1
|
Gaougaou G, Zahra R, Morel S, Bélanger V, Knoth IS, Cousineau D, D'Arc BF, Grzywacz K, Rousseau G, Déziel E, Godbout R, Lippé S, Millette M, Marcil V. Acceptability and safety of a probiotic beverage supplementation (Bio-K +) and feasibility of the proposed protocol in children with a diagnosis of autism spectrum disorder. J Neurodev Disord 2025; 17:30. [PMID: 40413384 PMCID: PMC12102953 DOI: 10.1186/s11689-025-09617-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 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: 01/24/2025] [Accepted: 05/06/2025] [Indexed: 05/27/2025] Open
Abstract
BACKGROUND Autism spectrum disorder (ASD) is a group of neurodevelopmental disorders defined by stereotyped behavior and challenges in social communication and social interaction. ASD is associated with various comorbidities, including anxiety, gastrointestinal (GI) symptoms and sleep disorders. Evidence supports an association between intestinal dysbiosis and the severity of ASD-related symptoms. Probiotic intake was suggested to restore microbial homeostasis and decrease neurobehavioral, GI and sleep symptoms in individuals diagnosed with autism. METHODS This study aims to evaluate the acceptability and safety of a Bio-K + probiotics beverage in autistic children aged 4 to 11 years and the feasibility of the proposed research protocol to measure its impact on behaviors and comorbidities. The 30-week study consisted of daily supplementation with Bio-K + probiotics for 14 weeks. Acceptability and safety were monitored throughout the study. Feasibility was assessed by comparing recruitment and completion rates to pre-established thresholds. Preliminary impact of supplementation on behaviors (Autism Treatment Evaluation Checklist (ATEC) score), GI symptoms and sleep disorders was evaluated. RESULTS Of the 23 children recruited (mean age 6.7 ± 2.2 years, 70% males), 65% had GI problems and 91% had sleep disorders. Probiotic supplementation was accepted by all participants and no product-related adverse event was reported. Feasibility rates exceeded pre-established thresholds for almost all study outcomes including recruitment rate, compliance, electroencephalography, actigraphy and completion of questionnaires. Preliminary data suggest an improvement in behaviors associated with autism assessed with the total ATEC score, and in GI symptoms and sleep disorders. CONCLUSION This study demonstrates probiotic beverage acceptability and safety and protocol feasibility in autistic children. To further support our data, a double-blinded placebo-controlled study is needed to determine its efficacy.
Collapse
Affiliation(s)
- Ghizlane Gaougaou
- Department of Nutrition, Université de Montréal, Montreal, QC, H3 T 1 A8, Canada
- Centre de recherche Azrieli du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montreal, QC, H3 T 1 C5, Canada
| | - Riham Zahra
- Bio-K Plus International Inc., Kerry (Canada) Inc., 495 Armand-Frappier Boulevard, Laval, QC, H7 V 4B3, Canada
| | - Sophia Morel
- Department of Nutrition, Université de Montréal, Montreal, QC, H3 T 1 A8, Canada
- Centre de recherche Azrieli du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montreal, QC, H3 T 1 C5, Canada
| | - Véronique Bélanger
- Department of Nutrition, Université de Montréal, Montreal, QC, H3 T 1 A8, Canada
- Centre de recherche Azrieli du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montreal, QC, H3 T 1 C5, Canada
| | - Inga Sophia Knoth
- Department of Psychology, Université de Montréal, Montreal, QC, H3 T 1 A8, Canada
- Centre de recherche Azrieli du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montreal, QC, H3 T 1 C5, Canada
| | - Dominique Cousineau
- Department of Pediatrics, Université de Montréal, Montreal, QC, H3 T 1 C5, Canada
- Centre de recherche Azrieli du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montreal, QC, H3 T 1 C5, Canada
| | - Baudouin Forgeot D'Arc
- Department of Psychiatry, Université de Montréal, Montréal, QC, H3 C 3 J7, Canada
- Centre de recherche Azrieli du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montreal, QC, H3 T 1 C5, Canada
| | - Kelly Grzywacz
- Division of Gastroenterology, Hepatology and Nutrition, Department of Pediatrics, CHU Sainte-Justine, Montreal, QC, H3 T 1 C5, Canada
| | - Guy Rousseau
- Research Center, Centre Intégré Universitaire du Nord-de-L'Île-de-Montréal, Montreal, QC, H1E 1 A4, Canada
- Department of Pharmacology and Physiology, Université de Montréal, Montreal, QC, H3 C 3 J7, Canada
| | - Eric Déziel
- Centre Armand-Frappier Santé Biotechnologie, Institut National de La Recherche Scientifique (INRS), Laval, QC, H7 V 1B7, Canada
| | - Roger Godbout
- Department of Psychiatry, Université de Montréal, Montréal, QC, H3 C 3 J7, Canada
- Research Center, Centre Intégré Universitaire du Nord-de-L'Île-de-Montréal, Montreal, QC, H1E 1 A4, Canada
| | - Sarah Lippé
- Department of Psychology, Université de Montréal, Montreal, QC, H3 T 1 A8, Canada
- Centre de recherche Azrieli du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montreal, QC, H3 T 1 C5, Canada
| | - Mathieu Millette
- Bio-K Plus International Inc., Kerry (Canada) Inc., 495 Armand-Frappier Boulevard, Laval, QC, H7 V 4B3, Canada
| | - Valérie Marcil
- Department of Nutrition, Université de Montréal, Montreal, QC, H3 T 1 A8, Canada.
- Centre de recherche Azrieli du CHU Sainte-Justine, 3175 Côte Sainte-Catherine, Montreal, QC, H3 T 1 C5, Canada.
| |
Collapse
|
2
|
Wang Z, Mou R, Jin S, Wang Q, Ju Y, Sun P, Xie R, Wang K. Streptococcus anginosus promotes gastric cancer progression via GSDME-mediated pyroptosis pathway: Molecular mechanisms of action of GSDME, cleaved caspase-3, and NLRP3 proteins. Int J Biol Macromol 2025; 307:142341. [PMID: 40118413 DOI: 10.1016/j.ijbiomac.2025.142341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/13/2025] [Accepted: 03/18/2025] [Indexed: 03/23/2025]
Abstract
Streptococcus vasculosus is a common oral and intestinal symbiotic bacteria, but it can transform into a pathogen under certain conditions, affecting the host's immune response. Studies have shown that Streptococcus vasculosus may promote tumor growth and metastasis by activating host inflammatory responses. This study simulated the environment of Streptococcus vascularis infection through in vitro cell culture experiment, and observed the influence of streptococcus vascularis at different time points and different concentrations on cancer cells. The expression and activity of GSDME, cleaved caspase-3 and NLRP3 proteins were detected by Western blot, immunofluorescence and flow cytometry. By constructing gene knockout and overexpression cell models, the role of these protein molecules in promoting cancer progression of Streptococcus vascularis was further verified. It was found that GSDME activation is a key step in Pyroptosis occurrence, and cleaved caspase-3 plays an important role in GSDME cleavage activation. The activation of NLRP3 inflammatome is closely related to the inflammatory response induced by Streptococcus vasculosus, and thus affects the tumor microenvironment.
Collapse
Affiliation(s)
- Zeshen Wang
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150076, Heilongjiang, China
| | - Ruishu Mou
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150076, Heilongjiang, China
| | - Shiyang Jin
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150076, Heilongjiang, China
| | - Qiancheng Wang
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150076, Heilongjiang, China
| | - Yuming Ju
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150076, Heilongjiang, China
| | - Pengcheng Sun
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150076, Heilongjiang, China
| | - Rui Xie
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150076, Heilongjiang, China.
| | - Kuan Wang
- Department of Gastrointestinal Surgery, Harbin Medical University Cancer Hospital, Harbin 150076, Heilongjiang, China.
| |
Collapse
|
3
|
Drapkina OM, Bakirov BA, Bakirova AE, Klyaritskaya IL, Maksimova EV, Sklyannaya EV, Vatutin NT, Garbuzova EV, Livzan MA, Ershova AI. The Department of Medical Sciences of the RAS, Therapeutic Sciences Council meeting: Scientific projects marathon of Russian therapeutic departments "From innovative technologies and education to healthcare practice. Gut microbiota: clinical aspects". КАРДИОВАСКУЛЯРНАЯ ТЕРАПИЯ И ПРОФИЛАКТИКА 2025; 23:4234. [DOI: 10.15829/1728-8800-2024-4234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/04/2025] Open
Affiliation(s)
- O. M. Drapkina
- National Medical Research Center for Therapy and Preventive Medicine
| | | | | | - I. L. Klyaritskaya
- Vernadsky Crimean Federal University, S.I. Georgievsky Medical institute
| | - E. V. Maksimova
- Vernadsky Crimean Federal University, S.I. Georgievsky Medical institute
| | - E. V. Sklyannaya
- M. Gorky Donetsk State Medical University; V.K. Gusak Institute of Emergency and Reconstructive Surgery
| | - N. T. Vatutin
- M. Gorky Donetsk State Medical University; V.K. Gusak Institute of Emergency and Reconstructive Surgery
| | - E. V. Garbuzova
- National Medical Research Center for Therapy and Preventive Medicine
| | | | - A. I. Ershova
- National Medical Research Center for Therapy and Preventive Medicine
| |
Collapse
|
4
|
Joubran P, Roux FA, Serino M, Deschamps JY. Gut Microbiota Comparison in Rectal Swabs Versus Stool Samples in Cats with Kidney Stones. Microorganisms 2024; 12:2411. [PMID: 39770613 PMCID: PMC11677927 DOI: 10.3390/microorganisms12122411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 11/17/2024] [Accepted: 11/21/2024] [Indexed: 01/11/2025] Open
Abstract
To investigate the role of the intestinal bacterial microbiota in the pathogenesis of calcium oxalate nephrolithiasis in cats, a condition characterized by the formation of kidney stones, it is desirable to identify a sample collection method that accurately reflects the microbiota's composition. The objective of this study was to evaluate the impact of fecal sample collection methods on the intestinal microbiota composition in two cat populations: healthy cats and kidney stone-diseased cats. The study included eighteen cats from the same colony, comprising nine healthy cats and nine cats with spontaneously occurring presumed calcium oxalate kidney stones. Three fecal collection methods were compared: rectal swabs, the collection of fresh stool, and the collection of stool exposed to ambient air for 24 h. The bacterial microbiota was analyzed through the high-resolution sequencing of the V3-V4 region of the 16S rRNA gene. For all cats, within the same individual, a one-way PERMANOVA analysis showed a significant difference between the rectal swabs and fresh stool (p = 0.0003), as well as between the rectal swabs and stool exposed to ambient air for 24 h (p = 0.0003), but no significant difference was identified between the fresh stool and non-fresh stool (p = 0.0651). When comparing the two populations of cats, this study provides seemingly conflicting results. (1) A principal component analysis (PCA) comparison revealed a significant difference in the bacterial composition between the healthy cats and the cats with kidney stones only when the sample was a fresh fecal sample (p = 0.0037). This finding suggests that the intestinal bacteria involved in the pathogenesis of kidney stones in cats are luminal and strictly anaerobic bacteria. Consequently, exposure to ambient air results in a loss of information, preventing the identification of dysbiosis. For clinical studies, non-fresh stool samples provided by owners does not appear suitable for studying the gut microbiota of cats with kidney stones; fresh stool should be favored. (2) Interestingly, the rectal swabs alone highlighted significant differences in the proportion of major phyla between the two populations. These findings highlight the critical importance of carefully selecting fecal collection methods when studying feline gut microbiota. Combining rectal swabs and fresh stool sampling provides complementary insights, offering the most accurate understanding of the gut microbiota composition in the context of feline kidney stone pathogenesis.
Collapse
Affiliation(s)
- Patrick Joubran
- NP3, Nutrition, PathoPhysiology and Pharmacology Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44307 Nantes, France; (P.J.); (F.A.R.)
| | - Françoise A. Roux
- NP3, Nutrition, PathoPhysiology and Pharmacology Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44307 Nantes, France; (P.J.); (F.A.R.)
- Emergency and Critical Care Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44307 Nantes, France
| | - Matteo Serino
- IRSD, Institut de Recherche en Santé Digestive, Institut National de la Santé et de la Recherche Médicale (INSERM) U1220, Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Ecole Nationale Vétérinaire de Toulouse (ENVT), Université de Toulouse III-Paul Sabatier (UPS), CS 60039, 31024 Toulouse, France;
| | - Jack-Yves Deschamps
- NP3, Nutrition, PathoPhysiology and Pharmacology Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44307 Nantes, France; (P.J.); (F.A.R.)
- Emergency and Critical Care Unit, Oniris VetAgro Bio, Nantes-Atlantic College of Veterinary Medicine, Food Science and Engineering, La Chantrerie, CEDEX 03, 44307 Nantes, France
| |
Collapse
|
5
|
Li S, Li Y, Cai Y, Yan Z, Wei J, Zhang H, Yue F, Chen T. Lacticaseibacillus paracasei NCU-04 relieves constipation and the depressive-like behaviors induced by loperamide in mice through the microbiome-gut-brain axis. Curr Res Food Sci 2024; 9:100875. [PMID: 39429918 PMCID: PMC11490870 DOI: 10.1016/j.crfs.2024.100875] [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: 08/21/2024] [Revised: 09/23/2024] [Accepted: 10/01/2024] [Indexed: 10/22/2024] Open
Abstract
Constipation is a prevalent gastrointestinal condition that significantly affects patients' physical and mental well-being, yet current treatments often lack safety and efficacy. Emerging evidence highlights the critical role of the microbiota-gut-brain axis (MBGA) in managing constipation, paving the way for probiotics as an adjuvant treatment to improve constipation symptoms. In this study, we isolated a gut probiotic strain, Lacticaseibacillus paracasei NCU-04, and investigated its improvement effects on loperamide-induced constipation in mice. We demonstrated that L. paracasei NCU-04 exhibited excellent probiotic properties, including robust growth, strong antibacterial and antioxidant capacities, and a lack of hemolytic activity in vitro. The administration of L. paracasei NCU-04 effectively improved the defecation-related indicators such as the fecal water content, time to the first black stool defecation, and intestine transit rate, suggesting enhanced gut immobility in constipated mice. Additionally, L. paracasei NCU-04 significantly reduced colon inflammation induced by loperamide. Further, L. paracasei NCU-04 increased levels of colonic motilin, 5-hydroxytryptamine (5-HT), and c-kit, while decreased that of aquaporin 3, vasoactive intestinal peptide, and peptide YY. Notably, L. paracasei NCU-04 effectively upregulated the expression of 5-HT and its receptor (i.e., 5-HT4R) in the brains of constipated mice. High-throughput sequencing revealed that L. paracasei NCU-04 restored the diversity and composition of the gut microbiota disturbed by loperamide, and significantly increased the relative abundance of Prevotella and Lactobacillus genera in the stool, while decreased that of Odoribacter, Rikenella, and Parabacteroides. Importantly, L. paracasei NCU-04 also effectively improved the depression-like behaviors associated with constipation, possibly through 5-HT mediated MGBA. These results suggest that L. paracasei NCU-04 may offer a promising approach for treating constipation and its related depressive symptoms, supporting its potential as a functional food or adjuvant therapy for human health.
