1
|
Kang JW, Vemuganti V, Kuehn JF, Ulland TK, Rey FE, Bendlin BB. Gut microbial metabolism in Alzheimer's disease and related dementias. Neurotherapeutics 2024; 21:e00470. [PMID: 39462700 DOI: 10.1016/j.neurot.2024.e00470] [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: 07/29/2024] [Revised: 09/30/2024] [Accepted: 10/04/2024] [Indexed: 10/29/2024] Open
Abstract
Multiple studies over the last decade have established that Alzheimer's disease and related dementias (ADRD) are associated with changes in the gut microbiome. These alterations in organismal composition result in changes in the abundances of functions encoded by the microbial community, including metabolic capabilities, which likely impact host disease mechanisms. Gut microbes access dietary components and other molecules made by the host and produce metabolites that can enter circulation and cross the blood-brain barrier (BBB). In recent years, several microbial metabolites have been associated with or have been shown to influence host pathways relevant to ADRD pathology. These include short chain fatty acids, secondary bile acids, tryptophan derivatives (such as kynurenine, serotonin, tryptamine, and indoles), and trimethylamine/trimethylamine N-oxide. Notably, some of these metabolites cross the BBB and can have various effects on the brain, including modulating the release of neurotransmitters and neuronal function, inducing oxidative stress and inflammation, and impacting synaptic function. Microbial metabolites can also impact the central nervous system through immune, enteroendocrine, and enteric nervous system pathways, these perturbations in turn impact the gut barrier function and peripheral immune responses, as well as the BBB integrity, neuronal homeostasis and neurogenesis, and glial cell maturation and activation. This review examines the evidence supporting the notion that ADRD is influenced by gut microbiota and its metabolites. The potential therapeutic advantages of microbial metabolites for preventing and treating ADRD are also discussed, highlighting their potential role in developing new treatments.
Collapse
Affiliation(s)
- Jea Woo Kang
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Vaibhav Vemuganti
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Jessamine F Kuehn
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Tyler K Ulland
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, WI, USA
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI, USA
| | - Barbara B Bendlin
- Wisconsin Alzheimer's Disease Research Center, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA; Wisconsin Alzheimer's Institute, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI, USA.
| |
Collapse
|
2
|
Acharya B, Tofthagen M, Maciej-Hulme ML, Suissa MR, Karlsson NG. Limited support for a direct connection between prebiotics and intestinal permeability - a systematic review. Glycoconj J 2024; 41:323-342. [PMID: 39287885 PMCID: PMC11522178 DOI: 10.1007/s10719-024-10165-8] [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: 02/07/2024] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024]
Abstract
The intestinal barrier is a selective interface between the body´s external and the internal environment. Its layer of epithelial cells is joined together by tight junction proteins. In intestinal permeability (IP), the barrier is compromised, leading to increased translocation of luminal contents such as large molecules, toxins and even microorganisms. Numerous diseases including Inflammatory Bowel Disease (IBD), Coeliac disease (CD), autoimmune disorders, and diabetes are believed to be associated with IP. Dietary interventions, such as prebiotics, may improve the intestinal barrier. Prebiotics are non-digestible food compounds, that promote the growth and activity of beneficial bacteria in the gut. This systematic review assesses the connection between prebiotic usage and IP. PubMed and Trip were used to identify relevant studies conducted between 2010-2023. Only six studies were found, which all varied in the characteristics of the population, study design, and types of prebiotics interventions. Only one study showed a statistically significant effect of prebiotics on IP. Alteration of intestinal barrier function was measured by lactulose/mannitol, chromium-labelled Ethylenediaminetetraacetic acid (51Cr-EDTA), lactulose/rhamnose, and sucralose/erythritol excretion as well as zonulin and glucagon-like peptide 2 levels. Three studies also conducted gut microbiota assessment, and one of them showed statistically significant improvement of the gut microbiome. This study also reported a decrease in zonulin level. The main conclusion from this review is that there is a lack of human studies in this important field. Futhermore, large population studies and using standardized protocols, would be required to properly assess the impact of prebiotic intervention and improvement on IP.
Collapse
Affiliation(s)
- Binayak Acharya
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Marthe Tofthagen
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Marissa L Maciej-Hulme
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Michal Rachel Suissa
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway
| | - Niclas G Karlsson
- Department of Life Sciences and Health, Faculty of Health Sciences, Oslo Metropolitan University, St. Olavs Plass, P.O. Box 4, N-0130, Oslo, Norway.
| |
Collapse
|
3
|
Li Y, Xue J, Zhang Z, Wang W, Wang Y, Zhang W. Alteration of gut microbiota in Henoch-Schönlein purpura children with gastrointestinal involvement. Ir J Med Sci 2024; 193:2397-2406. [PMID: 38967706 DOI: 10.1007/s11845-024-03750-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Accepted: 06/29/2024] [Indexed: 07/06/2024]
Abstract
BACKGROUND The compositional and structural changes of gut microbiota were closely related to the status of Henoch-Schönlein purpura (HSP). AIMS To investigate if clinical indicators and gut microbiota differ between HSP patients with or without gastrointestinal (GI) involvement and to explore the alterations of fecal microbiota in HSP children with and without GI symptoms. METHODS A total of 22 children with HSP were enrolled in the study. Fecal microbiota composition was analyzed by 16S rRNA sequencing. Clinical indicators, fecal microbial diversity, and compositions were compared between the two groups. RESULTS Respectively, 9 patients with GI involvement (HSP-A) and 13 patients without GI involvement (HSP-N) were enrolled. Prealbumin (PA) and the ratio of immunoglobulin A (IgA) / complement (C)3 were significantly decreased in the HSP-A group and an elevated D-dimer was found in the HSP-N group. The relative abundances of Blautia, Lachnospira, and Haemophilus were significantly higher in the HSP-A group compared to HSP-N. Lower levels of unidentified Prevotellaceae, Parabacteroides, and Romboutsia were found in HSP-A patients. The linear discriminant analysis effect size (LEfSe) showed that the biomarkers for the HSP-A group included Blautia, Anaerostipes, Veillonella, Lachnospira, and Haemophilus. For the HSP-N group, unidentified Prevotellaceae, Intestinibacter, Romboutsia, and Akkermansia were the prominent biomarkers at the genus level. Additionally, the ratio of IgA/C3 exhibited a negative correlation with the genus Blautia. Meanwhile, PA showed negatively correlation with Veillonella. CONCLUSIONS These results provide a broader understanding for future microbial-based therapies to decrease the development of GI involvement and improve the clinical outcome of HSP in children.
Collapse
Affiliation(s)
- Ye Li
- Department of Pediatrics, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Avenue, Jinan, 250012, Shandong, China
| | - Jiang Xue
- Department of Pediatrics, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Avenue, Jinan, 250012, Shandong, China
| | - Zhaohua Zhang
- Department of Pediatrics, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Avenue, Jinan, 250012, Shandong, China
| | - Wei Wang
- Department of Respiratory Medicine, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China
| | - Yulong Wang
- Department of Pediatrics, The Second Hospital, Cheeloo College of Medicine, Shandong University, 247 Beiyuan Avenue, Jinan, 250012, Shandong, China.
| | - Weiquan Zhang
- Department of Thoracic Surgery, The Second Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
4
|
Lo SW, Hung TH, Lin YT, Lee CS, Chen CY, Fang CJ, Lai PC. Clinical efficacy and safety of faecal microbiota transplantation in the treatment of irritable bowel syndrome: a systematic review, meta-analysis and trial sequential analysis. Eur J Med Res 2024; 29:464. [PMID: 39289768 PMCID: PMC11409544 DOI: 10.1186/s40001-024-02046-5] [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: 02/19/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND The aim of this study is to evaluate the efficacy and safety of faecal microbiota transplantation (FMT) for the treatment of irritable bowel syndrome (IBS). METHODS We searched four databases for randomised controlled trials (RCTs) that compared FMT with a control intervention in patients with IBS. The revised Cochrane risk-of-bias (RoB) tool was chosen for appraisal. Meta-analysis with trial sequential analysis (TSA) was conducted. Grading of Recommendations Assessment Development and Evaluation (GRADE) methodology was used to assess the certainty of evidence (CoE). RESULTS We included 12 RCTs with a total of 615 participants. Meta-analyses showed no significant difference between the FMT and control groups in terms of clinical responses (relative risk [RR] = 1.44, 95% confidence interval [CI] 0.88-2.33) and changes in IBS Severity Scoring System (IBS-SSS) scores (standardised mean difference [SMD] = - 0.31, 95% CI - 0.72 to 0.09) and IBS Quality of Life (IBS-QOL) scores (SMD = 0.30, 95% CI - 0.09 to 0.69). Subgroup analysis revealed that in studies with low RoB and using endoscopy, nasojejunal tube and rectal enema delivery, FMT led to a significant improvement in clinical responses and changes in IBS-SSS and IBS-QOL scores. TSA suggested that the current evidence is inconclusive and that the CoE is very low. CONCLUSION This study suggests that patients with IBS may benefit from FMT especially when it is administered via endoscopy, nasojejunal tube or rectal enema. However, the certainty of evidence is very low. Further research is needed to confirm the efficacy and safety of FMT for IBS treatment. TRIAL REGISTRATION PROSPERO registration number CRD42020211002.
Collapse
Affiliation(s)
- Shao-Wei Lo
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan
| | - Tsung-Hsuan Hung
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan
| | - Yen-Tsen Lin
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan
| | - Chun-Shen Lee
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan
| | - Chiung-Yu Chen
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Ching-Ju Fang
- Medical Library, National Cheng Kung University, Tainan, Taiwan
- Department of Secretariat, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan
| | - Pei-Chun Lai
- Education Centre, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, No.138, Sheng Li Road, Tainan, 704, Taiwan.
- Department of Paediatrics, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
5
|
Kanika NH, Hou X, Liu H, Dong Y, Wang J, Wang C. Specific gut microbiome's role in skin pigmentation: insights from SCARB1 mutants in Oujiang colour common carp. J Appl Microbiol 2024; 135:lxae226. [PMID: 39243120 DOI: 10.1093/jambio/lxae226] [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: 12/06/2023] [Revised: 08/13/2024] [Accepted: 09/05/2024] [Indexed: 09/09/2024]
Abstract
AIMS Beyond the pivotal roles of the gut microbiome in initiating physiological processes and modulating genetic factors, a query persists: Can a single gene mutation alter the abundance of the gut microbiome community? Not only this, but the intricate impact of gut microbiome composition on skin pigmentation has been largely unexplored. METHODS AND RESULTS Based on these premises, our study examines the abundance of lipase-producing gut microbes about differential gene expression associated with bile acid synthesis and lipid metabolism-related blood metabolites in red (whole wild) and white (whole white wild and SCARB1-/- mutant) Oujiang colour common carp. Following the disruption of the SCARB1 gene in the resulting mutant fish with white body colour (SCARB1-/-), there is a notable decrease in the abundance of gut microbiomes (Bacillus, Staphylococcus, Pseudomonas, and Serratia) associated with lipase production. This reduction parallels the downregulation seen in wild-type white body colour fish (WW), as contrasting to the wild-type red body colour fish (WR). Meanwhile, in SCARB1-/- fish, there was a downregulation noted not only at the genetic and metabolic levels but also a decrease in lipase-producing bacteria. This consistency with WW contrasts significantly with WR. Similarly, genes involved in the bile acid synthesis pathway, along with blood metabolites related to lipid metabolism, exhibited downregulation in SCARB1-/- fish. CONCLUSIONS The SCARB1 knockout gene blockage led to significant alterations in the gut microbiome, potentially influencing the observed reduction in carotenoid-associated skin pigmentation. Our study emphasizes that skin pigmentation is not only impacted by genetic factors but also by the gut microbiome. Meanwhile, the gut microbiome's adaptability can be rapidly shaped and may be driven by specific single-gene variations.
Collapse
Affiliation(s)
- Nusrat Hasan Kanika
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Xin Hou
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Hao Liu
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Yue Dong
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Jun Wang
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| | - Chenghui Wang
- Key Laboratory of Freshwater Aquatic Genetic Resources Certificated by the Ministry of Agriculture and Rural Affairs, National Demonstration Centre for Experimental Fisheries Science Education, Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai 201306, China
| |
Collapse
|
6
|
Scheithauer TPM, Montijn RC, Mieremet A. Gut microbe-host interactions in post-COVID syndrome: a debilitating or restorative partnership? Gut Microbes 2024; 16:2402544. [PMID: 39287023 PMCID: PMC11409505 DOI: 10.1080/19490976.2024.2402544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/14/2024] [Accepted: 09/04/2024] [Indexed: 09/19/2024] Open
Abstract
Post-COVID syndrome (PCS) patients have reported a wide range of symptoms, including fatigue, shortness of breath, and diarrhea. Particularly, the presence of gastrointestinal symptoms has led to the hypothesis that the gut microbiome is involved in the development and severity of PCS. The objective of this review is to provide an overview of the role of the gut microbiome in PCS by describing the microbial composition and microbial metabolites in COVID-19 and PCS. Moreover, host-microbe interactions via the microbiota-gut-brain (MGB) and the microbiota-gut-lung (MGL) axes are described. Furthermore, we explore the potential of therapeutically targeting the gut microbiome to support the recovery of PCS by reviewing preclinical model systems and clinical studies. Overall, current studies provide evidence that the gut microbiota is affected in PCS; however, diversity in symptoms and highly individual microbiota compositions suggest the need for personalized medicine. Gut-targeted therapies, including treatments with pre- and probiotics, have the potential to improve the quality of life of affected individuals.
