1
|
Joishy TK, Bhattacharya A, Singh CT, Mukherjee AK, Khan MR. Probiotic and anti-inflammatory properties of Lactiplantibacillus plantarum MKTJ24 isolated from an artisanal fermented fish of North-east India. N Biotechnol 2024; 83:121-132. [PMID: 39111568 DOI: 10.1016/j.nbt.2024.07.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/21/2024] [Accepted: 07/27/2024] [Indexed: 08/13/2024]
Abstract
The study aimed to isolate and characterize lactic acid bacteria from various traditional fermented fish products from North East India, including Xindol, Hentak, and Ngari, which hold significant dietary importance for the indigenous tribes. Additionally, the study sought to examine their untargeted metabolomic profiles. A total of 43 strains of Bacillus, Priestia, Staphylococcus, Pediococcus, and Lactiplantibacillus were isolated, characterized by 16 S rRNA gene and tested for probiotic properties. Five strains passed pH and bile salt tests with strain dependent antimicrobial activity, which exhibited moderate autoaggregation and hydrophobicity properties. Lactiplantibacillus plantarum MKTJ24 exhibited the highest hydrophobicity (42 %), which was further confirmed by adhesion assay in HT-29 cell lines (100 %). Lactiplantibacillus plantarum MKTJ24 treatment in LPS-stimulated HT-29 cells up-regulated expression of mucin genes compared to LPS-treated cells. Treatment of RAW 264.7 cells with Lactiplantibacillus plantarum MKTJ24 decreased LPS-induced reactive oxygen species (ROS) and nitric oxide (NO) productions. Further, genome analysis of Lactiplantibacillus plantarum MKTJ24 revealed the presence of several probiotic markers and immunomodulatory genes. The genome was found to harbor plantaricin operon involved in bacteriocin production. A pangenome analysis using all the publicly available L. plantarum genomes specifically isolated from fermented fish products identified 120 unique genes in Lactiplantibacillus plantarum MKTJ24. Metabolomic analysis indicated dominance of ascorbic acids, pentafluropropionate, cyclopropaneacetic acid, florobenzylamine, and furanone in Xindol. This study suggests that Lactiplantibacillus plantarum MKTJ24 has potential probiotic and immunomodulatory properties that could be used in processing traditional fermented fish products on an industrial scale to improve their quality and enhance functional properties.
Collapse
Affiliation(s)
- Tulsi K Joishy
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Anupam Bhattacharya
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Chingtham Thanil Singh
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-202002, India
| | - Ashis K Mukherjee
- Microbial Biotechnology and Protein Research Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India
| | - Mojibur R Khan
- Molecular Biology and Microbial Biotechnology Laboratory, Life Sciences Division, Institute of Advanced Study in Science and Technology (IASST), Guwahati, Assam, India.
| |
Collapse
|
2
|
Zhu Q, Azad MAK, Li R, Li C, Liu Y, Yin Y, Kong X. Dietary probiotic and synbiotic supplementation starting from maternal gestation improves muscular lipid metabolism in offspring piglets by reshaping colonic microbiota and metabolites. mSystems 2024; 9:e0004824. [PMID: 38767377 PMCID: PMC11237649 DOI: 10.1128/msystems.00048-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 04/16/2024] [Indexed: 05/22/2024] Open
Abstract
Probiotics and synbiotics have been intensively used in animal husbandry due to their advantageous roles in animals' health. However, there is a paucity of research on probiotic and synbiotic supplementation from maternal gestation to the postnatal growing phases of offspring piglets. Thus, we assessed the effects of dietary supplementation of these two additives to sows and offspring piglets on skeletal muscle and body metabolism, colonic microbiota composition, and metabolite profiles of offspring piglets. Pregnant Bama mini-pigs and their offspring piglets (after weaning) were fed either a basal diet or a basal diet supplemented with antibiotics, probiotics, or synbiotics. At 65, 95, and 125 days old, eight pigs per group were euthanized and sampled for analyses. Probiotics increased the intramuscular fat content in the psoas major muscle (PMM) at 95 days old, polyunsaturated fatty acid (PUFA) and n-3 PUFA levels in the longissimus dorsi muscle (LDM) at 65 days old, C16:1 level in the LDM at 125 days old, and upregulated ATGL, CPT-1, and HSL expressions in the PMM at 65 days old. Synbiotics increased the plasma HDL-C level at 65 days old and TC level at 65 and 125 days old and upregulated the CPT-1 expression in the PMM at 125 days old. In addition, probiotics and synbiotics increased the plasma levels of HDL-C at 65 days old, CHE at 95 days old, and LDL-C at 125 days old, while decreasing the C18:1n9t level in the PMM at 65 days old and the plasma levels of GLU, LDH, and TG at 95 days old. Microbiome