Collapse
Affiliation(s)
- Shengjie Li
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Province Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Yi Li
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Province Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Yujie Cai
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Province Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Zizhou Yan
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Province Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Jing Wei
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Province Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Hongyan Zhang
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
| | - Fenfang Yue
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Province Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| | - Tingtao Chen
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China
- National Engineering Research Center for Bioengineering Drugs and the Technologies, Jiangxi Province Key Laboratory of Bioengineering Drugs, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
- Jiangxi Province Key Laboratory of Bioengineering Drugs, School of Pharmacy, Jiangxi Medical College, Nanchang University, Nanchang, 330031, China
| |
Collapse
|
6
|
Rahmannia M, Poudineh M, Mirzaei R, Aalipour MA, Shahidi Bonjar AH, Goudarzi M, Kheradmand A, Aslani HR, Sadeghian M, Nasiri MJ, Sechi LA. Strain-specific effects of probiotics on depression and anxiety: a meta-analysis. Gut Pathog 2024; 16:46. [PMID: 39245752 PMCID: PMC11382490 DOI: 10.1186/s13099-024-00634-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/17/2024] [Indexed: 09/10/2024] Open
Abstract
INTRODUCTION Depression and anxiety are pervasive mental health disorders with substantial global burdens. Probiotics, live microorganisms known for their health benefits, have emerged as a potential therapeutic intervention for these conditions. This systematic review and meta-analysis aim to evaluate the strain-specific effects of probiotics on relieving depressive and anxiety symptoms while elucidating underlying mechanisms. METHODS EMBASE, Cochrane CENTRAL and PubMed/Medline were systematically queried to identify studies released until May 15, 2024. Randomized Controlled Trials (RCTs) that employed standardized assessment tools for depression and anxiety namely Beck Depression Inventory (BDI), Hamilton Depression Rating Scale (HAMD), Depression Anxiety Stress Scales (DASS), or Montgomery-Asberg Depression Rating Scale (MADRS) were included. RESULTS 12 RCTs involving 707 participants were included. Seven RCTs utilizing the BDI questionnaire demonstrated a significant decrease in depressive symptoms favoring probiotics containing strains such as Lactobacillus acidophilus, Lactobacillus paracasei, Lactobacillus casei, Lactobacillus plantarum, Lactobacillus salivarius, Bifidobacterium bifidum, Bifidobacterium lactis, Bifidobacterium breve, and Bifidobacterium longum (MD: -2.69, CI95%: -4.22/-1.16, p value: 0.00). Conversely, RCTs using HAMD showed a non-significant reduction in depressive symptoms (MD: -1.40, CI95%: -3.29/0.48, p value: 0.14). RCTs employing DASS and MADRS scales also showed no significant differences. CONCLUSION This meta-analysis offers valuable insights into the strain-specific effects of probiotics containing Lactobacillus and Bifidobacterium species on depressive and anxiety symptoms. While our findings suggest a significant reduction in depressive symptoms based on the BDI scale favoring probiotics, the lack of significant effects observed on the HAMD, DASS, and MADRS scales underscores the complexity inherent in these conditions. It is imperative to acknowledge the mixed results across different measurement scales, indicating the need for cautious interpretation. Therefore, we advocate for a nuanced understanding of probiotics' impacts on various dimensions of mood, emphasizing the necessity for further research.
Collapse
Affiliation(s)
- Maryam Rahmannia
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohadeseh Poudineh
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Roya Mirzaei
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Amin Aalipour
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Hashem Shahidi Bonjar
- Scientist of Dental Materials and Restorative Dentistry, School of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Goudarzi
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Kheradmand
- Department of Psychiatry, Taleghani Hospital Clinical Research Development Unit, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Reza Aslani
- Department of Clinical Pharmacy, School of Pharmacy, Iran University of Medical Sciences, Tehran, Iran
| | - Majid Sadeghian
- Department of Psychiatry, Taleghani Hospital Clinical Research Development Unit, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Javad Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | | |
Collapse
|
7
|
Li S, Zhao L, Xiao J, Guo Y, Fu R, Zhang Y, Xu S. The gut microbiome: an important role in neurodegenerative diseases and their therapeutic advances. Mol Cell Biochem 2024; 479:2217-2243. [PMID: 37787835 DOI: 10.1007/s11010-023-04853-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/06/2023] [Indexed: 10/04/2023]
Abstract
There are complex interactions between the gut and the brain. With increasing research on the relationship between gut microbiota and brain function, accumulated clinical and preclinical evidence suggests that gut microbiota is intimately involved in the pathogenesis of neurodegenerative diseases (NDs). Increasingly studies are beginning to focus on the association between gut microbiota and central nervous system (CNS) degenerative pathologies to find potential therapies for these refractory diseases. In this review, we summarize the changes in the gut microbiota in Alzheimer's disease, Parkinson's disease, multiple sclerosis, and amyotrophic lateral sclerosis and contribute to our understanding of the function of the gut microbiota in NDs and its possible involvement in the pathogenesis. We subsequently discuss therapeutic approaches targeting gut microbial abnormalities in these diseases, including antibiotics, diet, probiotics, and fecal microbiota transplantation (FMT). Furthermore, we summarize some completed and ongoing clinical trials of interventions with gut microbes for NDs, which may provide new ideas for studying NDs.
Collapse
Affiliation(s)
- Songlin Li
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Linna Zhao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Jie Xiao
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yuying Guo
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China
| | - Rong Fu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yunsha Zhang
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Shixin Xu
- Medical Experiment Center, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China.
- National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China.
- Tianjin Key Laboratory of Translational Research of TCM Prescription and Syndrome, Tianjin, China.
| |
Collapse
|
8
|
Di Chiano M, Sallustio F, Fiocco D, Rocchetti MT, Spano G, Pontrelli P, Moschetta A, Gesualdo L, Gadaleta RM, Gallone A. Psychobiotic Properties of Lactiplantibacillus plantarum in Neurodegenerative Diseases. Int J Mol Sci 2024; 25:9489. [PMID: 39273435 PMCID: PMC11394828 DOI: 10.3390/ijms25179489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Neurodegenerative disorders are the main cause of cognitive and physical disabilities, affect millions of people worldwide, and their incidence is on the rise. Emerging evidence pinpoints a disturbance of the communication of the gut-brain axis, and in particular to gut microbial dysbiosis, as one of the contributors to the pathogenesis of these diseases. In fact, dysbiosis has been associated with neuro-inflammatory processes, hyperactivation of the neuronal immune system, impaired cognitive functions, aging, depression, sleeping disorders, and anxiety. With the rapid advance in metagenomics, metabolomics, and big data analysis, together with a multidisciplinary approach, a new horizon has just emerged in the fields of translational neurodegenerative disease. In fact, recent studies focusing on taxonomic profiling and leaky gut in the pathogenesis of neurodegenerative disorders are not only shedding light on an overlooked field but are also creating opportunities for biomarker discovery and development of new therapeutic and adjuvant strategies to treat these disorders. Lactiplantibacillus plantarum (LBP) strains are emerging as promising psychobiotics for the treatment of these diseases. In fact, LBP strains are able to promote eubiosis, increase the enrichment of bacteria producing beneficial metabolites such as short-chain fatty acids, boost the production of neurotransmitters, and support the homeostasis of the gut-brain axis. In this review, we summarize the current knowledge on the role of the gut microbiota in the pathogenesis of neurodegenerative disorders with a particular focus on the benefits of LBP strains in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, autism, anxiety, and depression.
Collapse
Affiliation(s)
- Mariagiovanna Di Chiano
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Fabio Sallustio
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Daniela Fiocco
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Maria Teresa Rocchetti
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy
| | - Giuseppe Spano
- Department of Agriculture Food Natural Science Engineering (DAFNE), University of Foggia, 71122 Foggia, Italy
| | - Paola Pontrelli
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Antonio Moschetta
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Piazza Giulio Cesare n. 11, 70124 Bari, Italy
- National Institute for Biostructure and Biosystems (INBB), Viale delle Medaglie d'Oro n. 305, 00136 Roma, Italy
| | - Loreto Gesualdo
- Department of Precision and Regenerative Medicine and Ionian Area, University of Bari Aldo Moro, Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| | - Raffaella Maria Gadaleta
- Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Piazza Giulio Cesare n. 11, 70124 Bari, Italy
- National Institute for Biostructure and Biosystems (INBB), Viale delle Medaglie d'Oro n. 305, 00136 Roma, Italy
| | - Anna Gallone
- Department of Translational Biomedicine and Neuroscience, University of Bari Aldo Moro, Piazza Giulio Cesare n. 11, 70124 Bari, Italy
| |
Collapse
|
9
|
Aslam R, Herrles L, Aoun R, Pioskowik A, Pietrzyk A. Link between gut microbiota dysbiosis and childhood asthma: Insights from a systematic review. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. GLOBAL 2024; 3:100289. [PMID: 39105129 PMCID: PMC11298874 DOI: 10.1016/j.jacig.2024.100289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2023] [Revised: 03/13/2024] [Accepted: 03/19/2024] [Indexed: 08/07/2024]
Abstract
Asthma, a chronic inflammatory disorder of the airways, is a prevalent childhood chronic disease with a substantial global health burden. The complex etiology and pathogenesis of asthma involve genetic and environmental factors, posing challenges in diagnosis, severity prediction, and therapeutic strategies. Recent studies have highlighted the significant role of the gut microbiota and its interaction with the immune system in the development of asthma. Dysbiosis, an imbalance in microbial composition, has been associated with respiratory diseases through the gut-lung axis. This axis is an interaction between the gut and lungs, allowing microbial metabolites to influence the host immune system. This systematic review examines the association between gut microbiota composition, measured using 16S rRNA sequencing, during infancy and childhood, and the subsequent development of atopic wheeze and asthma. The results suggest that higher alpha diversity of bacteria such as Bifidobacterium, Faecalibacterium, and Roseburia may have protective effects against asthmatic outcomes. Conversely, lower relative abundances of bacteria like Bacteroides and certain fungi, including Malassezia, were associated with asthma. These findings highlight the potential of early screening and risk assessment of gut microbiota to identify individuals at risk of asthma. Furthermore, investigations targeting gut microbiota, such as dietary modifications and probiotic supplementation, may hold promise for asthma prevention and management. Future research should focus on identifying specific microbial signatures associated with asthma susceptibility and further investigate approaches like fecal microbiota transplantation. Understanding the role of gut microbiota in asthma pathogenesis can contribute to early detection and development of interventions to mitigate the risk of asthmatic pathogenesis in childhood.
Collapse
Affiliation(s)
- Rabbiya Aslam
- Scientific Group of Microbiology and Parasitology and the Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Laura Herrles
- Scientific Group of Microbiology and Parasitology and the Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Raquel Aoun
- Scientific Group of Microbiology and Parasitology and the Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Anna Pioskowik
- Scientific Group of Microbiology and Parasitology and the Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| | - Agata Pietrzyk
- Scientific Group of Microbiology and Parasitology and the Department of Microbiology, Faculty of Medicine, Jagiellonian University Medical College, Kraków, Poland
| |
Collapse
|
10
|
Pires L, González-Paramás AM, Heleno SA, Calhelha RC. Exploring Therapeutic Advances: A Comprehensive Review of Intestinal Microbiota Modulators. Antibiotics (Basel) 2024; 13:720. [PMID: 39200020 PMCID: PMC11350912 DOI: 10.3390/antibiotics13080720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 07/18/2024] [Accepted: 07/29/2024] [Indexed: 09/01/2024] Open
Abstract
The gut microbiota establishes a mutually beneficial relationship with the host starting from birth, impacting diverse metabolic and immunological processes. Dysbiosis, characterized by an imbalance of microorganisms, is linked to numerous medical conditions, including gastrointestinal disorders, cardiovascular diseases, and autoimmune disorders. This imbalance promotes the proliferation of toxin-producing bacteria, disrupts the host's equilibrium, and initiates inflammation. Genetic factors, dietary choices, and drug use can modify the gut microbiota. However, there is optimism. Several therapeutic approaches, such as probiotics, prebiotics, synbiotics, postbiotics, microbe-derived products, and microbial substrates, aim to alter the microbiome. This review thoroughly explores the therapeutic potential of these microbiota modulators, analysing recent studies to evaluate their efficacy and limitations. It underscores the promise of microbiota-based therapies for treating dysbiosis-related conditions. This article aims to ensure practitioners feel well-informed and up to date on the most influential methods in this evolving field by providing a comprehensive review of current research.
Collapse
Affiliation(s)
- Lara Pires
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (L.P.); (R.C.C.)
- Laboratório Associado para Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
- Grupo de Investigación en Polifenoles, Área de Nutrición y Bromatología, Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain;
| | - Ana M. González-Paramás
- Grupo de Investigación en Polifenoles, Área de Nutrición y Bromatología, Facultad de Farmacia, Universidad de Salamanca, Campus Miguel de Unamuno s/n, 37007 Salamanca, Spain;
| | - Sandrina A. Heleno
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (L.P.); (R.C.C.)
- Laboratório Associado para Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| | - Ricardo C. Calhelha
- Centro de Investigação de Montanha (CIMO), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal; (L.P.); (R.C.C.)