Collapse
Affiliation(s)
- Torsten P M Scheithauer
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Roy C Montijn
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, The Netherlands
| | - Arnout Mieremet
- Department of Microbiology and Systems Biology, Netherlands Organisation for Applied Scientific Research (TNO), Leiden, The Netherlands
| |
Collapse
|
7
|
Cho NA, Strayer K, Dobson B, McDonald B. Pathogenesis and therapeutic opportunities of gut microbiome dysbiosis in critical illness. Gut Microbes 2024; 16:2351478. [PMID: 38780485 PMCID: PMC11123462 DOI: 10.1080/19490976.2024.2351478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
For many years, it has been hypothesized that pathological changes to the gut microbiome in critical illness is a driver of infections, organ dysfunction, and other adverse outcomes in the intensive care unit (ICU). The advent of contemporary microbiome methodologies and multi-omics tools have allowed researchers to test this hypothesis by dissecting host-microbe interactions in the gut to better define its contribution to critical illness pathogenesis. Observational studies of patients in ICUs have revealed that gut microbial communities are profoundly altered in critical illness, characterized by markedly reduced alpha diversity, loss of commensal taxa, and expansion of potential pathogens. These key features of ICU gut dysbiosis have been associated with adverse outcomes including life-threatening hospital-acquired (nosocomial) infections. Current research strives to define cellular and molecular mechanisms connecting gut dysbiosis with infections and other outcomes, and to identify opportunities for therapeutic modulation of host-microbe interactions. This review synthesizes evidence from studies of critically ill patients that have informed our understanding of intestinal dysbiosis in the ICU, mechanisms linking dysbiosis to infections and other adverse outcomes, as well as clinical trials of microbiota-modifying therapies. Additionally, we discuss novel avenues for precision microbial therapeutics to combat nosocomial infections and other life-threatening complications of critical illness.
Collapse
Affiliation(s)
- Nicole A Cho
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Kathryn Strayer
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Breenna Dobson
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Braedon McDonald
- Department of Critical Care Medicine, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
8
|
Buttar J, Kon E, Lee A, Kaur G, Lunken G. Effect of diet on the gut mycobiome and potential implications in inflammatory bowel disease. Gut Microbes 2024; 16:2399360. [PMID: 39287010 PMCID: PMC11409510 DOI: 10.1080/19490976.2024.2399360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Revised: 07/31/2024] [Accepted: 08/28/2024] [Indexed: 09/19/2024] Open
Abstract
The gut microbiome is a complex, unique entity implicated in the prevention, pathogenesis, and progression of common gastrointestinal diseases. While largely dominated by bacterial populations, advanced sequencing techniques have identified co-inhabiting fungal communities, collectively referred to as the mycobiome. Early studies identified that gut inflammation is associated with altered microbial composition, known as gut dysbiosis. Altered microbial profiles are implicated in various pathological diseases, such as inflammatory bowel disease (IBD), though their role as a cause or consequence of systemic inflammation remains the subject of ongoing research. Diet plays a crucial role in the prevention and management of various diseases and is considered to be an essential regulator of systemic inflammation. This review compiles current literature on the impact of dietary modulation on the mycobiome, showing that dietary changes can alter the fungal architecture of the gut. Further research is required to understand the impact of diet on gut fungi, including the metabolic pathways and enzymes involved in fungal fermentation. Additionally, investigating whether dietary modulation of the gut mycobiome could be utilized as a therapy in IBD is essential.
Collapse
Affiliation(s)
- J Buttar
- Department of Medicine, University of British Columbia, Vancouver, Canada
| | - E Kon
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
| | - A Lee
- Faculty of Land and Food Systems, University of British Columbia, Vancouver, Canada
| | - G Kaur
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
| | - G Lunken
- Department of Medicine, University of British Columbia, Vancouver, Canada
- Department of Pediatrics, University of British Columbia, Vancouver, Canada
- BC Children's Hospital Research Institute, Vancouver, Canada
| |
Collapse
|
9
|
Ciccone MM, Lepera ME, Guaricci AI, Forleo C, Cafiero C, Colella M, Palmirotta R, Santacroce L. Might Gut Microbiota Be a Target for a Personalized Therapeutic Approach in Patients Affected by Atherosclerosis Disease? J Pers Med 2023; 13:1360. [PMID: 37763128 PMCID: PMC10532785 DOI: 10.3390/jpm13091360] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/01/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
In recent years, the increasing number of studies on the relationship between the gut microbiota and atherosclerosis have led to significant interest in this subject. The gut microbiota, its metabolites (metabolome), such as TMAO, and gut dysbiosis play an important role in the development of atherosclerosis. Furthermore, inflammation, originating from the intestinal tract, adds yet another mechanism by which the human ecosystem is disrupted, resulting in the manifestation of metabolic diseases and, by extension, cardiovascular diseases. The scientific community must understand and elucidate these mechanisms in depth, to gain a better understanding of the relationship between atherosclerosis and the gut microbiome and to promote the development of new therapeutic targets in the coming years. This review aims to present the knowledge acquired so far, to trigger others to further investigate this intriguing topic.
Collapse
Affiliation(s)
- Marco Matteo Ciccone
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Mario Erminio Lepera
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Andrea Igoren Guaricci
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Cinzia Forleo
- Cardiology Unit, Interdisciplinary Department of Medicine, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.M.C.); (M.E.L.); (A.I.G.); (C.F.)
| | - Concetta Cafiero
- Area of Molecular Pathology, Anatomic Pathology Unit, Fabrizio Spaziani Hospital, 03100 Frosinone, Italy;
| | - Marica Colella
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.C.); (L.S.)
| | - Raffele Palmirotta
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.C.); (L.S.)
| | - Luigi Santacroce
- Interdisciplinary Department of Medicine, Section of Microbiology and Virology, School of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (M.C.); (L.S.)
| |
Collapse
|
10
|
da Silva Soares NF, Quagliariello A, Yigitturk S, Martino ME. Gut microbes predominantly act as living beneficial partners rather than raw nutrients. Sci Rep 2023; 13:11981. [PMID: 37488173 PMCID: PMC10366161 DOI: 10.1038/s41598-023-38669-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 07/12/2023] [Indexed: 07/26/2023] Open
Abstract
Animals and their gut microbes mutually benefit their health. Nutrition plays a central role in this, directly influencing both host and microbial fitness and the nature of their interactions. This makes nutritional symbioses a complex and dynamic tri-system of diet-microbiota-host. Despite recent discoveries on this field, full control over the interplay among these partners is challenging and hinders the resolution of fundamental questions, such as how to parse the gut microbes' effect as raw nutrition or as symbiotic partners? To tackle this, we made use of the well-characterized Drosophila melanogaster/Lactiplantibacillus plantarum experimental model of nutritional symbiosis to generate a quantitative framework of gut microbes' effect on the host. By coupling experimental assays and Random Forest analysis, we show that the beneficial effect of L. plantarum strains primarily results from the active relationship as symbionts rather than raw nutrients, regardless of the bacterial strain. Metabolomic analysis of both active and inactive bacterial cells further demonstrated the crucial role of the production of beneficial bacterial metabolites, such as N-acetylated-amino-acids, as result of active bacterial growth and function. Altogether, our results provide a ranking and quantification of the main bacterial features contributing to sustain animal growth. We demonstrate that bacterial activity is the predominant and necessary variable involved in bacteria-mediated benefit, followed by strain-specific properties and the nutritional potential of the bacterial cells. This contributes to elucidate the role of beneficial bacteria and probiotics, creating a broad quantitative framework for host-gut microbiome that can be expanded to other model systems.
Collapse
Affiliation(s)
| | - Andrea Quagliariello
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
| | - Seren Yigitturk
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy
- Food Quality and Design Group, Wageningen University and Research, Wageningen, The Netherlands
| | - Maria Elena Martino
- Department of Comparative Biomedicine and Food Science, University of Padova, Padova, Italy.
| |
Collapse
|
11
|
Zhang MM, Gong ZC, Zhao Q, Xu DQ, Fu RJ, Tang YP, Chen YY. Time-dependent laxative effect of sennoside A, the core functional component of rhubarb, is attributed to gut microbiota and aquaporins. JOURNAL OF ETHNOPHARMACOLOGY 2023; 311:116431. [PMID: 37003403 DOI: 10.1016/j.jep.2023.116431] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2023] [Revised: 03/09/2023] [Accepted: 03/22/2023] [Indexed: 06/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sennoside A is a natural anthraquinone component mainly derived from rhubarb and has been routinely used as a clinical stimulant laxative. However, long-term application of sennoside A may lead to drug resistance and even adverse reactions, thus limiting its clinical use. Therefore, to reveal the time-dependent laxative effect and potential mechanism of sennoside A is of critical importance. AIM OF THE STUDY This study was conducted to investigate the time-dependent laxative effect of sennoside A and unveil its underlying mechanism from the perspective of gut microbiota and aquaporins (AQPs). MATERIALS AND METHODS Based on a mouse constipation model, 2.6 mg/kg sennoside A was administered orally for 1, 3, 7, 14 and 21 days, respectively. The laxative effect was assessed by the fecal index and fecal water content, the histopathology of the small intestine and colon was evaluated by hematoxylin-eosin staining. Gut microbiota changes was observed by 16S rDNA sequencing, and colonic AQPs expression was analyzed by quantitative real-time polymerase chain reaction and western blotting. Partial least-squares regression (PLSR) was used to screen out the effective indicators contributing to the laxative effect of sennoside A. The effective indicators were then fitted to time by a drug-time curve model to analyze the trend of efficacy of sennoside A, and the optimal time of administration was derived by comprehensive analysis with a three-dimensional (3D) time-effect image. RESULTS Sennoside A had a significant laxative effect at 7 days of administration with no pathological changes in the small intestine or colon; however, at 14 or 21 days of administration, the laxative effect diminished and slight damage to the colon was observed. Sennoside A affects the structure and function of gut microbes. The alpha diversity showed that the abundance and diversity of gut microorganisms reached the highest value after 7 days of administration. Partial least squares discriminant analysis showed that the composition of the flora was close to normal when administered for less than 7 days, but was closest to the composition of constipation over 7 days. The expression of aquaporin 3 (AQP3) and aquaporin 7 (AQP7) decreased gradually after the administration of sennoside A, with the lowest expression at 7 days, and then increased gradually afterwards, while the expression of aquaporin 1 (AQP1) was the opposite. The PLSR results showed that AQP1, AQP3, Lactobacillus, Romboutsia, Akkermansia and UCG_005 contributed more to the laxative effect of the fecal index, and after fitting with the drug-time curve model, each index showed a trend of increasing and then decreasing. The comprehensive evaluation of the 3D time-effect image concluded that the laxative effect of sennoside A reached its best after 7 days of administration. CONCLUSION Sennoside A should be used in regular dosages for less than one week, as it provides significant relief of constipation and exhibits no colonic damage within 7 days of administration. In addition, Sennoside A exerts its laxative effect by regulating gut microbiota of Lactobacillus Romboutsia, Akkermansia and UCG_005 and water channels of AQP1 and AQP3.
Collapse
Affiliation(s)
- Mei-Mei Zhang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Zhi-Cheng Gong
- Wuxi Institute of Chinese and Western Integrative Medicine, and Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu Province, China
| | - Qi Zhao
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Ding-Qiao Xu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Rui-Jia Fu
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Yu-Ping Tang
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China
| | - Yan-Yan Chen
- Key Laboratory of Shaanxi Administration of Traditional Chinese Medicine for TCM Compatibility, Shaanxi Key Laboratory of New Drugs and Chinese Medicine Foundation Research, State Key Laboratory of Research & Development of Characteristic Qin Medicine Resources (Cultivation), Shaanxi Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Shaanxi University of Chinese Medicine, Xi'an, 712046, Shaanxi Province, China; Wuxi Institute of Chinese and Western Integrative Medicine, and Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, Wuxi, 214062, Jiangsu Province, China.
| |
Collapse
|
12
|
Colonetti T, Saggioratto MC, Grande AJ, Colonetti L, Junior JCD, Ceretta LB, Roever L, Silva FR, da Rosa MI. Gut and Vaginal Microbiota in the Endometriosis: Systematic Review and Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2023; 2023:2675966. [PMID: 38601772 PMCID: PMC11006450 DOI: 10.1155/2023/2675966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 03/21/2023] [Accepted: 05/05/2023] [Indexed: 04/12/2024]
Abstract
Background Endometriosis is a clinical condition associated with genetic, endocrine, and immunological factors, present in 6 to 10% of women of reproductive age. Currently, the human microbiota has been studied and associated with the evolution of diseases due to its influence on pathogenesis, indicating that changes in the colonization of microorganisms in the genitourinary and gastrointestinal systems can promote physiological changes that can trigger inflammatory and immunological processes and hormonal dysregulation, which can be linked to endometriosis. Thus, this systematic review and meta-analysis evaluated microbiota changes in women with endometriosis. Methods The following electronic databases were searched up to April 2022: Medline, Embase, Web of Science, Cochrane Library, and gray literature (Google Scholar), using the keywords "dysbiosis", "microbiome" and "endometriosis", combined with their synonyms. The observational studies conducted with women diagnosed with endometriosis and women without endometriosis as controls were included. For the analyses, a standard mean difference with a 95% confidence interval was used using RevMan software (version 5.4), and for methodological quality assessment, the Newcastle-Ottawa scale was used. Results A total of 16 studies were found in the literature assessing the composition of the microbiota in women with endometriosis, and no significant difference were found for changes in alpha diversity analysis in gut microbiota (SMD = -0.28; 95% CI = -0.70 to 0.14; P = 0.19; I2 = 52%; four studies, 357 participants) or vaginal microbiota (SMD = -0.68; 95% CI = -1.72 to 0.35; P = 0.19; I2 = 66%; two studies, 49 participants). Conclusion In intestinal and vaginal samples from women with endometriosis, alpha-diversity did not present a significant difference when compared to the control population. However, each study individually showed a possible relationship between the female microbiota and endometriosis. This trial is registered with CRD42021260972.
