
- Hyunji Cho
- Department of Food and Nutrition, Hanyang University, Seoul 04763, Korea
| | - Miyea Jo
- Department of Food and Nutrition, Hanyang University, Seoul 04763, Korea
| | - Haemin Oh
- Department of Food and Nutrition, Hanyang University, Seoul 04763, Korea
| | - Yunjung Lee
- Department of Food and Nutrition, Hanyang University, Seoul 04763, Korea
| | - Yongsoon Park
- Department of Food and Nutrition, Hanyang University, Seoul 04763, Korea.
| |
Collapse
|
38
|
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
|
39
|
Martín-Hernández D, Martínez M, Robledo-Montaña J, Muñoz-López M, Virto L, Ambrosio N, Marín MJ, Montero E, Herrera D, Sanz M, Leza JC, Figuero E, García-Bueno B. Neuroinflammation related to the blood-brain barrier and sphingosine-1-phosphate in a pre-clinical model of periodontal diseases and depression in rats. J Clin Periodontol 2023; 50:642-656. [PMID: 36644813 DOI: 10.1111/jcpe.13780] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/17/2023]
Abstract
AIM To explore the potential mechanisms of neuroinflammation (microglia, blood-brain barrier [BBB] permeability, and the sphingosine-1-phosphate [S1P] pathways) resulting from the association between periodontitis and depression in rats. MATERIALS AND METHODS This pre-clinical in vivo experimental study used Wistar rats, in which experimental periodontitis (P) was induced by using oral gavages with Porphyromonas gingivalis and Fusobacterium nucleatum. Then, a chronic mild stress (CMS) model was implemented to induce a depressive-like behaviour, resulting in four groups: P with CMS (P+CMS+), P without CMS (P+CMS-), CMS without P (P-CMS+), and control (P-CMS-). After harvesting brain samples, protein/mRNA expression analyses and fluorescence immunohistochemistry were performed in the frontal cortex (FC). Results were analysed by ANOVA. RESULTS CMS exposure increased the number of microglia (an indicator of neuroinflammation) in the FC. In the combined model (P+CMS+), there was a decrease in the expression of tight junction proteins (zonula occludens-1 [ZO-1], occludin) and an increase in intercellular and vascular cell adhesion molecules (ICAM-1, VCAM-1) and matrix metalloproteinase 9 (MMP9), suggesting a more severe disruption of the BBB. The enzymes and receptors of S1P were also differentially regulated. CONCLUSIONS Microglia, BBB permeability, and S1P pathways could be relevant mechanisms explaining the association between periodontitis and depression.
Collapse
Affiliation(s)
- David Martín-Hernández
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), Hospital 12 de Octubre Research Institute (Imas12), Neurochemistry Research Institute UCM (IUIN), Madrid, Spain.,Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - María Martínez
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Javier Robledo-Montaña
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), Hospital 12 de Octubre Research Institute (Imas12), Neurochemistry Research Institute UCM (IUIN), Madrid, Spain.,Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - Marina Muñoz-López
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), Hospital 12 de Octubre Research Institute (Imas12), Neurochemistry Research Institute UCM (IUIN), Madrid, Spain.,Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - Leire Virto
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Anatomy and Embryology, Faculty of Optics, UCM, Madrid, Spain
| | - Nagore Ambrosio
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Maria José Marín
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain
| | - Eduardo Montero
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - David Herrera
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Mariano Sanz
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Juan C Leza
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), Hospital 12 de Octubre Research Institute (Imas12), Neurochemistry Research Institute UCM (IUIN), Madrid, Spain.,Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| | - Elena Figuero
- ETEP (Etiology and Therapy of Periodontal and Peri-implant Diseases) Research Group, UCM, Madrid, Spain.,Department of Dental Clinical Specialties, Faculty of Dentistry, UCM, Madrid, Spain
| | - Borja García-Bueno