- Laboratório Associado para Sustentabilidade e Tecnologia em Regiões de Montanha (SusTEC), Instituto Politécnico de Bragança, Campus de Santa Apolónia, 5300-253 Bragança, Portugal
| |
Collapse
|
11
|
Wolska M, Wypych TP, Rodríguez-Viso P. The Influence of Premature Birth on the Development of Pulmonary Diseases: Focus on the Microbiome. Metabolites 2024; 14:382. [PMID: 39057705 PMCID: PMC11279213 DOI: 10.3390/metabo14070382] [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: 06/06/2024] [Revised: 06/28/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Globally, around 11% of neonates are born prematurely, comprising a highly vulnerable population with a myriad of health problems. Premature births are often accompanied by an underdeveloped immune system biased towards a Th2 phenotype and microbiota dysbiosis. Typically, a healthy gut microbiota interacts with the host, driving the proper maturation of the host immunity. However, factors like cesarean section, formula milk feeding, hospitalization in neonatal intensive care units (NICU), and routine antibiotic treatments compromise microbial colonization and increase the risk of developing related diseases. This, along with alterations in the innate immune system, could predispose the neonates to the development of respiratory diseases later in life. Currently, therapeutic strategies are mainly focused on restoring gut microbiota composition using probiotics and prebiotics. Understanding the interactions between the gut microbiota and the immature immune system in premature neonates could help to develop novel therapeutic strategies for treating or preventing gut-lung axis disorders.
Collapse
Affiliation(s)
| | - Tomasz Piotr Wypych
- Laboratory of Host-Microbiota Interactions, Nencki Institute of Experimental Biology, Polish Academy of Sciences, Ludwika Pasteura 3, 02-093 Warsaw, Poland; (M.W.); (P.R.-V.)
| | | |
Collapse
|
12
|
Bibbò S, Porcari S, Del Vecchio LE, Severino A, Mullish BH, Ianiro G, Gasbarrini A, Cammarota G. Gut microbiota and immunotherapy of renal cell carcinoma. Hum Vaccin Immunother 2023; 19:2268982. [PMID: 37955340 PMCID: PMC10653624 DOI: 10.1080/21645515.2023.2268982] [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: 01/25/2023] [Accepted: 10/06/2023] [Indexed: 11/14/2023] Open
Abstract
The gut microbiome has recently been proposed as a key player in cancer development and progression. Several studies have reported that the composition of the gut microbiome plays a role in the response to immune checkpoint inhibitors (ICIs). The gut microbiome modulation has been investigated as a potential therapeutic strategy for cancer, mainly in patients undergoing therapy with ICIs. In particular, modulation through probiotics, FMT or other microbiome-related approaches have proven effective to improve the response to ICIs. In this review, we examine the role of the gut microbiome in enhancing clinical responses to ICIs in the treatment of renal cancer.
Collapse
Affiliation(s)
- Stefano Bibbò
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Serena Porcari
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Livio Enrico Del Vecchio
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Andrea Severino
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Benjamin H. Mullish
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, St Mary’s Hospital Campus, Imperial College London, London, UK
- Departments of Gastroenterology and Hepatology, St Mary’s Hospital, Imperial College Healthcare NHS Trust, London, UK
| | - Gianluca Ianiro
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Antonio Gasbarrini
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Giovanni Cammarota
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
13
|
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
|
14
|
Chen X, Chi J, Liu Y, Du R, Guo M, Xu W. Synthetic symbiotic bacteria reduces the toxicity of mercury ingested via contaminated food. Food Chem Toxicol 2023:113937. [PMID: 37433354 DOI: 10.1016/j.fct.2023.113937] [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: 05/04/2023] [Revised: 06/22/2023] [Accepted: 07/07/2023] [Indexed: 07/13/2023]
Abstract
Mercury contamination in food poses a significant threat to human health. In this article, we propose a novel approach to solve this problem by enhancing the function of gut microbiota against mercury using a synthetically engineered bacterial strain. An engineered Escherichia coli biosensor MerR with mercury binding function was introduced into the intestines of mice for colonization, whereafter the mice were challenged with oral mercury. Compared with the control mice and mice colonized with unengineered Escherichia coli, the mice with biosensor MerR cells in their gut showed significantly stronger mercury resistance. Furthermore, mercury distribution analysis revealed that biosensor MerR cells promoted the excretion of oral mercury with feces, thereby blocking the entry of mercury into the mice, decreasing the concentration of mercury in the circulatory system and organs, and, thus, attenuating the toxicity of mercury to the liver, kidneys and intestines. Colonization with the biosensor MerR did not result in significant health problems in the mice, nor were genetic circuit mutations or lateral transfers identified during the experiments, thus demonstrating the safety of this approach. This study elucidates the remarkable promise of synthetic biology for modulating gut microbiota function.
Collapse
Affiliation(s)
- Xiaolin Chen
- School of Food and Health, Beijing Technology and Business University, Beijing, 100048, China
| | - Jiani Chi
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Yanger Liu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Ruoxi Du
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China
| | - Mingzhang Guo
- School of Food and Health, Beijing Technology and Business University, Beijing, 100048, China; Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China.
| | - Wentao Xu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, 100083, China.
| |
Collapse
|
15
|
Elechi JOG, Sirianni R, Conforti FL, Cione E, Pellegrino M. Food System Transformation and Gut Microbiota Transition: Evidence on Advancing Obesity, Cardiovascular Diseases, and Cancers-A Narrative Review. Foods 2023; 12:2286. [PMID: 37372497 PMCID: PMC10297670 DOI: 10.3390/foods12122286] [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: 04/06/2023] [Revised: 05/22/2023] [Accepted: 05/23/2023] [Indexed: 06/29/2023] Open
Abstract
Food, a vital component of our daily life, is fundamental to our health and well-being, and the knowledge and practices relating to food have been passed down from countless generations of ancestors. Systems may be used to describe this extremely extensive and varied body of agricultural and gastronomic knowledge that has been gathered via evolutionary processes. The gut microbiota also underwent changes as the food system did, and these alterations had a variety of effects on human health. In recent decades, the gut microbiome has gained attention due to its health benefits as well as its pathological effects on human health. Many studies have shown that a person's gut microbiota partially determines the nutritional value of food and that diet, in turn, shapes both the microbiota and the microbiome. The current narrative review aims to explain how changes in the food system over time affect the makeup and evolution of the gut microbiota, advancing obesity, cardiovascular disease (CVD), and cancer. After a brief discussion of the food system's variety and the gut microbiota's functions, we concentrate on the relationship between the evolution of food system transformation and gut microbiota system transition linked to the increase of non-communicable diseases (NCDs). Finally, we also describe sustainable food system transformation strategies to ensure healthy microbiota composition recovery and maintain the host gut barrier and immune functions to reverse advancing NCDs.
Collapse
Affiliation(s)
- Jasper Okoro Godwin Elechi
- Department of Pharmacy and Health and Nutritional Sciences, University of Calabria, 87036 Arcavacata di Rende, Italy; (R.S.); (F.L.C.); (E.C.); (M.P.)
| | | | | | | | | |
Collapse
|
16
|
Rakotonirina A, Galperine T, Audry M, Kroemer M, Baliff A, Carrez L, Sadeghipour F, Schrenzel J, Guery B, Allémann É. Dry alginate beads for fecal microbiota transplantation: from model strains to fecal samples. Int J Pharm 2023; 639:122961. [PMID: 37075927 DOI: 10.1016/j.ijpharm.2023.122961] [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: 03/07/2023] [Revised: 04/05/2023] [Accepted: 04/11/2023] [Indexed: 04/21/2023]
Abstract
Clostridioides difficile infection (CDI) is a critical nosocomial infection with more than 124,000 cases per year in Europe and a mortality rate of 15-17%. The standard of care (SoC) is antibiotic treatment. Unfortunately, the relapse rate is high (∼35%) and SoC is significantly less effective against recurrent infection (rCDI). Fecal microbiota transplantation (FMT) is a recommended treatment against rCDI from the second recurrence episode and has an efficacy of 90%. The formulation of diluted donor stool deserves innovation because its actual administration routes deserve optimization (naso-duodenal/jejunal tubes, colonoscopy, enema or several voluminous oral capsules). Encapsulation of model bacteria strains in gel beads were first investigated. Then, the encapsulation method was applied to diluted stools. Robust spherical gel beads were obtained. The mean particle size was around 2 mm. A high loading of viable microorganisms was obtained for model strains and fecal samples. For plate-counting, values ranged from 1015 to 1017 CFU/g for single and mixed model strains, and 106 to 108 CFU/g for fecal samples. This corresponded to a viability of 30% to 60% as assessed by flow cytometry. This novel formulation is promising as the technology is applicable to both model strains and bacteria contained in the gut microbiota.
Collapse
Affiliation(s)
- Adèle Rakotonirina
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland
| | - Tatiana Galperine
- Infectious Diseases Service, Department of Medicine, University Hospital and University of Lausanne, 1011 & 1015 Lausanne, Switzerland; French Group of Faecal Microbiota Transplantation, Paris, France
| | - Maxime Audry
- Service of Pharmacy, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Marie Kroemer
- Service of Pharmacy, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Aurélie Baliff
- Infectious Diseases Service, Department of Medicine, University Hospital and University of Lausanne, 1011 & 1015 Lausanne, Switzerland
| | - Laurent Carrez
- Service of Pharmacy, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Farshid Sadeghipour
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland; Service of Pharmacy, Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Jacques Schrenzel
- Genomic Research Lab, Service of Infectious Diseases, Geneva University Hospitals and University of Geneva, 1211 Geneva, Switzerland
| | - Benoît Guery
- Infectious Diseases Service, Department of Medicine, University Hospital and University of Lausanne, 1011 & 1015 Lausanne, Switzerland
| | - Éric Allémann
- School of Pharmaceutical Sciences, University of Geneva, 1211 Geneva, Switzerland; Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, 1211 Geneva, Switzerland.
| |
Collapse
|
17
|
Feng P, Zhao S, Zhang Y, Li E. A review of probiotics in the treatment of autism spectrum disorders: Perspectives from the gut–brain axis. Front Microbiol 2023; 14:1123462. [PMID: 37007501 PMCID: PMC10060862 DOI: 10.3389/fmicb.2023.1123462] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 02/07/2023] [Indexed: 03/18/2023] Open
Abstract
Autism spectrum disorders (ASD) are a class of neurodevelopmental conditions with a large societal impact. Despite existing evidence suggesting a link between ASD pathogenesis and gut–brain axis dysregulation, there is no systematic review of the treatment of probiotics on ASD and its associated gastrointestinal abnormalities based on the gut–brain axis. Therefore, we performed an analysis for ASD based on preclinical and clinical research to give a comprehensive synthesis of published evidence of a potential mechanism for ASD. On the one hand, this review aims to elucidate the link between gastrointestinal abnormalities and ASD. Accordingly, we discuss gut microbiota dysbiosis regarding gut–brain axis dysfunction. On the other hand, this review suggests that probiotic administration to regulate the gut–brain axis might improve gastrointestinal symptoms, restore ASD-related behavioral symptoms, restore gut microbiota composition, reduce inflammation, and restore intestinal barrier function in human and animal models. This review suggests that targeting the microbiota through agents such as probiotics may represent an approach for treating subsets of individuals with ASD.
Collapse
Affiliation(s)
- Pengya Feng
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Key Laboratory of Helicobacter pylori, Microbiota and Gastrointestinal Cancer of Henan Province, Marshall Medical Research Center, Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shuai Zhao
- College of Bioengineering, Henan University of Technology, Zhengzhou, China
| | - Yangyang Zhang
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Enyao Li
- Department of Children Rehabilitation, Key Laboratory of Rehabilitation Medicine in Henan, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- *Correspondence: Enyao Li,
| |
Collapse
|
18
|
Dhanya Raj CT, Kandaswamy S, Suryavanshi MV, Ramasamy KP, Rajasabapathy R, Arthur James R. Genomic and metabolic properties of Staphylococcus gallinarum FCW1 MCC4687 isolated from naturally fermented coconut water towards GRAS assessment. Gene 2023; 867:147356. [PMID: 36907276 DOI: 10.1016/j.gene.2023.147356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 03/02/2023] [Accepted: 03/07/2023] [Indexed: 03/13/2023]
Abstract
Staphylococcus gallinarum FCW1 was isolated from naturally fermented coconut-water and identified by biochemical and molecular methods. Probiotic characterization and safety assessment were conducted through a series of in vitro tests. A high survival rate was observed when the strain was tested for resistance to bile, lysozyme, simulated gastric and intestinal fluid, phenol, and different temperature and salt concentrations. The strain showed antagonism against some pathogens, was susceptible to all antibiotics tested except penicillin, and showed no hemolytic and DNase activity. Hydrophobicity, autoaggregation, biofilm formation, and antioxidation tests indicated that the strain possessed a high adhesive and antioxidant ability. Enzymatic activity was used to evaluate the metabolic capacities of the strain. In-vivo experiment on zebrafish was performed to check its safety status. The whole-genome sequencing indicated that the genome contained 2,880,305 bp with a GC content of 33.23%. The genome annotation confirmed the presence of probiotic-associated genes and genes for oxalate degradation, sulfate reduction, acetate metabolism, and ammonium transport in the FCW1 strain, adding to the theory that this strain may be helpful in treating kidney stones. This study revealed that the strain FCW1 might be an excellent potential probiotic in developing fermented coconut beverages and treating and preventing kidney stone disease.
Collapse
Affiliation(s)
- C T Dhanya Raj
- Department of Marine Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | - Surabhi Kandaswamy
- School of Pharmacy and Biomedical Sciences, University of Central Lancashire, Preston, Lancashire PR1 2HE, United Kingdom; Manchester Centre for Genomic Medicine, School of Biological Sciences, Faculty of Biology Medicine and Health, The University of Manchester, 6(th) Floor, St Mary's Hospital, Oxford Road, Manchester M13 9WL, United Kingdom
| | - Mangesh V Suryavanshi
- Cardiovascular and Metabolic Sciences Department, Lerner Research Institute, Cleveland Clinic, OH 44195, United States.
| | | | - Raju Rajasabapathy
- Department of Marine Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India
| | - Rathinam Arthur James
- Department of Marine Science, Bharathidasan University, Tiruchirappalli 620024, Tamil Nadu, India.
| |
Collapse
|
19
|
Kim IB, Park SC, Kim YK. Microbiota-Gut-Brain Axis in Major Depression: A New Therapeutic Approach. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1411:209-224. [PMID: 36949312 DOI: 10.1007/978-981-19-7376-5_10] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
Major depression is impacted by the disruption of gut microbiota. Defects in gut microbiota can lead to microbiota-gut-brain axis dysfunction and increased vulnerability to major depression. While traditional chemotherapeutic approaches, such as antidepressant use, produce an overall partial therapeutic effect on depression, the gut microbiome has emerged as an effective target for better therapeutic outcomes. Recent representative studies on the microbiota hypothesis to explore the association between gut pathophysiology and major depression have indicated that restoring gut microbiota and microbiota-gut-brain axis could alleviate depression. We reviewed studies that supported the gut microbiota hypothesis to better understand the pathophysiology of depression; we also explored reports suggesting that gut microbiota restoration is an effective approach for improving depression. These findings indicate that gut microbiota and microbiota-gut-brain axis are appropriate new therapeutic targets for major depression.
