1
|
Ahrend H, Buchholtz A, Stope MB. Microbiome and Mucosal Immunity in the Intestinal Tract. In Vivo 2025; 39:17-24. [PMID: 39740876 PMCID: PMC11705094 DOI: 10.21873/invivo.13801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/11/2024] [Accepted: 11/06/2024] [Indexed: 01/02/2025]
Abstract
The human bowel is exposed to numerous biotic and abiotic external noxious agents. Accordingly, the digestive tract is frequently involved in malfunctions within the organism. Together with the commensal intestinal flora, it regulates the immunological balance between inflammatory defense processes and immune tolerance. Pathological changes in this system often cause chronic inflammatory bowel diseases including Crohn's disease and ulcerative colitis. This review article highlights the complex interaction between commensal microorganisms, the intestinal microbiome, and the intestinal epithelium-localized local immune system. The main functions of the human intestinal microbiome include (i) protection against pathogenic microbial colonization, (ii) maintenance of the barrier function of the intestinal epithelium, (iii) degradation and absorption of nutrients and (iv) active regulation of the intestinal immunity. The local intestinal immune system consists primarily of macrophages, antigen-presenting cells, and natural killer cells. These cells regulate the commensal intestinal microbiome and are in turn regulated by signaling factors of the epithelial cells and the microbiome. Deregulated immune responses play an important role and can lead to both reduced activity of the commensal microbiome and pathologically increased activity of harmful microorganisms. These aspects of chronic inflammatory bowel disease have become the focus of attention in recent years. It is therefore important to consider the immunological-microbial context in both the diagnosis and treatment of inflammatory bowel diseases. A promising holistic approach would include the most comprehensive possible diagnosis of the immune and microbiome status of the patient, both at the time of diagnostics and during therapy.
Collapse
Affiliation(s)
- Hannes Ahrend
- Department of Medicine, Israelite Hospital Hamburg, Hamburg, Germany
| | - Anja Buchholtz
- Department of Medicine, Israelite Hospital Hamburg, Hamburg, Germany
| | - Matthias B Stope
- Department of Gynecology and Gynecological Oncology, Research Laboratories, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
2
|
Brenchley JM, Serrano-Villar S. From dysbiosis to defense: harnessing the gut microbiome in HIV/SIV therapy. MICROBIOME 2024; 12:113. [PMID: 38907315 PMCID: PMC11193286 DOI: 10.1186/s40168-024-01825-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 04/26/2024] [Indexed: 06/23/2024]
Abstract
BACKGROUND Although the microbiota has been extensively associated with HIV pathogenesis, the majority of studies, particularly those using omics techniques, are largely correlative and serve primarily as a basis for hypothesis generation. Furthermore, most have focused on characterizing the taxonomic composition of the bacterial component, often overlooking other levels of the microbiome. The intricate mechanisms by which the microbiota influences immune responses to HIV are still poorly understood. Interventional studies on gut microbiota provide a powerful tool to test the hypothesis of whether we can harness the microbiota to improve health outcomes in people with HIV. RESULTS Here, we review the multifaceted role of the gut microbiome in HIV/SIV disease progression and its potential as a therapeutic target. We explore the complex interplay between gut microbial dysbiosis and systemic inflammation, highlighting the potential for microbiome-based therapeutics to open new avenues in HIV management. These include exploring the efficacy of probiotics, prebiotics, fecal microbiota transplantation, and targeted dietary modifications. We also address the challenges inherent in this research area, such as the difficulty in inducing long-lasting microbiome alterations and the complexities of study designs, including variations in probiotic strains, donor selection for FMT, antibiotic conditioning regimens, and the hurdles in translating findings into clinical practice. Finally, we speculate on future directions for this rapidly evolving field, emphasizing the need for a more granular understanding of microbiome-immune interactions, the development of personalized microbiome-based therapies, and the application of novel technologies to identify potential therapeutic agents. CONCLUSIONS Our review underscores the importance of the gut microbiome in HIV/SIV disease and its potential as a target for innovative therapeutic strategies.
Collapse
Affiliation(s)
- Jason M Brenchley
- Barrier Immunity Section, Lab of Viral Diseases, NIAID, NIH, Bethesda, MA, USA.
| | - Sergio Serrano-Villar
- Department of Infectious Diseases, Hospital Universitario Ramon y Cajal, IRYCIS and CIBERInfec, Madrid, Spain.
| |
Collapse
|
3
|
Cai J, Auster A, Cho S, Lai Z. Dissecting the human gut microbiome to better decipher drug liability: A once-forgotten organ takes center stage. J Adv Res 2023; 52:171-201. [PMID: 37419381 PMCID: PMC10555929 DOI: 10.1016/j.jare.2023.07.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 05/25/2023] [Accepted: 07/03/2023] [Indexed: 07/09/2023] Open
Abstract
BACKGROUND The gut microbiome is a diverse system within the gastrointestinal tract composed of trillions of microorganisms (gut microbiota), along with their genomes. Accumulated evidence has revealed the significance of the gut microbiome in human health and disease. Due to its ability to alter drug/xenobiotic pharmacokinetics and therapeutic outcomes, this once-forgotten "metabolic organ" is receiving increasing attention. In parallel with the growing microbiome-driven studies, traditional analytical techniques and technologies have also evolved, allowing researchers to gain a deeper understanding of the functional and mechanistic effects of gut microbiome. AIM OF REVIEW From a drug development perspective, microbial drug metabolism is becoming increasingly critical as new modalities (e.g., degradation peptides) with potential microbial metabolism implications emerge. The pharmaceutical industry thus has a pressing need to stay up-to-date with, and continue pursuing, research efforts investigating clinical impact of the gut microbiome on drug actions whilst integrating advances in analytical technology and gut microbiome models. Our review aims to practically address this need by comprehensively introducing the latest innovations in microbial drug metabolism research- including strengths and limitations, to aid in mechanistically dissecting the impact of the gut microbiome on drug metabolism and therapeutic impact, and to develop informed strategies to address microbiome-related drug liability and minimize clinical risk. KEY SCIENTIFIC CONCEPTS OF REVIEW We present comprehensive mechanisms and co-contributing factors by which the gut microbiome influences drug therapeutic outcomes. We highlight in vitro, in vivo, and in silico models for elucidating the mechanistic role and clinical impact of the gut microbiome on drugs in combination with high-throughput, functionally oriented, and physiologically relevant techniques. Integrating pharmaceutical knowledge and insight, we provide practical suggestions to pharmaceutical scientists for when, why, how, and what is next in microbial studies for improved drug efficacy and safety, and ultimately, support precision medicine formulation for personalized and efficacious therapies.
Collapse
Affiliation(s)
- Jingwei Cai
- Drug Metabolism & Pharmacokinetics, Genentech Inc., South San Francisco, CA 94080, USA.
| | - Alexis Auster
- Drug Metabolism & Pharmacokinetics, Genentech Inc., South San Francisco, CA 94080, USA
| | - Sungjoon Cho
- Drug Metabolism & Pharmacokinetics, Genentech Inc., South San Francisco, CA 94080, USA
| | - Zijuan Lai
- Drug Metabolism & Pharmacokinetics, Genentech Inc., South San Francisco, CA 94080, USA
| |
Collapse
|
4
|
Singh RP, Kumari K. Bacterial type VI secretion system (T6SS): an evolved molecular weapon with diverse functionality. Biotechnol Lett 2023; 45:309-331. [PMID: 36683130 DOI: 10.1007/s10529-023-03354-2] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 12/14/2022] [Accepted: 01/05/2023] [Indexed: 01/24/2023]
Abstract
Bacterial secretion systems are nanomolecular complexes that release a diverse set of virulence factors/or proteins into its surrounding or translocate to their target host cells. Among these systems, type VI secretion system 'T6SS' is a recently discovered molecular secretion system which is widely distributed in Gram-negative (-ve) bacteria, and shares structural similarity with the puncturing device of bacteriophages. The presence of T6SS is an advantage to many bacteria as it delivers toxins to its neighbour pathogens for competitive survival, and also translocates protein effectors to the host cells, leading to disruption of lipid membranes, cell walls, and cytoskeletons etc. Recent studies have characterized both anti-prokaryotic and anti-eukaryotic effectors, where T6SS is involved in diverse cellular functions including favouring colonization, enhancing the survival, adhesive modifications, internalization, and evasion of the immune system. With the evolution of advanced genomics and proteomics tools, there has been an increase in the number of characterized T6SS effector arsenals and also more clear information about the adaptive significance of this complex system. The functions of T6SS are generally regulated at the transcription, post-transcription and post-translational levels through diverse mechanisms. In the present review, we aimed to provide information about the distribution of T6SS in diverse bacteria, any structural similarity/or dissimilarity, effectors proteins, functional significance, and regulatory mechanisms. We also tried to provide information about the diverse roles played by T6SS in its natural environments and hosts, and further any changes in the microbiome.
Collapse
Affiliation(s)
- Rajnish Prakash Singh
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India.
| | - Kiran Kumari
- Department of Bioengineering and Biotechnology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| |
Collapse
|
5
|
Rodrigues A, Gonçalves A, Morais J, Araujo R, Falcão-Pires I. Diet-Induced Microbiome's Impact on Heart Failure: A Double-Edged Sword. Nutrients 2023; 15:1223. [PMID: 36904222 PMCID: PMC10004801 DOI: 10.3390/nu15051223] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/06/2023] Open
Abstract
Heart failure (HF) is a debilitating disease with a significant clinical and economic impact worldwide. Multiple factors seem to increase the risk of developing HF, such as hypertension, obesity and diabetes. Since chronic inflammation plays a significant role in HF pathophysiology and gut dysbiosis is associated with low-grade chronic inflammation, the risk of cardiovascular diseases is likely modulated by the gut microbiome (GM). Considerable progress has been made in HF management. However, there is a need to find new strategies to reduce mortality and increase the quality of life, mainly of HFpEF patients, since its prevalence continues to rise. Recent studies validate that lifestyle changes, such as diet modulation, represent a potential therapeutic approach to improve several cardiometabolic diseases, although their effects on the GM and its indirect cardiac impact still warrant further research. Hence, in this paper, we aim to clarify the link between HF and the human microbiome.
Collapse
Affiliation(s)
- Alexandre Rodrigues
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- i3S-Institute for Research & Innovation in Health, University of Porto, 4200-135 Porto, Portugal
- Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
| | - Alexandre Gonçalves
- Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
| | - Juliana Morais
- Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- Cintesis@RISE, Center for Health Technology and Services Research, 4200-450 Porto, Portugal
| | - Ricardo Araujo
- INEB-Institute of Biomedical Engineering, University of Porto, 4200-135 Porto, Portugal
- i3S-Institute for Research & Innovation in Health, University of Porto, 4200-135 Porto, Portugal
| | - Inês Falcão-Pires
- Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
- UnIC@RISE, Department of Surgery and Physiology, Faculty of Medicine of the University of Porto, 4200-319 Porto, Portugal
| |
Collapse
|
6
|
Pandey H, Tang DWT, Wong SH, Lal D. Gut Microbiota in Colorectal Cancer: Biological Role and Therapeutic Opportunities. Cancers (Basel) 2023; 15:cancers15030866. [PMID: 36765824 PMCID: PMC9913759 DOI: 10.3390/cancers15030866] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/23/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Colorectal cancer (CRC) is the second-leading cause of cancer-related deaths worldwide. While CRC is thought to be an interplay between genetic and environmental factors, several lines of evidence suggest the involvement of gut microbiota in promoting inflammation and tumor progression. Gut microbiota refer to the ~40 trillion microorganisms that inhabit the human gut. Advances in next-generation sequencing technologies and metagenomics have provided new insights into the gut microbial ecology and have helped in linking gut microbiota to CRC. Many studies carried out in humans and animal models have emphasized the role of certain gut bacteria, such as Fusobacterium nucleatum, enterotoxigenic Bacteroides fragilis, and colibactin-producing Escherichia coli, in the onset and progression of CRC. Metagenomic studies have opened up new avenues for the application of gut microbiota in the diagnosis, prevention, and treatment of CRC. This review article summarizes the role of gut microbiota in CRC development and its use as a biomarker to predict the disease and its potential therapeutic applications.
