1
|
Erdrich S, Gelissen IC, Vuyisich M, Toma R, Harnett JE. An association between poor oral health, oral microbiota, and pain identified in New Zealand women with central sensitisation disorders: a prospective clinical study. FRONTIERS IN PAIN RESEARCH 2025; 6:1577193. [PMID: 40270934 PMCID: PMC12014678 DOI: 10.3389/fpain.2025.1577193] [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: 02/15/2025] [Accepted: 03/17/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction The portal to the gastrointestinal tract is the oral cavity, with transient and permanent microbial residents. Oral pathogens are implicated in the aetiology of several chronic conditions. To date, the role of oral health and the oral microbiota in the aetiology of pain in sensitisation disorders have not been explored. Here, we examined associations between self-reported oral health, the oral microbiome, and various pain presentations in women. Methods Oral health in women was assessed using the WHO oral health questionnaire. Body pain, migraine, and abdominal pain were determined using validated instruments. Saliva samples were evaluated using metatranscriptomics for relative gene abundance. Demographic and clinical characteristics data were evaluated for relationships between oral health scores, pain measures, and the oral microbiota at three taxa levels. Results Participants in the lowest quintiles for oral health were more likely to suffer migraine headaches (χ 2 = 23.24, df 4, p < 0.001) and higher body pain scores. Four oral pathogenic species were significantly associated with SF36 bodily pain (q < 0.05) after controlling for confounders. Relative abundance of Gardnerella (genus) correlated moderately with oral health scores (ρ = -0.346, q = 0.001), while Lancefieldella (genus) and Mycoplasma salivarius were associated with migraine. Discussion Low oral health scores correlated with higher pain scores. Both were associated with higher relative abundance of oral pathobionts. This suggests a potential role for the oral microbiota in the aetiology of pain experienced by women with migraine headache and abdominal and body pain. These findings prompt consideration of an oral microbiome-nervous system axis. Trial registration The study was registered with the Australia and New Zealand Clinical Trials Registry (ANZCTR), registration number ACTRN12620001337965, on 11/12/2020 https://www.anzctr.org.au/, and with the World Health Organisation, UTN: U1111-1258-5108.
Collapse
Affiliation(s)
- Sharon Erdrich
- Faculty of Medicine and Health, Sydney Pharmacy School, The University of Sydney, Sydney, NSW, Australia
| | - Ingrid C. Gelissen
- Faculty of Medicine and Health, Sydney Pharmacy School, The University of Sydney, Sydney, NSW, Australia
| | | | - Ryan Toma
- Viome Life Sciences, Bothell, WA, United States
| | - Joanna E. Harnett
- Faculty of Medicine and Health, Sydney Pharmacy School, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
2
|
Nista EC, Parello S, Brigida M, Amadei G, Saviano A, De Lucia SS, Petruzziello C, Migneco A, Ojetti V. Exploring the Role of Gut Microbiota and Probiotics in Acute Pancreatitis: A Comprehensive Review. Int J Mol Sci 2025; 26:3433. [PMID: 40244415 PMCID: PMC11989318 DOI: 10.3390/ijms26073433] [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: 03/09/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/18/2025] Open
Abstract
Acute pancreatitis (AP) is a common and potentially severe gastrointestinal condition characterized by acute inflammation of the pancreas. The pathophysiology of AP is multifactorial and intricate, involving a cascade of events that lead to pancreatic injury and systemic inflammation. The progression of AP is influenced by many factors, including genetic predispositions, environmental triggers, and immune dysregulation. Recent studies showed a critical involvement of the gut microbiota in shaping the immune response and modulating inflammatory processes during AP. This review aims to provide a comprehensive overview of the emerging role of gut microbiota and probiotics in AP. We analyzed the implication of gut microbiota in pathogenesis of AP and the modification during an acute attack. The primary goals of microbiome-based therapies, which include probiotics, prebiotics, antibiotics, fecal microbiota transplantation, and enteral nutrition, are to alter the composition of the gut microbial community and the amount of metabolites derived from the microbiota. By resetting the entire flora or supplementing it with certain beneficial organisms and their byproducts, these therapeutic approaches aim to eradicate harmful microorganisms, reducing inflammation and avoiding bacterial translocation and the potential microbiota-based therapeutic target for AP from nutrition to pre- and probiotic supplementation to fecal transplantation.
Collapse
Affiliation(s)
- Enrico Celestino Nista
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | - Simone Parello
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | - Mattia Brigida
- Gastroenterology Unit, Policlinico Universitario Tor Vergata, 00133 Rome, Italy;
| | - Giulio Amadei
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | - Angela Saviano
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | - Sara Sofia De Lucia
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | | | - Alessio Migneco
- Fondazione Policlinico Gemelli, Istituiti di Ricovero e Cura a Carattere Scientifico (IRCCS), 00168 Rome, Italy; (E.C.N.); (S.P.); (G.A.); (A.S.); (S.S.D.L.); (A.M.)
| | - Veronica Ojetti
- Ospedale San Carlo di Nancy, GVM Research, 00165 Rome, Italy
- Department of Internal Medicine, UniCamillus International Medical University of Rome, 00131 Rome, Italy
| |
Collapse
|
3
|
Liu J, Guo M, Yuan X, Fan X, Wang J, Jiao X. Gut Microbiota and Their Metabolites: The Hidden Driver of Diabetic Nephropathy? Unveiling Gut Microbe's Role in DN. J Diabetes 2025; 17:e70068. [PMID: 40189872 PMCID: PMC11973130 DOI: 10.1111/1753-0407.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/21/2025] [Accepted: 02/17/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Diabetic nephropathy (DN) is a severe microvascular complication of diabetes with a complex pathogenesis. METHODS Recent studies were reviewed to explore the role of gut microbiota and its metabolites in DN development. RESULTS Dysbiosis of gut bacteria contributes to pathological changes such as glomerular sclerosis and renal tubule injury. Microbial metabolites are involved in DN through immune and inflammatory pathways. CONCLUSIONS Understanding the relationship between gut microbiota, its metabolites, and DN may offer potential implications for DN diagnosis, prevention, and treatment. Translating this knowledge into clinical practice presents challenges and opportunities.