Collapse
Affiliation(s)
- Il Bin Kim
- Department of Psychiatry, Hanyang University Guri Hospital, Guri, Republic of Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Seon-Cheol Park
- Department of Psychiatry, Hanyang University Guri Hospital, Guri, Republic of Korea.
- Department of Psychiatry, Hanyang University College of Medicine, Seoul, Republic of Korea.
| | - Yong-Ku Kim
- Department of Psychiatry, Korea University Ansan Hospital, Ansan, Republic of Korea
| |
Collapse
|
20
|
Elokil AA, Chen W, Mahrose K, Elattrouny MM, Abouelezz KFM, Ahmad HI, Liu HZ, Elolimy AA, Mandouh MI, Abdelatty AM, Li S. Early life microbiota transplantation from highly feed-efficient broiler improved weight gain by reshaping the gut microbiota in laying chicken. Front Microbiol 2022; 13:1022783. [PMID: 36466637 PMCID: PMC9715608 DOI: 10.3389/fmicb.2022.1022783] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/10/2022] [Indexed: 08/27/2023] Open
Abstract
Starting phase of laying chicken life is the building stone for rearing and production stages. Since, fecal microbial transplantation (FMT) regulates the gut microbial diversity and affects the productive performance of the bird. The aim of this study is to evaluate the effect of FMT from feed-efficient broiler chicken could program the diversity of gut microbiota and growth of recipient native slow growing egg-laying chicks. For this, a total of 150 (one-day-old) Jing Hong chicks were randomly assigned into two groups, each group consisted of 5 replicates (n = 15 bird/ replicate). The control group (CON) and FMT recipient birds (FMT) fed on basal diet, the FMT group received an oral daily dose of FMT prepared from Cobb-500 chickens. The FMT performed from the 1d to 28d of age, through the experimental period, feed intake and body weight were recorded weekly. At the end of a 28-day trial, carcass traits were assessed and cecal samples were collected for microbiome assessment via 16S rRNA-based metagenomic analysis to characterize the diversity and functions of microbial communities. The data were statistically analyzed using R software. Body weight and body weight gain increased, and FCR decreased (p = 0.01) in FMT group. The relative abundance of Firmicutes and the Firmicutes/Bacteroidetes (F/B) ratio were increased due to FMT administration (p = 0.01). A higher relative abundance of Lactobacillus, Lactococcus, and Bifidobacterium were presented in the FMT group. Meanwhile, Enterococcus, Helicobacter, and Bacteroides were more abundant in the CON group (p < 0.01). Kyoto encyclopedia of genes and genomes (KEGG) pathways for microbial functions regarding amino acid metabolism, secondary metabolites biosynthesis, carbohydrate metabolism, energy metabolism, and enzyme families, cofactors, and vitamins were significantly annotated in the FMT group. Overall, FMT administration from the donor of highly feed-efficient broilers improved weight gain by reshaping a distinct gut microbiome, which may be related to the metabolism and health in the recipients laying chicks, providing new insight on the application of the FMT technique for early life programming of laying chickens.
Collapse
Affiliation(s)
- Abdelmotaleb A. Elokil
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- Animal Production Department, Faculty of Agriculture, Moshtohor, Benha University, Mushthar, Egypt
| | - Wei Chen
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Key Laboratory of Animal Nutrition and Feed Science in South China, Ministry of Agriculture, Key Laboratory of Poultry Genetics and Breeding, Ministry of Agriculture, Guangdong Key Laboratory of Animal Breeding and Nutrition, Guangdong Public Laboratory of Animal Breeding and Nutrition, Guangzhou, China
| | - Khalid Mahrose
- Animal and Poultry Production Department, Faculty of Technology and Development, Zagazig University, Zagazig, Egypt
| | - Mahmoud M. Elattrouny
- Animal Production Department, Faculty of Agriculture, Moshtohor, Benha University, Mushthar, Egypt
| | - Khaled F. M. Abouelezz
- Department of Poultry Production, Faculty of Agriculture, Assiut University, Assiut, Egypt
| | - Hafiz Ishfaq Ahmad
- Department of Animal Breeding and Genetics, Faculty of Veterinary and Animal Sciences, The Islamia University of Bahawalpur, Bahawalpur, Pakistan
| | - Hua-Zhen Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Ahmed A. Elolimy
- Department of Animal Production, National Research Centre, Giza, Egypt
| | - Mahmoud I. Mandouh
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Alzahraa M. Abdelatty
- Department of Nutrition and Clinical Nutrition, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| | - Shijun Li
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, College of Animal Science and Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
21
|
Almeida C, Oliveira R, Baylina P, Fernandes R, Teixeira FG, Barata P. Current Trends and Challenges of Fecal Microbiota Transplantation-An Easy Method That Works for All? Biomedicines 2022; 10:2742. [PMID: 36359265 PMCID: PMC9687574 DOI: 10.3390/biomedicines10112742] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 09/10/2023] Open
Abstract
The gut microbiota refers to bacteria lodges in the gastrointestinal tract (GIT) that interact through various complex mechanisms. The disturbance of this ecosystem has been correlated with several diseases, such as neurologic, respiratory, cardiovascular, and metabolic diseases and cancer. Therefore, the modulation of the gut microbiota has emerged as a potential therapeutic tool; of the various forms of gut microbiota modulation, fecal microbiota transplantation (FMT) is the most approached. This recent technique involves introducing fecal material from a healthy donor into the patient's gastrointestinal tract, aiming to restore the gut microbiota and lead to the resolution of symptoms. This procedure implies a careful donor choice, fine collection and handling of fecal material, and a balanced preparation of the recipient and consequent administration of the prepared content. Although FMT is considered a biological therapy with promising effects, side effects such as diarrhea and abdominal pain have also been claimed, making this a significant challenge in the application of FMT. Bearing this in mind, the present review aims to summarize the recent advances in understanding FMT mechanisms, their impact across different pathological conditions, and the associated side effects, emphasizing the most recent published data.
Collapse
Affiliation(s)
- Cátia Almeida
- LaBMI—Laboratory of Medical & Industrial Biotechnology, Porto Polytechnic Institute, 4200-375 Porto, Portugal
- Department of Biomedicine, Unit of Biochemistry, Faculty of Medicine of Porto University, 4200-319 Porto, Portugal
- FP-i3ID, HEFP, FCS-UFP—Fernando Pessoa Hospital, Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
| | - Rita Oliveira
- FP-i3ID, HEFP, FCS-UFP—Fernando Pessoa Hospital, Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
| | - Pilar Baylina
- LaBMI—Laboratory of Medical & Industrial Biotechnology, Porto Polytechnic Institute, 4200-375 Porto, Portugal
- ESS-IPP—Health School, Porto Polytechnic Institute, 4200-072 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Rúben Fernandes
- LaBMI—Laboratory of Medical & Industrial Biotechnology, Porto Polytechnic Institute, 4200-375 Porto, Portugal
- FP-i3ID, HEFP, FCS-UFP—Fernando Pessoa Hospital, Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| | - Fábio G. Teixeira
- LaBMI—Laboratory of Medical & Industrial Biotechnology, Porto Polytechnic Institute, 4200-375 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- ICVS/3B’s-PT Government Associated Lab, 4710-057/4805-107 Braga/Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, 4710-057 Braga, Portugal
| | - Pedro Barata
- LaBMI—Laboratory of Medical & Industrial Biotechnology, Porto Polytechnic Institute, 4200-375 Porto, Portugal
- FP-i3ID, HEFP, FCS-UFP—Fernando Pessoa Hospital, Faculty of Health Sciences, Fernando Pessoa University, 4200-150 Porto, Portugal
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
| |
Collapse
|
22
|
De Benedittis G. Hypnobiome: A New, Potential Frontier of Hypnotherapy in the Treatment of Irritable Bowel Syndrome-A Narrative Review of the Literature. Int J Clin Exp Hypn 2022; 70:286-299. [PMID: 35792903 DOI: 10.1080/00207144.2022.2094269] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Increasing evidence suggests that the gut-brain axis may play a key role in health and disease via a bidirectional communication network involving neural and immunoendocrine pathways. This complex interplay deeply influences both gut microbiota and brain behavior. Pathobiome or gut dysbiosis is relevant for the pathogenesis of functional gastrointestinal disorders, such as IBS, chronic pain syndromes, and neurological and mental disorders. As a consequence, targeting the gut microbiota is emerging as a novel, effective therapeutic perspective. Among many treatment options, psychological interventions, including hypnosis, have been used to target the so-called psychobiome and its hypnotic analogue, i.e., hypnobiome, referring to their potential efficacy to modulate the mind-gut axis in IBS patients. A narrative review of the recent literature is provided and circumstantial evidence suggests that hypnobiome may represent a new promising frontier of hypnotherapy.
Collapse
|
23
|
Rakotonirina A, Galperine T, Allémann E. Fecal microbiota transplantation: a review on current formulations in Clostridioides difficile infection and future outlooks. Expert Opin Biol Ther 2022; 22:929-944. [PMID: 35763604 DOI: 10.1080/14712598.2022.2095901] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The role of the gut microbiota in health and the pathogenesis of several diseases has been highlighted in recent years. Even though the precise mechanisms involving the microbiome in these ailments are still unclear, microbiota-modulating therapies have been developed. Fecal microbiota transplantation (FMT) has shown significant results against Clostridioides difficile infection (CDI), and its potential has been investigated for other diseases. Unfortunately, the technical aspects of the treatment make it difficult to implement. Pharmaceutical technology approaches to encapsulate microorganisms could play an important role in providing this treatment and render the treatment modalities easier to handle. AREAS COVERED After an overview of CDI, this narrative review aims to discuss the current formulations for FMT and specifically addresses the technical aspects of the treatment. This review also distinguishes itself by focusing on the hurdles and emphasizing the possible improvements using pharmaceutical technologies. EXPERT OPINION FMT is an efficient treatment for recurrent CDI. However, its standardization is overlooked. The approach of industrial and hospital preparations of FMT are different, but both show promise in their respective methodologies. Novel FMT formulations could enable further research on dysbiotic diseases in the future.
Collapse
Affiliation(s)
- Adèle Rakotonirina
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| | - Tatiana Galperine
- Infectious Diseases Service, Department of Medicine, University Hospital and University of Lausanne, Lausanne, Switzerland.,French Group of Faecal Microbiota Transplantation
| | - Eric Allémann
- School of Pharmaceutical Sciences, University of Geneva, Geneva, Switzerland.,Institute of Pharmaceutical Sciences of Western Switzerland, University of Geneva, Geneva, Switzerland
| |
Collapse
|
24
|
Bibbò S, Ianiro G, Giambò F, Settanni CR, Cammarota G, Gasbarrini A. Role of gut microbiome on immunotherapy efficacy in melanoma. Hum Vaccin Immunother 2022; 18:1926759. [PMID: 34190675 PMCID: PMC9122303 DOI: 10.1080/21645515.2021.1926759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 04/14/2021] [Indexed: 11/16/2022] Open
Abstract
The gut microbiota is considered a key component in many aspects of cancer pathophysiology and response to therapy. In particular, in recent years intriguing evidences has been emerging regarding the role of the intestinal microbiota in the response to immunotherapy and in promoting the development of adverse events, such as colitis. For this reason, studies are being carried out both on pre-clinical models and on humans to study how to predict the response to immunotherapy through the study of the microbiota or how to improve its clinical response through modulation. Promising data have recently been reported through modulation by probiotics or prebiotics, and in particular by fecal microbiota transplantation. The aim of this review is to analyze the evidence regarding the role of the microbiota in immunotherapy with a particular focus on melanoma.
Collapse
Affiliation(s)
- Stefano Bibbò
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Gianluca Ianiro
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Federica Giambò
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Carlo Romano Settanni
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Giovanni Cammarota
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| | - Antonio Gasbarrini
- CEMAD Digestive Disease Center, Fondazione Policlinico Universitario “A. Gemelli” IRCCS, Università Cattolica del Sacro Cuore, Roma, Italy
| |
Collapse
|
25
|
Mohamed TM, Sun W, Bumbie GZ, Elokil AA, Mohammed KAF, Zebin R, Hu P, Wu L, Tang Z. Feeding Bacillus subtilis ATCC19659 to Broiler Chickens Enhances Growth Performance and Immune Function by Modulating Intestinal Morphology and Cecum Microbiota. Front Microbiol 2022; 12:798350. [PMID: 35281307 PMCID: PMC8904210 DOI: 10.3389/fmicb.2021.798350] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 12/13/2021] [Indexed: 12/18/2022] Open
Abstract
This study investigated dietary supplementation with Bacillus subtilis (BS) ATCC19659 on growth performance, biochemical indices, intestinal morphology, and cecum microflora in broiler chicks. A total of 600 Arbor 1-day Acres broilers of either sex were allotted to 5 treatments: chicks were fed a corn- and soybean-based diet (CON); chicks were fed basal diet containing 500 mg ZnB/kg (ZnB); chicks were fed basal diet containing 1 × 108 CFU/g feed of BS-ATCC19659 (BS-1); chicks were fed basal diet containing 3 × 108 CFU/g feed of BS-ATCC19659 (BS-3); and chicks were fed basal diet containing 5 × 108 CFU/g feed of BS-ATCC19659 (BS-5). Each treatment comprised 6 replicates with 20 birds for each replicate pen. Chicks in the BS-5 and BS-3 groups had higher body weight at the 21st and 42nd days and average daily gain from 1 to 21 days than that in the CON group (p < 0.05). Chicks in the BS-5 and ZnB groups had higher serum antioxidant activities and immunity response than those in the CON group (p < 0.05). Compared with the CON group, the liver mRNA abundance of GHR, TGF-β, IGF-1, IFN-γ, SOD, CAT, and GPX of chicks in three BS groups and the ileum villus length (μm) of chicks in BS-3 and ZnB groups was increased (p < 0.05). Compared with the CON group, the villus height-to-crypt depth ratio of the ileum of chicks in the BS-5 and BS-3 groups and the crypt depth and villus height-to-crypt depth ratio of the jejunum in the BS-5 and ZnB groups were increased (p < 0.05). The abundance of the Cyanobacteria phyla in the cecum decreased in response to treatment with both BS-ATCC19659 and ZnB groups (p < 0.05). Compared with the CON group, the cecum abundance of genera GCA-900066575 (Lachnospiraceae), Anaerofustis, and Papillibacter (Firmicutes phylum) in three BS groups were increased (p < 0.05); The abundance of genus Escherichia–Shigella reduced in the BS-3 group (p < 0.05). Compared with the CON group, the cecum abundance of genus Clostridia_unclassified in ZnB and BS-5 groups was decreased (p < 0.05) of broilers. Generally, Bacillus subtilis ATCC19659 as feed additive positively affected growth performance, immunity response, and cecal microflora of broilers.