Collapse
Affiliation(s)
- Himani Pandey
- Redcliffe Labs, Electronic City, Noida 201301, India
| | - Daryl W. T. Tang
- School of Biological Sciences, Nanyang Technological University, Singapore 308232, Singapore
| | - Sunny H. Wong
- Centre for Microbiome Medicine, Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore 308232, Singapore
- Correspondence: (S.H.W.); (D.L.)
| | - Devi Lal
- Department of Zoology, Ramjas College, University of Delhi, Delhi 110007, India
- Correspondence: (S.H.W.); (D.L.)
| |
Collapse
|
7
|
Verberk JDM, van Rooden SM, Hetem DJ, Wunderink HF, Vlek ALM, Meijer C, van Ravensbergen EAH, Huijskens EGW, Vainio SJ, Bonten MJM, van Mourik MSM. Reliability and validity of multicentre surveillance of surgical site infections after colorectal surgery. Antimicrob Resist Infect Control 2022; 11:10. [PMID: 35063009 PMCID: PMC8780777 DOI: 10.1186/s13756-022-01050-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 12/23/2021] [Indexed: 01/23/2023] Open
Abstract
Background Surveillance is the cornerstone of surgical site infection prevention programs. The validity of the data collection and awareness of vulnerability to inter-rater variation is crucial for correct interpretation and use of surveillance data. The aim of this study was to investigate the reliability and validity of surgical site infection (SSI) surveillance after colorectal surgery in the Netherlands. Methods In this multicentre prospective observational study, seven Dutch hospitals performed SSI surveillance after colorectal surgeries performed in 2018 and/or 2019. When executing the surveillance, a local case assessment was performed to calculate the overall percentage agreement between raters within hospitals. Additionally, two case-vignette assessments were performed to estimate intra-rater and inter-rater reliability by calculating a weighted Cohen’s Kappa and Fleiss’ Kappa coefficient. To estimate the validity, answers of the two case-vignettes questionnaires were compared with the answers of an external medical panel. Results 1111 colorectal surgeries were included in this study with an overall SSI incidence of 8.8% (n = 98). From the local case assessment it was estimated that the overall percent agreement between raters within a hospital was good (mean 95%, range 90–100%). The Cohen’s Kappa estimated for the intra-rater reliability of case-vignette review varied from 0.73 to 1.00, indicating substantial to perfect agreement. The inter-rater reliability within hospitals showed more variation, with Kappa estimates ranging between 0.61 and 0.94. In total, 87.9% of the answers given by the raters were in accordance with the medical panel. Conclusions This study showed that raters were consistent in their SSI-ascertainment (good reliability), but improvements can be made regarding the accuracy (moderate validity). Accuracy of surveillance may be improved by providing regular training, adapting definitions to reduce subjectivity, and by supporting surveillance through automation.
Collapse
|
8
|
Huang Z, Huang Y, Chen J, Tang Z, Chen Y, Liu H, Huang M, Qing L, Li L, Wang Q, Jia B. The role and therapeutic potential of gut microbiome in severe burn. Front Cell Infect Microbiol 2022; 12:974259. [PMID: 36467727 PMCID: PMC9714625 DOI: 10.3389/fcimb.2022.974259] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 10/21/2022] [Indexed: 05/07/2025] Open
Abstract
Severe burn is a serious acute trauma that can lead to significant complications such as sepsis, multiple organ failure, and high mortality worldwide. The gut microbiome, the largest microbial reservoir in the human body, plays a significant role in this pathogenic process. Intestinal dysbiosis and disruption of the intestinal mucosal barrier are common after severe burn, leading to bacterial translocation to the bloodstream and other organs of the body, which is associated with many subsequent severe complications. The progression of some intestinal diseases can be improved by modulating the composition of gut microbiota and the levels of its metabolites, which also provides a promising direction for post-burn treatment. In this article, we summarised the studies describing changes in the gut microbiome after severe burn, as well as changes in the function of the intestinal mucosal barrier. Additionally, we presented the potential and challenges of microbial therapy, which may provide microbial therapy strategies for severe burn.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Qin Wang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| | - Bo Jia
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
9
|
Nibali L, Gkranias N, Mainas G, Di Pino A. Periodontitis and implant complications in diabetes. Periodontol 2000 2022; 90:88-105. [PMID: 35913467 DOI: 10.1111/prd.12451] [Citation(s) in RCA: 72] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Epidemiologic evidence indicates that periodontitis is more frequent in patients with uncontrolled diabetes mellitus than in healthy controls, suggesting that it could be considered the "sixth complication" of diabetes. Actually, diabetes mellitus and periodontitis are two extraordinarily prevalent chronic diseases that share a number of comorbidities all converging toward an increased risk of cardiovascular disease. Periodontal treatment has recently been shown to have the potential to improve the metabolic control of diabetes, although long-term studies are lacking. Uncontrolled diabetes also seems to affect the response to periodontal treatment, as well as the risk to develop peri-implant diseases. Mechanisms of associations between diabetes mellitus and periodontal disease include the release of advanced glycation end products as a result of hyperglycemia and a range of shared predisposing factors of genetic, microbial, and lifestyle nature. This review discusses the evidence for the risk of periodontal and peri-implant disease in diabetic patients and the potential role of the dental professional in the diabetes-periodontal interface.
Collapse
Affiliation(s)
- Luigi Nibali
- Periodontology Unit, Centre for Host Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Nikolaos Gkranias
- Centre for Immunobiology and Regenerative Medicine and Centre for Oral Clinical Research, Institute of Dentistry, Queen Mary University London (QMUL), London, UK
| | - Giuseppe Mainas
- Periodontology Unit, Centre for Host Microbiome Interactions, Faculty of Dentistry, Oral & Craniofacial Sciences, King's College London, London, UK
| | - Antonino Di Pino
- Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| |
Collapse
|
10
|
Luck ME, Tao J, Lake EP. The Skin and Gut Microbiome in Hidradenitis Suppurativa: Current Understanding and Future Considerations for Research and Treatment. Am J Clin Dermatol 2022; 23:841-852. [DOI: 10.1007/s40257-022-00724-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/11/2022] [Indexed: 11/30/2022]
|
11
|
Bacterial Compositional Shifts of Gut Microbiomes in Patients with Rheumatoid Arthritis in Association with Disease Activity. Microorganisms 2022; 10:microorganisms10091820. [PMID: 36144422 PMCID: PMC9505928 DOI: 10.3390/microorganisms10091820] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/02/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is a chronic inflammatory disabling autoimmune disorder. Little is known regarding the association between the gut microbiome and etiopathogenesis of RA. We aimed to dissect the differences in gut microbiomes associated with RA in comparison to healthy individuals and, in addition, to identify the shifts in the bacterial community in association with disease activity; Methods: In order to identify compositional shifts in gut microbiomes of RA patients, V3-V4 hypervariable regions of 16S rRNA were sequenced using Illumina MiSeq. In total, sixty stool samples were collected from 45 patients with RA besides 15 matched healthy subjects; Results: Notably, RA microbiomes were significantly associated with diverse bacterial communities compared with healthy individuals. Likewise, a direct association between bacterial diversity and disease activity was detected in RA patients (Kruskal Wallis; p = 0.00047). In general, genus-level analysis revealed a positive coexistence between RA and Megasphaera, Adlercreutzia, Ruminococcus, Bacteroides, Collinsella, and Acidaminococcus. Furthermore, Spearman correlation analysis significantly stratified the most dominant genera into distinct clusters that were mainly based on disease activity (r ≥ 0.6; p ≤ 0.05). The predictive metabolic profile of bacterial communities associated with RA could support the potential impact of gut microbiomes in either the development or recovery of RA; Conclusions: The overall shifts in bacterial composition at different disease statuses could confirm the cross-linking of certain genera either to causation or progression of RA.
Collapse
|
12
|
Ulusan Bagci O, Caner A. The interaction of gut microbiota with parasitic protozoa. J Parasit Dis 2022; 46:8-11. [PMID: 35299914 PMCID: PMC8901934 DOI: 10.1007/s12639-021-01443-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022] Open
Abstract
The human intestinal microbiota is composed of a complex combination of microorganisms including bacteria, virus, and eukaryotes. The microbiota plays a critical role in homeostasis through creating a mucosal barrier, providing protective responses to pathogens, and affecting the immune system and metabolism of the host. Molecules secreted by parasites can alter composition of microbiota both by acting directly on the microbial community and indirectly by affecting the host physiology. On the other hand, the microbiota composition can affect the survival, physiology, and virulence of many parasitic protozoa. Explanation of possible interactions between the microbiota, immune response, and protozoa may further clarify the underlying mechanisms of infectivity, clinical variations, and life-cycle of protozoa.
Collapse
Affiliation(s)
- Ozlem Ulusan Bagci
- grid.8302.90000 0001 1092 2592Department of Parasitology, Faculty of Medicine, Ege University Medical School, 35100 Bornova, Izmir, Turkey
| | - Ayse Caner
- grid.8302.90000 0001 1092 2592Department of Parasitology, Faculty of Medicine, Ege University Medical School, 35100 Bornova, Izmir, Turkey ,grid.8302.90000 0001 1092 2592Cancer Research Center, Ege University, Izmir, Turkey ,grid.240145.60000 0001 2291 4776Departments of Experimental Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX USA
| |
Collapse
|
13
|
Neophytou C, Pitsouli C. How Gut Microbes Nurture Intestinal Stem Cells: A Drosophila Perspective. Metabolites 2022; 12:169. [PMID: 35208243 PMCID: PMC8878600 DOI: 10.3390/metabo12020169] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 02/07/2022] [Accepted: 02/08/2022] [Indexed: 11/16/2022] Open
Abstract
Host-microbiota interactions are key modulators of host physiology and behavior. Accumulating evidence suggests that the complex interplay between microbiota, diet and the intestine controls host health. Great emphasis has been given on how gut microbes have evolved to harvest energy from the diet to control energy balance, host metabolism and fitness. In addition, many metabolites essential for intestinal homeostasis are mainly derived from gut microbiota and can alleviate nutritional imbalances. However, due to the high complexity of the system, the molecular mechanisms that control host-microbiota mutualism, as well as whether and how microbiota affects host intestinal stem cells (ISCs) remain elusive. Drosophila encompasses a low complexity intestinal microbiome and has recently emerged as a system that might uncover evolutionarily conserved mechanisms of microbiota-derived nutrient ISC regulation. Here, we review recent studies using the Drosophila model that directly link microbiota-derived metabolites and ISC function. This research field provides exciting perspectives for putative future treatments of ISC-related diseases based on monitoring and manipulating intestinal microbiota.