- Department of Pharmacology and Toxicology, Faculty of Medicine, Complutense University of Madrid (UCM), Hospital 12 de Octubre Research Institute (Imas12), Neurochemistry Research Institute UCM (IUIN), Madrid, Spain.,Biomedical Network Research Center of Mental Health (CIBERSAM), Institute of Health Carlos III, Madrid, Spain
| |
Collapse
|
40
|
Pal D, Sahu P, Mishra AK, Hagelgans A, Sukocheva O. Histone Deacetylase Inhibitors as Cognitive Enhancers and Modifiers of Mood and Behavior. Curr Drug Targets 2023; 24:728-750. [PMID: 36475351 DOI: 10.2174/1389450124666221207090108] [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: 05/25/2022] [Revised: 09/29/2022] [Accepted: 10/10/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Epigenetic regulation of gene signalling is one of the fundamental molecular mechanisms for the generation and maintenance of cellular memory. Histone acetylation is a common epigenetic mechanism associated with increased gene transcription in the central nervous system (CNS). Stimulation of gene transcription by histone acetylation is important for the development of CNS-based long-term memory. Histone acetylation is a target for cognitive enhancement via the application of histone deacetylase (HDAC) inhibitors. The promising potential of HDAC inhibitors has been observed in the treatment of several neurodevelopmental and neurodegenerative diseases. OBJECTIVE This study assessed the current state of HDAC inhibition as an approach to cognitive enhancement and treatment of neurodegenerative diseases. Our analysis provides insights into the mechanism of action of HDAC inhibitors, associated epigenetic priming, and describes the therapeutic success and potential complications after unsupervised use of the inhibitors. RESULTS AND CONCLUSION Several chromatin-modifying enzymes play key roles in the regulation of cognitive processes. The importance of HDAC signaling in the brain is highlighted in this review. Recent advancements in the field of cognitive epigenetics are supported by the successful development of various HDAC inhibitors, demonstrating effective treatment of mood-associated disorders. The current review discusses the therapeutic potential of HDAC inhibition and observed complications after mood and cognitive enhancement therapies.
Collapse
Affiliation(s)
- Dilipkumar Pal
- Department of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, C.G., 495 009, India
| | - Pooja Sahu
- Department of Pharmaceutical Sciences, Guru Ghasidas Vishwavidyalaya (A Central University), Bilaspur, C.G., 495 009, India
| | | | - Albert Hagelgans
- Institute of Clinical Chemistry and Laboratory Medicine, University Hospital `Carl Gustav Carus`, Technical University of Dresden, Dresden 01307, Germany
| | - Olga Sukocheva
- College of Nursing and Health Sciences, Flinders University of South Australia, Bedford Park, 5042, SA, Australia
| |
Collapse
|
41
|
Han W, Wang N, Han M, Ban M, Sun T, Xu J. Reviewing the role of gut microbiota in the pathogenesis of depression and exploring new therapeutic options. Front Neurosci 2022; 16:1029495. [PMID: 36570854 PMCID: PMC9772619 DOI: 10.3389/fnins.2022.1029495] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 11/24/2022] [Indexed: 12/14/2022] Open
Abstract
The relationship between gut microbiota (GM) and mental health is one of the focuses of psychobiology research. In recent years, the microbial-gut-brain axis (MGBA) concept has gradually formed about this bidirectional communication between gut and brain. But how the GM is involved in regulating brain function and how they affect emotional disorders these mechanisms are tenuous and limited to animal research, and often controversial. Therefore, in this review, we attempt to summarize and categorize the latest advances in current research on the mechanisms of GM and depression to provide valid information for future diagnoses and therapy of mental disorders. Finally, we introduced some antidepressant regimens that can help restore gut dysbiosis, including classic antidepressants, Chinese materia medica (CMM), diet, and exogenous strains. These studies provide further insight into GM's role and potential pathways in emotion-related diseases, which holds essential possible clinical outcomes for people with depression or related psychiatric disorders. Future research should focus on clarifying the causal role of GM in disease and developing microbial targets, applying these findings to the prevention and treatment of depression.