analysis showed that probiotic and synbiotic supplementation increased colonic Actinobacteria, Firmicutes, Verrucomicrobia, Faecalibacterium, Pseudobutyrivibrio, and Turicibacter abundances. However, antibiotic supplementation decreased colonic Actinobacteria, Bacteroidetes, Prevotella, and Unclassified_Lachnospiraceae abundances. Furthermore, probiotic and synbiotic supplementation was associated with alterations in 8, 7, and 10 differential metabolites at three different age stages. Both microbiome and metabolome analyses showed that the differential metabolic pathways were associated with carbohydrate, amino acid, and lipid metabolism. However, antibiotic supplementation increased the C18:1n9t level in the PMM at 65 days old and xenobiotic biodegradation and metabolism at 125 days old. In conclusion, sow-offspring's diets supplemented with these two additives showed conducive effects on meat flavor, nutritional composition of skeletal muscles, and body metabolism, which may be associated with the reshaping of colonic microbiota and metabolites. However, antibiotic supplementation has negative effects on colonic microbiota composition and fatty acid composition in the PMM. IMPORTANCE The integral sow-offspring probiotic and synbiotic supplementation improves the meat flavor and the fatty acid composition of the LDM to some extent. Sow-offspring probiotic and synbiotic supplementation increases the colonic beneficial bacteria (including Firmicutes, Verrucomicrobia, Actinobacteria, Faecalibacterium, Turicibacter, and Pseudobutyrivibrio) and alters the colonic metabolite profiles, such as guanidoacetic acid, beta-sitosterol, inosine, cellobiose, indole, and polyamine. Antibiotic supplementation in sow-offspring's diets decreases several beneficial bacteria (including Bacteroidetes, Actinobacteria, Unclassified_Lachnospiraceae, and Prevotella) and has a favorable effect on improving the fatty acid composition of the LDM to some extent, while presenting the opposite effect on the PMM.
Collapse
Affiliation(s)
- Qian Zhu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Md Abul Kalam Azad
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Ruixuan Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chenjian Li
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Yang Liu
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
| | - Yulong Yin
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiangfeng Kong
- Key Laboratory of Agro-ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, Hunan, China
- College of Advanced Agricultural Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
3
|
Bozzi Cionci N, Reggio M, Baffoni L, Di Gioia D. Probiotic Administration for the Prevention and Treatment of Gastrointestinal, Metabolic and Neurological Disorders. ADVANCES IN PREDICTIVE, PREVENTIVE AND PERSONALISED MEDICINE 2023:219-250. [DOI: 10.1007/978-3-031-19564-8_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
4
|
Probiotics in the Management of Mental and Gastrointestinal Post-COVID Symptomes. J Clin Med 2022; 11:jcm11175155. [PMID: 36079082 PMCID: PMC9457065 DOI: 10.3390/jcm11175155] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2022] [Revised: 08/17/2022] [Accepted: 08/29/2022] [Indexed: 01/30/2023] Open
Abstract
Patients with “post-COVID” syndrome manifest with a variety of signs and symptoms that continue/develop after acute COVID-19. Among the most common are gastrointestinal (GI) and mental symptoms. The reason for symptom occurrence lies in the SARS-CoV-2 capability of binding to exact receptors, among other angiotensin converting enzyme 2 (ACE2) receptors in gastrointestinal lining and neuropilin-1 (NRP-1) in the nervous system, which leads to loss of gastrointestinal and blood-brain barriers integrity and function. The data are mounting that SARS-CoV-2 can trigger systemic inflammation and lead to disruption of gut-brain axis (GBA) and the development of disorders of gut brain interaction (DGBIs). Functional dyspepsia (FD) and irritable bowel syndrome (IBS) are the most common DGBIs syndromes. On the other hand, emotional disorders have also been demonstrated as DGBIs. Currently, there are no official recommendations or recommended procedures for the use of probiotics in patients with COVID-19. However, it can be assumed that many doctors, pharmacists, and patients will want to use a probiotic in the treatment of this disease. In such cases, strains with documented activity should be used. There is a constant need to plan and conduct new trials on the role of probiotics and verify their clinical efficacy for counteracting the negative consequences of COVID-19 pandemic. Quality control is another important but often neglected aspect in trials utilizing probiotics in various clinical entities. It determines the safety and efficacy of probiotics, which is of utmost importance in patients with post-acute COVID-19 syndrome.