Collapse
Affiliation(s)
| | - Chrysoula Pitsouli
- Department of Biological Sciences, University of Cyprus, 1 University Avenue, Aglantzia, Nicosia 2109, Cyprus;
| |
Collapse
|
14
|
Love CJ, Masson BA, Gubert C, Hannan AJ. The microbiota-gut-brain axis in Huntington's disease. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2022; 167:141-184. [DOI: 10.1016/bs.irn.2022.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
15
|
Intestinal microbiota and their metabolic contribution to type 2 diabetes and obesity. J Diabetes Metab Disord 2021; 20:1855-1870. [PMID: 34900829 PMCID: PMC8630233 DOI: 10.1007/s40200-021-00858-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023]
Abstract
Obesity and type 2 diabetes mellitus (T2DM) are common, chronic metabolic disorders with associated significant long-term health problems at global epidemic levels. It is recognised that gut microbiota play a central role in maintaining host homeostasis and through technological advances in both animal and human models it is becoming clear that gut microbiota are heavily involved in key pathophysiological roles in the aetiology and progression of both conditions. This review will focus on current knowledge regarding microbiota interactions with short chain fatty acids, the host inflammatory response, signaling pathways, integrity of the intestinal barrier, the interaction of the gut-brain axis and the subsequent impact on the metabolic health of the host.
Collapse
|
16
|
Diao H, Yan J, Li S, Kuang S, Wei X, Zhou M, Zhang J, Huang C, He P, Tang W. Effects of Dietary Zinc Sources on Growth Performance and Gut Health of Weaned Piglets. Front Microbiol 2021; 12:771617. [PMID: 34858378 PMCID: PMC8631109 DOI: 10.3389/fmicb.2021.771617] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/11/2021] [Indexed: 12/21/2022] Open
Abstract
The present study aimed to investigate the effects of dietary zinc sources on the growth performance and gut health of weaned piglets. In total, 96 Duroc × Landrace × Yorkshire (DLY) weaned piglets with an initial average body weight of 8.81±0.42kg were divided into four groups, with six replicates per treatment and four pigs per replicate. The dietary treatment groups were as follows: (1) control group, basal diet; (2) zinc sulphate (ZnSO4) group, basal diet +100mg/kg ZnSO4; (3) glycine zinc (Gly-Zn) group, basal diet +100mg/kg Gly-Zn and (4) zinc lactate group, and basal diet +100mg/kg zinc lactate. The whole trial lasted for 28days. Decreased F/G was noted in the Gly-Zn and zinc lactate groups (p<0.05). The zinc lactate group had a lower diarrhea rate than the control group (p<0.05). Moreover, the ZnSO4, Gly-Zn, and zinc lactate groups had significantly higher apparent total tract digestibility of dry matter (DM), crude protein (CP), ether extract (EE), crude ash, and zinc than the control group (p<0.05). The Gly-Zn and zinc lactate groups had higher jejunal villus height and a higher villus height:crypt depth ratio than the control group (p<0.05). In addition, the ZnSO4, Gly-Zn and zinc lactate groups had a significantly lower mRNA expression level of jejunal ZRT/IRT-like protein 4 (ZIP4) and higher mRNA expression level of jejunal interleukin-1β (IL-1β) than the control group (p<0.05). The mRNA expression level of jejunal zinc transporter 2 (ZNT2) was higher and that of jejunal Bcl-2-associated X protein (Bax) was lower in the Gly-Zn and zinc lactate groups than in the control group (p<0.05). Moreover, the zinc lactate group had a higher count of Lactobacillus spp. in the cecal digesta and higher mRNA expression levels of jejunal occludin and mucin 2 (MUC2) than the control group (p<0.05). In conclusion, dietary supplementation with 100mg/kg ZnSO4, Gly-Zn, or zinc lactate could improve the growth performance and gut barrier function of weaned piglets. Dietary supplementation with organic zinc, particularly zinc lactate, had the best effect.
Collapse
Affiliation(s)
- Hui Diao
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Academy of Animal Science, Chengdu, China
| | - Jiayou Yan
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Academy of Animal Science, Chengdu, China
| | - Shuwei Li
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Academy of Animal Science, Chengdu, China.,Sichuan Animtech Biology Development Co., Ltd, Chengdu, China
| | - Shengyao Kuang
- Livestock and Poultry Biological Products Key Laboratory of Sichuan Province, Sichuan Animtech Feed Co., Ltd, Chengdu, China
| | - Xiaolan Wei
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Academy of Animal Science, Chengdu, China
| | - Mengjia Zhou
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Academy of Animal Science, Chengdu, China
| | - Jinxiu Zhang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Academy of Animal Science, Chengdu, China
| | - Chongbo Huang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Academy of Animal Science, Chengdu, China
| | - Peng He
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Academy of Animal Science, Chengdu, China
| | - Wenjie Tang
- Animal Breeding and Genetics Key Laboratory of Sichuan Province, Sichuan Academy of Animal Science, Chengdu, China
| |
Collapse
|
17
|
Gallegos-Monterrosa R, Coulthurst SJ. The ecological impact of a bacterial weapon: microbial interactions and the Type VI secretion system. FEMS Microbiol Rev 2021; 45:fuab033. [PMID: 34156081 PMCID: PMC8632748 DOI: 10.1093/femsre/fuab033] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 06/20/2021] [Indexed: 12/13/2022] Open
Abstract
Bacteria inhabit all known ecological niches and establish interactions with organisms from all kingdoms of life. These interactions are mediated by a wide variety of mechanisms and very often involve the secretion of diverse molecules from the bacterial cells. The Type VI secretion system (T6SS) is a bacterial protein secretion system that uses a bacteriophage-like machinery to secrete a diverse array of effectors, usually translocating them directly into neighbouring cells. These effectors display toxic activity in the recipient cell, making the T6SS an effective weapon during inter-bacterial competition and interactions with eukaryotic cells. Over the last two decades, microbiology research has experienced a shift towards using systems-based approaches to study the interactions between diverse organisms and their communities in an ecological context. Here, we focus on this aspect of the T6SS. We consider how our perspective of the T6SS has developed and examine what is currently known about the impact that bacteria deploying the T6SS can have in diverse environments, including niches associated with plants, insects and mammals. We consider how T6SS-mediated interactions can affect host organisms by shaping their microbiota, as well as the diverse interactions that can be established between different microorganisms through the deployment of this versatile secretion system.
Collapse
Affiliation(s)
| | - Sarah J Coulthurst
- School of Life Sciences, University of Dundee, Dow Street, Dundee DD1 5EH, UK
| |
Collapse
|
18
|
Chen YT, Chiou SY, Hsu AH, Lin YC, Lin JS. Lactobacillus rhamnosus Strain LRH05 Intervention Ameliorated Body Weight Gain and Adipose Inflammation via Modulating the Gut Microbiota in High-Fat Diet-Induced Obese Mice. Mol Nutr Food Res 2021; 66:e2100348. [PMID: 34796638 DOI: 10.1002/mnfr.202100348] [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: 04/13/2021] [Revised: 10/13/2021] [Indexed: 12/21/2022]
Abstract
SCOPE This study aims to investigate the underlying mechanism of a specific probiotic strain on suppression of adipogenesis and inflammatory response in white adipose tissue (WAT) of high-fat diet (HFD)-fed mice. METHODS AND RESULTS Eight strains are screened in vitro for candidates of potential probiotics. Lactobacillus rhamnosus LRH05 (LRH05) and Lactobacillus reuteri LR47 (LR47) are screened out with lower triglyceride expression in vitro. The mice are fed a control diet (CD), HFD, or HFD supplemented with a dose of LRH05 or LR47 at 109 CFU per mouse per day for 10 weeks (n = 8), respectively. The results demonstrate that LRH05, but not LR47, significantly reduce body weight gain and the weight of WAT, as well as improve hepatic steatosis and glucose intolerance. LRH05 regulates the Mogat1, Igf-1, Mcp-1, and F4/80 mRNA expression and decreases macrophage infiltration in WAT. LRH05 shows an increase in butyric and propionic acid-producing bacteria, including Lachnoclostridium, Romboutsia, and Fusobacterium that is coincident with the increased fecal propionic acid and butyric acid levels. CONCLUSION LRH05 shows a strain-specific effect on ameliorating the pro-inflammatory process by reducing inflammatory macrophage infiltration and the expression of inflammation-related genes in mice. Thus, LRH05 can be considered a potential probiotic strain to prevent obesity.
Collapse
Affiliation(s)
- Yung-Tsung Chen
- Culture Collection & Research Institute, SYNBIO TECH INC., Kaohsiung City, Taiwan
| | - Shiou-Yun Chiou
- Culture Collection & Research Institute, SYNBIO TECH INC., Kaohsiung City, Taiwan
| | - Ai-Hua Hsu
- Culture Collection & Research Institute, SYNBIO TECH INC., Kaohsiung City, Taiwan
| | - Yu-Chun Lin
- Livestock Research Institute, Council of Agriculture, Executive Yuan, Tainan, Taiwan
| | - Jin-Seng Lin
- Culture Collection & Research Institute, SYNBIO TECH INC., Kaohsiung City, Taiwan
| |
Collapse
|
19
|
Enterocytes in Food Hypersensitivity Reactions. Animals (Basel) 2021; 11:ani11092713. [PMID: 34573679 PMCID: PMC8466009 DOI: 10.3390/ani11092713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 09/05/2021] [Accepted: 09/10/2021] [Indexed: 11/18/2022] Open
Abstract
Simple Summary Hypersensitivity to food, affecting both animals and humans, is increasing. Until a decade ago, it was thought that enterocytes, the most abundant constituent of the intestinal surface mucosa layer, served only to absorb digested food and prevent foreign and non-digested substances from passing below the intestinal layer. Growing evidence supports the involvement of enterocytes in immunological responses. Here, we present a comprehensive review of the new roles of enterocytes in food hypersensitivity conducted in animal models in order to better understand complicated immune pathological conditions. In addition, resources for further work in this area are suggested, along with a literature overview of the specific roles of enterocytes in maintaining oral tolerance. Lastly, it will be beneficial to investigate the various animal models involved in food hypersensitivity to reach the needed momentum necessary for the complete and profound understanding of the mechanisms of the ever-growing number of food allergies in animal and human populations. Abstract Food hypersensitivity reactions are adverse reactions to harmless dietary substances, whose causes are hidden within derangements of the complex immune machinery of humans and mammals. Until recently, enterocytes were considered as solely absorptive cells providing a physical barrier for unwanted lumen constituents. This review focuses on the enterocytes, which are the hub for innate and adaptive immune reactions. Furthermore, the ambiguous nature of enterocytes is also reflected in the fact that enterocytes can be considered as antigen-presenting cells since they constitutively express major histocompatibility complex (MHC) class II molecules. Taken together, it becomes clear that enterocytes have an immense role in maintaining oral tolerance to foreign antigens. In general, the immune system and its mechanisms underlying food hypersensitivity are still unknown and the involvement of components belonging to other anatomical systems, such as enterocytes, in these mechanisms make their elucidation even more difficult. The findings from studies with animal models provide us with valuable information about allergic mechanisms in the animal world, while on the other hand, these models are used to extrapolate results to the pathological conditions occurring in humans. There is a constant need for studies that deal with this topic and can overcome the glitches related to ethics in working with animals.
Collapse
|
20
|
Abstract
ABSTRACT Burn injuries are a common form of traumatic injury that leads to significant morbidity and mortality worldwide. Burn injuries are characterized by inflammatory processes and alterations in numerous organ systems and functions. Recently, it has become apparent that the gastrointestinal bacterial microbiome is a key component of regulating the immune response and recovery from burn and can also contribute to significant detrimental sequelae after injury, such as sepsis and multiple organ failure. Microbial dysbiosis has been linked to multiple disease states; however, its role in exacerbating acute traumatic injuries, such as burn, is poorly understood. In this article, we review studies that document changes in the intestinal microbiome after burn injury, assess the implications in post-burn pathogenesis, and the potential for further discovery and research.