Collapse
Affiliation(s)
- Wenjie Han
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Pharmacology, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China
| | - Na Wang
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Pharmacology, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China
| | - Mengzhen Han
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Pharmacology, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China
| | - Meng Ban
- Liaoning Microhealth Biotechnology Co., Ltd., Shenyang, China
| | - Tao Sun
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China
| | - Junnan Xu
- Department of Breast Medicine, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Pharmacology, Liaoning Cancer Hospital, Cancer Hospital of China Medical University, Shenyang, China,Department of Breast Medicine, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital, Shenyang, China,*Correspondence: Junnan Xu,
| |
Collapse
|
42
|
Intestinal Flora Affect Alzheimer's Disease by Regulating Endogenous Hormones. Neurochem Res 2022; 47:3565-3582. [DOI: 10.1007/s11064-022-03784-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/13/2022] [Accepted: 10/01/2022] [Indexed: 11/25/2022]
|
43
|
Anthocyanins from Opuntia ficus-indica Modulate Gut Microbiota Composition and Improve Short-Chain Fatty Acid Production. BIOLOGY 2022; 11:biology11101505. [PMID: 36290409 PMCID: PMC9598542 DOI: 10.3390/biology11101505] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Revised: 10/02/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
Opuntia ficus-indica is rich in a variety of active substances, such as anthocyanins, flavonoids, and polysaccharides. Some studies have shown that anthocyanins extracted from natural plants can regulate intestinal flora. The fruit was used as raw material, and anthocyanins were extracted from it. In vivo experiments were used to study the effect of Opuntia ficus-indica anthocyanins on the mouse intestine by 16S rRNA high-throughput sequencing (NovaSeq 6000 platform) and gas chromatography (hydrogen flame ionization detector (FID)) methods. Microbiota and effects of short-chain fatty acids (SCFAs). The results showed that after feeding anthocyanins, the diversity of intestinal microorganisms in mice was significantly increased (p < 0.05), the ratio of Firmicutes/Bacteroidetes (F/B value) was significantly decreased (p < 0.05), the relative abundances of beneficial bacteria Lactobacillus, Bifidobacterium, Prevotella, and Akkermansia in the intestinal tract of mice were significantly increased (p < 0.05), and the relative abundance of pathogenic bacteria Escherichia-Shigella and Desulfovibrio decreased significantly (p < 0.05). Furthermore, anthocyanins significantly increased the content of short-chain fatty acids in the cecum of mice, among which the content of acetic acid, propionic acid, and butyric acid increased the most. Opuntia ficus-indica anthocyanins can change the microbial diversity and flora composition of the mouse gut and promote the production of short-chain fatty acids. The findings provide a theoretical basis for the use of Opuntia ficus-indica anthocyanins as dietary supplements to regulate human intestinal flora.
Collapse
|
44
|
Zhu W, Peng K, Zhao Y, Xu C, Tao X, Liu Y, Huang Y, Yang X. Sodium butyrate attenuated diet-induced obesity, insulin resistance and inflammation partly by promoting fat thermogenesis via intro-adipose sympathetic innervation. Front Pharmacol 2022; 13:938760. [PMID: 36263123 PMCID: PMC9574364 DOI: 10.3389/fphar.2022.938760] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 09/12/2022] [Indexed: 11/13/2022] Open
Abstract
Emerging evidence suggests that butyrate, a short-chain fatty acid, may have beneficial effects on obesity and its associated metabolic comorbidities, but the related molecular mechanism is largely unknown. This study aims to investigate the role of butyrate in diet-induced obesity and metabolic disorders and the relevant regulatory mechanisms. Here, dietary supplementation with Sodium butyrate (NaB) was carried out in mice fed with a high-fat diet (HFD) or chow diet. At week 14, mice on HFD displayed an obese phenotype and down-regulated expression of thermogenic regulators including Ucp-1 and Pgc-1α in adipose tissue. Excitingly, NaB add-on treatment abolished these detrimental effects. Moreover, the obesity-induced insulin resistance, inflammation, fatty liver, and intestinal dysfunction were also attenuated by NaB administration. Mechanistically, NaB can promote fat thermogenesis via the increased local sympathetic innervation of adipose tissue, and blocking the β3-adrenergic signaling pathway by 6-hydroxydopamine abolished NaB-induced thermogenesis. Our study reveals a potential pharmacological target for NaB to combat obesity and metabolic disorders.
Collapse
Affiliation(s)
- Wanlong Zhu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Ke Peng
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yan Zhao
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Changjing Xu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xuemei Tao
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuanzhi Liu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yilan Huang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
- *Correspondence: Yilan Huang, ; Xuping Yang,
| | - Xuping Yang
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- School of Pharmacy, Southwest Medical University, Luzhou, China
- *Correspondence: Yilan Huang, ; Xuping Yang,
| |
Collapse
|
45
|
Tarawneh R, Penhos E. The gut microbiome and Alzheimer's disease: Complex and bidirectional interactions. Neurosci Biobehav Rev 2022; 141:104814. [PMID: 35934087 PMCID: PMC9637435 DOI: 10.1016/j.neubiorev.2022.104814] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 07/16/2022] [Accepted: 08/01/2022] [Indexed: 11/20/2022]
Abstract
Structural and functional alterations to the gut microbiome, referred to as gut dysbiosis, have emerged as potential key mediators of neurodegeneration and Alzheimer disease (AD) pathogenesis through the "gut -brain" axis. Emerging data from animal and clinical studies support an important role for gut dysbiosis in mediating neuroinflammation, central and peripheral immune dysregulation, abnormal brain protein aggregation, and impaired intestinal and brain barrier permeability, leading to neuronal loss and cognitive impairment. Gut dysbiosis has also been shown to directly influence various mechanisms involved in neuronal growth and repair, synaptic plasticity, and memory and learning functions. Aging and lifestyle factors including diet, exercise, sleep, and stress influence AD risk through gut dysbiosis. Furthermore, AD is associated with characteristic gut microbial signatures which offer value as potential markers of disease severity and progression. Together, these findings suggest the presence of a complex bidirectional relationship between AD and the gut microbiome and highlight the utility of gut modulation strategies as potential preventative or therapeutic strategies in AD. We here review the current literature regarding the role of the gut-brain axis in AD pathogenesis and its potential role as a future therapeutic target in AD treatment and/or prevention.