Collapse
Affiliation(s)
- Jinzhou Liu
- Department of PhysiologyThe Key Laboratory of Physiology of Shanxi Province, the Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical UniversityTaiyuanChina
| | - Min Guo
- Department of PhysiologyThe Key Laboratory of Physiology of Shanxi Province, the Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical UniversityTaiyuanChina
| | - Xiaobin Yuan
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Xiao Fan
- Department of UrologyFirst Hospital of Shanxi Medical UniversityTaiyuanChina
| | - Jin Wang
- Department of PhysiologyThe Key Laboratory of Physiology of Shanxi Province, the Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical UniversityTaiyuanChina
| | - Xiangying Jiao
- Department of PhysiologyThe Key Laboratory of Physiology of Shanxi Province, the Key Laboratory of Cellular Physiology of Ministry of Education, Shanxi Medical UniversityTaiyuanChina
| |
Collapse
|
4
|
Noureldein MH, Rumora AE, Teener SJ, Rigan DM, Hayes JM, Mendelson FE, Carter AD, Rubin WG, Savelieff MG, Feldman EL. Dietary Fatty Acid Composition Alters Gut Microbiome in Mice with Obesity-Induced Peripheral Neuropathy. Nutrients 2025; 17:737. [PMID: 40005065 PMCID: PMC11858455 DOI: 10.3390/nu17040737] [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: 01/23/2025] [Revised: 02/10/2025] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Peripheral neuropathy (PN), a complication of diabetes and obesity, progresses through a complex pathophysiology. Lifestyle interventions to manage systemic metabolism are recommended to prevent or slow PN, given the multifactorial risks of diabetes and obesity. A high-fat diet rich in saturated fatty acids (SFAs) induces PN, which a diet rich in monounsaturated fatty acids (MUFAs) rescues, independent of weight loss, suggesting factors beyond systemic metabolism impact nerve health. Interest has grown in gut microbiome mechanisms in PN, which is characterized by a distinct microbiota signature that correlates with sciatic nerve lipidome. METHODS Herein, we postulated that SFA- versus MUFA-rich diet would impact gut microbiome composition and correlate with PN development. To assess causality, we performed fecal microbiota transplantation (FMT) from donor mice fed SFA- versus MUFA-rich diet to lean recipient mice and assessed metabolic and PN phenotypes. RESULTS We found that the SFA-rich diet altered the microbiome community structure, which the MUFA-rich diet partially reversed. PN metrics correlated with several microbial families, some containing genera with feasible mechanisms of action for microbiome-mediated effects on PN. SFA and MUFA FMT did not impact metabolic phenotypes in recipient mice although SFA FMT marginally induced motor PN. CONCLUSIONS The involvement of diet-mediated changes in the microbiome on PN and gut-nerve axis may warrant further study.
Collapse
Affiliation(s)
- Mohamed H. Noureldein
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amy E. Rumora
- Department of Neurology, Columbia University, New York, NY 10032, USA
| | - Samuel J. Teener
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Diana M. Rigan
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - John M. Hayes
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Faye E. Mendelson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Andrew D. Carter
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Whitney G. Rubin
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| | - Masha G. Savelieff
- Department of Biomedical Sciences, University of North Dakota, Grand Forks, ND 58202, USA
| | - Eva L. Feldman
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
- NeuroNetwork for Emerging Therapies, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
5
|
Xie X, Li W, Xiong Z, Xu J, Liao T, Sun L, Xu H, Zhang M, Zhou J, Xiong W, Fu Z, Li Z, Han Q, Cui D, Anthony DC. Metformin reprograms tryptophan metabolism via gut microbiome-derived bile acid metabolites to ameliorate depression-Like behaviors in mice. Brain Behav Immun 2025; 123:442-455. [PMID: 39303815 DOI: 10.1016/j.bbi.2024.09.014] [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: 10/27/2023] [Revised: 09/08/2024] [Accepted: 09/13/2024] [Indexed: 09/22/2024] Open
Abstract
As an adjunct therapy, metformin enhances the efficacy of conventional antidepressant medications. However, its mode of action remains unclear. Here, metformin was found to ameliorate depression-like behaviors in mice exposed to chronic restraint stress (CRS) by normalizing the dysbiotic gut microbiome. Fecal transplants from metformin-treated mice ameliorated depressive behaviors in stressed mice. Microbiome profiling revealed that Akkermansia muciniphila (A. muciniphila), in particular, was markedly increased in the gut by metformin and that oral administration of this species alone was sufficient to reverse CRS-induced depressive behaviors and normalize aberrant stress-induced 5-hydroxytryptamine (5-HT) metabolism in the brain and gut. Untargeted metabolomic profiling further identified the bile acid metabolites taurocholate and deoxycholic acid as direct A. muciniphila-derived molecules that are, individually, sufficient to rescue the CRS-induced impaired 5-HT metabolism and depression-like behaviors. Thus, we report metformin reprograms 5-HT metabolism via microbiome-brain interactions to mitigate depressive syndromes, providing novel insights into gut microbiota-derived bile acids as potential therapeutic candidates for depressive mood disorders from bench to bedside.
Collapse
Affiliation(s)
- Xiaoxian Xie
- Shanghai Mental Health Center, Shanghai Jiao Tong University, School of Medicine, Shanghai 201109, PR China; Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT Oxford, UK; College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Wenwen Li
- Affiliated Mental Health Center and Hangzhou Seventh People's Hospital, School of Brain Science and Brain Medicine, Zhejiang University School of Medicine, Hangzhou 310058, PR China
| | - Ze Xiong
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Junyu Xu
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, PR China
| | - Tailin Liao
- NHC and CAMS Key Laboratory of Medical Neurobiology, Ministry of Education Frontier Science Center for Brain Research and Brain Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, PR China
| | - Lei Sun
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Haoshen Xu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Mengya Zhang
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Jiafeng Zhou
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Wenzheng Xiong
- Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT Oxford, UK
| | - Zhengwei Fu
- College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310032, PR China
| | - Zezhi Li
- The Affiliated Brain Hospital, Guangzhou Medical University, Guangzhou 510370, PR China; Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou Medical University, Guangzhou, PR China.