Collapse
Affiliation(s)
- Marisa E. Luck
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Alcohol Research Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Integrative Cell Biology Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| | - Caroline J. Herrnreiter
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Alcohol Research Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Biochemistry and Molecular Biology Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| | - Mashkoor A. Choudhry
- Burn & Shock Trauma Research Institute, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Alcohol Research Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Department of Surgery, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Department of Microbiology and Immunology, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Integrative Cell Biology Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
- Biochemistry and Molecular Biology Program, Stritch School of Medicine, Loyola University Chicago Health Sciences Division, Maywood, IL 60153, USA
| |
Collapse
|
21
|
Feng J, Tang YN, Zhou LX, Pan JS. Standardized Nursing Procedures for Fecal Microbiota Transplantation via Upper Endoscopy. Gastroenterol Nurs 2021; 44:227-232. [PMID: 34176888 DOI: 10.1097/sga.0000000000000577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 10/20/2020] [Indexed: 11/25/2022] Open
Abstract
Fecal microbiota transplantation is an emerging treatment option that lacks a standardized nursing procedure. In our department, fecal microbiota transplantation has been undertaken to treat chronic hepatitis B and inflammatory bowel diseases since 2015. The fecal microbiota transplantation process involves various nursing measures that are critical for the successful completion of the procedures. In our center, a set of standardized nursing procedures has been established and has proved effective and operable. Standardized nursing procedures enhance the efficacy of fecal microbiota transplantation and alleviate the risk of treatment-related complications.
Collapse
Affiliation(s)
- Juan Feng
- Juan Feng, BSc, is Specialist Nurse of Endoscopy, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Yun-Na Tang, BSc, is Specialist Nurse of Endoscopy, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Li-Xiang Zhou, BSc, is Head Nurse of Endoscopy and Associate Professor of Nursing, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Jin-Shui Pan, PhD, MD, is Associate Professor of Hepatology, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Yun-Na Tang
- Juan Feng, BSc, is Specialist Nurse of Endoscopy, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Yun-Na Tang, BSc, is Specialist Nurse of Endoscopy, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Li-Xiang Zhou, BSc, is Head Nurse of Endoscopy and Associate Professor of Nursing, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Jin-Shui Pan, PhD, MD, is Associate Professor of Hepatology, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Li-Xiang Zhou
- Juan Feng, BSc, is Specialist Nurse of Endoscopy, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Yun-Na Tang, BSc, is Specialist Nurse of Endoscopy, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Li-Xiang Zhou, BSc, is Head Nurse of Endoscopy and Associate Professor of Nursing, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Jin-Shui Pan, PhD, MD, is Associate Professor of Hepatology, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| | - Jin-Shui Pan
- Juan Feng, BSc, is Specialist Nurse of Endoscopy, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Yun-Na Tang, BSc, is Specialist Nurse of Endoscopy, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Li-Xiang Zhou, BSc, is Head Nurse of Endoscopy and Associate Professor of Nursing, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
- Jin-Shui Pan, PhD, MD, is Associate Professor of Hepatology, Department of Gastroenterology, Zhongshan Hospital Affiliated to Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
22
|
AB-Kefir Reduced Body Weight and Ameliorated Inflammation in Adipose Tissue of Obese Mice Fed a High-Fat Diet, but Not a High-Sucrose Diet. Nutrients 2021; 13:nu13072182. [PMID: 34202894 PMCID: PMC8308298 DOI: 10.3390/nu13072182] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/17/2021] [Accepted: 06/22/2021] [Indexed: 12/18/2022] Open
Abstract
Consumption of different types of high-calorie foods leads to the development of various metabolic disorders. However, the effects of multi-strain probiotics on different types of diet-induced obesity and intestinal dysbiosis remain unclear. In this study, mice were fed a control diet, high-fat diet (HFD; 60% kcal fat and 20% kcal carbohydrate), or western diet (WD; 40% kcal fat and 43% kcal carbohydrate) and administered with multi-strain AB-Kefir containing six strains of lactic acid bacteria and a Bifidobacterium strain, at 109 CFU per mouse for 10 weeks. Results demonstrated that AB-Kefir reduced body weight gain, glucose intolerance, and hepatic steatosis with a minor influence on gut microbiota composition in HFD-fed mice, but not in WD-fed mice. In addition, AB-Kefir significantly reduced the weight and size of adipose tissues by regulating the expression of CD36, Igf1, and Pgc1 in HFD-fed mice. Although AB-Kefir did not reduce the volume of white adipose tissue, it markedly regulated CD36, Dgat1 and Mogat1 mRNA expression. Moreover, the abundance of Eubacterium_coprostanoligenes_group and Ruminiclostridium significantly correlated with changes in body weight, liver weight, and fasting glucose in test mice. Overall, this study provides important evidence to understand the interactions between probiotics, gut microbiota, and diet in obesity treatment.
Collapse
|
23
|
Jia PP, Junaid M, Wen PP, Yang YF, Li WG, Yang XG, Pei DS. Role of germ-free animal models in understanding interactions of gut microbiota to host and environmental health: A special reference to zebrafish. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 279:116925. [PMID: 33744636 DOI: 10.1016/j.envpol.2021.116925] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 02/22/2021] [Accepted: 03/03/2021] [Indexed: 05/07/2023]
Abstract
Numerous pieces of evidence documented the importance of gut microbiota in regulating human health and evaluating the toxicity of environmental pollutants, which are closely related to the host health in various aspects, including nutrition, energy translation, metabolism, pathogen resistance, and immune function. A variety of environmental factors can disrupt gut microbiota and their functions, and inevitably cause immune diseases, obesity and diabetes. However, deciphering the inner mechanisms involved in the functional interaction of gut microbes with host health is still needed extensive investigations. This review focused on the essential roles of intestinal microbes in host-related diseases and highlighted the development and applications of germ-free (GF) animal models, mainly zebrafish. Moreover, the generation, immunity characters, advantages and challenges of GF zebrafish models were also summarized. Importantly, the composition and isolation of zebrafish gut bacteria for further application and toxicity evaluation of aquatic environmental pollutants were also discussed. In conclusion, GF zebrafish play irreplaceable roles in understanding the potential functions and responses of customized microbiota towards human and environmental health implications.
Collapse
Affiliation(s)
- Pan-Pan Jia
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Muhammad Junaid
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Ping-Ping Wen
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Yi-Fan Yang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Wei-Guo Li
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - Xian-Guang Yang
- College of Life Science, Henan Normal University, Xinxiang, 453007, China
| | - De-Sheng Pei
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China; College of Life Science, Henan Normal University, Xinxiang, 453007, China.
| |
Collapse
|
24
|
Chang TE, Luo JC, Yang UC, Huang YH, Hou MC, Lee FY. Fecal microbiota profile in patients with inflammatory bowel disease in Taiwan. J Chin Med Assoc 2021; 84:580-587. [PMID: 33871395 DOI: 10.1097/jcma.0000000000000532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Inflammatory bowel disease (IBD) is a chronic inflammatory disease associated with complicated interaction between immune, gut microbiota, and environmental factors in a genetically vulnerable host. Dysbiosis is often seen in patients with IBD. We aimed to investigate the fecal microbiota in patients with IBD and compared them with a control group in Taiwan. METHODS In this cross-sectional study, we investigated fecal microbiota in 20 patients with IBD and 48 healthy controls. Fecal samples from both IBD patients and controls were analyzed by the next-generation sequencing method and relevant software. RESULTS The IBD group showed lower bacterial richness and diversity compared with the control group. The principal coordinate analysis also revealed the significant structural differences between the IBD group and the control group. These findings were consistent whether the analysis was based on an operational taxonomic unit or amplicon sequence variant. However, no significant difference was found when comparing the composition of fecal microbiota between ulcerative colitis (UC) and Crohn's disease (CD). Further analysis showed that Lactobacillus, Enterococcus, and Bifidobacterium were dominant in the IBD group, whereas Faecalibacterium and Subdoligranulum were dominant in the control group at the genus level. When comparing UC, CD, and control group, Lactobacillus, Bifidobacterium, and Enterococcus were identified as dominant genera in the UC group. Fusobacterium and Escherichia_Shigella were dominant in the CD group. CONCLUSION Compared with the healthy control, the IBD group showed dysbiosis with a significant decrease in both richness and diversity of gut microbiota.
Collapse
Affiliation(s)
- Tien-En Chang
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- Endoscopic Center for Diagnosis and Therapy, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- National Yang Ming Chiao Tung University, School of Medicine, Taipei, Taiwan, ROC
| | - Jiing-Chyuan Luo
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- National Yang Ming Chiao Tung University, School of Medicine, Taipei, Taiwan, ROC
- Keelung Hospital, Ministry of Health Welfare, Keelung, Taiwan, ROC
| | - Ueng-Cheng Yang
- National Yang Ming Chiao Tung University, School of Medicine, Institute of Biomedical Informatics, Taipei, Taiwan, ROC
| | - Yi-Hsiang Huang
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- National Yang Ming Chiao Tung University, School of Medicine, Taipei, Taiwan, ROC
| | - Ming-Chih Hou
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- National Yang Ming Chiao Tung University, School of Medicine, Taipei, Taiwan, ROC
| | - Fa-Yauh Lee
- Division of Gastroenterology and Hepatology, Taipei Veterans General Hospital, Taipei, Taiwan, ROC
- National Yang Ming Chiao Tung University, School of Medicine, Taipei, Taiwan, ROC
| |
Collapse
|
25
|
Bui TPN, de Vos WM. Next-generation therapeutic bacteria for treatment of obesity, diabetes, and other endocrine diseases. Best Pract Res Clin Endocrinol Metab 2021; 35:101504. [PMID: 33785319 DOI: 10.1016/j.beem.2021.101504] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The human gut microbiota has appeared as an important factor affecting host health and intestinal bacteria have recently emerged as potential therapeutics to treat diabetes and other endocrine diseases. These mainly anaerobic bacteria have been identified either via comparative "omics" analysis of the intestinal microbiota in healthy and diseased subjects or of data collected by fecal microbiota transplantation studies. Both approaches require advanced and in-depth sequencing technologies to perform massive genomic screening to select bacteria with potential benefits. It has been shown that these potentially therapeutic bacteria can either produce bioactive products that directly influence the host patho-physiology and endocrine systems or produce specific signaling molecules that may do so. These bioactive compounds can be formed via degradation of dietary or host-derived components or the conversion of intermediate compounds produced by fermentation of intestinal bacteria. Several of these bacteria have shown causality in preclinical models and entered clinical phase studies, while their mode of action is being analyzed. In this review, we summarize the research on the most promising bacterial candidates with therapeutic properties with a specific focus on diabetes.