Collapse
|
5
|
Patel BK, Patel KH, Huang RY, Lee CN, Moochhala SM. The Gut-Skin Microbiota Axis and Its Role in Diabetic Wound Healing-A Review Based on Current Literature. Int J Mol Sci 2022; 23:ijms23042375. [PMID: 35216488 PMCID: PMC8880500 DOI: 10.3390/ijms23042375] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 12/18/2022] Open
Abstract
Diabetic foot ulcers (DFU) are a growing concern worldwide as they pose complications in routine clinical practices such as diagnosis and management. Bacterial interactions on the skin surface are vital to the pathophysiology of DFU and may control delayed wound healing. The microbiota from our skin directly regulates cutaneous health and disease by interacting with the numerous cells involved in the wound healing mechanism. Commensal microbiota, in particular, interact with wound-repairing skin cells to enhance barrier regeneration. The observed microbes in DFU include Staphylococcus, Streptococcus, Corynebacterium, Pseudomonas, and several anaerobes. Skin commensal microbes, namely S. epidermidis, can regulate the gamma delta T cells and induce Perforin-2 expression. The increased expression of Perforin-2 by skin cells destroyed S. aureus within the cells, facilitating wound healing. Possible crosstalk between the human commensal microbiome and different cell types involved in cutaneous wound healing promotes the immune response and helps to maintain the barrier function in humans. Wound healing is a highly well-coordinated, complex mechanism; it can be devastating if interrupted. Skin microbiomes are being studied in relation to the gut-skin axis along with their effects on dermatologic conditions. The gut-skin axis illustrates the connection wherein the gut can impact skin health due to its immunological and metabolic properties. The precise mechanism underlying gut-skin microbial interactions is still unidentified, but the immune and endocrine systems are likely to be involved. Next-generation sequencing and the development of bioinformatics pipelines may considerably improve the understanding of the microbiome-skin axis involved in diabetic wound healing in a much more sophisticated way. We endeavor to shed light on the importance of these pathways in the pathomechanisms of the most prevalent inflammatory conditions including the diabetes wound healing, as well as how probiotics may intervene in the gut-skin axis.
Collapse
Affiliation(s)
- Bharati Kadamb Patel
- Department of Surgery, National University of Singapore, Singapore 119228, Singapore; (B.K.P.); (C.N.L.)
| | | | - Ryan Yuki Huang
- Canyon Crest Academy, San Diego, CA 92130, USA;
- Department of Mechanical and Aerospace Engineering, University of California, San Diego, CA 92093, USA
| | - Chuen Neng Lee
- Department of Surgery, National University of Singapore, Singapore 119228, Singapore; (B.K.P.); (C.N.L.)
| | - Shabbir M. Moochhala
- Department of Surgery, National University of Singapore, Singapore 119228, Singapore; (B.K.P.); (C.N.L.)