Collapse
Affiliation(s)
- Tamy Colonetti
- Laboratory of Biomedicine Translational, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105-Bairro Universitário CEP, 88806-000 Criciúma, SC, Brazil
| | - Maria Carolina Saggioratto
- Laboratory of Biomedicine Translational, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105-Bairro Universitário CEP, 88806-000 Criciúma, SC, Brazil
| | - Antonio José Grande
- Laboratory of Evidence-Based Practice, Universidade Estadual de Mato Grosso do Sul (UEMS), Campo Grande, Mato Grosso do Sul, Brazil
| | - Laura Colonetti
- Laboratory of Biomedicine Translational, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105-Bairro Universitário CEP, 88806-000 Criciúma, SC, Brazil
| | - João Carlos Denoni Junior
- Laboratory of Biomedicine Translational, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105-Bairro Universitário CEP, 88806-000 Criciúma, SC, Brazil
| | - Luciane Bisognin Ceretta
- Postgraduate Program in Collective Health, Universidade do Extremo Sul Catarinense (UNESC), Criciúma, Santa Catarina, Brazil
| | - Leonardo Roever
- Department of Clinical Research, Federal University of Uberlândia, Uberlândia, Brazil
| | - Fábio Rosa Silva
- Laboratory of Biomedicine Translational, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105-Bairro Universitário CEP, 88806-000 Criciúma, SC, Brazil
| | - Maria Inês da Rosa
- Laboratory of Biomedicine Translational, Universidade do Extremo Sul Catarinense, Av. Universitária, 1105-Bairro Universitário CEP, 88806-000 Criciúma, SC, Brazil
| |
Collapse
|
13
|
Musazadeh V, Mohammadi Anilou M, Vajdi M, Karimi A, Sedgh Ahrabi S, Dehghan P. Effects of synbiotics supplementation on anthropometric and lipid profile parameters: Finding from an umbrella meta-analysis. Front Nutr 2023; 10:1121541. [PMID: 36908920 PMCID: PMC9995782 DOI: 10.3389/fnut.2023.1121541] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/06/2023] [Indexed: 02/25/2023] Open
Abstract
Introduction Several systematic reviews and meta-analyses have been carried out to assess the impact of synbiotics on lipid profiles and anthropometric parameters. In this regard, an umbrella meta-analysis was performed to provide a more accurate view of the overall impacts of synbiotic supplementation on lipid profile and anthropometric parameters. Methods Databases such as PubMed, Scopus, Embase, Web of Science, and Google Scholar were searched for this study from inception to January 2022. A random-effects model was applied to evaluate the effects of synbiotic supplementation on lipid profile and anthropometric parameters. The methodological quality of eligible articles was evaluated using the AMSTAR2 questionnaire. The GRADE approach was used to evaluate the overall certainty of the evidence in the meta-analyses. Results Meta-analyses of 17 studies revealed significant decreases in body mass index (BMI) (ES: -0.13 kg/m2; 95% CI: -0.19, -0.06, p < 0.001, I2 = 0.0%, p = 0.870), BW (ES: -1.30 kg; 95% CI: -2.19, -0.41, p = 0.004, I2 = 88.9%, p < 0.001), waist circumference (WC) (ES: -1.80 cm; 95% CI: -3.26, -0.34, p = 0.016, I2 = 94.1%, p < 0.001), low-density lipoprotein cholesterol (LDL-C) (ES: -2.81 mg/dl; 95% CI: -3.90, -1.72, p < 0.001, I2 = 95.1%, p < 0.001), total cholesterol (TC) (ES = -2.24 mg/dl; 95% CI: -3.18, -1.30, p < 0.001, I2 = 94.5%, p < 0.001), and triglyceride (TG) (ES: -0.43 mg/dl; 95% CI: -0.79, -0.07, p = 0.019, I2 = 78.0%, p < 0.001) but not high-density lipoprotein cholesterol (HDL-C) (ES: 0.23 mg/dl; 95% CI: -0.11, 0.56, p = 0.193, I2 = 45.2%, p = 0.051) following synbiotic supplementation. Discussion The present umbrella meta-analysis suggests synbiotic supplementation can slightly improve lipid profile and anthropometric indices and might be a therapeutic option for obesity and its related disorders. Systematic review registration www.crd.york.ac.uk/prospero, identifier CRD42022304376.
Collapse
Affiliation(s)
- Vali Musazadeh
- Student Research Committee, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Maryam Mohammadi Anilou
- Department of Emergency Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mahdi Vajdi
- Student Research Committee, Isfahan University of Medical Sciences, Isfahan, Iran
- Department of Community Nutrition, School of Nutrition and Food Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Arash Karimi
- Nutrition Research Center, Department of Clinical Nutrition, School of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sana Sedgh Ahrabi
- Student Research Committee, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parvin Dehghan
- Nutrition Research Center, Faculty of Nutrition and Food Science, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
14
|
Naseri K, Dabiri H, Olfatifar M, Shahrbaf MA, Yadegar A, Soheilian-Khorzoghi M, Sadeghi A, Saadati S, Rostami-Nejad M, Verma AK, Zali MR. Evaluation of gut microbiota of iranian patients with celiac disease, non-celiac wheat sensitivity, and irritable bowel syndrome: are there any similarities? BMC Gastroenterol 2023; 23:15. [PMID: 36647022 PMCID: PMC9841652 DOI: 10.1186/s12876-023-02649-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 01/11/2023] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND AND AIMS Individuals with celiac disease (CD), non-celiac wheat sensitivity (NCWS), and irritable bowel syndrome (IBS), show overlapping clinical symptoms and experience gut dysbiosis. A limited number of studies so far compared the gut microbiota among these intestinal conditions. This study aimed to investigate the similarities in the gut microbiota among patients with CD, NCWS, and IBS in comparison to healthy controls (HC). MATERIALS AND METHODS In this prospective study, in total 72 adult subjects, including CD (n = 15), NCWS (n = 12), IBS (n = 30), and HC (n = 15) were recruited. Fecal samples were collected from each individual. A quantitative real-time PCR (qPCR) test using 16S ribosomal RNA was conducted on stool samples to assess the relative abundance of Firmicutes, Bacteroidetes, Bifidobacterium spp., and Lactobacillus spp. RESULTS In all groups, Firmicutes and Lactobacillus spp. had the highest and lowest relative abundance respectively. The phylum Firmicutes had a higher relative abundance in CD patients than other groups. On the other hand, the phylum Bacteroidetes had the highest relative abundance among healthy subjects but the lowest in patients with NCWS. The relative abundance of Bifidobacterium spp. was lower in subjects with CD (P = 0.035) and IBS (P = 0.001) compared to the HCs. Also, the alteration of Firmicutes to Bacteroidetes ratio (F/B ratio) was statistically significant in NCWS and CD patients compared to the HCs (P = 0.05). CONCLUSION The principal coordinate analysis (PCoA), as a powerful multivariate analysis, suggested that the investigated gut microbial profile of patients with IBS and NCWS share more similarities to the HCs. In contrast, patients with CD had the most dissimilarity compared to the other groups in the context of the studied gut microbiota.
Collapse
Affiliation(s)
- Kaveh Naseri
- grid.1017.70000 0001 2163 3550School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC Australia
| | - Hossein Dabiri
- grid.411600.2Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Meysam Olfatifar
- grid.444830.f0000 0004 0384 871XGastroenterology and Hepatology Diseases Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Mohammad Amin Shahrbaf
- grid.411600.2Celiac Disease Department, Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Abbas Yadegar
- grid.411600.2Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Soheilian-Khorzoghi
- grid.411600.2Celiac Disease Department, Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amir Sadeghi
- grid.411600.2Celiac Disease Department, Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeede Saadati
- grid.411600.2Celiac Disease Department, Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Rostami-Nejad
- grid.411600.2Celiac Disease Department, Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Anil K. Verma
- grid.7010.60000 0001 1017 3210Celiac Disease Research Laboratory, Department of Pediatrics, Università Politecnica Delle Marche, 60123 Ancona, Italy
| | - Mohammad Reza Zali
- grid.411600.2Celiac Disease Department, Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Xie S, Zhang R, Li Z, Liu C, Chen Y, Yu Q. Microplastics perturb colonic epithelial homeostasis associated with intestinal overproliferation, exacerbating the severity of colitis. ENVIRONMENTAL RESEARCH 2023; 217:114861. [PMID: 36410465 DOI: 10.1016/j.envres.2022.114861] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 11/10/2022] [Accepted: 11/18/2022] [Indexed: 06/16/2023]
Abstract
A great amount of the population died due to living or working in an unhealthy environment, highlighting the critical role of environmental pollutants in inducing diseases. Microplastics are widespread environmental pollutants and have been found in various tissues of human beings, yet the risk of microplastics in the occurrence of disease, especially environmentally-related colitis, is unclear. This study focused on the effects of microplastics exposure on intestinal homeostasis and the initiation of colitis. We noticed that microplastics exposure had a limited impact on mice, as verified by no difference observed in bodyweight change, IL-1β and IL-6 levels in jejunum and liver. Nevertheless, in the colon, the IL-1β and IL-6 levels were slightly increased and the goblet cell number was decreased. Interestingly, we observed that crypt number and depth, the levels of intestinal stem cell markers, combined with the expression of proliferating cell nuclear antigen and proto-oncogene c-Myc were all significantly increased with microplastics treatment, indicating the overproliferation of colonic mucosa. The effect of microplastics on proliferation and differentiation of crypt was further demonstrated to be regulated by the overactivation of the Notch signaling pathway in intestinal organoids. Furthermore, microplastics exposure accelerated the development of colitis with severe bodyweight loss, diarrhea and bloody stools, macroscopic and pathological damage, and inflammation levels. Worsened liver pathological damage and inflammation in mice with colitis under microplastics exposure also were found. These results suggested that microplastics disrupted the balance between colonic epithelium self-renewal and differentiation, exacerbating the colitis, and might be an environmental-related disease risk factor.
Collapse
Affiliation(s)
- Shuang Xie
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Rui Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Zhaoyan Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Chunru Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China
| | - Yanyu Chen
- Laboratory of Microbiology, Immunology and Metabolism, Diprobio (Shanghai) Co., Limited, Shanghai, 200335, PR China
| | - Qinghua Yu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, Jiangsu, 210095, PR China; Laboratory of Microbiology, Immunology and Metabolism, Diprobio (Shanghai) Co., Limited, Shanghai, 200335, PR China.
| |
Collapse
|
16
|
Wang T, Yu R, Zhu L, Wang X, Yang B. Differences in the Intestinal Flora of Patients with Inflammatory Bowel Disease in Southwest China. Indian J Microbiol 2022; 62:384-392. [PMID: 35974916 PMCID: PMC9375786 DOI: 10.1007/s12088-022-01014-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/05/2022] [Indexed: 11/27/2022] Open
Abstract
To study changes in the intestinal flora associated with inflammatory bowel disease (IBD) in the Han population of southwest China, 48 participants were enrolled, 18 of whom had been diagnosed with IBD. Stool samples were collected from the participants. Sequencing of 16S rRNA gene was used to measure and identify the components of the intestinal flora. Diversity analysis and multivariate statistical analysis were conducted to study differences in intestinal flora between patients with IBD and healthy controls. The goods coverage, observed species, Shannon, and Simpson indices of alpha diversity were different (p < 0.05). Beta diversity analysis yielded significant differences between groups (R = 0.5668, p = 0.001 < 0.05). Compared with the composition of the intestinal flora in healthy controls, the relative abundances of Proteobacteria (18.56% vs. 3.56%, p = 0.001) and Fusobacterium (2.08% vs. 0.35%, p = 0.005) were higher in patients with IBD. Therefore, this study provides insight into the role of the microbiome in IBD. Supplementary Information The online version contains supplementary material available at 10.1007/s12088-022-01014-z.
Collapse
Affiliation(s)
- Tingting Wang
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Renlin Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lingling Zhu
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Xuean Wang
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Bin Yang
- Department of Laboratory Medicine, West China Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Gut Microbiota Manipulation in Irritable Bowel Syndrome. Microorganisms 2022; 10:microorganisms10071332. [PMID: 35889051 PMCID: PMC9319495 DOI: 10.3390/microorganisms10071332] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 06/24/2022] [Accepted: 06/29/2022] [Indexed: 02/05/2023] Open
Abstract
Increased knowledge suggests that disturbed gut microbiota, termed dysbiosis, might promote the development of irritable bowel syndrome (IBS) symptoms. Accordingly, gut microbiota manipulation has evolved in the last decade as a novel treatment strategy in order to improve IBS symptoms. In using different approaches, dietary management stands first in line, including dietary fiber supplements, prebiotics, and probiotics that are shown to change the composition of gut microbiota, fecal short-chain fatty acids and enteroendocrine cells densities and improve IBS symptoms. However, the exact mixture of beneficial bacteria for each individual remains to be identified. Prescribing nonabsorbable antibiotics still needs confirmation, although using rifaximin has been approved for diarrhea-predominant IBS. Fecal microbiota transplantation (FMT) has recently gained a lot of attention, and five out of seven placebo-controlled trials investigating FMT in IBS obtain promising results regarding symptom reduction and gut microbiota manipulation. However, more data, including larger cohorts and studying long-term effects, are needed before FMT can be regarded as a treatment for IBS in clinical practice.