| | - Qi Han
- Center for Brain Science Shanghai Children s Medical Center, Department of Anatomy and Physiology, Shanghai Frontiers Science Center of Cellular Homeostasis and Human Disease, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China; Shanghai Center for Brain Science and Brain-inspired Technology, Shanghai 200031, PR China.
| | - Donghong Cui
- Shanghai Mental Health Center, Shanghai Jiao Tong University, School of Medicine, Shanghai 201109, PR China.
| | - Daniel C Anthony
- Department of Pharmacology, University of Oxford, Mansfield Road, OX1 3QT Oxford, UK
| |
Collapse
|
6
|
Wei X, Wang H, Liu S, Bao K, Ke S, Zhou Z. Surfactin's impact on gut microbiota and intestinal tumor cells. Int J Biol Macromol 2025; 287:138607. [PMID: 39662559 DOI: 10.1016/j.ijbiomac.2024.138607] [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: 09/13/2024] [Revised: 11/24/2024] [Accepted: 12/08/2024] [Indexed: 12/13/2024]
Abstract
Surfactin exhibits multifunctional properties, including antimicrobial, anti-inflammatory, and antitumor activities, positioning it as a promising novel food additive. However, its specific effects on the gut environment remain largely unexplored. To investigate this, we conducted in vitro fermentation simulations to assess the impact of surfactin on gut microbiota. Additionally, we evaluated surfactin's inhibitory effects on HCT-116 cells. The results of the fermentation simulation indicated that surfactin significantly increased butyrate production and enhanced the relative abundance of Megamonas, Alistipes, the Oscillospiraceae NK4A214 group, and Methanobrevibacter smithii, while markedly inhibiting Desulfobacterota, potentially facilitating amino acid and lipid metabolism. Furthermore, surfactin significantly inhibited the proliferation of HCT-116 cells and promoted their apoptosis, without exhibiting cytotoxicity towards normal human intestinal epithelial crypt (HIEC) cells. In summary, surfactin demonstrates a favorable safety profile in the intestinal environment. This study contributes to our understanding of the interaction between surfactin and the gut environment.
Collapse
Affiliation(s)
- Xinyue Wei
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Huifang Wang
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Shijia Liu
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Kexin Bao
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Sheng Ke
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Zhongkai Zhou
- College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China; College of Food Science, Shihezi University, Shihezi 832003, China; ARC Functional Grains Centre, Charles Sturt University, Wagga Wagga, NSW 2678, Australia.
| |
Collapse
|
7
|
Zhou F, Liu Y, Shi Y, Wu N, Xie Y, Zhou X. Association between gut microbiota and acute pancreatitis: a bidirectional Mendelian randomization study. J Gastroenterol Hepatol 2024; 39:1895-1902. [PMID: 38888069 DOI: 10.1111/jgh.16658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/07/2024] [Accepted: 05/28/2024] [Indexed: 06/20/2024]
Abstract
BACKGROUND AND AIM The dysbiosis of gut microbiota has been reported in acute pancreatitis. However, the direction and magnitude between host microbiota and pancreas remains to be established. This study investigated the association between gut microbiota and acute pancreatitis using Mendelian randomization (MR) methods. METHODS Summary statistics of gut microbiota abundance and acute pancreatitis were extracted from genome-wide association studies (GWAS). The two-sample bidirectional MR design was employed to assess genetic association between the microbiota and pancreatitis, followed by a comprehensive sensitivity analysis to verify the robustness of the results. RESULTS Seven microbiota taxa have been identified as significantly associated with the development of pancreatitis. Host genetic-driven order Bacteroidales and class Bacteroidia are associated with an increased risk of pancreatitis. The genera Coprococcus and Eubacterium fissicatena group also exhibit a positive effect on the development of pancreatitis, while the genera Prevotella, Ruminiclostridium, and Ruminococcaceae act as protective factors against pancreatitis. In contrast, acute pancreatitis was positively correlated with phylum Proteobacteria and genus Lachnospiraceae and negatively correlated with genus Holdemania. CONCLUSIONS The bidirectional relationship between gut microbiota and acute pancreatitis suggests a critical role for host-microbiota crosstalk in the development of the disease. Targeted modulation of specific gut microbiota enables the prevention and treatment of acute pancreatitis.
Collapse
Affiliation(s)
- Feng Zhou
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yang Liu
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Yanqing Shi
- Department of Gastroenterology, Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi Province, China
| | - Nanzhen Wu
- Department of Gastrointestinal Surgery, Fengcheng People's Hospital, Fengcheng, Jiangxi Province, China
| | - Yong Xie
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| | - Xiaojiang Zhou
- Department of Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
8
|
Hellysaz A, Hagbom M. Rotavirus Sickness Symptoms: Manifestations of Defensive Responses from the Brain. Viruses 2024; 16:1086. [PMID: 39066248 PMCID: PMC11281384 DOI: 10.3390/v16071086] [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: 05/20/2024] [Revised: 07/01/2024] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
Rotavirus is infamous for being extremely contagious and for causing diarrhea and vomiting in infants. However, the symptomology is far more complex than what could be expected from a pathogen restricted to the boundaries of the small intestines. Other rotavirus sickness symptoms like fever, fatigue, sleepiness, stress, and loss of appetite have been clinically established for decades but remain poorly studied. A growing body of evidence in recent years has strengthened the idea that the evolutionarily preserved defensive responses that cause rotavirus sickness symptoms are more than just passive consequences of illness and rather likely to be coordinated events from the central nervous system (CNS), with the aim of maximizing the survival of the individual as well as the collective group. In this review, we discuss both established and plausible mechanisms of different rotavirus sickness symptoms as a series of CNS responses coordinated from the brain. We also consider the protective and the harmful nature of these events and highlight the need for further and deeper studies on rotavirus etiology.