Collapse
Affiliation(s)
- Taha M Mohamed
- Laboratory for Bio-Feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China.,Department of Animal and Fish Production, Faculty of Agriculture (Saba Basha), Alexandria University, Alexandria, Egypt
| | - Weizhong Sun
- Laboratory for Bio-Feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Gifty Z Bumbie
- Laboratory for Bio-Feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Abdelmotaleb A Elokil
- Animal Production Department, Faculty of Agriculture, Benha University, Moshtohor, Egypt
| | | | - Rao Zebin
- Laboratory for Bio-Feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Ping Hu
- Laboratory for Bio-Feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Liuting Wu
- Laboratory for Bio-Feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| | - Zhiru Tang
- Laboratory for Bio-Feed and Molecular Nutrition, College of Animal Science and Technology, Southwest University, Chongqing, China
| |
Collapse
|
26
|
de Sire A, de Sire R, Curci C, Castiglione F, Wahli W. Role of Dietary Supplements and Probiotics in Modulating Microbiota and Bone Health: The Gut-Bone Axis. Cells 2022; 11:cells11040743. [PMID: 35203401 PMCID: PMC8870226 DOI: 10.3390/cells11040743] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2021] [Revised: 02/04/2022] [Accepted: 02/09/2022] [Indexed: 02/04/2023] Open
Abstract
Osteoporosis is characterized by an alteration of bone microstructure with a decreased bone mineral density, leading to the incidence of fragility fractures. Around 200 million people are affected by osteoporosis, representing a major health burden worldwide. Several factors are involved in the pathogenesis of osteoporosis. Today, altered intestinal homeostasis is being investigated as a potential additional risk factor for reduced bone health and, therefore, as a novel potential therapeutic target. The intestinal microflora influences osteoclasts’ activity by regulating the serum levels of IGF-1, while also acting on the intestinal absorption of calcium. It is therefore not surprising that gut dysbiosis impacts bone health. Microbiota alterations affect the OPG/RANKL pathway in osteoclasts, and are correlated with reduced bone strength and quality. In this context, it has been hypothesized that dietary supplements, prebiotics, and probiotics contribute to the intestinal microecological balance that is important for bone health. The aim of the present comprehensive review is to describe the state of the art on the role of dietary supplements and probiotics as therapeutic agents for bone health regulation and osteoporosis, through gut microbiota modulation.
Collapse
Affiliation(s)
- Alessandro de Sire
- Physical Medicine and Rehabilitation Unit, Department of Medical and Surgical Sciences, University of Catanzaro “Magna Graecia”, 88100 Catanzaro, Italy
- Correspondence: (A.d.S.); (W.W.)
| | - Roberto de Sire
- Gastroenterology Unit, Department of Clinical Medicine and Surgery, University Federico II of Naples, 80126 Naples, Italy; (R.d.S.); (F.C.)
| | - Claudio Curci
- Physical Medicine and Rehabilitation Unit, Department of Neurosciences, ASST Carlo Poma, 46100 Mantova, Italy;
| | - Fabiana Castiglione
- Gastroenterology Unit, Department of Clinical Medicine and Surgery, University Federico II of Naples, 80126 Naples, Italy; (R.d.S.); (F.C.)
| | - Walter Wahli
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Clinical Sciences Building, Singapore 308232, Singapore
- Toxalim Research Center in Food Toxicology (UMR 1331), French National Research Institute for Agriculture, Food, and the Environment (INRAE), F-31300 Toulouse, France
- Center for Integrative Genomics, University of Lausanne, Le Génopode, CH-1015 Lausanne, Switzerland
- Correspondence: (A.d.S.); (W.W.)
| |
Collapse
|
27
|
Mu X, Zhang X, Gao H, Gao L, Li Q, Zhao C. Crosstalk between peripheral and the brain-resident immune components in epilepsy. J Integr Neurosci 2022; 21:9. [PMID: 35164445 DOI: 10.31083/j.jin2101009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 03/30/2021] [Accepted: 05/31/2021] [Indexed: 01/05/2025] Open
Abstract
Epilepsy is one of the most common neurology diseases. It is characterized by recurrent, spontaneous seizures and accompanied by various comorbidities which can significantly affect a person's life. Accumulating evidence indicates an essential pathophysiological role for neuroinflammation in epilepsy, which involves activation of microglia and astrocytes, recruitment of peripheral leukocytes into the central nervous system, and release of some inflammatory mediators, including pro-inflammatory factors and anti-inflammatory cytokines. There is complex crosstalk between the central nervous system and peripheral immune responses associated with the progression of epilepsy. This review provides an update of current knowledge about the contribution of this crosstalk associated with epilepsy. Additionally, how gut microbiota is involved in epilepsy and its possible influence on crosstalk is also discussed. Such recent advances in understanding suggest innovative methods for targeting the molecules correlated with the crosstalk and may provide a better prognosis for patients diagnosed with epilepsy.
Collapse
Affiliation(s)
- Xiaopeng Mu
- Neurology, The First Hospital of China Medical University, 110001 Shenyang, Liaoning, China
- Neurology, The Fourth Hospital of China Medical University, 110032 Shenyang, Liaoning, China
| | - Xiuchun Zhang
- Neurology, The First Hospital of China Medical University, 110001 Shenyang, Liaoning, China
| | - Honghua Gao
- Neurology, The Fourth Hospital of China Medical University, 110032 Shenyang, Liaoning, China
| | - Lianbo Gao
- Neurology, The Fourth Hospital of China Medical University, 110032 Shenyang, Liaoning, China
| | - Qingchang Li
- Department of Pathology, The First Hospital of China Medical University, 110001 Shenyang, Liaoning, China
| | - Chuansheng Zhao
- Neurology, The First Hospital of China Medical University, 110001 Shenyang, Liaoning, China
- Stroke Center, The First Hospital of China Medical University, 110001 Shenyang, Liaoning, China
| |
Collapse
|
28
|
Neag MA, Mitre AO, Catinean A, Buzoianu AD. Overview of the microbiota in the gut-liver axis in viral B and C hepatitis. World J Gastroenterol 2021; 27:7446-7461. [PMID: 34887642 PMCID: PMC8613744 DOI: 10.3748/wjg.v27.i43.7446] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 08/13/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
Viral B and C hepatitis are a major current health issue, both diseases having a chronic damaging effect on the liver and its functions. Chronic liver disease can lead to even more severe and life-threatening conditions, such as liver cirrhosis and hepatocellular carcinoma. Recent years have uncovered an important interplay between the liver and the gut microbiome: the gut-liver axis. Hepatitis B and C infections often cause alterations in the gut microbiota by lowering the levels of ‘protective’ gut microorganisms and, by doing so, hinder the microbiota ability to boost the immune response. Treatments aimed at restoring the gut microbiota balance may provide a valuable addition to current practice therapies and may help limit the chronic changes observed in the liver of hepatitis B and C patients. This review aims to summarize the current knowledge on the anato-functional axis between the gut and liver and to highlight the influence that hepatitis B and C viruses have on the microbiota balance, as well as the influence of treatments aimed at restoring the gut microbiota on infected livers and disease progression.
Collapse
Affiliation(s)
- Maria Adriana Neag
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy of Cluj-Napoca, Cluj-Napoca 400337, Romania
| | - Andrei Otto Mitre
- Faculty of Medicine, Iuliu Hatieganu University of Medicine and Pharmacy of Cluj-Napoca, Cluj-Napoca 400012, Romania
| | - Adrian Catinean
- Department of Internal Medicine, Iuliu Hatieganu University of Medicine and Pharmacy of Cluj-Napoca, Cluj-Napoca 400006, Romania
| | - Anca Dana Buzoianu
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Hatieganu University of Medicine and Pharmacy of Cluj-Napoca, Cluj-Napoca 400337, Romania
| |
Collapse
|
29
|
Kim J, Hlaing SP, Lee J, Saparbayeva A, Kim S, Hwang DS, Lee EH, Yoon IS, Yun H, Kim MS, Moon HR, Jung Y, Yoo JW. Exfoliated bentonite/alginate nanocomposite hydrogel enhances intestinal delivery of probiotics by resistance to gastric pH and on-demand disintegration. Carbohydr Polym 2021; 272:118462. [PMID: 34420722 DOI: 10.1016/j.carbpol.2021.118462] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/14/2021] [Accepted: 07/18/2021] [Indexed: 12/11/2022]
Abstract
In this study, we developed Lactobacillus rhamnosus GG (LGG)-encapsulating exfoliated bentonite/alginate nanocomposite hydrogels for protecting probiotics by delaying gastric fluid penetration into the nanocomposite and their on-demand release in the intestine. The pore size of the bentonite/alginate nanocomposite hydrogels (BA15) was two-fold smaller than that of alginate hydrogel (BA00). Following gastric pH challenge, the survival of LGG in BA15 decreased by only 1.43 log CFU/g as compared to the 6.25 log CFU/g decrease in alginate (BA00). Further, the internal pH of BA15 decreased more gradually than that of BA00. After oral administration in mice, BA15 maintained shape integrity during gastric passage, followed by appropriate disintegration within the target intestinal area. Additionally, a fecal recovery experiment in mice showed that the viable counts of LGG in BA15 were six-fold higher than those in BA00. The findings suggest the exfoliated bentonite/alginate nanocomposite hydrogel as a promising platform for intestinal delivery of probiotics.
Collapse
Affiliation(s)
- Jihyun Kim
- College of Pharmacy, Pusan National University, Busan 46241, South Korea
| | - Shwe Phyu Hlaing
- College of Pharmacy, Pusan National University, Busan 46241, South Korea
| | - Juho Lee
- College of Pharmacy, Pusan National University, Busan 46241, South Korea
| | | | - Sangsik Kim
- Department of Biosystems Engineering, The University of Arizona, Tucson, AZ 85721, United States
| | - Dong Soo Hwang
- Division of Environmental Science and Engineering, Pohang University of Science and Technology, Pohang 37673, South Korea
| | - Eun Hee Lee
- College of Pharmacy, Korea University, Sejong 30019, South Korea
| | - In-Soo Yoon
- College of Pharmacy, Pusan National University, Busan 46241, South Korea
| | - Hwayoung Yun
- College of Pharmacy, Pusan National University, Busan 46241, South Korea
| | - Min-Soo Kim
- College of Pharmacy, Pusan National University, Busan 46241, South Korea
| | - Hyung Ryong Moon
- College of Pharmacy, Pusan National University, Busan 46241, South Korea
| | - Yunjin Jung
- College of Pharmacy, Pusan National University, Busan 46241, South Korea
| | - Jin-Wook Yoo
- College of Pharmacy, Pusan National University, Busan 46241, South Korea.
| |
Collapse
|
30
|
Czajeczny D, Kabzińska K, Wójciak RW. Effects of Bifidobacterium Lactis BS01 and Lactobacillus Acidophilus LA02 on cognitive functioning in healthy women. APPLIED NEUROPSYCHOLOGY-ADULT 2021:1-9. [PMID: 34493130 DOI: 10.1080/23279095.2021.1967155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
A growing body of research studies the relationship between probiotic bacteria in the gut and the host organism, including the impact on cognitive functioning. Data from human studies are scarce; however, recent studies point toward the beneficial role of probiotics for cognitive functioning. One of the mechanisms involved in this relationship is the probiotic's ability to influence inflammation and immune response. The aim of this initial study was to investigate the effects of probiotic supplementation with Bifidobacterium Lactis BS01 and Lactobacillus Acidophilus LA02 on cognitive functioning in healthy, young adult females. A total of 53 participants aged 19-31 were enrolled, and 38 completed the trial. A 6-week probiotic or placebo treatment was conducted. Five measures of cognitive functioning were applied pre- and post-treatment. Both groups showed general improvement at the second assessment. Contrary to our hypothesis, the placebo group improved slightly, but significantly, in four out of five measures of cognitive functioning, with the exception of the Wisconsin Card Sorting Test (WCST). The supplementation group improved significantly in two measures of the WCST, compared to the placebo group. Similar results have been previously reported. Probiotic supplementation, while not harmful, might not be beneficial for cognition in the healthy population, or at least not universally.