- Department of Pharmacology, National University of Singapore, Singapore 117600, Singapore
- Correspondence:
| |
Collapse
|
6
|
Requena T, Pérez Martínez G. Probiotics, Prebiotics, Synbiotics, Postbiotics and Other Biotics. What's Next? COMPREHENSIVE GUT MICROBIOTA 2022:197-210. [DOI: 10.1016/b978-0-12-819265-8.00094-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
7
|
Davoodvandi A, Fallahi F, Tamtaji OR, Tajiknia V, Banikazemi Z, Fathizadeh H, Abbasi-Kolli M, Aschner M, Ghandali M, Sahebkar A, Taghizadeh M, Mirzaei H. An Update on the Effects of Probiotics on Gastrointestinal Cancers. Front Pharmacol 2021; 12:680400. [PMID: 34992527 PMCID: PMC8724544 DOI: 10.3389/fphar.2021.680400] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 11/26/2021] [Indexed: 12/28/2022] Open
Abstract
Because of their increasing prevalence, gastrointestinal (GI) cancers are regarded as an important global health challenge. Microorganisms residing in the human GI tract, termed gut microbiota, encompass a large number of living organisms. The role of the gut in the regulation of the gut-mediated immune responses, metabolism, absorption of micro- and macro-nutrients and essential vitamins, and short-chain fatty acid production, and resistance to pathogens has been extensively investigated. In the past few decades, it has been shown that microbiota imbalance is associated with the susceptibility to various chronic disorders, such as obesity, irritable bowel syndrome, inflammatory bowel disease, asthma, rheumatoid arthritis, psychiatric disorders, and various types of cancer. Emerging evidence has shown that oral administration of various strains of probiotics can protect against cancer development. Furthermore, clinical investigations suggest that probiotic administration in cancer patients decreases the incidence of postoperative inflammation. The present review addresses the efficacy and underlying mechanisms of action of probiotics against GI cancers. The safety of the most commercial probiotic strains has been confirmed, and therefore these strains can be used as adjuvant or neo-adjuvant treatments for cancer prevention and improving the efficacy of therapeutic strategies. Nevertheless, well-designed clinical studies are still needed for a better understanding of the properties and mechanisms of action of probiotic strains in mitigating GI cancer development.
Collapse
Affiliation(s)
- Amirhossein Davoodvandi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Farzaneh Fallahi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Omid Reza Tamtaji
- Students’ Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Vida Tajiknia
- Department of Surgery, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Zarrin Banikazemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hadis Fathizadeh
- Department of Laboratory Sciences, Sirjan Faculty of Medicine Sciences, Sirjan, Iran
| | - Mohammad Abbasi-Kolli
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, United States
| | - Maryam Ghandali
- School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
8
|
Marlicz W, Skonieczna-Żydecka K, Krynicka P, Łoniewski I, Rydzewska G. Probiotics in irritable bowel syndrome - is the quest for the right strain over? Rapid review of existing guidelines and recommendations. PRZEGLAD GASTROENTEROLOGICZNY 2021; 16:369-382. [PMID: 34976247 PMCID: PMC8690954 DOI: 10.5114/pg.2021.111766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/30/2021] [Indexed: 02/07/2023]
Abstract
Irritable bowel syndrome (IBS) - functional gastrointestinal disorder (FGIDs) and disorder of gut-brain interaction (DGBIs) - has emerged as an important medical problem with an impact on health care systems, affecting patients' quality of life. The management of IBS consists of pharmacological and non-pharmacological treatments; however, the data of their long-term efficacy are scarce. Modulation of gastrointestinal microbiota, by means of probiotics and prebiotics, is often sought and advertised as a popular treatment modality in IBS. Faecal microbiota transplantation (FMT) awaits recommendations for IBS treatment and requires more methodological assessments. To date, numerous guidelines and recommendations have been published on the role of probiotics in IBS. Because no probiotic claim for probiotics in foods has yet been granted by the European Food and Safety Authority (EFSA), medical practitioners still recommend probiotics on the basis of available literature and recommendations released by independent health authorities. We aimed to summarize published formal recommendations and guidelines regarding the clinical effectiveness of available probiotic strains and conduct a random-effects meta-analysis of outcomes for which ≥ 2 studies contributed data on the same probiotic strain recommended to adults with IBS. Based on available and most recent guidelines, we report that probiotics, as a group, may be an effective treatment for global symptoms and abdominal pain in IBS, with the strongest effect for genus Lactobacillus. Our current and updated meta-analysis is in line with several reports documenting significant effects of Lactobacillus plantarum (Lp299v) in reducing the risk of global symptoms and their persistence, which could assist clinicians in making the choice for the right probiotic strain in IBS patients.