Collapse
Affiliation(s)
| | - Marie Hagbom
- Division of Molecular Medicine and Virology, Department of Biomedical and Clinical Sciences, Linköping University, 581 85 Linköping, Sweden;
| |
Collapse
|
9
|
Zhang YW, Wu Y, Liu XF, Chen X, Su JC. Targeting the gut microbiota-related metabolites for osteoporosis: The inextricable connection of gut-bone axis. Ageing Res Rev 2024; 94:102196. [PMID: 38218463 DOI: 10.1016/j.arr.2024.102196] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 12/24/2023] [Accepted: 01/09/2024] [Indexed: 01/15/2024]
Abstract
Osteoporosis is a systemic skeletal disease characterized by decreased bone mass, destruction of bone microstructure, raised bone fragility, and enhanced risk of fractures. The correlation between gut microbiota and bone metabolism has gradually become a widespread research hotspot in recent years, and successive studies have revealed that the alterations of gut microbiota and its-related metabolites are related to the occurrence and progression of osteoporosis. Moreover, several emerging studies on the relationship between gut microbiota-related metabolites and bone metabolism are also underway, and extensive research evidence has indicated an inseparable connection between them. Combined with latest literatures and based on inextricable connection of gut-bone axis, this review is aimed to summarize the relation, potential mechanisms, application strategies, clinical application prospects, and existing challenges of gut microbiota and its-related metabolites on osteoporosis, thus updating the knowledge in this research field and providing certain reference for future researches.
Collapse
Affiliation(s)
- Yuan-Wei Zhang
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Yan Wu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China
| | - Xiang-Fei Liu
- Department of Orthopaedics, Shanghai Zhongye Hospital, Shanghai 200941, China.
| | - Xiao Chen
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai 200092, China.
| | - Jia-Can Su
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai JiaoTong University School of Medicine, Shanghai 200092, China; Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
10
|
Elangovan A, Dahiya B, Kirola L, Iyer M, Jeeth P, Maharaj S, Kumari N, Lakhanpal V, Michel TM, Rao KRSS, Cho SG, Yadav MK, Gopalakrishnan AV, Kadhirvel S, Kumar NS, Vellingiri B. Does gut brain axis has an impact on Parkinson's disease (PD)? Ageing Res Rev 2024; 94:102171. [PMID: 38141735 DOI: 10.1016/j.arr.2023.102171] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 12/04/2023] [Accepted: 12/13/2023] [Indexed: 12/25/2023]
Abstract
Parkinson's Disease (PD) is becoming a growing global concern by being the second most prevalent disease next to Alzheimer's Disease (AD). Henceforth new exploration is needed in search of new aspects towards the disease mechanism and origin. Evidence from recent studies has clearly stated the role of Gut Microbiota (GM) in the maintenance of the brain and as a root cause of various diseases and disorders including other neurological conditions. In the case of PD, with an unknown etiology, the GM is said to have a larger impact on the disease pathophysiology. Although GM and its metabolites are crucial for maintaining the normal physiology of the host, it is an undeniable fact that there is an influence of GM in the pathophysiology of PD. As such the Enteroendocrine Cells (EECs) in the epithelium of the intestine are one of the significant regulators of the gut-brain axis and act as a communication mediator between the gut and the brain. The communication is established via the molecules of neuroendocrine which are said to have a crucial part in neurological diseases such as AD, PD, and other psychiatry-related disorders. This review is focused on understanding the proper role of GM and EECs in PD. Here, we also focus on some of the metabolites and compounds that can interact with the PD genes causing various dysfunctions in the cell and facilitating the disease conditions using bioinformatical tools. Various mechanisms concerning EECs and PD, their identification, the latest studies, and available current therapies have also been discussed.
Collapse
Affiliation(s)
- Ajay Elangovan
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Bhawna Dahiya
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Laxmi Kirola
- Department of Biotechnology, School of Health Sciences and Technology (SoHST), UPES University, Dehradun, Uttarakhand 248007, India
| | - Mahalaxmi Iyer
- Department of Microbiology, Central University of Punjab, Bathinda 151401, Punjab, India; Department of Biotechnology, Karpagam Academy of Higher Education (Deemed to be University), Coimbatore 641021, Tamil Nadu, India
| | - Priyanka Jeeth
- Department of Computational Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Sakshi Maharaj
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Nikki Kumari
- Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Vikas Lakhanpal
- Department of Neurology, All India Institute of Medical Sciences, Bathinda 151005, Punjab, India
| | - Tanja Maria Michel
- Research Unit of Psychiatry, Dept. of Psychiatry Odense, Clinical Institute, University of Southern Denmark, J.B. Winslowsvej 20, Indg. 220B, Odense, Denmark
| | - K R S Sambasiva Rao
- Mangalayatan University - Jabalpur, Jabalpur - 481662, Madhya Pradesh, India
| | - Ssang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, Molecular & Cellular Reprogramming Center and Institute of Advanced Regenerative Science, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Mukesh Kumar Yadav
- Department of Microbiology, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology, Vellore 632 014, India
| | - Saraboji Kadhirvel
- Department of Computational Sciences, Central University of Punjab, Bathinda 151401, Punjab, India
| | - Nachimuthu Senthil Kumar
- Department of Biotechnology, Mizoram University (A Central University), Aizawl, 796 004 Mizoram, India
| | - Balachandar Vellingiri
- Human Cytogenetics and Stem Cell Laboratory, Department of Zoology, School of Basic Sciences, Central University of Punjab, Bathinda 151401, Punjab, India.
| |
Collapse
|
11
|
Corriero A, Giglio M, Inchingolo F, Moschetta A, Varrassi G, Puntillo F. Gut Microbiota Modulation and Its Implications on Neuropathic Pain: A Comprehensive Literature Review. Pain Ther 2024; 13:33-51. [PMID: 38087070 PMCID: PMC10796891 DOI: 10.1007/s40122-023-00565-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Accepted: 11/13/2023] [Indexed: 01/19/2024] Open
Abstract
Neuropathic pain (NP) is a chronic pain disorder arising from somatosensory nervous system impairment. Extensive evidence supports the notion that the gut microbiota (GM) is crucial in maintaining human health by performing vital tasks. At the same time, its disruption has been linked to the emergence and advancement of an expanding range of disorders, including NP, in which GM could play a role in its pathophysiology. The crosstalk between the nervous system and GM happens through immune mediators, metabolites, and nervous structures and involves both central and peripheral nervous systems. This literature review aims to thoroughly investigate the function of modulating GM in the treatment of NP. It will achieve this by integrating existing knowledge, identifying underlying mechanisms, and evaluating the possible clinical consequences of exploiting the gut-brain axis. We will cover the main therapeutic applications of the described GM-modulators, such as probiotics, faecal microbiota transplantation, dietary supplements and emotional support, to the main kinds of NP in which any evidence, even if only pre-clinical, has been unravelled in recent years. The explored NP areas include chemotherapy-induced peripheral neuropathy, diabetic neuropathy, trauma-induced neuropathic pain, trigeminal neuralgia, postherpetic neuralgia and low back pain.