Collapse
Affiliation(s)
- Dominik Czajeczny
- Department of Clinical Psychology, Poznan University of Medical Sciences, Poznań, Poland
| | - Karolina Kabzińska
- Department of Clinical Psychology, Poznan University of Medical Sciences, Poznań, Poland
| | - Rafał Wojciech Wójciak
- Department of Clinical Psychology, Poznan University of Medical Sciences, Poznań, Poland
| |
Collapse
|
31
|
Does probiotic supplementation aid weight loss? A randomized, single-blind, placebo-controlled study with Bifidobacterium lactis BS01 and Lactobacillus acidophilus LA02 supplementation. Eat Weight Disord 2021; 26:1719-1727. [PMID: 32797375 PMCID: PMC8292266 DOI: 10.1007/s40519-020-00983-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/03/2020] [Indexed: 12/15/2022] Open
Abstract
PURPOSE Probiotic supplements are gaining popularity worldwide. This trend is especially present in females, and a common motivation for consumption is weight loss, no matter the BMI. The aim of this study was to investigate the effects of probiotic supplementation on weight loss in healthy, young adult females and to put claims made by manufacturers of such products to the test. METHODS The study utilizes a randomized, single-blind, placebo-control design. 53 females aged 19-33 were enrolled, and 38 completed the trial. A 6 week supplementation with Bifidobacterium lactis BS01 and Lactobacillus acidophilus LA02 or placebo was conducted. Anthropometric measures (body mass, BMI, body fat percentage, arm skinfold fat, waist circumference, and WHR) were applied pre and post-treatment. RESULTS No significant changes in anthropometric measures were observed in both supplementation and placebo groups. CONCLUSION The results of this investigation do not support claims made by probiotic products manufacturers, that they aid weight loss. Our results seem to support an argument that weight loss is mostly associated with food habits and dietary behaviors, not probiotic intake. It is possible that probiotic supplementation may play a facilitating weight loss but has no effect without dietary intervention. Another possible explanation is that due to strain specificity-bacteria strains used in this study are not effective for weight loss. LEVEL OF EVIDENCE I: randomized controlled trial.
Collapse
|
32
|
Khalil NA, Eltahan NR, Elaktash HM, Aly S, Sarbini SR. Prospective evaluation of probiotic and prebiotic supplementation on diabetic health associated with gut microbiota. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
33
|
Lee HJ, Hong JK, Kim JK, Kim DH, Jang SW, Han SW, Yoon IY. Effects of Probiotic NVP-1704 on Mental Health and Sleep in Healthy Adults: An 8-Week Randomized, Double-Blind, Placebo-Controlled Trial. Nutrients 2021; 13:2660. [PMID: 34444820 PMCID: PMC8398773 DOI: 10.3390/nu13082660] [Citation(s) in RCA: 75] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 12/19/2022] Open
Abstract
The human gut microbiome is closely linked to mental health and sleep. We aimed to verify the efficacy and safety of probiotic NVP-1704, a mixture of Lactobacillus reuteri NK33 and Bifidobacterium adolescentis NK98, in improving stress, depression, anxiety, and sleep disturbances, along with the measurement of some blood biomarkers. A total of 156 healthy adults with subclinical symptoms of depression, anxiety, and insomnia were retrospectively registered and randomly assigned to receive either NVP-1704 (n = 78) or a placebo (n = 78) for eight weeks. Participants completed the Stress Response Inventory, Beck's Depression and Anxiety Inventory, Pittsburg Sleep Quality Index, and Insomnia Severity Index at baseline, at four and eight weeks of treatment. Pre- and post-treatment blood tests for biomarkers were conducted. After intervention, gut microbiota composition was quantified by pyrosequencing the bacterial 16S rRNA gene. The NVP-1704 group had a more significant reduction in depressive symptoms at four and eight weeks of treatment, and anxiety symptoms at four weeks compared to the placebo group. Those receiving NVP-1704 also experienced an improvement in sleep quality. NVP-1704 treatment led to a decrease in serum interleukin-6 levels. Furthermore, NVP-1704 increased Bifidobacteriaceae and Lactobacillacea, whereas it decreased Enterobacteriaceae in the gut microbiota composition. Our findings suggest that probiotic NVP-1704 could be beneficial for mental health and sleep.
Collapse
Affiliation(s)
- Hyuk Joo Lee
- Department of Psychiatry, Uijeongbu Eulji Medical Center, Eulji University, Gyeonggi 11759, Korea;
- Department of Neuropsychiatry, School of Medicine, Eulji University, Daejeon 34824, Korea
| | - Jung Kyung Hong
- Department of Psychiatry, Bundang Hospital, Seoul National University, Seongnam 13620, Korea;
| | - Jeon-Kyung Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea; (J.-K.K.); (D.-H.K.)
| | - Dong-Hyun Kim
- Neurobiota Research Center, College of Pharmacy, Kyung Hee University, Seoul 02447, Korea; (J.-K.K.); (D.-H.K.)
| | - Seok Won Jang
- PB Department, Navipharm Inc., Suwon 16209, Korea; (S.W.J.); (S.-W.H.)
| | - Seung-Won Han
- PB Department, Navipharm Inc., Suwon 16209, Korea; (S.W.J.); (S.-W.H.)
| | - In-Young Yoon
- Department of Psychiatry, Bundang Hospital, Seoul National University, Seongnam 13620, Korea;
- Department of Psychiatry, School of Medicine, Seoul National University, Seoul 03080, Korea
| |
Collapse
|
34
|
Kim JH, Kim K, Kim W. Gut microbiota restoration through fecal microbiota transplantation: a new atopic dermatitis therapy. Exp Mol Med 2021; 53:907-916. [PMID: 34017060 PMCID: PMC8178377 DOI: 10.1038/s12276-021-00627-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 02/07/2023] Open
Abstract
The pathogenesis of atopic dermatitis (AD) involves complex factors, including gut microbiota and immune modulation, which remain poorly understood. The aim of this study was to restore gut microbiota via fecal microbiota transplantation (FMT) to ameliorate AD in mice. FMT was performed using stool from donor mice. The gut microbiota was characterized via 16S rRNA sequencing and analyzed using Quantitative Insights into Microbial Ecology 2 with the DADA2 plugin. Gut metabolite levels were determined by measuring fecal short-chain fatty acid (SCFA) contents. AD-induced allergic responses were evaluated by analyzing blood parameters (IgE levels and eosinophil percentage, eosinophil count, basophil percentage, and monocyte percentage), the levels of Th1 and Th2 cytokines, dermatitis score, and the number of mast cells in the ileum and skin tissues. Calprotectin level was measured to assess gut inflammation after FMT. FMT resulted in the restoration of gut microbiota to the donor state and increases in the levels of SCFAs as gut metabolites. In addition, FMT restored the Th1/Th2 balance, modulated Tregs through gut microbiota, and reduced IgE levels and the numbers of mast cells, eosinophils, and basophils. FMT is associated with restoration of gut microbiota and immunologic balance (Th1/Th2) along with suppression of AD-induced allergic responses and is thus a potential new therapy for AD.
Collapse
Affiliation(s)
- Jong-Hwa Kim
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Kiyoung Kim
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul, South Korea
| | - Wonyong Kim
- Department of Microbiology, Chung-Ang University College of Medicine, Seoul, South Korea.
| |
Collapse
|
35
|
Zommiti M, Feuilloley MGJ, Connil N. Update of Probiotics in Human World: A Nonstop Source of Benefactions till the End of Time. Microorganisms 2020; 8:E1907. [PMID: 33266303 PMCID: PMC7760123 DOI: 10.3390/microorganisms8121907] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 11/23/2020] [Accepted: 11/23/2020] [Indexed: 02/06/2023] Open
Abstract
Lactic acid bacteria (LAB) are known for their biotechnological potential. Moreover, LAB are distinguished by amazing criteria: Adjusting the intestinal environment, inhibiting pathogenic microbes in the gastrointestinal tract, ability to reduce pathogen adhesion activity, improving the balance of the microbiota inside the intestine, capabilities of regulating intestinal mucosal immunity, and maintaining intestinal barrier function. The escalating number of research and studies about beneficial microorganisms and their impact on promoting health has attracted a big interest in the last decades. Since antiquity, various based fermented products of different kinds have been utilized as potential probiotic products. Nevertheless, the current upsurge in consumers' interest in bioalternatives has opened new horizons for the probiotic field in terms of research and development. The present review aims at shedding light on the world of probiotics, a continuous story of astonishing success in various fields, in particular, the biomedical sector and pharmaceutical industry, as well as to display the importance of probiotics and their therapeutic potential in purpose to compete for sturdy pathogens and to struggle against diseases and acute infections. Shadows and future trends of probiotics use are also discussed.
Collapse
Affiliation(s)
- Mohamed Zommiti
- Unité de Protéomique Fonctionnelle et Potentiel Nutraceutique de la Biodiversité de Tunisie, Institut Supérieur des Sciences Biologiques Appliquées de Tunis, Université Tunis El-Manar, Tunis 1006, Tunisia
| | - Marc G. J. Feuilloley
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Université de Rouen Normandie, Normandie Université, F-27000 Evreux, France; (M.G.J.F.); (N.C.)
| | - Nathalie Connil
- Laboratoire de Microbiologie Signaux et Microenvironnement (LMSM) EA 4312, Université de Rouen Normandie, Normandie Université, F-27000 Evreux, France; (M.G.J.F.); (N.C.)
| |
Collapse
|
36
|
Wlazło Ł, Łopuszyński W, Nowakowicz-Dębek B, Ossowski M, Bis-Wencel H. Translocation of intestinal bacteria as a cause of subcutaneous abscesses of the neck and head in American mink (Neovison vison) - a case report. BMC Vet Res 2020; 16:434. [PMID: 33167981 PMCID: PMC7654155 DOI: 10.1186/s12917-020-02654-3] [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: 04/15/2020] [Accepted: 10/28/2020] [Indexed: 11/20/2022] Open
Abstract
Background The problem of transmission of intestinal microorganisms to tissues occurs when intestinal epithelial cells do not adhere tightly (tight junction), which is caused by improper nutrition, usually associated with poor mucosal status. The impact on maintaining its proper condition in the case of animals also depends on the proper preparation and fragmentation of the ingredients of the feed. Intestinal microbiota disorders are increasingly indicated as one of the causes of many autoimmune, neurodevelopmental and metabolic diseases. However, there are no studies indicating damage to the intestinal barrier of animals resulting in the penetration of microorganisms from the gastrointestinal tract directly into the bloodstream which may result in the development of chronic inflammation. Case presentation On a mink (Neovison vison) farm with a foundation stock of 4,000 females, abscesses were observed in the head, followed by progressive deaths. Antibiotic treatment with amoxicillin and clavulanic acid added to the animals’ feed was not successful. Macroscopic and microscopic changes indicated local suppurative inflammation of the skin and subcutaneous tissue with the presence of purulent fistulas. Microbiological analysis showed a significant increase in Escherichia coli in all samples taken from the abscesses. The results indicate the migration of intestinal bacteria through disturbance of the permeability of the intestinal barrier and their transfer to the blood. Symptoms were alleviated in all animals following changes in the feed components and in feed particle size. Conclusions It is necessary to take into account the possibility of transmission of intestinal bacteria in the etiology of inflammatory diseases in animals. Conducting more research in this field will improve the understanding of the relationship between intestinal microbes and the health of the body as a whole.
Collapse
Affiliation(s)
- Łukasz Wlazło
- Department of Animal Hygiene and Environmental Hazards, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland
| | - Wojciech Łopuszyński
- Sub-Department of Pathomorphology and Forensic Veterinary Medicine, Department and Clinic of Animal Internal Diseases, University of Life Sciences in Lublin, Głęboka 30, 20-612, Lublin, Poland
| | - Bożena Nowakowicz-Dębek
- Department of Animal Hygiene and Environmental Hazards, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland.
| | - Mateusz Ossowski
- Department of Animal Hygiene and Environmental Hazards, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland
| | - Hanna Bis-Wencel
- Department of Animal Hygiene and Environmental Hazards, University of Life Sciences in Lublin, Akademicka 13, 20-950, Lublin, Poland
| |
Collapse
|
37
|
Arulsamy A, Tan QY, Balasubramaniam V, O’Brien TJ, Shaikh MF. Gut Microbiota and Epilepsy: A Systematic Review on Their Relationship and Possible Therapeutics. ACS Chem Neurosci 2020; 11:3488-3498. [PMID: 33064448 DOI: 10.1021/acschemneuro.0c00431] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Dysbiosis of gut microbiota may lead to a range of diseases including neurological disorders. Thus, it is hypothesized that regulation of the intestinal microbiota may prevent or treat epilepsy. The purpose of this systematic review is to evaluate the evidence investigating the relationship between gut microbiota and epilepsy and possible interventions. A systematic review of the literature was done on four databases (PubMed, Scopus, EMBASE, and Web of Science). Study selection was restricted to original research articles while following the PRISMA guidelines. Six studies were selected. These studies cohesively support the interaction between gut microbiota and epileptic seizures. Gut microbiota analysis identified increases in Firmicutes, Proteobacteria, Verrucomicrobia, and Fusobacteria with decreases in Bacteroidetes and Actinobacteria in epileptic patients. Ketogenic diet, probiotics, and fecal microbiota transplantation (FMT) improved the dysbiosis of the gut microbiota and seizure activity. However, the studies either had a small sample size, lack of subject variability, or short study or follow-up period, which may question their reliability. Nevertheless, these limited studies conclusively suggest that gut microbiota diversity and dysbiosis may be involved in the pathology of epilepsy. Future studies providing more reliable and in depth insight into the gut microbial community will spark promising alternative therapies to current epilepsy treatment.
Collapse
Affiliation(s)
- Alina Arulsamy
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Selangor, Malaysia
| | - Qian Ying Tan
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Selangor, Malaysia
| | - Vinod Balasubramaniam
- Infection and Immunity Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Selangor Malaysia
| | - Terence J. O’Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, 3004 VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, 3010 VIC, Australia
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, 47500 Selangor, Malaysia
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, 3004 VIC, Australia
- Department of Medicine, Royal Melbourne Hospital, The University of Melbourne, Parkville, 3010 VIC, Australia
| |
Collapse
|
38
|
Pop OL, Vodnar DC, Diaconeasa Z, Istrati M, Bințințan A, Bințințan VV, Suharoschi R, Gabbianelli R. An Overview of Gut Microbiota and Colon Diseases with a Focus on Adenomatous Colon Polyps. Int J Mol Sci 2020; 21:7359. [PMID: 33028024 PMCID: PMC7582333 DOI: 10.3390/ijms21197359] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 09/30/2020] [Accepted: 10/02/2020] [Indexed: 12/24/2022] Open
Abstract
It is known and accepted that the gut microbiota composition of an organism has an impact on its health. Many studies deal with this topic, the majority discussing gastrointestinal health. Adenomatous colon polyps have a high prevalence as colon cancer precursors, but in many cases, they are hard to diagnose in their early stages. Gut microbiota composition correlated with the presence of adenomatous colon polyps may be a noninvasive and efficient tool for diagnosis with a high impact on human wellbeing and favorable health care costs. This review is meant to analyze the gut microbiota correlated with the presence of adenomatous colon polyps as the first step for early diagnosis, prophylaxis, and treatment.