Collapse
Affiliation(s)
- Rawan Tarawneh
- Department of Neurology, Center for Memory and Aging, Alzheimer Disease Research Center, The University of New Mexico, Albuquerque, NM 87106, USA.
| | - Elena Penhos
- College of Medicine, The Ohio State University, Columbus, OH, USA 43210
| |
Collapse
|
46
|
Central and peripheral regulations mediated by short-chain fatty acids on energy homeostasis. Transl Res 2022; 248:128-150. [PMID: 35688319 DOI: 10.1016/j.trsl.2022.06.003] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 05/16/2022] [Accepted: 06/01/2022] [Indexed: 11/24/2022]
Abstract
The human gut microbiota influences obesity, insulin resistance, and the subsequent development of type 2 diabetes (T2D). The gut microbiota digests and ferments nutrients resulting in the production of short-chain fatty acids (SCFAs), which generate various beneficial metabolic effects on energy and glucose homeostasis. However, their roles in the central nervous system (CNS)-mediated outputs on the metabolism have only been minimally studied. Here, we explore what is known and future directions that may be worth exploring in this emerging area. Specifically, we searched studies or data in English by using PubMed, Google Scholar, and the Human Metabolome Database. Studies were filtered by time from 1978 to March 2022. As a result, 195 studies, 53 reviews, 1 website, and 1 book were included. One hundred and sixty-five of 195 studies describe the production and metabolism of SCFAs or the effects of SCFAs on energy homeostasis, glucose balance, and mental diseases through the gut-brain axis or directly by a central pathway. Thirty of 195 studies show that inappropriate metabolism and excessive of SCFAs are metabolically detrimental. Most studies suggest that SCFAs exert beneficial metabolic effects by acting as the energy substrate in the TCA cycle, regulating the hormones related to satiety regulation and insulin secretion, and modulating immune cells and microglia. These functions have been linked with AMPK signaling, GPCRs-dependent pathways, and inhibition of histone deacetylases (HDACs). However, the studies focusing on the central effects of SCFAs are still limited. The mechanisms by which central SCFAs regulate appetite, energy expenditure, and blood glucose during different physiological conditions warrant further investigation.
Collapse
|
47
|
Hua H, Huang C, Liu H, Xu X, Xu X, Wu Z, Liu C, Wang Y, Yang C. Depression and antidepressant effects of ketamine and its metabolites: The pivotal role of gut microbiota. Neuropharmacology 2022; 220:109272. [PMID: 36170927 DOI: 10.1016/j.neuropharm.2022.109272] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 09/19/2022] [Accepted: 09/20/2022] [Indexed: 02/07/2023]
Abstract
The discovery of the robust antidepressant actions of ketamine is regarded as one of the greatest advancements in depression treatment in the past 60 years. Recent findings have provided strong evidence for the presence of bidirectional communication networks between the gastrointestinal tract and the brain in depression. Moreover, increasing evidence supports the antidepressant role of ketamine in regulating the gut microbiome and microbiota-derived molecules; however, the mechanisms underpinning such effects are still ambiguous. This review summarizes the current understanding of the anti-depressant mechanisms of ketamine and its metabolites regarding the bidirectional regulation by microbiota-gut-brain axis. We review the relationship between gut microbiota and the antidepressant mechanisms of ketamine, and discuss the role of stress response, brain-derived neurotrophic factor (BDNF)-mediated neurogenesis, anti-inflammatory effect and neurotransmitters.