Collapse
Affiliation(s)
- Alberto Corriero
- Department of Interdisciplinary Medicine - ICU Section, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy.
| | - Mariateresa Giglio
- Department of Interdisciplinary Medicine - ICU Section, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine - Dental Medicine Section, University of Bari "Aldo Moro", 70124, Bari, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", Piazza Giulio Cesare 11, 70124, Bari, Italy
| | | | - Filomena Puntillo
- Department of Interdisciplinary Medicine - ICU Section, University of Bari "Aldo Moro", Piazza G. Cesare 11, 70124, Bari, Italy.
| |
Collapse
|
12
|
Cescon M, Gambarotta G, Calabrò S, Cicconetti C, Anselmi F, Kankowski S, Lang L, Basic M, Bleich A, Bolsega S, Steglich M, Oliviero S, Raimondo S, Bizzotto D, Haastert-Talini K, Ronchi G. Gut microbiota depletion delays somatic peripheral nerve development and impairs neuromuscular junction maturation. Gut Microbes 2024; 16:2363015. [PMID: 38845453 PMCID: PMC11164225 DOI: 10.1080/19490976.2024.2363015] [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: 08/02/2023] [Accepted: 05/29/2024] [Indexed: 06/12/2024] Open
Abstract
Gut microbiota is responsible for essential functions in human health. Several communication axes between gut microbiota and other organs via neural, endocrine, and immune pathways have been described, and perturbation of gut microbiota composition has been implicated in the onset and progression of an emerging number of diseases. Here, we analyzed peripheral nerves, dorsal root ganglia (DRG), and skeletal muscles of neonatal and young adult mice with the following gut microbiota status: a) germ-free (GF), b) gnotobiotic, selectively colonized with 12 specific gut bacterial strains (Oligo-Mouse-Microbiota, OMM12), or c) natural complex gut microbiota (CGM). Stereological and morphometric analyses revealed that the absence of gut microbiota impairs the development of somatic median nerves, resulting in smaller diameter and hypermyelinated axons, as well as in smaller unmyelinated fibers. Accordingly, DRG and sciatic nerve transcriptomic analyses highlighted a panel of differentially expressed developmental and myelination genes. Interestingly, the type III isoform of Neuregulin1 (NRG1), known to be a neuronal signal essential for Schwann cell myelination, was overexpressed in young adult GF mice, with consequent overexpression of the transcription factor Early Growth Response 2 (Egr2), a fundamental gene expressed by Schwann cells at the onset of myelination. Finally, GF status resulted in histologically atrophic skeletal muscles, impaired formation of neuromuscular junctions, and deregulated expression of related genes. In conclusion, we demonstrate for the first time a gut microbiota regulatory impact on proper development of the somatic peripheral nervous system and its functional connection to skeletal muscles, thus suggesting the existence of a novel 'Gut Microbiota-Peripheral Nervous System-axis.'
Collapse
Affiliation(s)
- Matilde Cescon
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| | - Sonia Calabrò
- Department of Molecular Medicine, University of Padova, Padova, Italy
- Department of Biology, University of Padova, Padova, Italy
| | - Chiara Cicconetti
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Francesca Anselmi
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Svenja Kankowski
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Lower-Saxony, Germany
| | - Luisa Lang
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Lower-Saxony, Germany
| | - Marijana Basic
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Lower-Saxony, Germany
| | - Andre Bleich
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Lower-Saxony, Germany
| | - Silvia Bolsega
- Institute for Laboratory Animal Science and Central Animal Facility, Hannover Medical School, Hannover, Lower-Saxony, Germany
| | - Matthias Steglich
- Research Core Unit Genomics, Hannover Medical School, Hannover, Lower-Saxony, Germany
| | - Salvatore Oliviero
- Department of Life Sciences and Systems Biology, University of Torino, Torino, Italy
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| | - Dario Bizzotto
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Kirsten Haastert-Talini
- Institute of Neuroanatomy and Cell Biology, Hannover Medical School, Hannover, Lower-Saxony, Germany
- Centre for Systems Neuroscience (ZSN), Hannover Medical School, Hannover, Lower-Saxony, Germany
| | - Giulia Ronchi
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Orbassano, Italy
| |
Collapse
|
13
|
Khan R, Di Gesù CM, Lee J, McCullough LD. The contribution of age-related changes in the gut-brain axis to neurological disorders. Gut Microbes 2024; 16:2302801. [PMID: 38237031 PMCID: PMC10798364 DOI: 10.1080/19490976.2024.2302801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 01/04/2024] [Indexed: 01/22/2024] Open
Abstract
Trillions of microbes live symbiotically in the host, specifically in mucosal tissues such as the gut. Recent advances in metagenomics and metabolomics have revealed that the gut microbiota plays a critical role in the regulation of host immunity and metabolism, communicating through bidirectional interactions in the microbiota-gut-brain axis (MGBA). The gut microbiota regulates both gut and systemic immunity and contributes to the neurodevelopment and behaviors of the host. With aging, the composition of the microbiota changes, and emerging studies have linked these shifts in microbial populations to age-related neurological diseases (NDs). Preclinical studies have demonstrated that gut microbiota-targeted therapies can improve behavioral outcomes in the host by modulating microbial, metabolomic, and immunological profiles. In this review, we discuss the pathways of brain-to-gut or gut-to-brain signaling and summarize the role of gut microbiota and microbial metabolites across the lifespan and in disease. We highlight recent studies investigating 1) microbial changes with aging; 2) how aging of the maternal microbiome can affect offspring health; and 3) the contribution of the microbiome to both chronic age-related diseases (e.g., Parkinson's disease, Alzheimer's disease and cerebral amyloidosis), and acute brain injury, including ischemic stroke and traumatic brain injury.