Collapse
Affiliation(s)
- Thi Phuong Nam Bui
- Laboratory of Microbiology, Wageningen University, Stippeneng 4, 6708, WE, Wageningen, the Netherlands; Caelus Pharmaceuticals BV, 3474, KG, Zegveld, the Netherlands
| | - Willem M de Vos
- Laboratory of Microbiology, Wageningen University, Stippeneng 4, 6708, WE, Wageningen, the Netherlands; Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
26
|
Sharma VK, Singh TG, Garg N, Dhiman S, Gupta S, Rahman MH, Najda A, Walasek-Janusz M, Kamel M, Albadrani GM, Akhtar MF, Saleem A, Altyar AE, Abdel-Daim MM. Dysbiosis and Alzheimer's Disease: A Role for Chronic Stress? Biomolecules 2021; 11:678. [PMID: 33946488 PMCID: PMC8147174 DOI: 10.3390/biom11050678] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/23/2021] [Accepted: 04/26/2021] [Indexed: 02/06/2023] Open
Abstract
Alzheimer's disease (AD) is an incurable, neuropsychiatric, pathological condition that deteriorates the worth of geriatric lives. AD is characterized by aggregated senile amyloid plaques, neurofibrillary tangles, neuronal loss, gliosis, oxidative stress, neurotransmitter dysfunction, and bioenergetic deficits. The changes in GIT composition and harmony have been recognized as a decisive and interesting player in neuronal pathologies including AD. Microbiota control and influence the oxidoreductase status, inflammation, immune system, and the endocrine system through which it may have an impact on the cognitive domain. The altered and malfunctioned state of microbiota is associated with minor infections to complicated illnesses that include psychosis and neurodegeneration, and several studies show that microbiota regulates neuronal plasticity and neuronal development. The altered state of microbiota (dysbiosis) may affect behavior, stress response, and cognitive functions. Chronic stress-mediated pathological progression also has a well-defined role that intermingles at various physiological levels and directly impacts the pathological advancement of AD. Chronic stress-modulated alterations affect the well-established pathological markers of AD but also affect the gut-brain axis through the mediation of various downstream signaling mechanisms that modulate the microbial commensals of GIT. The extensive literature reports that chronic stressors affect the composition, metabolic activities, and physiological role of microbiota in various capacities. The present manuscript aims to elucidate mechanistic pathways through which stress induces dysbiosis, which in turn escalates the neuropathological cascade of AD. The stress-dysbiosis axis appears a feasible zone of work in the direction of treatment of AD.
Collapse
Affiliation(s)
- Vivek Kumar Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (V.K.S.); (N.G.); (S.D.); (S.G.)
- Goverment College of Pharmacy, District Shimla, Rohru 171207, India
| | - Thakur Gurjeet Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (V.K.S.); (N.G.); (S.D.); (S.G.)
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (V.K.S.); (N.G.); (S.D.); (S.G.)
| | - Sonia Dhiman
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (V.K.S.); (N.G.); (S.D.); (S.G.)
| | - Saurabh Gupta
- Chitkara College of Pharmacy, Chitkara University, Punjab 140401, India; (V.K.S.); (N.G.); (S.D.); (S.G.)
| | - Md. Habibur Rahman
- Department of Pharmacy, Southeast University, Banani, Dhaka 1213, Bangladesh;
| | - Agnieszka Najda
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland; (A.N.); (M.W.-J.)
| | - Magdalena Walasek-Janusz
- Laboratory of Quality of Vegetables and Medicinal Plants, Department of Vegetable Crops and Medicinal Plants, University of Life Sciences in Lublin, 15 Akademicka Street, 20-950 Lublin, Poland; (A.N.); (M.W.-J.)
| | - Mohamed Kamel
- Department of Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt;
| | - Ghadeer M. Albadrani
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11474, Saudi Arabia;
| | - Muhammad Furqan Akhtar
- Riphah Institute of Pharmaceutical Sciences, Riphah International University, Lahore Campus, Lahore 54950, Pakistan;
| | - Ammara Saleem
- Department of Pharmacology, Faculty of Pharmaceutical Sciences, Government College University Faisalabad, Faisalabad 38000, Pakistan;
| | - Ahmed E. Altyar
- Department of Pharmacy Practice, Faculty of Pharmacy, King Abdulaziz University, P.O. Box 80260, Jeddah 21589, Saudi Arabia;
| | - Mohamed M. Abdel-Daim
- Pharmacology Department, Faculty of Veterinary Medicine, Suez Canal University, Ismailia 41522, Egypt
| |
Collapse
|
27
|
Effects of bioactive components of Pu-erh tea on gut microbiomes and health: A review. Food Chem 2021; 353:129439. [PMID: 33743430 DOI: 10.1016/j.foodchem.2021.129439] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/19/2021] [Accepted: 02/21/2021] [Indexed: 12/20/2022]
Abstract
Pu-erh tea is a post-fermentation tea with unique flavor and multiple health benefits. Due to the various microorganisms involved in the post-fermentation process, Pu-erh tea contains highly complex components, which have rich interactions with the gut microbiomes (GMs). Because the structure and homeostasis of GMs are closely related to human wellness and the various diseases progress, the beneficial effects of Pu-erh tea on GMs have a great potential for application in health care. However, there is no systematic summary of the bioactive components of Pu-erh tea, and their effects on the GMs. Here, we review the current studies on the effects of Pu-erh tea and its bioactive components on the structure of GMs as well as on health improvement, and further discuss the relevant quality indicators. This "components - function - indicators" clue will hopefully stimulate the standardization of Pu-erh tea fermentation process and the development of its functional products.
Collapse
|
28
|
Rashidinejad A, Bahrami A, Rehman A, Rezaei A, Babazadeh A, Singh H, Jafari SM. Co-encapsulation of probiotics with prebiotics and their application in functional/synbiotic dairy products. Crit Rev Food Sci Nutr 2020; 62:2470-2494. [PMID: 33251846 DOI: 10.1080/10408398.2020.1854169] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Oral administration of live probiotics along with prebiotics has been suggested with numerous beneficial effects for several conditions including certain infectious disorders, diarrheal illnesses, some inflammatory bowel diseases, and most recently, irritable bowel syndrome. Though, delivery of such viable bacteria to the host intestine is a major challenge, due to the poor survival of the ingested probiotic bacteria during the gastric transit, especially within the stomach where the pH is highly acidic. Although microencapsulation has been known as a promising approach for improving the viability of probiotics in the human digestive tract, the success rate is not satisfactory. For this reason, co-encapsulation of probiotics with probiotics has been practised as a novel alternative approach for further improvement of the oral delivery of viable probiotics toward their targeted release in the host intestine. This paper discusses the co-encapsulation technologies used for delivery of probiotics toward better stability and viability, as well the incorporation of co-encapsulated probiotics and prebiotics in functional/synbiotic dairy foods. The common encapsulation technologies (and the materials) used for this purpose, the stability and survival of co-encapsulated probiotics in the food, and the release behavior of the co-encapsulated probiotics in the gastrointestinal tract have also been explained. Most studies reported a significant improvement particularly in the viability of bacteria associated with the presence of prebiotics. Nevertheless, the previous research has mostly been carried out in the simulated digestion, meaning that future systematic research is to be carried out to investigate the efficacy of the co-encapsulation on the survival of the bacteria in the gut in vivo.
Collapse
Affiliation(s)
- Ali Rashidinejad
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Akbar Bahrami
- Program of Applied Science and Technology, Center for Excellence in Post-Harvest Technologies, North Carolina Agricultural and Technical State University, North Carolina Research Campus, Kannapolis, North Carolina, USA
| | - Abdur Rehman
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Jiangsu, People's Republic of China
| | - Atefe Rezaei
- Department of Food Science and Technology, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran.,Food Security Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Afshin Babazadeh
- Center for Motor Neuron Disease Research, Faculty of medicine, health and human sciences, Macquarie University, Sydney, New South Wales, Australia
| | - Harjinder Singh
- Riddet Institute, Massey University, Palmerston North, New Zealand
| | - Seid Mahdi Jafari
- Department of Food Materials & Process Design Engendering, Gorgan University of Agricultural Sciences and Natural Resources, Gorgan, Iran
| |
Collapse
|
29
|
Watson KM, Gaulke CA, Tsikitis VL. Understanding the microbiome: a primer on the role of the microbiome in colorectal neoplasia. Ann Gastroenterol 2020; 33:223-236. [PMID: 32382225 PMCID: PMC7196612 DOI: 10.20524/aog.2020.0467] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 01/24/2020] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer is a leading cause of cancer-related death internationally, with mounting evidence pointing to the role of the microbiome in adenoma and cancer development. This article aims to provide clinicians with a foundation for understanding the field of research into the microbiome. We also illustrate the various ways in which the microbiota have been linked to colorectal cancer, with a specific focus on microbiota with identified virulence factors, and also on the ways that byproducts of microbiota metabolism may result in oncogenesis. We also review strategies for manipulating the microbiome for therapeutic effects.
Collapse
Affiliation(s)
- Katherine M. Watson
- Department of Surgery, Oregon Health & Science University, Portland, OR (Katherine M. Watson, Vassiliki Liana Tsikitis)
| | | | - Vassiliki Liana Tsikitis
- Department of Surgery, Oregon Health & Science University, Portland, OR (Katherine M. Watson, Vassiliki Liana Tsikitis)
| |
Collapse
|
30
|
Kang B, Alvarado LJ, Kim T, Lehmann ML, Cho H, He J, Li P, Kim BH, Larochelle A, Kelsall BL. Commensal microbiota drive the functional diversification of colon macrophages. Mucosal Immunol 2020; 13:216-229. [PMID: 31772323 PMCID: PMC7039809 DOI: 10.1038/s41385-019-0228-3] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2019] [Revised: 10/01/2019] [Accepted: 10/24/2019] [Indexed: 02/04/2023]
Abstract
Mononuclear phagocytes are a heterogeneous population of leukocytes essential for immune homeostasis that develop tissue-specific functions due to unique transcriptional programs driven by local microenvironmental cues. Single cell RNA sequencing (scRNA-seq) of colonic myeloid cells from specific pathogen free (SPF) and germ-free (GF) C57BL/6 mice revealed extensive heterogeneity of both colon macrophages (MPs) and dendritic cells (DCs). Modeling of developmental pathways combined with inference of gene regulatory networks indicate two major trajectories from common CCR2+ precursors resulting in colon MP populations with unique transcription factors and downstream target genes. Compared to SPF mice, GF mice had decreased numbers of total colon MPs, as well as selective proportional decreases of two major CD11c+CD206intCD121b+ and CD11c-CD206hiCD121b- colon MP populations, whereas DC numbers and proportions were not different. Importantly, these two major colon MP populations were clearly distinct from other colon MP populations regarding their gene expression profile, localization within the lamina propria (LP) and ability to phagocytose macromolecules from the blood. These data uncover the diversity of intestinal myeloid cell populations at the molecular level and highlight the importance of microbiota on the unique developmental as well as anatomical and functional fates of colon MPs.