Collapse
Affiliation(s)
- Hao Hua
- Department of Anesthesiology, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, 214062, China
| | - Chaoli Huang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Hanyu Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Xiangyang Xu
- Nhwa Institute of Pharmaceutical Research, Jiangsu Nhwa Pharmaceutical Co., Ltd & Jiangsu Key Laboratory of Central Nervous System Drug Research and Development, Xuzhou, 221116, China
| | - Xiangqing Xu
- Nhwa Institute of Pharmaceutical Research, Jiangsu Nhwa Pharmaceutical Co., Ltd & Jiangsu Key Laboratory of Central Nervous System Drug Research and Development, Xuzhou, 221116, China
| | - Zifeng Wu
- Department of Anesthesiology, Wuxi 9th People's Hospital Affiliated to Soochow University, Wuxi, 214062, China
| | - Cunming Liu
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Yuanyuan Wang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| | - Chun Yang
- Department of Anesthesiology and Perioperative Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
48
|
Kumar Palepu MS, Dandekar MP. Remodeling of microbiota gut-brain axis using psychobiotics in depression. Eur J Pharmacol 2022; 931:175171. [PMID: 35926568 DOI: 10.1016/j.ejphar.2022.175171] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 07/14/2022] [Accepted: 07/21/2022] [Indexed: 12/11/2022]
Abstract
Depression is a multifaceted psychiatric disorder mainly orchestrated by dysfunction of neuroendocrine, neurochemical, immune, and metabolic systems. The interconnection of gut microbiota perturbation with the central nervous system disorders has been well documented in recent times. Indeed, alteration of commensal intestinal microflora is noted in several psychiatric disorders such as anxiety and depression, which are presumed to be routed through the enteric nervous system, autonomic nervous system, endocrine, and immune system. This review summarises the new mechanisms underlying the crosstalk between gut microbiota and brain involved in the management of depression. Depression-induced changes in the commensal intestinal microbiota are majorly linked with the disruption of gut integrity, hyperinflammation, and modulation of short-chain fatty acids, neurotransmitters, kynurenine metabolites, endocannabinoids, brain-derived neurotropic factors, hypothalamic-pituitary-adrenal axis, and gut peptides. The restoration of gut microbiota with prebiotics, probiotics, postbiotics, synbiotics, and fermented foods (psychobiotics) has gained a considerable attention for the management of depression. Recent evidence also propose the role of gut microbiota in the process of treatment-resistant depression. Thus, remodeling of the microbiota-gut-brain axis using psychobiotics appears to be a promising therapeutic approach for the reversal of psychiatric disorders, and it is imperative to decipher the underlying mechanisms for gut-brain crosstalk.
Collapse
Affiliation(s)
- Mani Surya Kumar Palepu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India
| | - Manoj P Dandekar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Balanagar, Hyderabad, Telangana, India.
| |
Collapse
|
49
|
Kim S, Park S, Choi TG, Kim SS. Role of Short Chain Fatty Acids in Epilepsy and Potential Benefits of Probiotics and Prebiotics: Targeting “Health” of Epileptic Patients. Nutrients 2022; 14:nu14142982. [PMID: 35889939 PMCID: PMC9322917 DOI: 10.3390/nu14142982] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 07/17/2022] [Accepted: 07/19/2022] [Indexed: 11/16/2022] Open
Abstract
The WHO’s definition of health transcends the mere absence of disease, emphasizing physical, mental, and social well-being. As this perspective is being increasingly applied to the management of chronic diseases, research on gut microbiota (GM) is surging, with a focus on its potential for persistent and noninvasive dietary therapeutics. In patients with epilepsy (PWE), a chronic lack of seizure control along with often neglected psychiatric comorbidities greatly disrupt the quality of life. Evidence shows that GM-derived short chain fatty acids (SCFAs) may impact seizure susceptibility through modulating (1) excitatory/inhibitory neurotransmitters, (2) oxidative stress and neuroinflammation, and (3) psychosocial stress. These functions are also connected to shared pathologies of epilepsy and its two most common psychiatric consequences: depression and anxiety. As the enhancement of SCFA production is enabled through direct administration, as well as probiotics and prebiotics, related dietary treatments may exert antiseizure effects. This paper explores the potential roles of SCFAs in the context of seizure control and its mental comorbidities, while analyzing existing studies on the effects of pro/prebiotics on epilepsy. Based on currently available data, this study aims to interpret the role of SCFAs in epileptic treatment, extending beyond the absence of seizures to target the health of PWE.