Collapse
Affiliation(s)
- Oana Lelia Pop
- Department of Food Science, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (O.L.P.); (D.C.V.); (Z.D.)
| | - Dan Cristian Vodnar
- Department of Food Science, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (O.L.P.); (D.C.V.); (Z.D.)
| | - Zorita Diaconeasa
- Department of Food Science, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (O.L.P.); (D.C.V.); (Z.D.)
| | - Magdalena Istrati
- Regional Institute of Gastroenterology and Hepatology “Prof. Dr. Octavian Fodor”, 400158 Cluj-Napoca, Romania;
| | - Adriana Bințințan
- 1st Medical Clinic, Department of Gastroenterology, Emergency County Hospital, 400006 Cluj Napoca, Romania;
| | - Vasile Virgil Bințințan
- 1st Surgical Clinic, Department of Surgery, University of Medicine and Pharmacy Cluj Napoca, 400006 Cluj Napoca, Romania;
| | - Ramona Suharoschi
- Department of Food Science, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (O.L.P.); (D.C.V.); (Z.D.)
| | - Rosita Gabbianelli
- Unit of Molecular Biology, School of Pharmacy, University of Camerino, Via Gentile III da Varano, 62032 Camerino, Italy
| |
Collapse
|
39
|
Tabashsum Z, Peng M, Alvarado-Martinez Z, Aditya A, Bhatti J, Romo PB, Young A, Biswas D. Competitive reduction of poultry-borne enteric bacterial pathogens in chicken gut with bioactive Lactobacillus casei. Sci Rep 2020; 10:16259. [PMID: 33004922 PMCID: PMC7530658 DOI: 10.1038/s41598-020-73316-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 09/15/2020] [Indexed: 01/10/2023] Open
Abstract
In this study, the effect of sustainable probiotics on Campylobacter jejuni colonization and gut microbiome composition was evaluated using chicken as a model organism. Chickens were given Lactobacillus casei over-expressing myosin-cross-reactive antigen (LC+mcra). LC+mcra can generate bioactive compounds in larger quantity including conjugated linoleic acid. A total of 120 chickens were used in duplicate trials to investigate the effectiveness of LC+mcra in decreasing C. jejuni colonization by means of kanamycin resistant strain compared to the control group. We observed that LC+mcra can efficiently colonize various parts of the chicken gut and competitively reduce colonization of natural and challenged C. jejuni and natural Salmonella enterica. LC+mcra was found to reduce C. jejuni colonization in cecum, ileum and jejunum, by more than one log CFU/g when compared to the no-probiotic control group. Furthermore, 16S rRNA compositional analysis revealed lower abundance of Proteobacteria, higher abundance of Firmicutes, along with enriched bacterial genus diversity in gut of LC+mcra fed chicken. Decreased contamination of drinking water by C. jejuni and S. enterica was also observed, suggesting a potential function of reducing horizontal transfer of enteric bacteria in poultry. Outcomes of this study reveal high potential of LC+mcra as sustainable approach to decrease colonization of C. jejuni and S. enterica in poultry gut along with other beneficial attributes.
Collapse
Affiliation(s)
- Zajeba Tabashsum
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, MD, 20742, USA
| | - Mengfei Peng
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Zabdiel Alvarado-Martinez
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, MD, 20742, USA
| | - Arpita Aditya
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Jacob Bhatti
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, MD, 20742, USA
| | - Paulina Bravo Romo
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Alana Young
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA
| | - Debabrata Biswas
- Biological Sciences Program - Molecular and Cellular Biology, University of Maryland, College Park, MD, 20742, USA.
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, 20742, USA.
- Center for Food Safety and Security Systems, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
40
|
Rusu IG, Suharoschi R, Vodnar DC, Pop CR, Socaci SA, Vulturar R, Istrati M, Moroșan I, Fărcaș AC, Kerezsi AD, Mureșan CI, Pop OL. Iron Supplementation Influence on the Gut Microbiota and Probiotic Intake Effect in Iron Deficiency-A Literature-Based Review. Nutrients 2020; 12:E1993. [PMID: 32635533 PMCID: PMC7400826 DOI: 10.3390/nu12071993] [Citation(s) in RCA: 92] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/15/2022] Open
Abstract
Iron deficiency in the human body is a global issue with an impact on more than two billion individuals worldwide. The most important functions ensured by adequate amounts of iron in the body are related to transport and storage of oxygen, electron transfer, mediation of oxidation-reduction reactions, synthesis of hormones, the replication of DNA, cell cycle restoration and control, fixation of nitrogen, and antioxidant effects. In the case of iron deficiency, even marginal insufficiencies may impair the proper functionality of the human body. On the other hand, an excess in iron concentration has a major impact on the gut microbiota composition. There are several non-genetic causes that lead to iron deficiencies, and thus, several approaches in their treatment. The most common methods are related to food fortifications and supplements. In this review, following a summary of iron metabolism and its health implications, we analyzed the scientific literature for the influence of iron fortification and supplementation on the gut microbiome and the effect of probiotics, prebiotics, and/or synbiotics in iron absorption and availability for the organism.
Collapse
Affiliation(s)
- Ioana Gabriela Rusu
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Ramona Suharoschi
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Dan Cristian Vodnar
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Carmen Rodica Pop
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Sonia Ancuța Socaci
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Romana Vulturar
- Department of Molecular Sciences, University of Medicine and Pharmacy Iuliu Hatieganu, 400349 Cluj-Napoca, Romania;
- Cognitive Neuroscience Laboratory, University Babes-Bolyai, 400327 Cluj-Napoca, Romania
| | - Magdalena Istrati
- Regional Institute of Gastroenterology and Hepatology “Prof. Dr. Octavian Fodor”, 400158 Cluj-Napoca, Romania;
| | - Ioana Moroșan
- Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400349 Cluj-Napoca, Romania;
| | - Anca Corina Fărcaș
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Andreea Diana Kerezsi
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Carmen Ioana Mureșan
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| | - Oana Lelia Pop
- Department of Food Science, University of Agricultural Science and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (I.G.R.); (R.S.); (D.C.V.); (C.R.P.); (S.A.S.); (A.C.F.); (A.D.K.); (C.I.M.)
| |
Collapse
|
41
|
Moles L, Otaegui D. The Impact of Diet on Microbiota Evolution and Human Health. Is Diet an Adequate Tool for Microbiota Modulation? Nutrients 2020; 12:nu12061654. [PMID: 32498430 PMCID: PMC7352211 DOI: 10.3390/nu12061654] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 05/29/2020] [Accepted: 05/31/2020] [Indexed: 02/08/2023] Open
Abstract
The human microbiome is emerging as an interesting field in research into the prevention of health problems and recovery from illness in humans. The complex ecosystem formed by the microbiota is continuously interacting with its host and the environment. Diet could be assumed to be one of the most prominent factors influencing the microbiota composition. Nevertheless, and in spite of numerous strategies proposed to modulate the human microbiota through dietary means, guidelines to achieve this goal have yet to be established. This review assesses the correlation between social and dietary changes over the course of human evolution and the adaptation of the human microbiota to those changes. In addition, it discusses the main dietary strategies for modulating the microbiota and the difficulties of putting them properly into practice.
Collapse
|
42
|
Hatanaka M, Morita H, Aoyagi Y, Sasaki K, Sasaki D, Kondo A, Nakamura T. Effective bifidogenic growth factors cyclo-Val-Leu and cyclo-Val-Ile produced by Bacillus subtilis C-3102 in the human colonic microbiota model. Sci Rep 2020; 10:7591. [PMID: 32372037 PMCID: PMC7200657 DOI: 10.1038/s41598-020-64374-w] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 04/16/2020] [Indexed: 12/20/2022] Open
Abstract
Bifidobacterium species are known to fulfill important functions within the human colon. Thus, stimulating the activity of bifidobacteria is important to maintain host health. We revealed that culture supernatants of Bacillus subtilis C-3102 (referred to as C-3102) stimulated the growth of Bifidobacterium species. In this study, we isolated and identified six bifidogenic growth factors, which were cyclo (D-Val-D-Ile), cyclo (L-Val-D-Ile), cyclo (D-Val-L-Ile), cyclo (L-Val-L-Ile), cyclo (D-Val-L-Leu) and cyclo (L-Val-L-Leu). These six cyclic dipeptides increased the growth of Bifidobacterium species and had no effect on potentially harmful gut organisms. Moreover, supplementation with a mixture of these six cyclic dipeptides significantly increased the abundance of microorganisms related to the genus Bifidobacterium in a human colonic microbiota model culture system, although supplementation with a single type of dipeptide had no effect. These results show that cyclic dipeptides containing Val-Leu and Val-Ile produced by C-3102 could serve as bifidogenic growth factors in the gut microbial community.
Collapse
Affiliation(s)
- Misaki Hatanaka
- Research & Development Dept, Asahi Calpis Wellness Co., Ltd., 4-1, 2-chome, Ebisu-Minami, Shibuya-ku, Tokyo, 150-0022, Japan.
| | - Hiroto Morita
- Department of Microbiological Flora Technology, Core Technology Laboratories, Asahi Quality and Innovations Co., Ltd. 5-11-10 Fuchinobe, Chuo-ku, Sagamihara-shi, 252-0206, Japan
| | - Yumi Aoyagi
- Research & Development Dept, Asahi Calpis Wellness Co., Ltd., 4-1, 2-chome, Ebisu-Minami, Shibuya-ku, Tokyo, 150-0022, Japan
| | - Kengo Sasaki
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo, 657-8501, Japan
| | - Daisuke Sasaki
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo, 657-8501, Japan
| | - Akihiko Kondo
- Graduate School of Science, Technology and Innovation, Kobe University, 1-1 Rokkodai-cho, Nada-ku, Kobe, Hyogo, 657-8501, Japan
| | - Teppei Nakamura
- Department of Microbiological Flora Technology, Core Technology Laboratories, Asahi Quality and Innovations Co., Ltd. 5-11-10 Fuchinobe, Chuo-ku, Sagamihara-shi, 252-0206, Japan
| |
Collapse
|
43
|
Gomes AC, Hoffmann C, Mota JF. Gut microbiota is associated with adiposity markers and probiotics may impact specific genera. Eur J Nutr 2019; 59:1751-1762. [PMID: 31250099 DOI: 10.1007/s00394-019-02034-0] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Accepted: 06/24/2019] [Indexed: 02/07/2023]
Abstract
PURPOSE It has been suggested that restoring gut microbiota alterations with probiotics represents a potential clinical target for the treatment of gut microbiota-related diseases, such as obesity. Here, we apply 16S rDNA microbiota profiling to establish which bacteria in the human gut are associated with obesity and cardiometabolic risk factors, and to evaluate whether probiotic supplementation modulates gut microbiota. METHODS We evaluated the effects of a probiotic mixture (2 × 1010 CFU/day of Lactobacillus acidophilus LA-14, Lactobacillus casei LC-11, Lactococcus lactis LL-23, Bifidobacterium bifidum BB-06, and Bifidobacterium lactis BL-4) in 32 overweight or obese women in a double-blind, randomized, placebo-controlled study. Using 16S rDNA sequencing, we characterized fecal samples and investigated the relationships between microbiome data and diet, body composition, antioxidant enzymes, and inflammatory profile. In addition, we characterized the degree of variation among fecal communities after the intervention. RESULTS BMI, weight, fat mass, lean mass, conicity index, protein intake, monounsaturated fat intake, glycated hemoglobin, TNF-α, and IL6/IL10 were significantly correlated with microbiome composition. The candidate division TM7 was strongly associated with all adiposity markers and Clostridiaceae associated negatively with TNF-α. The family Clostridiaceae increased and TM7 tended to decrease after the probiotic mixture supplementation. Subjects were clustered according to body composition, and a higher proportion of TM7 was observed in those with higher adiposity. CONCLUSIONS Ecosystem-wide analysis of probiotic use effects on the gut microbiota revealed a genera specific influence, and one of which (TM7) represents a promising novel target for obesity treatment. TRIAL REGISTRATION NUMBER U1111-1137-4566.
Collapse
Affiliation(s)
- Aline Corado Gomes
- Clinical and Sports Nutrition Research Laboratory (LABINCE), Faculty of Nutrition, Federal University of Goias, St. 227, Block 68, Setor Leste Universitário, Goiânia, GO, 74.605-080, Brazil
| | - Christian Hoffmann
- Department of Food Sciences and Experimental Nutrition, School of Pharmaceutical Sciences, University of São Paulo, São Paulo, SP, Brazil
| | - João Felipe Mota
- Clinical and Sports Nutrition Research Laboratory (LABINCE), Faculty of Nutrition, Federal University of Goias, St. 227, Block 68, Setor Leste Universitário, Goiânia, GO, 74.605-080, Brazil.
| |
Collapse
|
44
|
Tsiaoussis J, Antoniou MN, Koliarakis I, Mesnage R, Vardavas CI, Izotov BN, Psaroulaki A, Tsatsakis A. Effects of single and combined toxic exposures on the gut microbiome: Current knowledge and future directions. Toxicol Lett 2019; 312:72-97. [PMID: 31034867 DOI: 10.1016/j.toxlet.2019.04.014] [Citation(s) in RCA: 84] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 04/08/2019] [Accepted: 04/11/2019] [Indexed: 12/12/2022]
Abstract
Human populations are chronically exposed to mixtures of toxic chemicals. Predicting the health effects of these mixtures require a large amount of information on the mode of action of their components. Xenobiotic metabolism by bacteria inhabiting the gastrointestinal tract has a major influence on human health. Our review aims to explore the literature for studies looking to characterize the different modes of action and outcomes of major chemical pollutants, and some components of cosmetics and food additives, on gut microbial communities in order to facilitate an estimation of their potential mixture effects. We identified good evidence that exposure to heavy metals, pesticides, nanoparticles, polycyclic aromatic hydrocarbons, dioxins, furans, polychlorinated biphenyls, and non-caloric artificial sweeteners affect the gut microbiome and which is associated with the development of metabolic, malignant, inflammatory, or immune diseases. Answering the question 'Who is there?' is not sufficient to define the mode of action of a toxicant in predictive modeling of mixture effects. Therefore, we recommend that new studies focus to simulate real-life exposure to diverse chemicals (toxicants, cosmetic/food additives), including as mixtures, and which combine metagenomics, metatranscriptomics and metabolomic analytical methods achieving in that way a comprehensive evaluation of effects on human health.