Collapse
Affiliation(s)
- Byunghyun Kang
- National Institute of Allergy and Infectious Diseases, Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA
| | - Luigi J Alvarado
- National Heart, Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA
| | - Teayong Kim
- San Diego State University, 5500 Campanile Dr., San Diego, CA, 92182, USA
| | | | - Hyeseon Cho
- National Institute of Allergy and Infectious Diseases, Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA
| | - Jianping He
- National Institute of Allergy and Infectious Diseases, Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA
| | - Peng Li
- National Heart, Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA
| | - Bong-Hyun Kim
- National Laboratory of Cancer Research, NIH, Frederick, MD, 21702, USA
| | - Andre Larochelle
- National Heart, Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA
| | - Brian L Kelsall
- National Institute of Allergy and Infectious Diseases, Lung and Blood Institute, NIH, Bethesda, MD, 20892, USA.
| |
Collapse
|
31
|
Cochrane K, Robinson AV, Holt RA, Allen-Vercoe E. A survey of Fusobacterium nucleatum genes modulated by host cell infection. Microb Genom 2020; 6:e000300. [PMID: 31661053 PMCID: PMC7067209 DOI: 10.1099/mgen.0.000300] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 09/10/2019] [Indexed: 12/23/2022] Open
Abstract
Here, we report comprehensive transcriptomic profiles from Fusobacterium nucleatum under conditions that mimic the first stages of bacterial infection in a highly differentiated adenocarcinoma epithelial cell line. Our transcriptomic in vitro adenocarcinoma approach allows us to measure the expression dynamics and regulation of bacterial virulence and response factors in real time, and is a novel strategy for clarifying the role of F. nucleatum infection in colorectal cancer (CRC) progression. Our data show that: (i) infection alters metabolic and functional pathways in F. nucleatum, allowing the bacterium to adapt to the host-imposed milieu; (ii) infection also stimulates the expression of genes required to help induce and promote a hypoxic and inflammatory microenvironment in the host; and (iii) F. nucleatum invasion occurs by a haematogenous route of infection. Our study identifies novel gene targets from F. nucleatum that are activated during invasion and which may aid in determining how this species invades and promotes disease within the human gastrointestinal tract. These invasion-specific genes may be useful as biomarkers for CRC progression in a host and could also assist in the development of new diagnostic tools and treatments (such as vaccines or small molecule drug targets), which will be able to combat infection and inflammation in the host while circumventing the potential problem of F. nucleatum tolerization.
Collapse
Affiliation(s)
- Kyla Cochrane
- Genome Sciences Center, BC Cancer Agency, Vancouver, British Columbia, V5Z 1L3, Canada
- Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, N1G 2W1, Canada
| | - Avery V. Robinson
- Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, N1G 2W1, Canada
| | - Robert A. Holt
- Molecular Biology and Biochemistry, Simon Fraser University, Burnaby, British Columbia, Canada
- Medical Genetics, University of British Columbia, Vancouver, British Columbia, Canada
- Canada's Michael Smith Genome Sciences Centre, Vancouver, British Columbia, Canada
| | - Emma Allen-Vercoe
- Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, N1G 2W1, Canada
| |
Collapse
|
32
|
Shavandi A, Saeedi P, Gérard P, Jalalvandi E, Cannella D, Bekhit AED. The role of microbiota in tissue repair and regeneration. J Tissue Eng Regen Med 2020; 14:539-555. [PMID: 31845514 DOI: 10.1002/term.3009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2019] [Revised: 09/15/2019] [Accepted: 10/28/2019] [Indexed: 12/22/2022]
Abstract
A comprehensive understanding of the human body endogenous microbiota is essential for acquiring an insight into the involvement of microbiota in tissue healing and regeneration process in order to enable development of biomaterials with a better integration with human body environment. Biomaterials used for biomedical applications are normally germ-free, and the human body as the host of the biomaterials is not germ-free. The complexity and role of the body microbiota in tissue healing/regeneration have been underestimated historically. Traditionally, studies aiming at the development of novel biomaterials had focused on the effects of environment within the target tissue, neglecting the signals generated from the microbiota and their impact on tissue regeneration. The significance of the human body microbiota in relation to metabolism, immune system, and consequently tissue regeneration has been recently realised and is a growing research field. This review summarises recent findings on the role of microbiota and mechanisms involved in tissue healing and regeneration, in particular skin, liver, bone, and nervous system regrowth and regeneration highlighting the potential new roles of microbiota for development of a new generation of biomaterials.
Collapse
Affiliation(s)
- Amin Shavandi
- BioMatter-BTL, École interfacultaire de Bioingénieurs (EIB), Université Libre de Brussels, Brussels, Belgium
| | - Pouya Saeedi
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Philippe Gérard
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | - Esmat Jalalvandi
- School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, UK
| | - David Cannella
- PhotoBioCatalysis Unit - BTL - École interfacultaire de Bioingénieurs (EIB), Université Libre de Brussels, Brussels, Belgium
| | | |
Collapse
|
33
|
Abstract
Stress is a nonspecific response of the body to any demand imposed upon it, disrupting the body homoeostasis and manifested with symptoms such as anxiety, depression or even headache. These responses are quite frequent in the present competitive world. The aim of this review is to explore the effect of stress on gut microbiota. First, we summarize evidence of where the microbiota composition has changed as a response to a stressful situation, and thereby the effect of the stress response. Likewise, we review different interventions that can modulate microbiota and could modulate the stress according to the underlying mechanisms whereby the gut-brain axis influences stress. Finally, we review both preclinical and clinical studies that provide evidence of the effect of gut modulation on stress. In conclusion, the influence of stress on gut microbiota and gut microbiota on stress modulation is clear for different stressors, but although the preclinical evidence is so extensive, the clinical evidence is more limited. A better understanding of the mechanism underlying stress modulation through the microbiota may open new avenues for the design of therapeutics that could boost the pursued clinical benefits. These new designs should not only focus on stress but also on stress-related disorders such as anxiety and depression, in both healthy individuals and different populations.
Collapse
|
34
|
Oral Vaccine Delivery: The Coming Age of Particulate Vaccines to Elicit Mucosal Immunity. MUCOSAL DELIVERY OF DRUGS AND BIOLOGICS IN NANOPARTICLES 2020. [DOI: 10.1007/978-3-030-35910-2_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
35
|
El Hadad S, Zakareya A, Al-Hejin A, Aldahlawi A, Alharbi M. Sustaining exposure to high concentrations of bifidobacteria inhibits gene expression of Mouse's mucosal immunity. Heliyon 2019; 5:e02866. [PMID: 31890933 PMCID: PMC6926234 DOI: 10.1016/j.heliyon.2019.e02866] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Revised: 07/23/2019] [Accepted: 11/12/2019] [Indexed: 12/28/2022] Open
Abstract
Numerous dietary products are supplemented with probiotics that may be beneficial for human health. Recently, bifidobacteria have received increasing attention as a genus of probiotic bacteria with high efficiency and few side effects. To examine potential effects of different bifidobacteria concentrations on the mucosal immune response, we fed mice with (a) 108 colony-forming units (CFU) of bifidobacteria (group 108B), and (b) with 1012 CFU of bifidobacteria (group 1012B) over 42 days and assessed gene expression in intestinal mucosa and immune marker concentrations in serum samples; ten untreated female mice were used as a control. Continuous exposure to 108 CFU of bifidobacteria activated both macrophages and Treg immune cells through significantly increasing the expression of mucosal TLR2 and IL10-mRNA genes, but inhibited Th1 and Th2 cells via significant downregulation of IL4 and IFNγ gene expression, compared to untreated mice. Interestingly, group 1012B showed down-regulated expression of TLR2, IL10, and IL4 genes but up-regulated expression of IFNγ, compared to group 108B and to the control. Also, polyclonal immunoglobulins IgG, IgM, and IgA showed a significant increase in all treated mice compared to the control. We conclude that high concentrations of bifidobacteria reduced innate immune functions. Furthermore, adaptive immunity seemed to be enhanced by increasing stimulation of T and B lymphocytes, suggesting aberration of the immune system following intestinal inflammation due to constant exposure to high concentrations of bifidobacteria. Both experimental bifidobacteria concentrations increased the total levels of circulating Igs, particularly of IgA.
Collapse
Affiliation(s)
- Sahar El Hadad
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Research Center of Genetic Engineering and Bioinformatics, VACSERA, Cairo, Egypt.,Immunolgy Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ayeshah Zakareya
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Ahmed Al-Hejin
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Microbiology Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alia Aldahlawi
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia.,Immunolgy Unit, King Fahad Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia.,Inflammatory Bowel Disease Research Group, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mona Alharbi
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
36
|
Diaz Carrasco JM, Casanova NA, Fernández Miyakawa ME. Microbiota, Gut Health and Chicken Productivity: What Is the Connection? Microorganisms 2019; 7:microorganisms7100374. [PMID: 31547108 PMCID: PMC6843312 DOI: 10.3390/microorganisms7100374] [Citation(s) in RCA: 217] [Impact Index Per Article: 36.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2019] [Revised: 08/31/2019] [Accepted: 09/06/2019] [Indexed: 01/13/2023] Open
Abstract
Gut microbiota and its relationship to animal health and productivity in commercial broiler chickens has been difficult to establish due to high variability between flocks, which derives from plenty of environmental, nutritional, and host factors that influence the load of commensal and pathogenic microbes surrounding birds during their growth cycle in the farms. Chicken gut microbiota plays a key role in the maintenance of intestinal health through its ability to modulate host physiological functions required to maintain intestinal homeostasis, mainly through competitive exclusion of detrimental microorganisms and pathogens, preventing colonization and therefore decreasing the expense of energy that birds normally invest in keeping the immune system active against these pathogens. Therefore, a “healthy” intestinal microbiota implies energy saving for the host which translates into an improvement in productive performance of the birds. This review compiles information about the main factors that shape the process of gut microbiota acquisition and maturation, their interactions with chicken immune homeostasis, and the outcome of these interactions on intestinal health and productivity.
Collapse
Affiliation(s)
- Juan M Diaz Carrasco
- Instituto de Patobiología Veterinaria, Centro Nacional de Investigaciones Agropecuarias, Instituto Nacional de Tecnología Agropecuaria, Calle Las Cabañas y Los Reseros s/n, Casilla de Correo 25, 1712 Castelar, Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas, Godoy Cruz 2290, 1425 Ciudad Autónoma de Buenos Aires, Argentina.
| | - Natalia A Casanova
- Instituto de Patobiología Veterinaria, Centro Nacional de Investigaciones Agropecuarias, Instituto Nacional de Tecnología Agropecuaria, Calle Las Cabañas y Los Reseros s/n, Casilla de Correo 25, 1712 Castelar, Buenos Aires, Argentina.
| | - Mariano E Fernández Miyakawa
- Instituto de Patobiología Veterinaria, Centro Nacional de Investigaciones Agropecuarias, Instituto Nacional de Tecnología Agropecuaria, Calle Las Cabañas y Los Reseros s/n, Casilla de Correo 25, 1712 Castelar, Buenos Aires, Argentina.
- Consejo Nacional de Investigaciones Científicas y Técnicas, Godoy Cruz 2290, 1425 Ciudad Autónoma de Buenos Aires, Argentina.
| |
Collapse
|
37
|
Biofabrication of Bacterial Constructs: New Three-Dimensional Biomaterials. Bioengineering (Basel) 2019; 6:bioengineering6020044. [PMID: 31091672 PMCID: PMC6631521 DOI: 10.3390/bioengineering6020044] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/10/2019] [Accepted: 05/10/2019] [Indexed: 12/22/2022] Open
Abstract
An enormous number of bacteria live in almost every environment; from deep oceans to below the surface of the earth or in our gastrointestinal tract. Although biofabrication is growing and maturing very quickly, the involvement of bacteria in this process has not been developed at a similar pace. From the development of a new generation of biomaterials to green bioremediation for the removal of hazardous environmental pollutants or to develop innovative food products in a recent trend, researchers have used cutting-edge biofabrication techniques to reveal the great potential of 3D structured bacterial constructs. These 3D bacterial workhouses may fundamentally change our approach toward biomaterials.