Collapse
Affiliation(s)
- Wojciech Marlicz
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | | | - Patrycja Krynicka
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| | - Igor Łoniewski
- Department of Biochemical Sciences, Pomeranian Medical University, Szczecin, Poland
| | - Grażyna Rydzewska
- Clinical Department of Internal Medicine and Gastroenterology with Inflammatory Bowel Disease Unit, Central Clinical Hospital of the Ministry of the Inferior and Administration, Warsaw, Poland
- Collegium Medicum, Jan Kochanowski University, Kielce, Poland
| |
Collapse
|
9
|
Abstract
The probiotic Escherichia coli strain Nissle 1917 (DSM 6601, Mutaflor), generally considered beneficial and safe, has been used for a century to treat various intestinal diseases. However, Nissle 1917 hosts in its genome the pks pathogenicity island that codes for the biosynthesis of the genotoxin colibactin. Colibactin is a potent DNA alkylator, suspected to play a role in colorectal cancer development. We show in this study that Nissle 1917 is functionally capable of producing colibactin and inducing interstrand cross-links in the genomic DNA of epithelial cells exposed to the probiotic. This toxicity was even exacerbated with lower doses of the probiotic, when the exposed cells started to divide again but exhibited aberrant anaphases and increased gene mutation frequency. DNA damage was confirmed in vivo in mouse models of intestinal colonization, demonstrating that Nissle 1917 produces the genotoxin in the gut lumen. Although it is possible that daily treatment of adult humans with their microbiota does not produce the same effects, administration of Nissle 1917 as a probiotic or as a chassis to deliver therapeutics might exert long-term adverse effects and thus should be considered in a risk-versus-benefit evaluation. IMPORTANCE Nissle 1917 is sold as a probiotic and considered safe even though it has been known since 2006 that it harbors the genes for colibactin synthesis. Colibactin is a potent genotoxin that is now linked to causative mutations found in human colorectal cancer. Many papers concerning the use of this strain in clinical applications ignore or elude this fact or misleadingly suggest that Nissle 1917 does not induce DNA damage. Here, we demonstrate that Nissle 1917 produces colibactin in vitro and in vivo and induces mutagenic DNA damage. This is a serious safety concern that must not be ignored in the interests of patients, the general public, health care professionals, and ethical probiotic manufacturers.
Collapse
|
10
|
Jani K, Sharma A. Targeted amplicon sequencing reveals the probiotic potentials of microbial communities associated with traditional fermented foods of northeast India. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.111578] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
11
|
Nwachukwu CU, Aliyu KI, Ewuola EO. Growth indices, intestinal histomorphology, and blood profile of rabbits fed probiotics- and prebiotics-supplemented diets. Transl Anim Sci 2021; 5:txab096. [PMID: 34316539 PMCID: PMC8309950 DOI: 10.1093/tas/txab096] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Indexed: 12/28/2022] Open
Abstract
In a 12-week feeding trial, 32 rabbits (Chinchilla × New Zealand White; 56 days old; 691 ± 1 g body weight) were used to investigate the effect of pro- and prebiotics as growth enhancer on the growth performance, intestinal mucosal development, hematological and serum biochemical responses of rabbits. The dietary Biotronic® prebiotics and Biovet®-YC probiotics were added at 400 mg/kg and 50 mg/kg, respectively. The rabbits were housed individually and randomly assigned to four dietary treatments (n = 8/group; 50:50 bucks to does) including a control diet (diet 1), diet 2 (control + Biotronic® prebiotics), diet 3 (control + Biovet®-YC probiotics) and diet 4 (control + symbiotics [Biotronic® prebiotics and Biovet®-YC probiotics]). Body weight (BW), average daily gain (ADG), dry matter intake (DMI), and feed conversion ratio (FCR) were monitored. Five rabbits per treatment were used for organ assessment and intestinal histomorphology after feeding trial. Blood samples were collected for hematological and serum biochemical analysis. Results showed that supplementation of Biotronic® prebiotics and symbiotics in rabbit diet significantly (P < 0.05) increased final BW and ADG compared to Biovet®-YC probiotic and control diets. Kidney, lung, esophagus, gastro-intestinal tract, small and large intestines were significantly (P < 0.05) influenced by dietary treatments. Ileal mucosal assessment revealed that villus height (VH), villus width, villus density, crypt depth (CD), and VH:CD ratio of rabbits fed Biotronic® prebiotic and symbiotic diets were similar and significantly (P < 0.05) higher than those rabbits fed control and Biovet®-YC probiotic diets. Packed cell volume of rabbits fed symbiotic and control diets was significantly (P < 0.05) higher than those fed Biotronic® prebiotic and Biovet®-YC probiotic diets. This study suggests that Biotronic® prebiotics and its combination with Biovet®-YC probiotics are good alternative growth promoting feed additives in rabbit nutrition. They improved performance, intestinal development and blood profiles and aid feed digestion, nutrient absorption and utilization in rabbits.
Collapse
Affiliation(s)
- Chinwe Uchechi Nwachukwu
- Department of Agricultural Science, School of Agriculture and Vocational Studies, Alvan Ikoku Federal College of Education, Owerri, Imo State, 460281, Nigeria
| | - Karimat Imam Aliyu
- Livestock Department, Ministry of Agriculture and Natural Resources, Kwara State, Nigeria
| | - Emmanuel Olubisi Ewuola
- Animal Physiology Laboratory, Department of Animal Science, University of Ibadan, Ibadan, Nigeria
| |
Collapse
|
12
|
Pujo J, Petitfils C, Le Faouder P, Eeckhaut V, Payros G, Maurel S, Perez-Berezo T, Van Hul M, Barreau F, Blanpied C, Chavanas S, Van Immerseel F, Bertrand-Michel J, Oswald E, Knauf C, Dietrich G, Cani PD, Cenac N. Bacteria-derived long chain fatty acid exhibits anti-inflammatory properties in colitis. Gut 2021; 70:1088-1097. [PMID: 32978245 DOI: 10.1136/gutjnl-2020-321173] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 07/15/2020] [Accepted: 08/30/2020] [Indexed: 12/16/2022]
Abstract
OBJECTIVE Data from clinical research suggest that certain probiotic bacterial strains have the potential to modulate colonic inflammation. Nonetheless, these data differ between studies due to the probiotic bacterial strains used and the poor knowledge of their mechanisms of action. DESIGN By mass-spectrometry, we identified and quantified free long chain fatty acids (LCFAs) in probiotics and assessed the effect of one of them in mouse colitis. RESULTS Among all the LCFAs quantified by mass spectrometry in Escherichia coli Nissle 1917 (EcN), a probiotic used for the treatment of multiple intestinal disorders, the concentration of 3-hydroxyoctadecaenoic acid (C18-3OH) was increased in EcN compared with other E. coli strains tested. Oral administration of C18-3OH decreased colitis induced by dextran sulfate sodium in mice. To determine whether other bacteria composing the microbiota are able to produce C18-3OH, we targeted the gut microbiota of mice with prebiotic fructooligosaccharides (FOS). The anti-inflammatory properties of FOS were associated with an increase in colonic C18-3OH concentration. Microbiota analyses revealed that the concentration of C18-3OH was correlated with an increase in the abundance in Allobaculum, Holdemanella and Parabacteroides. In culture, Holdemanella biformis produced high concentration of C18-3OH. Finally, using TR-FRET binding assay and gene expression analysis, we demonstrated that the C18-3OH is an agonist of peroxisome proliferator activated receptor gamma. CONCLUSION The production of C18-3OH by bacteria could be one of the mechanisms implicated in the anti-inflammatory properties of probiotics. The production of LCFA-3OH by bacteria could be implicated in the microbiota/host interactions.