Collapse
Affiliation(s)
- Romeesa Khan
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Claudia M. Di Gesù
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Juneyoung Lee
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Louise D. McCullough
- Department of Neurology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| |
Collapse
|
14
|
Orozco-Cordoba J, Mazas C, Du Pont G, Lamoyi E, Cárdenas G, Fierro NA. Viral Biology and Immune Privilege in the Development of Extrahepatic Manifestations During Hepatitis E Virus Infection. Viral Immunol 2023; 36:627-641. [PMID: 38064537 DOI: 10.1089/vim.2023.0096] [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: 12/22/2023] Open
Abstract
Hepatitis E virus (HEV) exhibits tropism toward hepatocytes and thus affects the liver; however, HEV may also affect other tissues, including the heart, kidneys, intestines, testicles, and central nervous system. To date, the pathophysiological links between HEV infection and extrahepatic manifestations have not yet been established. Considering that HEV infects multiple types of cells, the direct effects of virus replication in peripheral tissues represent a plausible explanation for extrahepatic manifestations. In addition, since the immune response is crucial in the development of the disease, the immune characteristics of affected tissues should be revisited to identify commonalities explaining the effects of the virus. This review summarizes the most recent advances in understanding the virus biology and immune-privileged status of specific tissues as major elements for HEV replication in diverse organs. These discoveries may open avenues to explain the multiple extrahepatic manifestations associated with HEV infection and ultimately to design effective strategies for infection control.
Collapse
Affiliation(s)
- Javier Orozco-Cordoba
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Camila Mazas
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Gisela Du Pont
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Edmundo Lamoyi
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| | - Graciela Cárdenas
- Departamento de Neuroinfectología, Instituto Nacional de Neurología Manuel Velasco Suárez, Mexico City, Mexico
| | - Nora A Fierro
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad Universitaria, Mexico City, Mexico
| |
Collapse
|
15
|
Mansuy-Aubert V, Ravussin Y. Short chain fatty acids: the messengers from down below. Front Neurosci 2023; 17:1197759. [PMID: 37483350 PMCID: PMC10359501 DOI: 10.3389/fnins.2023.1197759] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 06/19/2023] [Indexed: 07/25/2023] Open
Abstract
Short-chain fatty acids (SCFAs), produced by the metabolism of dietary fibers in the gut, have wide-ranging effects locally and throughout the body. They modulate the enteric and central nervous systems, benefit anti-inflammatory pathways, and serve as energy sources. Recent research reveals SCFAs as crucial communicators between the gut and brain, forming the gut-brain axis. This perspective highlights key findings and discusses signaling mechanisms connecting SCFAs to the brain. By shedding light on this link, the perspective aims to inspire innovative research in this rapidly developing field.
Collapse
Affiliation(s)
- Virginie Mansuy-Aubert
- Department of Biomedical Sciences, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Yann Ravussin
- Laboratory of Energetics and Advanced Nutrition (LEAN), Department of Endocrinology, Metabolism and Cardiovascular Systems (EMC), Faculty of Science and Medicine, University of Fribourg (UNIFR), Fribourg, Switzerland
| |
Collapse
|
16
|
Calabrò S, Kankowski S, Cescon M, Gambarotta G, Raimondo S, Haastert-Talini K, Ronchi G. Impact of Gut Microbiota on the Peripheral Nervous System in Physiological, Regenerative and Pathological Conditions. Int J Mol Sci 2023; 24:ijms24098061. [PMID: 37175764 PMCID: PMC10179357 DOI: 10.3390/ijms24098061] [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: 04/03/2023] [Revised: 04/21/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
It has been widely demonstrated that the gut microbiota is responsible for essential functions in human health and that its perturbation is implicated in the development and progression of a growing list of diseases. The number of studies evaluating how the gut microbiota interacts with and influences other organs and systems in the body and vice versa is constantly increasing and several 'gut-organ axes' have already been defined. Recently, the view on the link between the gut microbiota (GM) and the peripheral nervous system (PNS) has become broader by exceeding the fact that the PNS can serve as a systemic carrier of GM-derived metabolites and products to other organs. The PNS as the communication network between the central nervous system and the periphery of the body and internal organs can rather be affected itself by GM perturbation. In this review, we summarize the current knowledge about the impact of gut microbiota on the PNS, with regard to its somatic and autonomic divisions, in physiological, regenerative and pathological conditions.