Collapse
|
38
|
Xi Y, Shuling N, Kunyuan T, Qiuyang Z, Hewen D, ChenCheng G, Tianhe Y, Liancheng L, Xin F. Characteristics of the intestinal flora of specific pathogen free chickens with age. Microb Pathog 2019; 132:325-334. [PMID: 31082529 DOI: 10.1016/j.micpath.2019.05.014] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Revised: 04/08/2019] [Accepted: 05/09/2019] [Indexed: 02/06/2023]
Abstract
Specific pathogen-free (SPF) experimental animals are recognized as standard laboratory animals in the fields of biomedical, animal husbandry and veterinary research and production. Intestinal flora plays a critical role in nutrient absorption, improving health and protecting the host from pathogens. We therefore explored the variation and maintenance of intestinal flora in SPF chicks in order to better understand the composition of intestinal microflorain SPF chickens, and provide reference for the study of intestinal flora of SPF experimental animals. Five chicks were randomly selected at each of 14, 28, and 42 days, and ceca were removed for DNA extraction. The Illumina Miseq platform was used for microbiome analysis of the V3-V4 region of the 16S rRNA gene. During the course of chick gut microbiome development, we observed major changes in diversity, especially between day 14 and day 28. Firmicutes, Proteobacteria, and Bacteroidetes were the main bacterial taxa, and Firmicutes increased significantly with age. The genus with the highest relative abundance was Lactobacillus, followed by Faecalibacterium. In addition, while abundance of Ruminococcaceae spp., Ruminococcus, and Blautia increased with age, Lactobacillus, Enterobacteriaceae spp., and Oscillospira decreased with age. Interestingly, the abundance of Faecalibacterium first increased and then decreased over time. The characteristics of SPF chicken gut flora at different ages establish a basis for the regulation of intestinal flora in the early stage of brooding, and also provide a theoretical foundation for controlling and preventing infections and poultry diseases in newborn chickens.
Collapse
Affiliation(s)
- Yu Xi
- College of Veterinary Medicine and College of Animal Science, Jilin University, Changchun, 130062, China
| | - Niu Shuling
- College of Animal Science and Technology, Changchun Sci-Tech University, Shuangyang, Jilin Province, 130600, China
| | - Tie Kunyuan
- College of Veterinary Medicine and College of Animal Science, Jilin University, Changchun, 130062, China
| | - Zhang Qiuyang
- College of Veterinary Medicine and College of Animal Science, Jilin University, Changchun, 130062, China
| | - Deng Hewen
- College of Veterinary Medicine and College of Animal Science, Jilin University, Changchun, 130062, China
| | - Gao ChenCheng
- College of Veterinary Medicine and College of Animal Science, Jilin University, Changchun, 130062, China
| | - Yu Tianhe
- College of Veterinary Medicine and College of Animal Science, Jilin University, Changchun, 130062, China
| | - Lei Liancheng
- College of Veterinary Medicine and College of Animal Science, Jilin University, Changchun, 130062, China
| | - Feng Xin
- College of Veterinary Medicine and College of Animal Science, Jilin University, Changchun, 130062, China.
| |
Collapse
|
39
|
Pichon M, Burucoa C. Impact of the Gastro-Intestinal Bacterial Microbiome on Helicobacter-Associated Diseases. Healthcare (Basel) 2019; 7:E34. [PMID: 30813360 PMCID: PMC6473412 DOI: 10.3390/healthcare7010034] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 02/18/2019] [Accepted: 02/20/2019] [Indexed: 12/15/2022] Open
Abstract
Helicobacter pylori is a bacterium that selectively infects the gastric epithelium of half of the world population. The microbiome, community of microorganisms gained major interest over the last years, due to its modification associated to health and disease states. Even if most of these descriptions have focused on chronic disorders, this review describes the impact of the intestinal bacterial microbiome on host response to Helicobacter associated diseases. Microbiome has a direct impact on host cells, major barrier of the gastro-intestinal tract, but also an indirect impact on immune system stimulation, by enhancing or decreasing non-specific or adaptive response. In microbial infections, especially in precancerous lesions induced by Helicobacter pylori infection, these modifications could lead to different outcome. Associated to data focusing on the microbiome, transcriptomic analyses of the eukaryote response would lead to a complete understanding of these complex interactions and will allow to characterize innovative biomarkers and personalized therapies.
Collapse
Affiliation(s)
- Maxime Pichon
- Bacteriology and Infection Control Laboratory, Infectious Agents Department, University Hospital of Poitiers, 86021 Poitiers, France.
- Laboratoire Inflammation, Tissus Épithéliaux et Cytokines, EA 4331, Faculté de Médecine et de Pharmacie, University of Poitiers, 86022 Poitiers, France.
| | - Christophe Burucoa
- Bacteriology and Infection Control Laboratory, Infectious Agents Department, University Hospital of Poitiers, 86021 Poitiers, France.
- Laboratoire Inflammation, Tissus Épithéliaux et Cytokines, EA 4331, Faculté de Médecine et de Pharmacie, University of Poitiers, 86022 Poitiers, France.
| |
Collapse
|
40
|
Dieterich W, Schink M, Zopf Y. Microbiota in the Gastrointestinal Tract. Med Sci (Basel) 2018; 6:medsci6040116. [PMID: 30558253 PMCID: PMC6313343 DOI: 10.3390/medsci6040116] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/13/2018] [Accepted: 12/13/2018] [Indexed: 02/07/2023] Open
Abstract
Gut microbiota are permanent residents of humans with the highest concentrations being found in human colon. Humans get the first contact with bacteria at delivery, and microbiota are subject of permanent change during the life. The individual microbiota pattern is highly variable and varying environmental conditions, e.g., diets, antigen exposure, infections, or medication, as well as genetics, age, or hygiene factors, strongly influence the bacterial community. A fine interaction between the host and microbiota determines the outcome of health or disease. The gut immune system is constantly challenged to distinguish between commensal non-invasive bacteria and potential pathogens. Goblet cells produce mucins that prevent most gut bacteria from penetrating through intestinal epithelial barrier, and Paneth cells are the main supplier of anti-microbial defensins. Gut epithelial and immune cells recognize bacteria via surface markers and they initiate an adequate immune answer. A dysbiosis is noticed in several diseases, but the crucial role in pathogenesis has to be proven. Prebiotics or probiotics are discussed as valuable tools to preserve or restore a healthy gut community.
Collapse
Affiliation(s)
- Walburga Dieterich
- Medical Clinic 1, Friedrich-Alexander-Universität Erlangen-Nürnberg; Ulmenweg 18, 91054 Erlangen, Germany.
- Hector Center of Excellence for Nutrition, Exercise and Sports, University of Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Monic Schink
- Medical Clinic 1, Friedrich-Alexander-Universität Erlangen-Nürnberg; Ulmenweg 18, 91054 Erlangen, Germany.
- Hector Center of Excellence for Nutrition, Exercise and Sports, University of Erlangen-Nürnberg, 91054 Erlangen, Germany.
| | - Yurdagül Zopf
- Medical Clinic 1, Friedrich-Alexander-Universität Erlangen-Nürnberg; Ulmenweg 18, 91054 Erlangen, Germany.
- Hector Center of Excellence for Nutrition, Exercise and Sports, University of Erlangen-Nürnberg, 91054 Erlangen, Germany.
| |
Collapse
|
41
|
In vitro and ex vivo systems at the forefront of infection modeling and drug discovery. Biomaterials 2018; 198:228-249. [PMID: 30384974 PMCID: PMC7172914 DOI: 10.1016/j.biomaterials.2018.10.030] [Citation(s) in RCA: 62] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2018] [Revised: 10/05/2018] [Accepted: 10/23/2018] [Indexed: 12/11/2022]
Abstract
Bacterial infections and antibiotic resistant bacteria have become a growing problem over the past decade. As a result, the Centers for Disease Control predict more deaths resulting from microorganisms than all cancers combined by 2050. Currently, many traditional models used to study bacterial infections fail to precisely replicate the in vivo bacterial environment. These models often fail to incorporate fluid flow, bio-mechanical cues, intercellular interactions, host-bacteria interactions, and even the simple inclusion of relevant physiological proteins in culture media. As a result of these inadequate models, there is often a poor correlation between in vitro and in vivo assays, limiting therapeutic potential. Thus, the urgency to establish in vitro and ex vivo systems to investigate the mechanisms underlying bacterial infections and to discover new-age therapeutics against bacterial infections is dire. In this review, we present an update of current in vitro and ex vivo models that are comprehensively changing the landscape of traditional microbiology assays. Further, we provide a comparative analysis of previous research on various established organ-disease models. Lastly, we provide insight on future techniques that may more accurately test new formulations to meet the growing demand of antibiotic resistant bacterial infections.
Collapse
|
42
|
Altamirano-Barrera A, Uribe M, Chávez-Tapia NC, Nuño-Lámbarri N. The role of the gut microbiota in the pathology and prevention of liver disease. J Nutr Biochem 2018; 60:1-8. [PMID: 29653359 DOI: 10.1016/j.jnutbio.2018.03.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2017] [Revised: 03/01/2018] [Accepted: 03/08/2018] [Indexed: 02/07/2023]
|
43
|
Coccia M. General properties of the evolution of research fields: a scientometric study of human microbiome, evolutionary robotics and astrobiology. Scientometrics 2018. [DOI: 10.1007/s11192-018-2902-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
44
|
Wang M, Chen Y, Wang Y, Li Y, Zhang X, Zheng H, Ma F, Ma C, Lu B, Xie Z, Liao Q. Beneficial changes of gut microbiota and metabolism in weaned rats with Lactobacillus acidophilus NCFM and Bifidobacterium lactis Bi-07 supplementation. J Funct Foods 2018. [DOI: 10.1016/j.jff.2018.07.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
45
|
Moreira GV, Azevedo FF, Ribeiro LM, Santos A, Guadagnini D, Gama P, Liberti EA, Saad M, Carvalho C. Liraglutide modulates gut microbiota and reduces NAFLD in obese mice. J Nutr Biochem 2018; 62:143-154. [PMID: 30292107 DOI: 10.1016/j.jnutbio.2018.07.009] [Citation(s) in RCA: 102] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2017] [Revised: 05/19/2018] [Accepted: 07/26/2018] [Indexed: 02/07/2023]
Abstract
Metabolic disorders such as insulin resistance and diabetes are associated with obesity and nonalcoholic fatty liver disease (NAFLD). The aggressive form of a fatty liver disease may progress to cirrhosis and hepatocellular carcinoma. Furthermore, recent studies demonstrated that there is a dysbiosis in the gut microbiota associated with early stages of metabolic disease. Therefore, the identification and repurposing of drugs already used to treat insulin resistance may be an excellent option for other disorders. We evaluated the effect of liraglutide on obesity, NAFLD and gut microbiota modulation in two different animal models of obesity: the ob/ob mice and the high-fat diet (HFD)-fed mice. Liraglutide treatment induced significant weight loss in both obesity models, showed improvements in glycemic parameters and reduced inflammatory cell infiltration in the cecum and the liver. In ob/ob mice, the liraglutide treatment was able to reduce the accumulation of liver fat by 78% and reversed steatosis in the HFD mice. The gut microbiota analysis showed that liraglutide changed the overall composition as well as the relative abundance of weight-relevant phylotypes such as a reduction of Proteobacteria and an increase of Akkermansia muciniphila in the treated HFD group. We show that liraglutide can lead to weight loss and gut microbiota modulations, and is associated with an improvement of NAFLD. Furthermore, by generating a profile of the intestinal microbiota, we compiled a list of potential bacterial targets that may modulate metabolism and induce a metabolic profile that is considered normal or clinically controlled.