Collapse
|
18
|
Zhang DD, Li HJ, Zhang HR, Ye XC. Poria cocos water-soluble polysaccharide modulates anxiety-like behavior induced by sleep deprivation by regulating the gut dysbiosis, metabolic disorders and TNF-α/NF-κB signaling pathway. Food Funct 2022; 13:6648-6664. [PMID: 35642970 DOI: 10.1039/d2fo00811d] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Poria cocos (P. cocos) has been traditionally used as folk medicine and functional food in China for more than 2000 years. The water-soluble polysaccharide is the main component of P. cocos decoction. The effects and mechanisms of the water-soluble polysaccharide from P. cocos (PCWP) were investigated in chronic sleep deprivation (CSD)-induced anxiety in rats. CSD induced anxiety, gut dysbiosis, and inflammatory responses, and reduced neurotransmitter levels, whereas PCWP intervention ameliorated anxiety-like behaviors, increased the levels of 5-hydroxytryptamine, dopamine, norepinephrine, and γ-aminobutyric acid in the hypothalamus, regulated gastrointestinal peptide levels, reduced inflammatory factors, and inhibited the tumor necrosis factor (TNF)-α/nuclear factor (NF)-κB signaling pathway in rats with CSD. The changes in the intestinal flora composition were determined using 16S rDNA sequencing, and indicated that PCWP significantly improved species richness and diversity in the intestinal flora of rats with anxiety, and adjusted the abundance of the following dysregulated bacteria closer to that of the normal group: Rikenellaceae_RC9_gut_group, Ruminococcus, Prevotellaceae_UCG-001, Prevotellaceae_NK3B31_group, Fusicatenibacter. Metabolomics was used to analyze fecal samples to identify significantly altered metabolites in the PCWP-treated groups. Thirty-eight PCWP-related metabolites and four metabolic pathways such as sphingolipid metabolism, taurine and hypotaurine metabolism, vitamin B6 metabolism, and glycerophospholipid metabolism were explored. The results of serum metabolomics showed that 26 biomarkers were significantly changed after PCWP intervention compared with the model group. The regulatory effects of metabolic pathway enrichment on sphingolipid, phenylalanine, and taurine and hypotaurine metabolism, and validation results showed that PCWP intervention regulated the activity of enzymes involved in the above metabolic pathways. A strong correlation between intestinal bacteria and potential biomarkers was found. Our findings present new evidence supporting the potential effect of PCWP in preventing the progression of anxiety by inhibiting the TNF-α/NF-κB signaling pathway, alleviating metabolic disorders, and ameliorating the gut microflora imbalance.
Collapse
Affiliation(s)
- Dan-Dan Zhang
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China.
| | - Hui-Jun Li
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China.
| | - Han-Rui Zhang
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China.
| | - Xiao-Chuan Ye
- Hubei Key Laboratory of Resources and Chemistry of Chinese Medicine, School of Pharmacy, Hubei University of Chinese Medicine, Wuhan, China.
| |
Collapse
|
19
|
Shor EK, Brown SP, Freeman DA. Bacteria and Bellicosity: Photoperiodic Shifts in Gut Microbiota Drive Seasonal Aggressive Behavior in Male Siberian Hamsters. J Biol Rhythms 2022; 37:296-309. [PMID: 35502701 DOI: 10.1177/07487304221092105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The existence of a microbiome-gut-brain axis has been established wherein gut microbiota significantly impacts host behavior and physiology, with increasing evidence suggesting a role for the gut microbiota in maintaining host homeostasis. Communication between the gut microbiota and the host is bidirectional, and shifts in the composition of the gut microbiota are dependent on both internal and external cues (host-derived signals, such as stress and immunity, and endocrine and environmental signals, such as photoperiod). Although there is host-driven seasonal variation in the composition of the microbiota, the mechanisms linking photoperiod, gut microbiota, and host behavior have not been characterized. The results of the present study suggest that seasonal changes in the gut microbiota drive seasonal changes in aggression. Implanting short-day Siberian hamsters (Phodopus sungorus) with fecal microbiota from long-day hamsters resulted in a reversal of seasonal aggression, whereby short-day hamsters displayed aggression levels typical of long-day hamsters. In addition, there are correlations between aggressive behavior and several bacterial taxa. These results implicate the gut microbiota as part of the photoperiodic mechanism regulating seasonal host behavior and contribute toward a more comprehensive understanding of the relationships between the microbiota, host, and environment.
Collapse
Affiliation(s)
- Elyan K Shor
- Department of Biological Sciences, Center for Biodiversity Research, The University of Memphis, Memphis, Tennessee, USA
| | - Shawn P Brown
- Department of Biological Sciences, Center for Biodiversity Research, The University of Memphis, Memphis, Tennessee, USA
| | - David A Freeman
- Department of Biological Sciences, Center for Biodiversity Research, The University of Memphis, Memphis, Tennessee, USA
| |
Collapse
|
20
|
Charitos IA, Topi S, Gagliano-Candela R, De Nitto E, Polimeno L, Montagnani M, Santacroce L. The toxic effects of endocrine disrupting chemicals (EDCs) on gut microbiota: Bisphenol A (BPA). A review. Endocr Metab Immune Disord Drug Targets 2022; 22:716-727. [PMID: 35339192 DOI: 10.2174/1871530322666220325114045] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 10/01/2021] [Accepted: 12/20/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Bisphenol A (BPA), an important industrial material widely applied in daily products, is considered an endocrine-disrupting chemical that may adversely affect humans. Growing evidence have shown that intestinal bacterial alterations caused by BPA exposure play an important role in several local and systemic diseases. AIM OF THE STUDY finding evidence that BPA-induced alterations in gut microbiota composition and activity may perturb its role on human health. RESULTS evidence from several experimental settings show that both low and high doses of BPA, interfere with the hormonal, homeostatic and reproductive systems in both animals and human systems. Moreover, it has recently been classified as an environmental obesogenic, with metabolic-disrupting effects on lipid metabolism and pancreatic b-cell functions. Several evidence characterize PBA as an environmental contributor to type II diabetes, metabolic syndrome, and obesity. However, the highest estimates of the exposure derived from foods alone or in combination with other sources are 3 to 5 times below the new tolerable daily intake (TDI) value, today reduced by the European Food Safety Authority (EFSA) experts from 50 micrograms per kilogramme of bodyweight per day (µg/kg bw/day) to 4 µg/kg bw/day. CONCLUSIONS Considering estimates for the total amount of BPA that can be ingested daily over a lifetime, many International Health Authorities conclude that dietary exposure of adult humans to BPA does not represent a risk to consumers' health, declaring its safety due to very-low established levels in food and water and declare any appreciable health risk.
Collapse
Affiliation(s)
- Ioannis Alexandros Charitos
- National Poison Center, OO. RR. University Hospital of Foggia, Foggia, Italy
- Interdepartmental Research Center for Pre-Latin, Latin and Oriental Rights and Culture Studies (CEDICLO), University of Bari, Bari, Italy
- Department of Clinical Disciplines, University of Elbasan, Elbasan, Albania
| | - Skender Topi
- Interdepartmental Research Center for Pre-Latin, Latin and Oriental Rights and Culture Studies (CEDICLO), University of Bari, Bari, Italy
- Department of Clinical Disciplines, University of Elbasan, Elbasan, Albania
| | - Roberto Gagliano-Candela
- Interdepartmental Research Center for Pre-Latin, Latin and Oriental Rights and Culture Studies (CEDICLO), University of Bari, Bari, Italy
- Department of Interdisciplinary Medicine, Microbiology and Virology Unit, School of Medicine, University of Bari, Bari, Italy
| | - Emanuele De Nitto
- Department of Medical Basic Sciences, Neurosciences and Sense Organs, Section of Biochemistry, School of Medicine, University of Bari, Bari, Italy
| | - Lorenzo Polimeno
- Department of Clinical Disciplines, University of Elbasan, Elbasan, Albania
- Polypheno Academic Spin Off, University of Bari, Bari, Italy
| | - Monica Montagnani
- Department of Biomedical Sciences and Human Oncology, Section of Pharmacology, School of Medicine, University of Bari, Bari, Italy
| | - Luigi Santacroce
- Interdepartmental Research Center for Pre-Latin, Latin and Oriental Rights and Culture Studies (CEDICLO), University of Bari, Bari, Italy
- Department of Clinical Disciplines, University of Elbasan, Elbasan, Albania
- Department of Interdisciplinary Medicine, Microbiology and Virology Unit, School of Medicine, University of Bari, Bari, Italy
- Polypheno Academic Spin Off, University of Bari, Bari, Italy
| |
Collapse
|
21
|
Halahlah A, Piironen V, Mikkonen KS, Ho TM. Polysaccharides as wall materials in spray-dried microencapsulation of bioactive compounds: Physicochemical properties and characterization. Crit Rev Food Sci Nutr 2022; 63:6983-7015. [PMID: 35213281 DOI: 10.1080/10408398.2022.2038080] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Natural bioactive compounds (BCs) are types of chemicals found in plants and certain foods that promote good health, however they are sensitive to processing and environmental conditions. Microencapsulation by spray drying is a widely used and cost-effective approach to create a coating layer to surround and protect BCs and control their release, enabling the production of high functional products/ingredients with extended shelf life. In this process, wall materials determine protection efficiency, and physical properties, bioavailability, and storage stability of microencapsulated products. Therefore, an understanding of physicochemical properties of wall materials is essential for the successful and effective spray-dried microencapsulation process. Typically, polysaccharide-based wall materials are generated from more sustainable sources and have a wider range of physicochemical properties and applications compared to their protein-based counterparts. In this review, we highlight the essential physicochemical properties of polysaccharide-based wall materials for spray-dried microencapsulation of BCs including solubility, thermal stability, and emulsifying properties, rheological and film forming properties. We provide further insight into possibilities for the chemical structure modification of native wall materials and their controlled release behaviors. Finally, we summarize the most recent studies involving polysaccharide biopolymers as wall materials and/or emulsifiers in spray-dried microencapsulation of BCs.
Collapse
Affiliation(s)
| | - Vieno Piironen
- Department of Food and Nutrition, University of Helsinki, Finland
| | - Kirsi S Mikkonen
- Department of Food and Nutrition, University of Helsinki, Finland
- Helsinki Institute of Sustainability Science (HELSUS), University of Helsinki, Finland
| | - Thao M Ho
- Department of Food and Nutrition, University of Helsinki, Finland
- Helsinki Institute of Sustainability Science (HELSUS), University of Helsinki, Finland
| |
Collapse
|
22
|
Bugrov N, Rudenko P, Lutsay V, Gurina R, Zharov A, Khairova N, Molchanova M, Krotova E, Shopinskaya M, Bolshakova M, Popova I. Fecal Microbiota Analysis in Cats with Intestinal Dysbiosis of Varying Severity. Pathogens 2022; 11:pathogens11020234. [PMID: 35215175 PMCID: PMC8875498 DOI: 10.3390/pathogens11020234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 02/04/2022] [Accepted: 02/07/2022] [Indexed: 01/27/2023] Open
Abstract
Recent studies have shown that the gut microbiota plays an important role in the pathogenesis of gastrointestinal diseases in various animal species. There are only limited data on the microbiome in cats with varying grades of dysbiosis. The purpose of the study was a detailed analysis of the quantitative and qualitative fecal microbiota spectrum in cats with intestinal dysbiosis of varying severity. The data obtained indicate that, depending on the dysbiosis severity in cats, the intestinal microbiome landscape changes significantly. It has been established that, depending on the dysbiosis severity, there is a shift in the balance between the Gram-positive and Gram-negative bacterial pools and in the nature of the isolation of specific bacteria forms, in the amount of obligate microbiota isolation, as well as individual facultative strains. When analyzing the serotyping of E. coli cultures isolated at various grades of intestinal dysbiosis severity, differences were found both in the isolation amount of various serotypes from one animal and in the prevalence of certain serotypes for each disease severity. A retrospective analysis of the fecal microbiota sensitivity in cats with dysbiosis to antibacterial drugs showed that, depending on the disease severity, the number of isolates sensitive to antibiotics increases significantly.
Collapse
Affiliation(s)
- Nikolay Bugrov
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (N.B.); (P.R.); (V.L.); (E.K.); (M.S.); (M.B.)
| | - Pavel Rudenko
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (N.B.); (P.R.); (V.L.); (E.K.); (M.S.); (M.B.)
- Biological Testing Laboratory, Branch of Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences (BIBCh RAS), 142290 Pushchino, Russia
| | - Vladimir Lutsay
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (N.B.); (P.R.); (V.L.); (E.K.); (M.S.); (M.B.)
| | - Regina Gurina
- Department of Technosphere Safety, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (R.G.); (A.Z.); (N.K.)
| | - Andrey Zharov
- Department of Technosphere Safety, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (R.G.); (A.Z.); (N.K.)
| | - Nadiya Khairova
- Department of Technosphere Safety, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (R.G.); (A.Z.); (N.K.)
| | - Maria Molchanova
- Department of Foreign Languages, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia;
| | - Elena Krotova
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (N.B.); (P.R.); (V.L.); (E.K.); (M.S.); (M.B.)
| | - Marina Shopinskaya
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (N.B.); (P.R.); (V.L.); (E.K.); (M.S.); (M.B.)
| | - Marina Bolshakova
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (N.B.); (P.R.); (V.L.); (E.K.); (M.S.); (M.B.)
| | - Irina Popova
- Department of Veterinary Medicine, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia; (N.B.); (P.R.); (V.L.); (E.K.); (M.S.); (M.B.)