Collapse
Affiliation(s)
- Sonia Calabrò
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
- Department of Biology, University of Padova, Viale G. Colombo 3, 35131 Padova, Italy
| | - Svenja Kankowski
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
| | - Matilde Cescon
- Department of Molecular Medicine, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy
| | - Giovanna Gambarotta
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Stefania Raimondo
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| | - Kirsten Haastert-Talini
- Hannover Medical School, Institute of Neuroanatomy and Cell Biology, Carl-Neuberg-Str. 1, 30625 Hannover, Germany
- Center for Systems Neuroscience Hannover (ZSN), Buenteweg 2, 30559 Hannover, Germany
| | - Giulia Ronchi
- Department of Clinical and Biological Sciences & Neuroscience Institute Cavalieri Ottolenghi (NICO), University of Torino, Regione Gonzole 10, Orbassano, 10043 Torino, Italy
| |
Collapse
|
17
|
Li X, Xu S, Zhang Y, Li K, Gao XJ, Guo MY. Berberine Depresses Inflammation and Adjusts Smooth Muscle to Ameliorate Ulcerative Colitis of Cats by Regulating Gut Microbiota. Microbiol Spectr 2022; 10:e0320722. [PMID: 36287004 PMCID: PMC9769923 DOI: 10.1128/spectrum.03207-22] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Accepted: 09/27/2022] [Indexed: 01/10/2023] Open
Abstract
Intestinal microbiota dysbiosis is a well established characteristic of ulcerative colitis (UC). Regulating the gut microbiota is an effective UC treatment strategy. Berberine (BBR), an alkaloid extracted from several Chinese herbs, is a common traditional Chinese medicine. To establish the efficacy and mechanism of action of BBR, we constructed a UC model using healthy adult shorthair cats to conduct a systematic study of colonic tissue pathology, inflammatory factor expression, and gut microbiota structure. We investigated the therapeutic capacity of BBR for regulating the gut microbiota and thus work against UC in cats using 16S rRNA genes amplicon sequencing technology. Our results revealed that dextran sulfate sodium (DSS)-induced cat models of UC showed weight loss, diarrhea accompanied by mucous and blood, histological abnormalities, and shortening of the colon, all of which were significantly alleviated by supplementation with BBR. A 16S rRNA gene-based microbiota analysis demonstrated that BBR could significantly benefit gut microbiota. Western blot, quantitative PCR, and enzyme-linked immunosorbent assays (ELISAs) showed that in DSS-induced cat models, the expression of the inflammatory factors was increased, activating the JAK2/STAT3 signaling pathway, and treatment with BBR reversed this effect. The myosin light chain (MLC) phosphorylation in the smooth muscle of the intestines is associated with motility of inflammation-related diarrhea in cats. This study used gut flora analyses to demonstrate the anti-UC effects of BBR and its potential therapeutic mechanisms and offers novel insights into the prevention of inflammatory diseases using natural products. IMPORTANCE Ulcerative colitis (UC) is common in clinics. Intestinal microbiota disorder is correlated with ulcerative colitis. Although there are many studies on ulcerative colitis in rats, there are few studies on colitis in cats. Therefore, this study explored the possibility of the use of BBR as a safe and efficient treatment for colitis in cats. The results demonstrated the therapeutic effects of BBR on UC based on the state of the intestinal flora. The study found BBR supplementation to be effective against dextran sulfate sodium (DSS)-induced colitis, smooth muscle damage, and gut microbiota dysbiosis.
Collapse
Affiliation(s)
- Xueying Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Shuang Xu
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Yanhe Zhang
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Kan Li
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Xue-Jiao Gao
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| | - Meng-yao Guo
- Department of Clinical Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People’s Republic of China
| |
Collapse
|
18
|
Narabayashi H, Koma C, Nakata K, Ikegami M, Nakanishi Y, Ogihara J, Tsuda M, Hosono A, Hanazawa S, Takahashi K. Gut microbiota-dependent adaptor molecule recruits DNA methyltransferase to the TLR4 gene in colonic epithelial cells to suppress inflammatory reactions. Front Mol Biosci 2022; 9:1005136. [PMID: 36339704 PMCID: PMC9634067 DOI: 10.3389/fmolb.2022.1005136] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 09/30/2022] [Indexed: 11/13/2022] Open
Abstract
The intestine is inhabited by a large number of commensal bacteria that are immunologically non-self, potentially causing inflammation. However, in a healthy intestine, inflammation is strictly controlled at low levels to maintain homeostasis. We previously reported that the gut microbiota induce DNA methylation of the gene encoding Toll-like receptor (TLR) 4, a pattern recognition receptor that recognizes lipopolysaccharides of gram-negative bacteria, in colonic epithelial cells, suggesting its role in controlling intestinal inflammation. However, there remains a question of how gut microbiota cause methylation of only specific genes including TLR4, despite the fact that DNA methyltransferase (DNMT) is common to all genes targeted for methylation. Here, we identified RBM14 as an adaptor molecule that recruits DNMT to the TLR4 gene. RBM14 was shown to bind DNMT3 and be expressed at significantly higher levels in an intestinal epithelial cell (IEC) line with hypermethylated TLR4 gene than in an IEC line with hypomethylated TLR4 gene. In addition, RBM14 interacted with DNA regions of the TLR4 gene, and knockdown of RBM14 suppressed DNA methylation of the TLR4 gene in IECs. Furthermore, RBM14 expression was higher in colonic epithelial cells of conventional mice than in those of germ-free mice. Collectively, these results indicate that the gut microbiota induce methylation of the TLR4 gene in colonic epithelial cells by upregulating RBM14, which can recruit DNMT3 to the gene. The regulation of adaptor molecules such as RBM14, which bind to specific target genes and recruit DNMT, can explain, at least in part, how gut microbiota contribute to the maintenance of intestinal homeostasis through epigenetic control of specific gene expression in IECs.
Collapse
Affiliation(s)
- Hikari Narabayashi
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Chiharu Koma
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Kazuaki Nakata
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Mion Ikegami
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Yusuke Nakanishi
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Jun Ogihara
- Department of Chemistry and Life Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Masato Tsuda
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Akira Hosono
- Department of Food Bioscience and Biotechnology, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Shigemasa Hanazawa
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
| | - Kyoko Takahashi
- Department of Applied Biological Science, College of Bioresource Sciences, Nihon University, Fujisawa, Japan
- *Correspondence: Kyoko Takahashi,
| |
Collapse
|
19
|
Bai J, Cai Y, Huang Z, Gu Y, Huang N, Sun R, Zhang G, Liu R. Shouhui Tongbian Capsule ameliorates constipation via gut microbiota-5-HT-intestinal motility axis. Biomed Pharmacother 2022; 154:113627. [PMID: 36058152 DOI: 10.1016/j.biopha.2022.113627] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/28/2022] [Accepted: 08/29/2022] [Indexed: 12/12/2022] Open
Abstract
Constipation has become an epidemic enteric medical problem, accompanied with increasing long-term sequelae. Gut microbiota and serotonin (5-HT) have been believed as predominant player in the treatment of constipation. In clinical practices, Shouhui Tongbian Capsule (SHTB) was found to effectively improve constipation symptoms and promote gastrointestinal motility. However, the specific mechanism of SHTB is not clearly elucidated. Our current study aims to explore the therapeutic effects of SHTB against the development of constipation and the underlying mechanisms related to gut bacterial and 5-HT. We established loperamide hydrochloride (LH)-induced experimental constipation mouse model to evaluate the effect of SHTB. 16S RNA sequencing, fecal microbiota transplants (FMT), high performance liquid chromatograph, and molecular biological analysis were performed to investigate the potential mechanisms of SHTB. Our data demonstrated that SHTB significantly ameliorated LH-induced experimental constipation and accelerated enteric motility via promoting 5-HT biosynthesis in enterochromaffin cells and enteric neuron growth of the enteric nervous system (ENS) in both the small intestine and colon. Additionally, SHTB significantly modulated gut microbiota dysbiosis and potentially altered microbiota metabolites to enhance intestinal 5-HT production. Finally, FMT study confirmed that the effects of SHTB on 5-HT production and constipation are dependent on modulating intestinal microbiota dysbiosis. In conclusion, our current study deciphered therapeutic mechanism of SHTB in the treatment of experimental constipation from perspectives of gut microbiota-5-HT-intetinal motility axis and provides novel insights into the appropriate and safe application of SHTB in the clinic.