Collapse
Affiliation(s)
- John Tsiaoussis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, 71110 Heraklion, Greece
| | - Michael N Antoniou
- Gene Expression and Therapy Group, King's College London, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, 8th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Ioannis Koliarakis
- Laboratory of Anatomy-Histology-Embryology, Medical School, University of Crete, 71110 Heraklion, Greece
| | - Robin Mesnage
- Gene Expression and Therapy Group, King's College London, Faculty of Life Sciences & Medicine, Department of Medical and Molecular Genetics, 8th Floor, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, United Kingdom
| | - Constantine I Vardavas
- Laboratory of Toxicology, Medical School, University of Crete, Voutes, 71409 Heraklion, Crete, Greece
| | - Boris N Izotov
- Department of Analytical, Toxicology, Pharmaceutical Chemistry and Pharmacognosy, Sechenov University, 119991 Moscow, Russia
| | - Anna Psaroulaki
- Department of Clinical Microbiology and Microbial Pathogenesis, Medical School, University of Crete, 71110 Heraklion, Greece
| | - Aristidis Tsatsakis
- Laboratory of Toxicology, Medical School, University of Crete, Voutes, 71409 Heraklion, Crete, Greece; Department of Analytical, Toxicology, Pharmaceutical Chemistry and Pharmacognosy, Sechenov University, 119991 Moscow, Russia.
| |
Collapse
|
45
|
The Influence of Maternal-Foetal Parameters on Concentrations of Zonulin and Calprotectin in the Blood and Stool of Healthy Newborns during the First Seven Days of Life. An Observational Prospective Cohort Study. J Clin Med 2019; 8:jcm8040473. [PMID: 30959960 PMCID: PMC6517987 DOI: 10.3390/jcm8040473] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/03/2019] [Accepted: 04/04/2019] [Indexed: 12/19/2022] Open
Abstract
Background: It can be hypothetically assumed that maternal and perinatal factors influence the intestinal barrier. Methods: The study was conducted with 100 healthy, full-term newborns breastfed in the first week of life, with similar analyses for their mothers. Zonulin and calprotectin levels were used as intestinal permeability markers. Results: The median (range) zonulin concentrations (ng/mL) were in mothers: serum, 21.39 (6.39–57.54); stool, 82.23 (42.52–225.74); and newborns: serum cord blood, 11.14 (5.82–52.34); meconium, 54.15 (1.36–700.65); and stool at age seven days, 114.41 (29.38–593.72). Calprotectin median (range) concentrations (µg/mL) in mothers were: stool, 74.79 (3.89–211.77); and newborns: meconium, 154.76 (6.93–8884.11); and stool at age seven days 139.12 (11.89–627.35). The use of antibiotics during pregnancy resulted in higher zonulin concentrations in umbilical-cord serum and calprotectin concentrations in newborn stool at seven days, while antibiotic therapy during labour resulted in higher zonulin concentrations in the stool of newborns at seven days. Zonulin concentrations in the stool of newborns (at seven days) who were born via caesarean section were higher compared to with vaginal birth. With further analyses, caesarean section was found to have a greater effect on zonulin concentrations than prophylactic administration of antibiotics in the perinatal period. Pregnancy mass gain >18 kg was associated with higher calprotectin concentrations in maternal stool. Body Mass Index (BMI) increase >5.7 during pregnancy was associated with decreased zonulin concentrations in maternal stool and increased calprotectin concentrations in stool of mothers and newborns at seven days. There was also a negative correlation between higher BMI increase in pregnancy and maternal zonulin stool concentrations and a positive correlation between BMI increase in pregnancy and maternal calprotectin stool concentrations. Conclusion: Maternal-foetal factors such as caesarean section, antibiotic therapy during pregnancy, as well as change in mother’s BMI during pregnancy may increase intestinal permeability in newborns. Changes in body mass during pregnancy can also affect intestinal permeability in mothers. However, health consequences associated with increased intestinal permeability during the first days of life are unknown. Additionally, before the zonulin and calprotectin tests can be adopted as universal diagnostic applications to assess increased intestinal permeability, validation of these tests is necessary.
Collapse
|
46
|
Mu J, Chen Q, Zhu L, Wu Y, Liu S, Zhao Y, Ma T. Influence of gut microbiota and intestinal barrier on enterogenic infection after liver transplantation. Curr Med Res Opin 2019; 35:241-248. [PMID: 29701490 DOI: 10.1080/03007995.2018.1470085] [Citation(s) in RCA: 12] [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] [Indexed: 12/13/2022]
Abstract
Liver transplantation is currently a standard therapy for patients with end-stage liver diseases and hepatocellular carcinoma. Given that liver transplantation has undergone a thriving development in these decades, the survival rates after liver transplantation have markedly improved as a result of the critical advancement in surgical techniques, immunosuppressive therapies, and post-operative care. However, infection remains a fatal complication after liver transplantation surgery. In particular, enterogenic infection represents a major complication in liver transplant recipients. This article gives an overview of infection cases after liver transplantation and focuses on the discussion of enterogenic infection in terms of its pathophysiology, risk factor, outcome, and treatment.
Collapse
Affiliation(s)
- Jingzhou Mu
- a College of Basic Medicine , Dalian Medical University , Dalian , Liaoning Province , PR China
| | - Qiuyu Chen
- a College of Basic Medicine , Dalian Medical University , Dalian , Liaoning Province , PR China
| | - Liang Zhu
- a College of Basic Medicine , Dalian Medical University , Dalian , Liaoning Province , PR China
| | - Yunhong Wu
- b College of Public Health , Dalian Medical University , Dalian , Liaoning Province , PR China
| | - Suping Liu
- a College of Basic Medicine , Dalian Medical University , Dalian , Liaoning Province , PR China
| | - Yufei Zhao
- a College of Basic Medicine , Dalian Medical University , Dalian , Liaoning Province , PR China
| | - Tonghui Ma
- a College of Basic Medicine , Dalian Medical University , Dalian , Liaoning Province , PR China
| |
Collapse
|
47
|
Giau VV, Wu SY, Jamerlan A, An SSA, Kim SY, Hulme J. Gut Microbiota and Their Neuroinflammatory Implications in Alzheimer's Disease. Nutrients 2018; 10:nu10111765. [PMID: 30441866 PMCID: PMC6266223 DOI: 10.3390/nu10111765] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Revised: 11/08/2018] [Accepted: 11/10/2018] [Indexed: 12/12/2022] Open
Abstract
The bidirectional communication between the central nervous system (CNS) and the gut microbiota plays a pivotal role in human health. Increasing numbers of studies suggest that the gut microbiota can influence the brain and behavior of patients. Various metabolites secreted by the gut microbiota can affect the cognitive ability of patients diagnosed with neurodegenerative diseases. Nearly one in every ten Korean senior citizens suffers from Alzheimer’s disease (AD), the most common form of dementia. This review highlights the impact of metabolites from the gut microbiota on communication pathways between the brain and gut, as well as the neuroinflammatory roles they may have in AD patients. The objectives of this review are as follows: (1) to examine the role of the intestinal microbiota in homeostatic communication between the gut microbiota and the brain, termed the microbiota–gut–brain (MGB) axis; (2) to determine the underlying mechanisms of signal dysfunction; and (3) to assess the impact of signal dysfunction induced by the microbiota on AD. This review will aid in understanding the microbiota of elderly people and the neuroinflammatory roles they may have in AD.
Collapse
Affiliation(s)
- Vo Van Giau
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Seongnam-si, Gyeonggi-do 461-701, Korea.
| | - Si Ying Wu
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Seongnam-si, Gyeonggi-do 461-701, Korea.
| | - Angelo Jamerlan
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Seongnam-si, Gyeonggi-do 461-701, Korea.
| | - Seong Soo A An
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Seongnam-si, Gyeonggi-do 461-701, Korea.
| | - Sang Yun Kim
- Department of Neurology, Seoul National University College of Medicine & Neurocognitive Behavior Center, Seoul National University Bundang Hospital, Seoul 100-011, Korea.
| | - John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Sungnam-daero, Seongnam-si, Gyeonggi-do 461-701, Korea.
| |
Collapse
|
48
|
Bourrat P. Have Causal Claims About the Gut Microbiome Been Over-Hyped? Bioessays 2018; 40:e1800178. [DOI: 10.1002/bies.201800178] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Indexed: 11/06/2022]
Affiliation(s)
- Pierrick Bourrat
- Department of Philosophy; Macquarie University; Building W6A North Ryde New South Wales Australia 2109
- Department of Philosophy; HPS School & Charles Perkins Centre; The University of Sydney; Camperdown New South Wales Australia 2006
| |
Collapse
|
49
|
De Wolfe TJ, Eggers S, Barker AK, Kates AE, Dill-McFarland KA, Suen G, Safdar N. Oral probiotic combination of Lactobacillus and Bifidobacterium alters the gastrointestinal microbiota during antibiotic treatment for Clostridium difficile infection. PLoS One 2018; 13:e0204253. [PMID: 30265691 PMCID: PMC6161886 DOI: 10.1371/journal.pone.0204253] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Accepted: 09/04/2018] [Indexed: 12/21/2022] Open
Abstract
Perturbations in the gastrointestinal microbiome caused by antibiotics are a major risk factor for Clostridium difficile infection (CDI). Probiotics are often recommended to mitigate CDI symptoms; however, there exists only limited evidence showing probiotic efficacy for CDI. Here, we examined changes to the GI microbiota in a study population where probiotic treatment was associated with significantly reduced duration of CDI diarrhea. Subjects being treated with standard of care antibiotics for a primary episode of CDI were randomized to probiotic treatment or placebo for 4 weeks. Probiotic treatment consisted of a daily multi-strain capsule (Lactobacillus acidophilus NCFM, ATCC 700396; Lactobacillus paracasei Lpc-37, ATCC SD5275; Bifidobacterium lactis Bi-07, ATCC SC5220; Bifidobacterium lactis B1-04, ATCC SD5219) containing 1.7 x 1010 CFUs. Stool was collected and analyzed using 16S rRNA sequencing. Microbiome analysis revealed apparent taxonomic differences between treatments and timepoints. Subjects administered probiotics had reduced Verrucomicrobiaceae at week 8 compared to controls. Bacteroides were significantly reduced between weeks 0 to 4 in probiotic treated subjects. Ruminococcus (family Lachnospiraceae), tended to be more abundant at week 8 than week 4 within the placebo group and at week 8 than week 0 within the probiotic group. Similar to these results, previous studies have associated these taxa with probiotic use and with mitigation of CDI symptoms. Compositional prediction of microbial community function revealed that subjects in the placebo group had microbiomes enriched with the iron complex transport system, while probiotic treated subjects had microbiomes enriched with the antibiotic transport system. Results indicate that probiotic use may impact the microbiome function in the face of a CDI; yet, more sensitive methods with higher resolution are warranted to better elucidate the roles associated with these changes. Continuing studies are needed to better understand probiotic effects on microbiome structure and function and the resulting impacts on CDI.
Collapse
Affiliation(s)
- T. J. De Wolfe
- Department of Biomedical Informatics, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Medicine, Division of Infectious Disease, University of Wisconsin – Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - S. Eggers
- Department of Population Health Sciences, University of Wisconsin – Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - A. K. Barker
- Department of Population Health Sciences, University of Wisconsin – Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
| | - A. E. Kates
- Department of Medicine, Division of Infectious Disease, University of Wisconsin – Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
| | - K. A. Dill-McFarland
- Department of Bacteriology, University of Wisconsin – Madison, Madison, Wisconsin, United States of America
| | - G. Suen
- Department of Bacteriology, University of Wisconsin – Madison, Madison, Wisconsin, United States of America
| | - N. Safdar
- Department of Medicine, Division of Infectious Disease, University of Wisconsin – Madison School of Medicine and Public Health, Madison, Wisconsin, United States of America
- William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin, United States of America
| |
Collapse
|
50
|
Liang S, Wu X, Jin F. Gut-Brain Psychology: Rethinking Psychology From the Microbiota-Gut-Brain Axis. Front Integr Neurosci 2018; 12:33. [PMID: 30271330 PMCID: PMC6142822 DOI: 10.3389/fnint.2018.00033] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 07/19/2018] [Indexed: 12/12/2022] Open
Abstract
Mental disorders and neurological diseases are becoming a rapidly increasing medical burden. Although extensive studies have been conducted, the progress in developing effective therapies for these diseases has still been slow. The current dilemma reminds us that the human being is a superorganism. Only when we take the human self and its partner microbiota into consideration at the same time, can we better understand these diseases. Over the last few centuries, the partner microbiota has experienced tremendous change, much more than human genes, because of the modern transformations in diet, lifestyle, medical care, and so on, parallel to the modern epidemiological transition. Existing research indicates that gut microbiota plays an important role in this transition. According to gut-brain psychology, the gut microbiota is a crucial part of the gut-brain network, and it communicates with the brain via the microbiota-gut-brain axis. The gut microbiota almost develops synchronously with the gut-brain, brain, and mind. The gut microbiota influences various normal mental processes and mental phenomena, and is involved in the pathophysiology of numerous mental and neurological diseases. Targeting the microbiota in therapy for these diseases is a promising approach that is supported by three theories: the gut microbiota hypothesis, the "old friend" hypothesis, and the leaky gut theory. The effects of gut microbiota on the brain and behavior are fulfilled by the microbiota-gut-brain axis, which is mainly composed of the nervous pathway, endocrine pathway, and immune pathway. Undoubtedly, gut-brain psychology will bring great enhancement to psychology, neuroscience, and psychiatry. Various microbiota-improving methods including fecal microbiota transplantation, probiotics, prebiotics, a healthy diet, and healthy lifestyle have shown the capability to promote the function of the gut-brain, microbiota-gut-brain axis, and brain. It will be possible to harness the gut microbiota to improve brain and mental health and prevent and treat related diseases in the future.
Collapse
Affiliation(s)
- Shan Liang
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| | - Xiaoli Wu
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Feng Jin
- Key Laboratory of Mental Health, Institute of Psychology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|