- Correspondence:
| |
Collapse
|
23
|
Lim CL, Raju CS, Mahboob T, Kayesth S, Gupta KK, Jain GK, Dhobi M, Nawaz M, Wilairatana P, de Lourdes Pereira M, Patra JK, Paul AK, Rahmatullah M, Nissapatorn V. Precision and Advanced Nano-Phytopharmaceuticals for Therapeutic Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:238. [PMID: 35055257 PMCID: PMC8778544 DOI: 10.3390/nano12020238] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/07/2022] [Accepted: 01/08/2022] [Indexed: 02/04/2023]
Abstract
Phytopharmaceuticals have been widely used globally since ancient times and acknowledged by healthcare professionals and patients for their superior therapeutic value and fewer side-effects compared to modern medicines. However, phytopharmaceuticals need a scientific and methodical approach to deliver their components and thereby improve patient compliance and treatment adherence. Dose reduction, improved bioavailability, receptor selective binding, and targeted delivery of phytopharmaceuticals can be likely achieved by molding them into specific nano-formulations. In recent decades, nanotechnology-based phytopharmaceuticals have emerged as potential therapeutic candidates for the treatment of various communicable and non-communicable diseases. Nanotechnology combined with phytopharmaceuticals broadens the therapeutic perspective and overcomes problems associated with plant medicine. The current review highlights the therapeutic application of various nano-phytopharmaceuticals in neurological, cardiovascular, pulmonary, and gastro-intestinal disorders. We conclude that nano-phytopharmaceuticals emerge as promising therapeutics for many pathological conditions with good compliance and higher acceptance.
Collapse
Affiliation(s)
- Chooi Ling Lim
- Division of Applied Biomedical Science and Biotechnology, School of Health Sciences, International Medical University, Kuala Lumpur 57000, Malaysia
| | - Chandramathi S. Raju
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Tooba Mahboob
- Department of Medical Microbiology, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia;
| | - Sunil Kayesth
- Department of Zoology, Deshbandhu College, University of Delhi, New Delhi 110019, India;
| | - Kamal K. Gupta
- Department of Zoology, Deshbandhu College, University of Delhi, New Delhi 110019, India;
| | - Gaurav Kumar Jain
- Department of Pharmacognosy and Phytochemistry, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India; (G.K.J.); (M.D.)
| | - Mahaveer Dhobi
- Department of Pharmacognosy and Phytochemistry, Delhi Pharmaceutical Sciences and Research University (DPSRU), New Delhi 110017, India; (G.K.J.); (M.D.)
| | - Muhammad Nawaz
- Department of Nano-Medicine, Institute for Research and Medical Consultations ((IRMC), Imam Abdulrahman Bin Faisal University, Dammam 34212, Saudi Arabia;
| | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Maria de Lourdes Pereira
- CICECO-Aveiro Institute of Materials & Department of Medical Sciences, University of Aveiro, 3810-193 Aveiro, Portugal;
| | - Jayanta Kumar Patra
- Research Institute of Biotechnology & Medical Converged Science, Dongguk University-Seoul, Goyang-si 10326, Korea;
| | - Alok K. Paul
- School of Pharmacy and Pharmacology, University of Tasmania, Private Bag 26, Hobart, TAS 7001, Australia;
| | - Mohammed Rahmatullah
- Department of Biotechnology & Genetic Engineering, University of Development Alternative, Lalmatia, Dhaka 1207, Bangladesh;
| | - Veeranoot Nissapatorn
- School of Allied Health Sciences and World Union for Herbal Drug Discovery (WUHeDD), Walailak University, Nakhon Si Thammarat 80160, Thailand
| |
Collapse
|
24
|
Lai W, Yang S, Lin X, Zhang X, Huang Y, Zhou J, Fu C, Li R, Zhang Z. Zingiber officinale: A Systematic Review of Botany, Phytochemistry and Pharmacology of Gut Microbiota-Related Gastrointestinal Benefits. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:1007-1042. [PMID: 35729087 DOI: 10.1142/s0192415x22500410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/15/2023]
Abstract
Ginger (Zingiber officinale Rosc.) is a traditional edible medicinal herb with a wide range of uses and long cultivation history. Fresh ginger (Zingiberis Recens Rhizoma; Sheng Jiang in Chinese, SJ) and dried ginger (Zingiberis Rhizoma; Gan Jiang in Chinese, GJ) are designated as two famous traditional Chinese herbal medicines, which are different in plant cultivation, appearances and functions, together with traditional applications. Previous researches mainly focused on the differences in chemical composition between them, but there was no systematical comparison on the similarity concerning research achievements of the two herbs. Meanwhile, ginger has traditionally been used for the treatment of gastrointestinal disorders, but so far, the possible interaction with human gut microbiota has hardly been considered. This review comprehensively presents similarities and differences between SJ and GJ retrospectively, particularly proposing them the significant differences in botany, phytochemistry and ethnopharmacology, which can be used as evidence for clinical application of SJ and GJ. Furthermore, the pharmacology of gut microbiota-related gastrointestinal benefits has also been discussed in order to explore better ways to prevent and treat gastrointestinal disorders, which can be used as a reference for further research.
Collapse
Affiliation(s)
- Wenjing Lai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Shasha Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Xia Lin
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Xing Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - You Huang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Jingwei Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Chaomei Fu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Rui Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
| | - Zhen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, P. R. China
- Key Laboratory of Quality Control and Efficacy Evaluation of Traditional Chinese Medicine Formula Granules, Sichuan New Green Medicine Science and Technology Development Co., Ltd., Pengzhou 610081, P. R. China
| |
Collapse
|
25
|
Gupta A, Singh V, Mani I. Dysbiosis of human microbiome and infectious diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 192:33-51. [DOI: 10.1016/bs.pmbts.2022.06.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
26
|
Mandalari G, Barreca D, Gervasi T, Roussell MA, Klein B, Feeney MJ, Carughi A. Pistachio Nuts ( Pistacia vera L.): Production, Nutrients, Bioactives and Novel Health Effects. PLANTS (BASEL, SWITZERLAND) 2021; 11:18. [PMID: 35009022 PMCID: PMC8747606 DOI: 10.3390/plants11010018] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 05/24/2023]
Abstract
Epidemiological and clinical studies have indicated positive outcomes related to tree nut consumption. Here, we review the production, nutrient, phytochemical composition and emerging research trends on the health benefits of pistachio nuts (Pistacia vera L.). Pistachios are a good source of protein, fiber, monounsaturated fatty acids, minerals and vitamins, as well as carotenoids, phenolic acids, flavonoids and anthocyanins. Polyphenols in pistachios are important contributors to the antioxidant and anti-inflammatory effect, as demonstrated in vitro and in vivo through animal studies and clinical trials. The antimicrobial and antiviral potential of pistachio polyphenols has also been assessed and could help overcome drug resistance. Pistachio consumption may play a role in cognitive function and has been associated with a positive modulation of the human gut microbiota and beneficial effects on skin health. Pistachio polyphenol extracts may affect enzymes involved in glucose regulation and so type 2 diabetes. Taken together, these data demonstrate the health benefits of including pistachios in the diet. Further studies are required to investigate the mechanisms involved.
Collapse
Affiliation(s)
- Giuseppina Mandalari
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy;
| | - Davide Barreca
- Department of Chemical, Biological, Pharmaceutical and Environmental Science, University of Messina, 98166 Messina, Italy;
| | - Teresa Gervasi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, 98125 Messina, Italy;
| | | | - Bob Klein
- California Pistachio Research Board, Fresno, CA 93727, USA;
| | - Mary Jo Feeney
- Consultant to the Food and Agriculture Industries, Los Altos Hills, CA 94024, USA;
| | | |
Collapse
|
27
|
Szigethy E, Tansel A, Pavlick AN, Marroquin MA, Serio CD, Silfee V, Wallace ML, Kingsley MJ, Levinthal DJ. A Coached Digital Cognitive Behavioral Intervention Reduces Anxiety and Depression in Adults With Functional Gastrointestinal Disorders. Clin Transl Gastroenterol 2021; 12:e00436. [PMID: 34874018 PMCID: PMC8751763 DOI: 10.14309/ctg.0000000000000436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 10/08/2021] [Indexed: 12/12/2022] Open
Abstract
INTRODUCTION Traditional cognitive behavioral interventions (CBIs) improve mood and gastrointestinal symptom severity in patients with functional gastrointestinal disorders (FGIDs) but face substantial barriers to implementation. Integrating behavioral health technology into medical clinic workflows could overcome these barriers. We evaluated the feasibility and impact of a coached digital CBI (dCBI) as a first-line intervention in a prospective cohort of emotionally distressed patients with FGID. METHODS Patients with anxiety and/or depressive symptoms were offered a dCBI (an app called RxWell) during routine clinic visits. RxWell provides cognitive behavioral techniques enhanced by within-app text messaging with a health coach. Both gastroenterology and behavioral health-care providers electronically prescribed RxWell. We tracked patient interactions with RxWell, and patients completed anxiety (General Anxiety Disorder-7) and depression (Personal Health Questionniare Depression Scale) measures through the app. Our primary study outcome was the change in General Anxiety Disorder-7 and Personal Health Questionniare Depression Scale scores. RESULTS Of 364 patients with FGID (mean age 43 years [SD 16 years]; 73.1% women) prescribed the dCBI, 48.4% enrolled (median use, 3 techniques [interquartile range 1-14]). About half of RxWell enrollees communicated with health coaches. The mean baseline anxiety score was 11.4 (SD 5.5), and the depression score was 11.5 (SD 6.1). RxWell users experienced improvements in anxiety (mean change 2.71 [t = 3.7, df = 58; P < 0.001]) and depression (mean change 2.9 [t = 4.2, df = 45; P < 0.001]) at 4 months. DISCUSSION Patients with FGIDs and moderately severe anxiety and depressive symptoms are willing to use dCBI tools recommended by their providers. Our pilot data demonstrate that dCBI usage is associated with clinically and statistically significant mood symptom reductions.
Collapse
Affiliation(s)
- Eva Szigethy
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA;
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA;
| | - Aylin Tansel
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA;
| | - Alexa N. Pavlick
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA;
| | - Maria A. Marroquin
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA;
| | - Catherine D. Serio
- University of Pittsburgh Medical Center Insurance Services Division, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Valerie Silfee
- University of Pittsburgh Medical Center Insurance Services Division, University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Meredith L. Wallace
- Department of Psychiatry, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA;
| | - Michael J. Kingsley
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA;
| | - David J. Levinthal
- Department of Medicine, Division of Gastroenterology, Hepatology, and Nutrition, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA;
| |
Collapse
|
28
|
Conti L, Borro M, Milani C, Simmaco M, Esposito G, Canali G, Pilozzi E, Ventura M, Annibale B, Lahner E. Gastric microbiota composition in patients with corpus atrophic gastritis. Dig Liver Dis 2021; 53:1580-1587. [PMID: 34116969 DOI: 10.1016/j.dld.2021.05.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/30/2021] [Accepted: 05/03/2021] [Indexed: 02/05/2023]
Abstract
BACKGROUND In corpus atrophic gastritis (CAG), hypochlorhydria makes plausible the overgrowth of intragastric bacteria, whose role in gastric carcinogenesis is under debate. AIMS To characterize the antrum/corpus composition of the gastric bacterial microbiota in CAG patients compared to controls without CAG. METHODS A cross-sectional monocentric study on consecutive patients with known histological diagnosis of CAG undergoing gastroscopy for gastric cancer surveillance and patients without CAG undergoing gastroscopy for dyspepsia or anemia (108 biopsies from 55 patients, median age 61.5). Genomic DNA from one antral and one corpus biopsy from each case (n = 23) and control (n = 32) was extracted. Gastric microbiota was assessed by sequencing hypervariable regions of the 16SrRNA gene. RESULTS Bacterial abundance and diversity were significantly lower in CAG cases than in controls (p < 0.001). Firmicutes were more frequent in cases, Bacteroidetes and Fusobacteria in controls (p < 0.0001). Streptococcaceae were more abundant in cases (p < 0.0001), Prevotellaceae in controls (p < 0.0001). The genus Streptococcus was positively correlated with severe OLGA/OLGIM stages linked to a higher risk of gastric cancer. CONCLUSION Gastric bacterial microbiota in CAG showed a reduced abundance and complexity but was characterized by higher colonization of Firmicutes, in particular Streptococcus, increased in subjects with severe atrophy/metaplasia stages at higher risk of gastric cancer.