Collapse
Affiliation(s)
- G V Moreira
- Institute Biomedical Sciences, University of Sao Paulo-Department of Physiology and Biophysical
| | - F F Azevedo
- State University of Campinas-School of Nursing
| | - L M Ribeiro
- Institute Biomedical Sciences, University of Sao Paulo-Department of Physiology and Biophysical
| | - A Santos
- Department of Internal Medicine, State University of Campinas
| | - D Guadagnini
- Department of Internal Medicine, State University of Campinas
| | - P Gama
- Institute Biomedical Sciences, University of Sao Paulo-Department of Cell and Developmental Biology
| | - E A Liberti
- Institute Biomedical Sciences, University of Sao Paulo-Department of Anatomy
| | - Mja Saad
- Department of Internal Medicine, State University of Campinas
| | - Cro Carvalho
- Institute Biomedical Sciences, University of Sao Paulo-Department of Physiology and Biophysical.
| |
Collapse
|
46
|
Zhang T, Yang Y, Liang Y, Jiao X, Zhao C. Beneficial Effect of Intestinal Fermentation of Natural Polysaccharides. Nutrients 2018; 10:E1055. [PMID: 30096921 PMCID: PMC6116026 DOI: 10.3390/nu10081055] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/27/2018] [Accepted: 08/07/2018] [Indexed: 12/11/2022] Open
Abstract
With the rapid development of modern society, many chronic diseases are increasing including diabetes, obesity, cardiovascular diseases, etc., which further cause an increased death rate worldwide. A high caloric diet with reduced natural polysaccharides, typically indigestible polysaccharides, is considered a health risk factor. With solid evidence accumulating that indigestible polysaccharides can effectively prevent and/or ameliorate symptoms of many chronic diseases, we give a narrative review of many natural polysaccharides extracted from various food resources which mainly contribute their health beneficial functions via intestinal fermentation.
Collapse
Affiliation(s)
- Tiehua Zhang
- College of Food Science and Engineering, Jilin University, Changchun 130062, Jilin, China.
| | - Yang Yang
- College of Food Science and Engineering, Jilin University, Changchun 130062, Jilin, China.
| | - Yuan Liang
- College of Food Science and Engineering, Jilin University, Changchun 130062, Jilin, China.
| | - Xu Jiao
- College of Food Science and Engineering, Jilin University, Changchun 130062, Jilin, China.
| | - Changhui Zhao
- College of Food Science and Engineering, Jilin University, Changchun 130062, Jilin, China.
| |
Collapse
|
47
|
Diao H, Yan HL, Xiao Y, Yu B, Zheng P, He J, Yu J, Mao XB, Chen DW. Modulation of intestine development by fecal microbiota transplantation in suckling pigs. RSC Adv 2018; 8:8709-8720. [PMID: 35539874 PMCID: PMC9078615 DOI: 10.1039/c7ra11234c] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 02/12/2018] [Indexed: 11/21/2022] Open
Abstract
The present study was conducted to investigate the effects of early fecal microbiota transplantation on gut development in sucking piglets. A total of 24 3 day-old DLY sucking piglets (2.11 ± 0.15) kg were randomly divided into four groups (TMP, YMP, RMP and control group (CON)), which were transplanted with intact fecal microbiota of Tibetan pig (TP), Yorkshire pig (YP), Rongchang pig (RP), and without transplantation, respectively. The whole trial lasted for 56 d. The results are as follows: when compared with the YMP and RMP treatments, TMP and CON had a lower diarrhea index (P < 0.05), TMP and CON had higher GLP-2 and ANG4 mRNA abundances in the ileum (P < 0.05), and the TMP had a higher jejunal villus height: crypt depth and a higher colonic GLP-2 mRNA abundance (P < 0.05). Moreover, when compared with the YMP and RMP treatments, TMP had an enhanced DMT1 mRNA abundance in the duodenum (P < 0.05), TMP and CON had a greater lactase activity and a higher DMT1 mRNA abundance in the jejunum (P < 0.05), and CON had a higher γ-GT activity in the jejunum (P < 0.05). The jejunal Ca2+, Mg2+-ATPase activity in TMP was higher than that in CON, and the jejunal Na+, K+-ATPase activity in TMP was higher than that in the other three treatments (P < 0.05). Besides, when compared with the YMP and RMP treatments, TMP had a lower MDA content and a higher MUC1 mRNA abundance in the jejunum (P < 0.05); CON had a higher SOD activity in the jejunum (P < 0.05), whereas TMP and CON had a higher butyric acid concentration in the colon and a lower LPS content in the serum (P < 0.05). Finally, when compared with the TMP treatment, the other three treatments had an enhanced IL-10 mRNA abundance in the colon (P < 0.05), YMP and CON had higher counts of Escherichia coli in the colonic digesta (P < 0.05), and the CON had lower counts of Lactobacillus spp in the cecal and colonic digesta (P < 0.05). These data indicated that early transplantation of the fecal microbiota from the Yorkshire pigs and Rongchang pigs to DLY suckling piglets would destroy the gut microbiota balance and thus damage intestinal health.
Collapse
Affiliation(s)
- H Diao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education Xinkang Road 46# Ya'an Sichuan Province 625014 People's Republic of China +86-835-288-5106 +86-835-288-5106
- Institute of Animal Nutrition, Sichuan Academy of Animal Science No. 7 Niusha Road Chengdu Sichuan 610066 People's Republic of China
| | - H L Yan
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education Xinkang Road 46# Ya'an Sichuan Province 625014 People's Republic of China +86-835-288-5106 +86-835-288-5106
| | - Y Xiao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education Xinkang Road 46# Ya'an Sichuan Province 625014 People's Republic of China +86-835-288-5106 +86-835-288-5106
| | - B Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education Xinkang Road 46# Ya'an Sichuan Province 625014 People's Republic of China +86-835-288-5106 +86-835-288-5106
| | - P Zheng
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education Xinkang Road 46# Ya'an Sichuan Province 625014 People's Republic of China +86-835-288-5106 +86-835-288-5106
| | - J He
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education Xinkang Road 46# Ya'an Sichuan Province 625014 People's Republic of China +86-835-288-5106 +86-835-288-5106
| | - J Yu
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education Xinkang Road 46# Ya'an Sichuan Province 625014 People's Republic of China +86-835-288-5106 +86-835-288-5106
| | - X B Mao
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education Xinkang Road 46# Ya'an Sichuan Province 625014 People's Republic of China +86-835-288-5106 +86-835-288-5106
| | - D W Chen
- Institute of Animal Nutrition, Sichuan Agricultural University, Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education Xinkang Road 46# Ya'an Sichuan Province 625014 People's Republic of China +86-835-288-5106 +86-835-288-5106
| |
Collapse
|
48
|
Serino M. Molecular Paths Linking Metabolic Diseases, Gut Microbiota Dysbiosis and Enterobacteria Infections. J Mol Biol 2018; 430:581-590. [PMID: 29374557 DOI: 10.1016/j.jmb.2018.01.010] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/19/2018] [Indexed: 02/07/2023]
Abstract
Alterations of both ecology and functions of gut microbiota are conspicuous traits of several inflammatory pathologies, notably metabolic diseases such as obesity and type 2 diabetes. Moreover, the proliferation of enterobacteria, subdominant members of the intestinal microbial ecosystem, has been shown to be favored by Western diet, the strongest inducer of both metabolic diseases and gut microbiota dysbiosis. The inner interdependence between the host and the gut microbiota is based on a plethora of molecular mechanisms by which host and intestinal microbes modify each other. Among these mechanisms are as follows: (i) the well-known metabolic impact of short chain fatty acids, produced by microbial fermentation of complex carbohydrates from plants; (ii) a mutual modulation of miRNAs expression, both on the eukaryotic (host) and prokaryotic (gut microbes) side; (iii) the production by enterobacteria of virulence factors such as the genotoxin colibactin, shown to alter the integrity of host genome and induce a senescence-like phenotype in vitro; (iv) the microbial excretion of outer-membrane vesicles, which, in addition to other functions, may act as a carrier for multiple molecules such as toxins to be delivered to target cells. In this review, I describe the major molecular mechanisms by which gut microbes exert their metabolic impact at a multi-organ level (the gut barrier being in the front line) and support the emerging triad of metabolic diseases, gut microbiota dysbiosis and enterobacteria infections.
Collapse
Affiliation(s)
- Matteo Serino
- IRSD, Université de Toulouse, INSERM, INRA, ENVT, UPS, Toulouse, France.
| |
Collapse
|
49
|
Gut microbiota in the pathogenesis of inflammatory bowel disease. Clin J Gastroenterol 2017; 11:1-10. [PMID: 29285689 DOI: 10.1007/s12328-017-0813-5] [Citation(s) in RCA: 884] [Impact Index Per Article: 110.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 12/20/2017] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel disease (IBD), including ulcerative colitis and Crohn's disease, is a chronic and relapsing inflammatory disorder of the intestine. Although its incidence is increasing globally, the precise etiology remains unclear and a cure for IBD has yet to be discovered. The most accepted hypothesis of IBD pathogenesis is that complex interactions between genetics, environmental factors, and the host immune system lead to aberrant immune responses and chronic intestinal inflammation. The human gut harbors a complex and abundant aggregation of microbes, collectively referred to as the gut microbiota. The gut microbiota has physiological functions associated with nutrition, the immune system, and defense of the host. Recent advances in next-generation sequencing technology have identified alteration of the composition and function of the gut microbiota, which is referred to as dysbiosis, in IBD. Clinical and experimental data suggest dysbiosis may play a pivotal role in the pathogenesis of IBD. This review is focused on the physiological function of the gut microbiota and the association between the gut microbiota and pathogenesis in IBD. In addition, we review the therapeutic options for manipulating the altered gut microbiota, such as probiotics and fecal microbiota transplantation.
Collapse
|
50
|
Smolentseva O, Gusarov I, Gautier L, Shamovsky I, DeFrancesco AS, Losick R, Nudler E. Mechanism of biofilm-mediated stress resistance and lifespan extension in C. elegans. Sci Rep 2017; 7:7137. [PMID: 28769037 PMCID: PMC5540977 DOI: 10.1038/s41598-017-07222-8] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Accepted: 06/26/2017] [Indexed: 01/16/2023] Open
Abstract
Bacteria naturally form communities of cells known as biofilms. However the physiological roles of biofilms produced by non-pathogenic microbiota remain largely unknown. To assess the impact of a biofilm on host physiology we explored the effect of several non-pathogenic biofilm-forming bacteria on Caenorhabditis elegans. We show that biofilm formation by Bacillus subtilis, Lactobacillus rhamnosus and Pseudomonas fluorescens induces C. elegans stress resistance. Biofilm also protects against pathogenic infection and prolongs lifespan. Total mRNA analysis identified a set of host genes that are upregulated in response to biofilm formation by B. subtilis. We further demonstrate that mtl-1 is responsible for the biofilm-mediated increase in oxidative stress resistance and lifespan extension. Induction of mtl-1 and hsp-70 promotes biofilm-mediated thermotolerance. ilys-2 activity accounts for biofilm-mediated resistance to Pseudomonas aeruginosa killing. These results reveal the importance of non-pathogenic biofilms for host physiology and provide a framework to study commensal biofilms in higher organisms.
Collapse
Affiliation(s)
- Olga Smolentseva
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Ivan Gusarov
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Laurent Gautier
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Ilya Shamovsky
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA
| | - Alicia S DeFrancesco
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, 02138, USA
| | - Richard Losick
- Department of Molecular and Cellular Biology, Harvard University, Cambridge, Massachusetts, 02138, USA
| | - Evgeny Nudler
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY, 10016, USA.
- Howard Hughes Medical Institute, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|