Collapse
Affiliation(s)
- Soomin Kim
- Department of Preliminary Medicine, School of Medicine, Kyung Hee University, Seoul 02447, Korea;
| | - Siyeon Park
- School of Pharmacy, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA;
| | - Tae Gyu Choi
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (T.G.C.); (S.S.K.); Tel.: +82-2-961-0287 (T.G.C.); +82-2-961-0524 (S.S.K.)
| | - Sung Soo Kim
- Department of Biochemistry and Molecular Biology, School of Medicine, Kyung Hee University, Seoul 02447, Korea
- Biomedical Science Institute, Kyung Hee University, Seoul 02447, Korea
- Correspondence: (T.G.C.); (S.S.K.); Tel.: +82-2-961-0287 (T.G.C.); +82-2-961-0524 (S.S.K.)
| |
Collapse
|
50
|
Luo H, Li W, Wu L, Zhong S, Du C, Liu Y, Xu Y, Huang X, Bahru AH, Tang X, Zhou J, Wang D, Lou X, Bin X, Xiao X. Differences in cognition, short-chain fatty acids and related metabolites in pregnant versus non-pregnant women: a cross-sectional study. BMC Pregnancy Childbirth 2022; 22:533. [PMID: 35778690 PMCID: PMC9248184 DOI: 10.1186/s12884-022-04853-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Accepted: 06/21/2022] [Indexed: 11/10/2022] Open
Abstract
Background Pregnancy induces cognitive reorganization which can lead to mental disorders. The aim of this study is to determine differences in cognitive scores, short-chain fatty acids (SCFAs) and related metabolites between pregnant and non-pregnant participants. Methods This cross-sectional study included 67 full-term pregnant women and 31 non-pregnant women. We compared scores of mental state and cognitive assessment tests, as well as serum concentrations of SCFAs, hormones, inflammatory factors, and neurotransmitters between these groups. Results Scores for information processing speed, immediate visual memory, motor response speed and accuracy, execution ability and verbal use ability in the pregnant group were lower than those in the non-pregnant group (p < 0.05 for all tests). Total serum SCFAs in the pregnant group were significantly lower than those in the non-pregnant group (P = 0.031). Among them, acetate and propionate were significantly decreased (P = 0.013 and 0.037, respectively) whereas butyrate was significantly increased (P = 0.035). Serum peptide YY, glucagon-like peptide-1, γ-aminobutyric acid, and dopamine showed no differences between the two groups. However, cortisol, adrenocorticotropic hormone, and acetylcholine were significantly increased in the pregnant group as compared with the non-pregnant group (P = 0.039, 0.016, and 0.012, respectively). Tumor necrosis factor-α was increased and interleukin-10 significantly decreased in the pregnant group (P = 0.045 and 0.019, respectively). Conclusion According to our study findings, cognitive reorganization in the third trimester of pregnancy showed that both the passive storage capacity of working memory and the executive function of online information processing were decreased to varying degrees. At the same time, the changes in total SCFAs, the proportions of SCFAs and related metabolites were also detected. These changes in the internal environment may be increasing the risk of perinatal mental illness.
Collapse
Affiliation(s)
- Huijuan Luo
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong, 510630, China
| | - Wengxiang Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong, 510630, China
| | - Lulu Wu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong, 510630, China
| | - Shuming Zhong
- Department of Psychology, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, 510630, China
| | - Chengrong Du
- Department of Clinical Medicine, International College, Jinan University, Guangzhou, Guangdong, 510630, China
| | - Yimeng Liu
- Department of Clinical Medicine, International College, Jinan University, Guangzhou, Guangdong, 510630, China
| | - Yating Xu
- Department of Clinical Medicine, International College, Jinan University, Guangzhou, Guangdong, 510630, China
| | - Xinyu Huang
- Department of Clinical Medicine, International College, Jinan University, Guangzhou, Guangdong, 510630, China
| | - Awol Hanan Bahru
- Department of Clinical Medicine, International College, Jinan University, Guangzhou, Guangdong, 510630, China
| | - Xiaomei Tang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong, 510630, China
| | - Juan Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong, 510630, China
| | - Dongju Wang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong, 510630, China
| | - Xiangying Lou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong, 510630, China
| | - Xuefan Bin
- Shanghai Medical College, Fudan University (SMCFU), 138 Yi xue yuan Road, Shanghai, 200032, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, No.601, West Huangpu Avenue, Guangzhou, Guangdong, 510630, China.
| |
Collapse
|