Collapse
Affiliation(s)
- Laura Conti
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | - Marina Borro
- Department of Neurosciences, Mental Health, and Sensory Organs, "Sapienza" University of Rome, via di Grottarossa 1035-1039, 00189 Rome, Italy
| | - Christian Milani
- Laboratory of Probiogenomics, Microbiome Research Hub, Dept. Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Maurizio Simmaco
- Department of Neurosciences, Mental Health, and Sensory Organs, "Sapienza" University of Rome, via di Grottarossa 1035-1039, 00189 Rome, Italy
| | - Gianluca Esposito
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | - Giulia Canali
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, University Sapienza, Rome, Italy
| | - Marco Ventura
- Laboratory of Probiogenomics, Microbiome Research Hub, Dept. Chemistry, Life Sciences, and Environmental Sustainability, University of Parma, Parma, Italy
| | - Bruno Annibale
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza, Rome, Italy
| | - Edith Lahner
- Medical-Surgical Department of Clinical Sciences and Translational Medicine, Sant'Andrea Hospital, University Sapienza, Rome, Italy.
| |
Collapse
|
29
|
Vicentini FA, Keenan CM, Wallace LE, Woods C, Cavin JB, Flockton AR, Macklin WB, Belkind-Gerson J, Hirota SA, Sharkey KA. Intestinal microbiota shapes gut physiology and regulates enteric neurons and glia. MICROBIOME 2021; 9:210. [PMID: 34702353 PMCID: PMC8549243 DOI: 10.1186/s40168-021-01165-z] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Accepted: 09/15/2021] [Indexed: 05/05/2023]
Abstract
BACKGROUND The intestinal microbiota plays an important role in regulating gastrointestinal (GI) physiology in part through interactions with the enteric nervous system (ENS). Alterations in the gut microbiome frequently occur together with disturbances in enteric neural control in pathophysiological conditions. However, the mechanisms by which the microbiota regulates GI function and the structure of the ENS are incompletely understood. Using a mouse model of antibiotic (Abx)-induced bacterial depletion, we sought to determine the molecular mechanisms of microbial regulation of intestinal function and the integrity of the ENS. Spontaneous reconstitution of the Abx-depleted microbiota was used to assess the plasticity of structure and function of the GI tract and ENS. Microbiota-dependent molecular mechanisms of ENS neuronal survival and neurogenesis were also assessed. RESULTS Adult male and female Abx-treated mice exhibited alterations in GI structure and function, including a longer small intestine, slower transit time, increased carbachol-stimulated ion secretion, and increased intestinal permeability. These alterations were accompanied by the loss of enteric neurons in the ileum and proximal colon in both submucosal and myenteric plexuses. A reduction in the number of enteric glia was only observed in the ileal myenteric plexus. Recovery of the microbiota restored intestinal function and stimulated enteric neurogenesis leading to increases in the number of enteric glia and neurons. Lipopolysaccharide (LPS) supplementation enhanced neuronal survival alongside bacterial depletion, but had no effect on neuronal recovery once the Abx-induced neuronal loss was established. In contrast, short-chain fatty acids (SCFA) were able to restore neuronal numbers after Abx-induced neuronal loss, demonstrating that SCFA stimulate enteric neurogenesis in vivo. CONCLUSIONS Our results demonstrate a role for the gut microbiota in regulating the structure and function of the GI tract in a sex-independent manner. Moreover, the microbiota is essential for the maintenance of ENS integrity, by regulating enteric neuronal survival and promoting neurogenesis. Molecular determinants of the microbiota, LPS and SCFA, regulate enteric neuronal survival, while SCFA also stimulates neurogenesis. Our data reveal new insights into the role of the gut microbiota that could lead to therapeutic developments for the treatment of enteric neuropathies. Video abstract.
Collapse
Affiliation(s)
- Fernando A. Vicentini
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1 Canada
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 4N1 Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
| | - Catherine M. Keenan
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1 Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
| | - Laurie E. Wallace
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1 Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
| | - Crystal Woods
- Department of Pediatrics, Section of Gastroenterology, Hepatology and Nutrition, University of Colorado, Aurora, CO 80045 USA
| | - Jean-Baptiste Cavin
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1 Canada
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 4N1 Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
| | - Amanda R. Flockton
- Department of Pediatrics, Section of Gastroenterology, Hepatology and Nutrition, University of Colorado, Aurora, CO 80045 USA
| | - Wendy B. Macklin
- Department of Cell and Developmental Biology, University of Colorado School of Medicine, Aurora, CO 80045 USA
| | - Jaime Belkind-Gerson
- Department of Pediatrics, Section of Gastroenterology, Hepatology and Nutrition, University of Colorado, Aurora, CO 80045 USA
- Neurogastroenterology and Motility Program, Digestive Health Institute, Children’s Hospital Colorado, Aurora, CO 80045 USA
| | - Simon A. Hirota
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1 Canada
- Inflammation Research Network, University of Calgary, Calgary, AB T2N 4N1 Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
- Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
| | - Keith A. Sharkey
- Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1 Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, AB T2N 4N1 Canada
- Department of Physiology and Pharmacology, Cumming School of Medicine, University of Calgary, 3330 Hospital Drive NW, Calgary, AB T2N 4N1 Canada
| |
Collapse
|
30
|
Bokoliya SC, Dorsett Y, Panier H, Zhou Y. Procedures for Fecal Microbiota Transplantation in Murine Microbiome Studies. Front Cell Infect Microbiol 2021; 11:711055. [PMID: 34621688 PMCID: PMC8490673 DOI: 10.3389/fcimb.2021.711055] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 08/24/2021] [Indexed: 12/11/2022] Open
Abstract
Fecal microbiota transplantation (FMT) has been widely recognized as an approach to determine the microbiome’s causal role in gut dysbiosis-related disease models and as a novel disease-modifying therapy. Despite potential beneficial FMT results in various disease models, there is a variation and complexity in procedural agreement among research groups for performing FMT. The viability of the microbiome in feces and its successful transfer depends on various aspects of donors, recipients, and lab settings. This review focuses on the technical practices of FMT in animal studies. We first document crucial factors required for collecting, handling, and processing donor fecal microbiota for FMT. Then, we detail the description of gut microbiota depletion methods, FMT dosages, and routes of FMT administrations in recipients. In the end, we describe assessments of success rates of FMT with sustainability. It is critical to work under the anaerobic condition to preserve as much of the viability of bacteria. Utilization of germ- free mice or depletion of recipient gut microbiota by antibiotics or polyethylene glycol are two common recipient preparation approaches to achieve better engraftment. Oral-gastric gavage preferred by most researchers for fast and effective administration of FMT in mice. Overall, this review highlights various methods that may lead to developing the standard and reproducible protocol for FMT.
Collapse
Affiliation(s)
- Suresh C Bokoliya
- Department of Medicine, University of Connecticut (UConn) Health, Farmington, CT, United States
| | - Yair Dorsett
- Department of Medicine, University of Connecticut (UConn) Health, Farmington, CT, United States
| | - Hunter Panier
- Department of Medicine, University of Connecticut (UConn) Health, Farmington, CT, United States
| | - Yanjiao Zhou
- Department of Medicine, University of Connecticut (UConn) Health, Farmington, CT, United States
| |
Collapse
|
31
|
Enteric Microbiota-Mediated Serotonergic Signaling in Pathogenesis of Irritable Bowel Syndrome. Int J Mol Sci 2021; 22:ijms221910235. [PMID: 34638577 PMCID: PMC8508930 DOI: 10.3390/ijms221910235] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/18/2021] [Accepted: 09/19/2021] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome (IBS) is a chronic functional disorder that affects the gastrointestinal tract. Details regarding the pathogenesis of IBS remain largely unknown, though the dysfunction of the brain-gut-microbiome (BGM) axis is a major etiological factor, in which neurotransmitters serve as a key communication tool between enteric microbiota and the brain. One of the most important neurotransmitters in the pathology of IBS is serotonin (5-HT), as it influences gastrointestinal motility, pain sensation, mucosal inflammation, immune responses, and brain activity, all of which shape IBS features. Genome-wide association studies discovered susceptible genes for IBS in serotonergic signaling pathways. In clinical practice, treatment strategies targeting 5-HT were effective for a certain portion of IBS cases. The synthesis of 5-HT in intestinal enterochromaffin cells and host serotonergic signaling is regulated by enteric resident microbiota. Dysbiosis can trigger IBS development, potentially through aberrant 5-HT signaling in the BGM axis; thus, the manipulation of the gut microbiota may be an alternative treatment strategy. However, precise information regarding the mechanisms underlying the microbiota-mediated intestinal serotonergic pathway related to the pathogenesis of IBS remains unclear. The present review summarizes current knowledge and recent progress in understanding microbiome–serotonin interaction in IBS cases.
Collapse
|
32
|
Neroni B, Evangelisti M, Radocchia G, Di Nardo G, Pantanella F, Villa MP, Schippa S. Relationship between sleep disorders and gut dysbiosis: what affects what? Sleep Med 2021; 87:1-7. [PMID: 34479058 DOI: 10.1016/j.sleep.2021.08.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 07/29/2021] [Accepted: 08/03/2021] [Indexed: 12/25/2022]
Abstract
Sleep plays a fundamental role in maintaining good psycho-physical health, it can influence hormone levels, mood, and weight. Recent studies, focused on the interconnection between intestinal microbiome and sleep disorders, have shown the growing importance of a healthy and balanced intestinal microbiome for the hosts health. Normally, gut microbiota and his host are linked by mutualistic relationship, that in some conditions, can be compromised by shifts in microbiota's composition, called dysbiosis. Both sleep problems and dysbiosis of the gut microbiome can lead to metabolic disorders and, in this review, we will explore what is present in literature on the link between sleep pathologies and intestinal dysbiosis.
Collapse
Affiliation(s)
- Bruna Neroni
- Department of Public Health and Infection Disease, Microbiology Section Sapienza University of Rome, Italy
| | | | - Giulia Radocchia
- Department of Public Health and Infection Disease, Microbiology Section Sapienza University of Rome, Italy
| | - Giovanni Di Nardo
- Sant'Andrea Hospital, NESMOS Department, Sapienza University of Rome, Italy
| | - Fabrizio Pantanella
- Department of Public Health and Infection Disease, Microbiology Section Sapienza University of Rome, Italy
| | - Maria Pia Villa
- Sant'Andrea Hospital, NESMOS Department, Sapienza University of Rome, Italy
| | - Serena Schippa
- Department of Public Health and Infection Disease, Microbiology Section Sapienza University of Rome, Italy.
| |
Collapse
|
33
|
Forouzan S, McGrew K, Kosten TA. Drugs and bugs: Negative affect, psychostimulant use and withdrawal, and the microbiome. Am J Addict 2021; 30:525-538. [PMID: 34414622 DOI: 10.1111/ajad.13210] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 07/12/2021] [Accepted: 07/14/2021] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND AND OBJECTIVES A growing body of literature demonstrates that the human microbiota plays a crucial role in health and disease states, as well as in the body's response to stress. In addition, the microbiome plays a role in psychological well-being and regulating negative affect. Regulation of negative affect is a factor in psychostimulant abuse disorders. We propose a risk chain in which stress leads to negative affect that places an individual at risk to develop or relapse to psychostimulant abuse disorder. Stress, negative affect, and psychostimulant use all alter the gut microbiome. METHODS This review brings together the literature on affective disorders, stress, and psychostimulant abuse disorders to assess possible modulatory actions of the gut-brain axis to regulate these conditions. RESULTS Studies reviewed across the various disciplines suggest that the dysbiosis resulting from drug use, drug withdrawal, or stress may cause an individual to be more susceptible to addiction and relapse. Probiotics and prebiotics reduce stress and negative affect. SCIENTIFIC SIGNIFICANCE Treatment during the withdrawal phase of psychostimulant abuse disorder, when the microbiome is altered, may ameliorate the symptoms of stress and negative affect leading to a reduced risk of relapse to psychostimulant use.
Collapse
Affiliation(s)
- Shadab Forouzan
- Department of Psychology, Texas Institute for Measurement, Evaluation and Statistics (TIMES), University of Houston, Houston, Texas, USA
| | - Keely McGrew
- Department of Psychology, Texas Institute for Measurement, Evaluation and Statistics (TIMES), University of Houston, Houston, Texas, USA
| | - Therese A Kosten
- Department of Psychology, Texas Institute for Measurement, Evaluation and Statistics (TIMES), University of Houston, Houston, Texas, USA
| |
Collapse
|
34
|
Multiomic Approach to Analyze Infant Gut Microbiota: Experimental and Analytical Method Optimization. Biomolecules 2021; 11:biom11070999. [PMID: 34356622 PMCID: PMC8301799 DOI: 10.3390/biom11070999] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/15/2021] [Accepted: 06/28/2021] [Indexed: 12/16/2022] Open
Abstract
Background: The human intestinal microbiome plays a central role in overall health status, especially in early life stages. 16S rRNA amplicon sequencing is used to profile its taxonomic composition; however, multiomic approaches have been proposed as the most accurate methods for study of the complexity of the gut microbiota. In this study, we propose an optimized method for bacterial diversity analysis that we validated and complemented with metabolomics by analyzing fecal samples. Methods: Forty-eight different analytical combinations regarding (1) 16S rRNA variable region sequencing, (2) a feature selection approach, and (3) taxonomy assignment methods were tested. A total of 18 infant fecal samples grouped depending on the type of feeding were analyzed by the proposed 16S rRNA workflow and by metabolomic analysis. Results: The results showed that the sole use of V4 region sequencing with ASV identification and VSEARCH for taxonomy assignment produced the most accurate results. The application of this workflow showed clear differences between fecal samples according to the type of feeding, which correlated with changes in the fecal metabolic profile. Conclusion: A multiomic approach using real fecal samples from 18 infants with different types of feeding demonstrated the effectiveness of the proposed 16S rRNA-amplicon sequencing workflow.
Collapse
|
35
|
Moon CM, Hong SN. Fecal Microbiota Transplantation beyond Clostridioides Difficile Infection. Clin Endosc 2021; 54:149-151. [PMID: 33765728 PMCID: PMC8039751 DOI: 10.5946/ce.2021.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Accepted: 02/25/2021] [Indexed: 11/14/2022] Open
Abstract
With advancing analytical methods for gut microbes, many studies have been conducted, revealing that gut microbes cause various diseases, including gastrointestinal and non-gastrointestinal diseases. Accordingly, studies have been actively conducted to analyze the effects on the prevention and treatment of these diseases through changes in intestinal microbes and control of dysbiosis. Fecal microbiota transplantation (FMT) is an effort and is currently being applied to Clostridioides difficile treatment in Korea. Many studies have demonstrated the application of FMT in inflammatory bowel disease, irritable bowel syndrome, non-alcoholic fatty liver disease, metabolic syndrome, obesity, and diabetes. With further studies and accumulation of evidence, FMT could help treat presently untreatable diseases in clinical practice.