Collapse
Affiliation(s)
- Jinzhao Bai
- Beijing University of Chinese Medicine, School of Materia Medica, Beijing 100029, China
| | - Yajie Cai
- Beijing University of Chinese Medicine, School of Materia Medica, Beijing 100029, China
| | - Zhiyan Huang
- Lunan Hope Pharmaceutical Co., Ltd., Linyi 276006, China; Lunan Pharmaceutical Group Co., Ltd., State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi 276006, China
| | - Yiqing Gu
- Beijing University of Chinese Medicine, School of Materia Medica, Beijing 100029, China
| | - Nana Huang
- The Second Hospital of Shandong University, Ji'nan 250033, China
| | - Rong Sun
- The Second Hospital of Shandong University, Ji'nan 250033, China.
| | - Guimin Zhang
- Lunan Hope Pharmaceutical Co., Ltd., Linyi 276006, China; Lunan Pharmaceutical Group Co., Ltd., State Key Laboratory of Generic Manufacture Technology of Chinese Traditional Medicine, Linyi 276006, China.
| | - Runping Liu
- Beijing University of Chinese Medicine, School of Materia Medica, Beijing 100029, China.
| |
Collapse
|
20
|
Mothersill C, Cocchetto A, Seymour C. Low Dose and Non-Targeted Radiation Effects in Environmental Protection and Medicine-A New Model Focusing on Electromagnetic Signaling. Int J Mol Sci 2022; 23:11118. [PMID: 36232421 PMCID: PMC9570230 DOI: 10.3390/ijms231911118] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 09/16/2022] [Accepted: 09/17/2022] [Indexed: 11/17/2022] Open
Abstract
The role of signalling in initiating and perpetuating effects triggered by deposition of ionising radiation energy in parts of a system is very clear. Less clear are the very early steps involved in converting energy to chemical and biological effects in non-targeted parts of the system. The paper aims to present a new model, which could aid our understanding of the role of low dose effects in determining ultimate disease outcomes. We propose a key role for electromagnetic signals resulting from physico-chemical processes such as excitation decay, and acoustic waves. These lead to the initiation of damage response pathways such as elevation of reactive oxygen species and membrane associated changes in key ion channels. Critically, these signalling pathways allow coordination of responses across system levels. For example, depending on how these perturbations are transduced, adverse or beneficial outcomes may predominate. We suggest that by appreciating the importance of signalling and communication between multiple levels of organisation, a unified theory could emerge. This would allow the development of models incorporating time, space and system level to position data in appropriate areas of a multidimensional domain. We propose the use of the term "infosome" to capture the nature of radiation-induced communication systems which include physical as well as chemical signals. We have named our model "the variable response model" or "VRM" which allows for multiple outcomes following exposure to low doses or to signals from low dose irradiated cells, tissues or organisms. We suggest that the use of both dose and infosome in radiation protection might open up new conceptual avenues that could allow intrinsic uncertainty to be embraced within a holistic protection framework.
Collapse
Affiliation(s)
- Carmel Mothersill
- Department of Biology, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Alan Cocchetto
- National CFIDS Foundation, 285 Beach Ave., Hull, MA 02045-1602, USA
| | - Colin Seymour
- Department of Biology, McMaster University, Hamilton, ON L8S 4K1, Canada
| |
Collapse
|
21
|
Exposure to Veterinary Antibiotics via Food Chain Disrupts Gut Microbiota and Drives Increased Escherichia coli Virulence and Drug Resistance in Young Adults. Pathogens 2022; 11:pathogens11091062. [PMID: 36145494 PMCID: PMC9500718 DOI: 10.3390/pathogens11091062] [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: 07/01/2022] [Revised: 08/15/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Exposure to veterinary antibiotics (VAs) and preferred as veterinary antibiotics (PVAs) via the food chain is unavoidable for their extensive use not only for treating bacterial infections, but also for use as growth promoters in livestock and aquaculture. One of the consequences is the disturbance of gut microbiota. However, its impact on the virulence and drug resistance of opportunistic pathogens is still unclear. In this study, a total of 26 antibiotics were detected in the urine of 300 young undergraduates in Anhui Province. We found that excessive intake of milk was positively correlated to high levels of VAs and PVAs. It led to the dysbiosis of gut microbiota characterized by high abundance of Bacteroidetes and Proteobacteria. The increase in Proteobacteria was mainly due to a single operational taxonomic unit (OTU) of Escherichia coli (E. coli). We isolated several E. coli strains from participants and compared their drug resistance and virulence using PCR assay and virulence-related assays. We observed that exposure to high levels of VAs and PVAs induced more resistant genes and drove E. coli strain to become more virulent. At last, we conducted transcriptome analysis to investigate the molecular mechanism of virulent and drug-resistant regulators in the highly virulent E. coli strain. We noted that there were multiple pathways involved in the drug resistance and virulence of the highly virulent strain. Our results demonstrated that participants with high-level VAs and PVAs exposure have a disrupted gut microbiota following the appearance of highly drug-resistant and virulent E. coli and, therefore may be at elevated risk for long-term health complications.
Collapse
|