Collapse
Affiliation(s)
- Chang Mo Moon
- Department of Internal Medicine, Ewha Womans University College of Medicine, Seoul, Korea
| | - Sung Noh Hong
- Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| |
Collapse
|
36
|
Kitamura K, Sasaki M, Matsumoto M, Shionoya H, Iida K. Protective effect of Bacteroides fragilis LPS on Escherichia coli LPS-induced inflammatory changes in human monocytic cells and in a rheumatoid arthritis mouse model. Immunol Lett 2021; 233:48-56. [PMID: 33741378 DOI: 10.1016/j.imlet.2021.03.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 03/14/2021] [Accepted: 03/15/2021] [Indexed: 11/26/2022]
Abstract
It has been reported that patients with rheumatoid arthritis (RA) have significantly less bacteria belonging to the Bacteroides group in their microbiota. We speculate that inhibition of cytokine production is impaired in patients with RA owing to their low levels of intestinal bacteria belonging to the Bacteroidetes group. Here we investigated the effect of Bacteroides fragilis lipopolysaccharide (B-LPS) on cytokine production in vitro and on the development of collagen antibody-induced arthritis (CAIA) in DBA/1 mice, an animal model of RA. in vitro culture experiments showed that Escherichia coli LPS (E-LPS)-induced cytokine production from THP-1 monocytic cells and peripheral blood mononuclear cells was significantly suppressed by B-LPS in a dose-dependent manner. A decrease in TNF-α and IL-1β production was also observed in LPS-tolerized macrophages induced by B-LPS at concentrations equal to and higher than that of E-LPS. Similar results were obtained when autoclaved feces were used to induce cytokine production instead of E-LPS. In in vivo experiments using CAIA models, B-LPS had no adverse effects even when administered at 10 times the concentration of E-LPS, which elicits severe arthritis. In addition, simultaneous administration of high dose B-LPS with E-LPS or administration of B-LPS prior to E-LPS significantly suppressed arthritis development in CAIA model animals when compared with administration of E-LPS alone. These results suggest that increasing certain bacterial groups such as Bacteroides is an effective strategy for preventing arthritis development in patients with RA.
Collapse
Affiliation(s)
- Kaori Kitamura
- Department of Food and Nutritional Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo, 112-8610, Japan; Research Lab Section 5, Asama Chemical Co Ltd, 20-6 Kodenmacho, Chuo-ku, Tokyo, 103-0001, Japan
| | - Mizuho Sasaki
- Department of Food and Nutritional Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo, 112-8610, Japan
| | - Moe Matsumoto
- Department of Food and Nutritional Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo, 112-8610, Japan
| | - Hiroshi Shionoya
- Research Lab Section 5, Asama Chemical Co Ltd, 20-6 Kodenmacho, Chuo-ku, Tokyo, 103-0001, Japan
| | - Kaoruko Iida
- Department of Food and Nutritional Sciences, Graduate School of Humanities and Sciences, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo, 112-8610, Japan; Institute for Human Life Innovation, Ochanomizu University, 2-1-1 Otsuka, Bunkyo-ku, Tokyo, 112-8610, Japan.
| |
Collapse
|
37
|
Belizário JE, Faintuch J, Malpartida MG. Breath Biopsy and Discovery of Exclusive Volatile Organic Compounds for Diagnosis of Infectious Diseases. Front Cell Infect Microbiol 2021; 10:564194. [PMID: 33520731 PMCID: PMC7839533 DOI: 10.3389/fcimb.2020.564194] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 11/16/2020] [Indexed: 01/13/2023] Open
Abstract
Exhaled breath contains thousand metabolites and volatile organic compounds (VOCs) that originated from both respiratory tract and internal organ systems and their microbiomes. Commensal and pathogenic bacteria and virus of microbiomes are capable of producing VOCs of different chemical classes, and some of them may serve as biomarkers for installation and progression of various common human diseases. Here we describe qualitative and quantitative methods for measuring VOC fingerprints generated by cellular and microbial metabolic and pathologic pathways. We describe different chemical classes of VOCs and their role in the host cell-microbial interactions and their impact on infection disease pathology. We also update on recent progress on VOC signatures emitted by isolated bacterial species and microbiomes, and VOCs identified in exhaled breath of patients with respiratory tract and gastrointestinal diseases, and inflammatory syndromes, including the acute respiratory distress syndrome and sepsis. The VOC curated databases and instrumentations have been developed through statistically robust breathomic research in large patient populations. Scientists have now the opportunity to find potential biomarkers for both triage and diagnosis of particular human disease.
Collapse
Affiliation(s)
- José E Belizário
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Joel Faintuch
- Department of Gastroenterology of Medical School, University of Sao Paulo, São Paulo, Brazil
| | | |
Collapse
|
38
|
Hajiagha MN, Taghizadeh S, Asgharzadeh M, Dao S, Ganbarov K, Köse Ş, Kafil HS. Gut microbiota And Human Body Interactions; Its Impact on Health: a review. Curr Pharm Biotechnol 2021; 23:4-14. [PMID: 33397232 DOI: 10.2174/1389201022666210104115836] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 11/17/2020] [Accepted: 11/19/2020] [Indexed: 12/27/2022]
Abstract
Gut microbiota (GM) as an organ of the human body has a particular and autonomous function that related to it. This review aimed to investigate human intestinal and gut microbiota interaction and its impact on health. As a creation referable database about this dynamic and complex organ, several comprehensive projects are implemented by using culture-dependent (culturomics), culture independent methods (e.g metagenomics, mathematics model), and Gnotobiological together. This study was done by searching PubMed, Scopus and Google scholar database in the gut, health microbiota and interaction keywords. The first acquired microbiota during pregnancy or childbirth is colonized in the gut by using specific and non-specific mechanisms. That`s structure and shape reach relative stability with selection pressure along with host development until adulthood and keep its resilience against external or internal variables depending on the host genetics and negative feedback. Due to several research individuals have 2 functional group microbiota including the core (common between vast majorities human) and flexible (transient population) microbiome. The most important role of the GM in the human body can be summarized in three basic landscapes: metabolic, immune system, and gut-brain axis interaction. So that loss of microbial population balance will lead to disorder and disease.
Collapse
Affiliation(s)
| | - Sepehr Taghizadeh
- Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Mohammad Asgharzadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz. Iran
| | - Sounkalo Dao
- Faculté de Médecine, de Pharmacie et d'Odonto-Stomatologie (FMPOS), University of Bamako, Bamako. Mali
| | | | - Şükran Köse
- Department of Infectious Diseases and Clinical Microbiology, University of Health Sciences, Tepecik Training and Research Hospital, İzmir. Turkey
| | - Hossein Samadi Kafil
- Drug Applied Research Center, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz. Iran
| |
Collapse
|
39
|
Kaur G, Behl T, Bungau S, Kumar A, Uddin MS, Mehta V, Zengin G, Mathew B, Shah MA, Arora S. Dysregulation of the Gut-Brain Axis, Dysbiosis and Influence of Numerous Factors on Gut Microbiota Associated Parkinson's Disease. Curr Neuropharmacol 2021; 19:233-247. [PMID: 32504503 PMCID: PMC8033978 DOI: 10.2174/1570159x18666200606233050] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/27/2020] [Accepted: 06/02/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Parkinson's disease (PD) has been one of the substantial social, medical concerns and, burdens of the present time. PD is a gradually devastating neurodegenerative disorder of the neurological function marked with α-synucleinopathy affecting numerous regions of the brain-gut axis, as well as the central, enteric, and autonomic nervous system. Its etiology is a widely disputed topic. OBJECTIVE This review emphasizes to find out the correlation among the microbial composition and the observable disturbances in the metabolites of the microbial species and its impact on the immune response, which may have a concrete implication on the occurrence, persistence and, pathophysiology of PD via the gut-brain axis. METHODS An in-depth research and the database was developed from the available peer-reviewed articles to date (March 2020) utilizing numerous search engines like PubMed, MEDLINE and, other internet sources. RESULTS Progressively increasing shreds of evidence have proved the fact that dysbiosis in the gut microbiome plays a central role in many neurological disorders, such as PD. Indeed, a disordered microbiome-gut-brain axis in PD could be focused on gastrointestinal afflictions that manifest primarily several years prior to the diagnosis, authenticating a concept wherein the pathological pathway progresses from the intestine reaching the brain. CONCLUSION The microbiota greatly affects the bidirectional interaction between the brain and the gut via synchronized neurological, immunological, and neuroendocrine mechanisms. It can be concluded that a multitude of factors discussed in this review steadily induce the onset of dysbacteriosis that may exacerbate the etiologic mechanism of Parkinson's disease.
Collapse
Affiliation(s)
| | - Tapan Behl
- Address correspondence to this author at the Chitkara College of Pharmacy, Chitkara University, Punjab, India; Tel: +91-8527517931;, E-mails: ;
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Azad MA, Gao J, Ma J, Li T, Tan B, Huang X, Yin J. Opportunities of prebiotics for the intestinal health of monogastric animals. ANIMAL NUTRITION (ZHONGGUO XU MU SHOU YI XUE HUI) 2020; 6:379-388. [PMID: 33364453 PMCID: PMC7750794 DOI: 10.1016/j.aninu.2020.08.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/16/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
The goal of prebiotic applications from different sources is to improve the gut ecosystem where the host and microbiota can benefit from prebiotics. It has already been recognized that prebiotics have potential roles in the gut ecosystem because gut microbiota ferment complex dietary macronutrients and carry out a broad range of functions in the host body, such as the production of nutrients and vitamins, protection against pathogens, and maintenance of immune system balance. The gut ecosystem is very crucial and can be affected by numerous factors consisting of dietary constituents and commensal bacteria. This review focuses on recent scientific evidence, confirming a beneficial effect of prebiotics on animal health, particularly in terms of protection against pathogenic bacteria and increasing the number of beneficial bacteria that may improve epithelial cell barrier functions. It has also been reviewed that modification of the gut ecosystem through the utilization of prebiotics significantly affects the intestinal health of animals. However, the identification and characterization of novel potential prebiotics remain a topical issue and elucidation of the metagenomics relationship between gut microbiota alteration and prebiotic substances is necessary for future prebiotic studies.
Collapse
Affiliation(s)
- Md A.K. Azad
- College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jing Gao
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, 410125, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Jie Ma
- College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Tiejun Li
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-ecological Processes in Subtropical Region, Institute of Subtropical Agriculture, Chinese Academy of Sciences, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Changsha, 410125, China
| | - Bie Tan
- College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Xingguo Huang
- College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Hunan Provincial Engineering Research Center of Applied Microbial Resources Development for Livestock and Poultry, Changsha, 410128, China
| |
Collapse
|
41
|
María Remes Troche J, Coss Adame E, Ángel Valdovinos Díaz M, Gómez Escudero O, Eugenia Icaza Chávez M, Antonio Chávez-Barrera J, Zárate Mondragón F, Antonio Ruíz Velarde Velasco J, Rafael Aceves Tavares G, Antonio Lira Pedrín M, Cerda Contreras E, Carmona Sánchez RI, Guerra López H, Solana Ortiz R. Lactobacillus acidophilus LB: a useful pharmabiotic for the treatment of digestive disorders. Therap Adv Gastroenterol 2020; 13:1756284820971201. [PMID: 33281937 PMCID: PMC7692339 DOI: 10.1177/1756284820971201] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 10/07/2020] [Indexed: 02/04/2023] Open
Abstract
Dysbiosis, a loss of balance between resident bacterial communities and their host, is associated with multiple diseases, including inflammatory bowel diseases (nonspecific chronic ulcerative colitis and Crohn's disease), and digestive functional disorders. Probiotics, prebiotics, synbiotic organisms and, more recently, pharmabiotics, have been shown to modulate the human microbiota. In this review, we provide an overview of the key concepts relating to probiotics, prebiotics, synbiotic organisms, and pharmabiotics, with a focus on available clinical evidence regarding the specific use of a unique pharmabiotic, the strain Lactobacillus acidophilus LB (Lactobacillus boucardii), for the management of gastrointestinal disorders. Since it does not contain living organisms, the administration of L. acidophilus LB is effective and safe as an adjuvant in the treatment of acute diarrhea, chronic diarrhea, and antibiotic-associated diarrhea, even in the presence of immunosuppression.
Collapse
Affiliation(s)
- José María Remes Troche
- Instituto e Investigaciones Médico Biológicas de la Universidad Veracruzana, Veracruz, Mexico
| | - Enrique Coss Adame
- Gastroenterology Department, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Miguel Ángel Valdovinos Díaz
- National Institute of Medical Sciences and Nutrition Salvador Zubirán, Vasco de Quiroga 15, Belisario Domínguez Secc 16, Mexico City 14080, Mexico
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Uhlig F, Grundy L, Garcia-Caraballo S, Brierley SM, Foster SJ, Grundy D. Identification of a Quorum Sensing-Dependent Communication Pathway Mediating Bacteria-Gut-Brain Cross Talk. iScience 2020; 23:101695. [PMID: 33163947 PMCID: PMC7607502 DOI: 10.1016/j.isci.2020.101695] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 07/03/2020] [Accepted: 10/14/2020] [Indexed: 12/12/2022] Open
Abstract
Despite recently established contributions of the intestinal microbiome to human health and disease, our understanding of bacteria-host communication pathways with regard to the gut-brain axis remains limited. Here we provide evidence that intestinal neurons are able to "sense" bacteria independently of the host immune system. Using supernatants from cultures of the opportunistic pathogen Staphylococcus aureus (S. aureus) we demonstrate the release of mediators with neuromodulatory properties at high population density. These mediators induced a biphasic response in extrinsic sensory afferent nerves, increased membrane permeability in cultured sensory neurons, and altered intestinal motility and secretion. Genetic manipulation of S. aureus revealed two key quorum sensing-regulated classes of pore forming toxins that mediate excitation and inhibition of extrinsic sensory nerves, respectively. As such, bacterial mediators have the potential to directly modulate gut-brain communication to influence intestinal symptoms and reflex function in vivo, contributing to homeostatic, behavioral, and sensory consequences of infection.
Collapse
Affiliation(s)
- Friederike Uhlig
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| | - Luke Grundy
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, SA, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Sonia Garcia-Caraballo
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, SA, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Stuart M. Brierley
- Visceral Pain Research Group, College of Medicine and Public Health, Flinders Health and Medical Research Institute (FHMRI), Flinders University, Bedford Park, SA, Australia
- Hopwood Centre for Neurobiology, Lifelong Health Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, Adelaide, SA, Australia
- Discipline of Medicine, University of Adelaide, Adelaide, SA, Australia
| | - Simon J. Foster
- Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK
- Florey Institute, University of Sheffield, Sheffield, UK
| | - David Grundy
- Department of Biomedical Science, University of Sheffield, Sheffield, UK
| |
Collapse
|
43
|
Perry IE, Sonu I, Scarpignato C, Akiyama J, Hongo M, Vega KJ. Potential proton pump inhibitor-related adverse effects. Ann N Y Acad Sci 2020; 1481:43-58. [PMID: 32761834 DOI: 10.1111/nyas.14428] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 05/31/2020] [Accepted: 06/12/2020] [Indexed: 12/11/2022]
Abstract
Proton pump inhibitors (PPIs) are one of the most common medications taken by patients worldwide. PPIs are used to treat acid-related disorders, including gastroesophageal reflux disease, peptic ulcer disease, Helicobacter pylori infection, and nonsteroidal anti-inflammatory drug/stress ulceration. For some of these diseases, long-term treatment is necessary. With such prolonged use, concern and investigation into potential adverse effects has increased. In addition, data are available regarding potential anticancer effects of PPIs, especially regarding solid tumors. The aim of this review is to assess the literature on PPIs with regard to common concerns, such as drug-drug interactions, the intestinal microbiome, dementia and central nervous system disease, and osteoporosis, as well as to highlight potential negative and positive impacts of the drug in cancer.
Collapse
Affiliation(s)
- Issac E Perry
- Division of Gastroenterology and Hepatology, Augusta University-Medical College of Georgia, Augusta, Georgia
| | - Irene Sonu
- Division of Gastroenterology and Hepatology, Stanford University, Redwood City, California
| | - Carmelo Scarpignato
- Department of Health Sciences, United Campus of Malta, Msida, Malta.,Faculty of Medicine, Chinese University of Hong Kong, ShaTin, Hong Kong
| | - Junichi Akiyama
- Division of Gastroenterology and Hepatology, National Center for Global Health and Medicine, Tokyo, Japan
| | - Michio Hongo
- Department of Comprehensive Medicine, Tohoku University School of Medicine, Sendai, Miyagi, Japan.,Department of Medicine, Kurokawa General Hospital, Kurokawa, Miyagi, Japan
| | - Kenneth J Vega
- Division of Gastroenterology and Hepatology, Augusta University-Medical College of Georgia, Augusta, Georgia
| |
Collapse
|
44
|
Effect of Gluten-Free Diet on Gut Microbiota Composition in Patients with Celiac Disease and Non-Celiac Gluten/Wheat Sensitivity. Nutrients 2020; 12:nu12061832. [PMID: 32575561 PMCID: PMC7353361 DOI: 10.3390/nu12061832] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/16/2020] [Accepted: 06/17/2020] [Indexed: 02/07/2023] Open
Abstract
Celiac disease (CD) and non-celiac gluten/wheat sensitivity (NCG/WS) are the two most frequent conditions belonging to gluten-related disorders (GRDs). Both these diseases are triggered and worsened by gluten proteins ingestion, although other components, such as amylase/trypsin inhibitors (ATI) and fermentable oligosaccharides, disaccharides, monosaccharides and polyols (FODMAPs), seem to be involved in the NCG/WS onset. Therefore, the only effective treatment to date is the long-life adherence to a strictly gluten-free diet. Recently, increasing attention has been paid to the intestinal barrier, a dynamic system comprising various components, which regulate the delicate crosstalk between metabolic, motor, neuroendocrine and immunological functions. Among the elements characterizing the intestinal barrier, the microbiota plays a key role, modulating the gut integrity maintenance, the immune response and the inflammation process, linked to the CD and NCG/WS outbreak. This narrative review addresses the most recent findings on the gut microbiota modulation induced by the gluten-free diet (GFD) in healthy, CD and NCG/WS patients.
Collapse
|
45
|
Hadi A, Ghaedi E, Khalesi S, Pourmasoumi M, Arab A. Effects of synbiotic consumption on lipid profile: a systematic review and meta-analysis of randomized controlled clinical trials. Eur J Nutr 2020; 59:2857-2874. [DOI: 10.1007/s00394-020-02248-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 04/01/2020] [Indexed: 12/17/2022]
|
46
|
Napoli E, Siracusa L, Ruberto G. New Tricks for Old Guys: Recent Developments in the Chemistry, Biochemistry, Applications and Exploitation of Selected Species from the Lamiaceae Family. Chem Biodivers 2020; 17:e1900677. [PMID: 31967708 DOI: 10.1002/cbdv.201900677] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 01/21/2020] [Indexed: 12/13/2022]
Abstract
Lamiaceae is one of the largest families of flowering plants comprising about 250 genera and over 7,000 species. Most of the plants of this family are aromatic and therefore important source of essential oils. Lamiaceae are widely used as culinary herbs and reported as medicinal plants in several folk traditions. In the Mediterranean area oregano, sage, rosemary, thyme and lavender stand out for geographical diffusion and variety of uses. The aim of this review is to provide recent data dealing with the phytochemical and pharmacological studies, and the more recent applications of the essential oils and the non-volatile phytocomplexes. This literature survey suggests how the deeper understanding of biomolecular processes in the health and food sectors as per as pest control bioremediation of cultural heritage, or interaction with human microbiome, fields, leads to the rediscovery and new potential applications of well-known plants.
Collapse
Affiliation(s)
- Edoardo Napoli
- Istituto del CNR di Chimica Biomolecolare, Via Paolo Gaifami, 18, IT-95126, Catania, Italy
| | - Laura Siracusa
- Istituto del CNR di Chimica Biomolecolare, Via Paolo Gaifami, 18, IT-95126, Catania, Italy
| | - Giuseppe Ruberto
- Istituto del CNR di Chimica Biomolecolare, Via Paolo Gaifami, 18, IT-95126, Catania, Italy
| |
Collapse
|
47
|
Alizadeh M, Shojadoost B, Astill J, Taha-Abdelaziz K, Karimi SH, Bavananthasivam J, Kulkarni RR, Sharif S. Effects of in ovo Inoculation of Multi-Strain Lactobacilli on Cytokine Gene Expression and Antibody-Mediated Immune Responses in Chickens. Front Vet Sci 2020; 7:105. [PMID: 32185187 PMCID: PMC7058628 DOI: 10.3389/fvets.2020.00105] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Accepted: 02/11/2020] [Indexed: 01/12/2023] Open
Abstract
This study was conducted to investigate the effects of various doses of a multi-strain lactobacilli mixture (Lactobacillus salivarius, Lactobacillus reuteri, Lactobacillus crispatus, and Lactobacillus johnsonii) on the innate and adaptive immune responses in broiler chickens. At embryonic day eighteen, 200 eggs were injected with PBS, or three different doses of a multi-strain lactobacilli mixture (1 × 105, 1 × 106, and 1 × 107 CFU/egg, P1, P2, and P3 respectively) along with a group of negative control. On days 5 and 10 post-hatch, cecal tonsil, bursa of fabricius, and spleen were collected for gene expression and cellular analysis. On days 14 and 21 post-hatch, birds were immunized intramuscularly with both sheep red blood cells (SRBC) and keyhole limpet hemocyanin (KLH). Serum samples were collected on days 0, 7, 14, and 21 after primary immunization. The results demonstrated that lactobacilli inoculation increased the splenic expression of cytokines, including interferon (IFN) - α, IFN-β, IFN-γ, interleukin (IL)-8, and IL-12 on day 5 post-hatch compared to the control group (PBS). However, in cecal tonsils, lactobacilli treatment downregulated the expression of IL-6 on day 5 post-hatch and IL-2 and IL-8 on day 10 post-hatch. No significant differences were observed in the expression of cytokine genes in the bursa except for IL-13 which was upregulated in lactobacilli-treated groups P2 and P3 on days 5 and 10 post-hatch. Flow cytometry analysis showed that the percentage of KUL01, CD4+ and CD8+ splenocytes was not affected by treatments. In addition, no significant differences were observed for antibody titers against SRBC. However, lactobacilli treatment (P1, P2, and P3) was found to increase IgM titers on day 21 post-primary immunization compared to controls. Furthermore, in ovo injection of the highest dose of probiotics (1 × 107, P3) increased serum IgG titers against KLH on day 7 post-primary immunization. In conclusion, this study demonstrated that that in ovo administration of lactobacilli can improve antibody-mediated immune responses and differentially modulate cytokine expression in mucosal and systemic lymphoid tissues of chickens.
Collapse
Affiliation(s)
- Mohammadali Alizadeh
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Bahram Shojadoost
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Jake Astill
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Khaled Taha-Abdelaziz
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
- Department of Pathology, Faculty of Veterinary Medicine, Beni-Suef University, Beni Suef, Egypt
| | - Seyed Hossein Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| | - Jegarubee Bavananthasivam
- Department of Pathology and Molecular Medicine, McMaster Immunology Research Centre, M. G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Raveendra R. Kulkarni
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, Canada
| |
Collapse
|
48
|
Bascuñán KA, Araya M, Roncoroni L, Doneda L, Elli L. Dietary Gluten as a Conditioning Factor of the Gut Microbiota in Celiac Disease. Adv Nutr 2020; 11:160-174. [PMID: 31399743 PMCID: PMC7442381 DOI: 10.1093/advances/nmz080] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/12/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022] Open
Abstract
The gut microbiota plays a relevant role in determining an individual's health status, and the diet is a major factor in modulating the composition and function of gut microbiota. Gluten constitutes an essential dietary component in Western societies and is the environmental trigger of celiac disease. The presence/absence of gluten in the diet can change the diversity and proportions of the microbial communities constituting the gut microbiota. There is an intimate relation between gluten metabolism and celiac disease pathophysiology and gut microbiota; their interrelation defines intestinal health and homeostasis. Environmental factors modify the intestinal microbiota and, in turn, its changes modulate the mucosal and immune responses. Current evidence from studies of young and adult patients with celiac disease increasingly supports that dysbiosis (i.e., compositional and functional alterations of the gut microbiome) is present in celiac disease, but to what extent this is a cause or consequence of the disease and whether the different intestinal diseases (celiac disease, ulcerative colitis, Crohn disease) have specific change patterns is not yet clear. The use of bacterial-origin enzymes that help completion of gluten digestion is of interest because of the potential application as coadjuvant in the current treatment of celiac disease. In this narrative review, we address the current knowledge on the complex interaction between gluten digestion and metabolism, celiac disease, and the intestinal microbiota.
Collapse
Affiliation(s)
- Karla A Bascuñán
- Department of Nutrition, School of Medicine, University of Chile, Santiago, Chile
- Centre for the Prevention and Diagnosis of Celiac Disease/Gastroenterology II, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, and Università degli Studi di Milano, Milan, Italy
| | - Magdalena Araya
- Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Leda Roncoroni
- Centre for the Prevention and Diagnosis of Celiac Disease/Gastroenterology II, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, and Università degli Studi di Milano, Milan, Italy
- Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Luisa Doneda
- Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Luca Elli
- Centre for the Prevention and Diagnosis of Celiac Disease/Gastroenterology II, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, and Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
49
|
Akhtar M, Haleem A, Anvari S, Nazli A, Sager M. An analysis of gut dysbiosis in obesity, diabetes, and chronic gut conditions. IBNOSINA JOURNAL OF MEDICINE AND BIOMEDICAL SCIENCES 2020. [DOI: 10.4103/ijmbs.ijmbs_102_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
50
|
Golofast B, Vales K. The connection between microbiome and schizophrenia. Neurosci Biobehav Rev 2019; 108:712-731. [PMID: 31821833 DOI: 10.1016/j.neubiorev.2019.12.011] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 12/01/2019] [Accepted: 12/06/2019] [Indexed: 12/15/2022]
Abstract
There has been an accumulation of knowledge about the human microbiome, some detailed investigations of the gastrointestinal microbiota and its functions, and the highlighting of complex interactions between the gut, the gut microbiota, and the central nervous system. That assumes the involvement of the microbiome in the pathogenesis of various CNS diseases, including schizophrenia. Given this information and the fact, that the gut microbiota is sensitive to internal and environmental influences, we have speculated that among the factors that influence the formation and composition of gut microbiota during life, possible key elements in the schizophrenia development chain are hidden where gut microbiota is a linking component. This article aims to describe and understand the developmental relationships between intestinal microbiota and the risk of developing schizophrenia.
Collapse
Affiliation(s)
- Bogdana Golofast
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Prague East, Czech Republic; Third Faculty of Medicine, Charles University, Ruská 87, 100 00 Prague 10, Czech Republic.
| | - Karel Vales
- National Institute of Mental Health, Topolova 748, 250 67 Klecany, Prague East, Czech Republic
| |
Collapse
|