Collapse
Affiliation(s)
- Julien Pujo
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France.,Farncombe Family Digestive Health Institute, McMaster University Faculty of Health Sciences, Hamilton, Ontario, Canada
| | - Camille Petitfils
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France
| | | | - Venessa Eeckhaut
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Gaelle Payros
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France
| | - Sarah Maurel
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France
| | - Teresa Perez-Berezo
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France
| | - Matthias Van Hul
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Louvain Drug Research Institute, Metabolism and Nutrition Research Group, UCLouvain, Université catholique de Louvain, Brussels, Belgium
| | - Frederick Barreau
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France
| | - Catherine Blanpied
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France
| | - Stephane Chavanas
- Centre for Pathophysiology Toulouse-Purpan (CPTP), INSERM, CNRS, University of Toulouse, Toulouse, France
| | - Filip Van Immerseel
- Department of Pathology, Bacteriology and Avian Diseases, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | - Eric Oswald
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France.,Service de bactériologie-hygiène, CHU Toulouse, Hôpital Purpan, Toulouse, France
| | - Claude Knauf
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France.,NeuroMicrobiota, European Associated Laboratory (EAL), INSERM/UCLouvain, Toulouse, Brussels, France, Belgium
| | - Gilles Dietrich
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France
| | - Patrice D Cani
- Walloon Excellence in Life Sciences and Biotechnology (WELBIO), Louvain Drug Research Institute, Metabolism and Nutrition Research Group, UCLouvain, Université catholique de Louvain, Brussels, Belgium.,NeuroMicrobiota, European Associated Laboratory (EAL), INSERM/UCLouvain, Toulouse, Brussels, France, Belgium
| | - Nicolas Cenac
- IRSD, INSERM, INRA, INP-ENVT, Toulouse University 3 Paul Sabatier, Toulouse, France
| |
Collapse
|
13
|
Liu J, Xu Y, Jiang B. Novel Insights Into Pathogenesis and Therapeutic Strategies of Hepatic Encephalopathy, From the Gut Microbiota Perspective. Front Cell Infect Microbiol 2021; 11:586427. [PMID: 33692964 PMCID: PMC7937792 DOI: 10.3389/fcimb.2021.586427] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 01/08/2021] [Indexed: 01/10/2023] Open
Abstract
Since the 1950s, gradual changes in the gut microbiota of patients with hepatic encephalopathy have been observed. Previous research has indicated potential associations between the gut and brain, and the gut microbiota is becoming a hot topic in research on diseases of the nervous system. However, for the past few decades, studies of hepatic encephalopathy have been restricted to controlling the gut microbiota during macroscopic manipulation, such as probiotic intervention, while its clinical use remains controversial, and the cellular mechanisms underlying this condition are still poorly understood. This thesis seeks to comprehensively understand and explain the role of gut microbiota in hepatic encephalopathy as well as analyze the effects of intervention by regulating the gut microbiota. Evidence is presented that shows that dysbiosis of the gut microbiota is the primary pathological driver of hepatic encephalopathy and impacts pathologic progression via complex regulatory networks. As a result, suggestions were identified for future mechanistic research and improvements in therapeutic strategies for hepatic encephalopathy.
Collapse
Affiliation(s)
- Jiachen Liu
- Xiangya Medical College of Central South University, Changsha, China
| | - Yantao Xu
- Xiangya Medical College of Central South University, Changsha, China
| | - Bimei Jiang
- Department of Pathophysiology, Sepsis Translational Medicine Key Laboratory of Hunan Province, Xiangya School of Medicine, Central South University, Changsha, China
| |
Collapse
|
14
|
Stasiak-Różańska L, Berthold-Pluta A, Pluta AS, Dasiewicz K, Garbowska M. Effect of Simulated Gastrointestinal Tract Conditions on Survivability of Probiotic Bacteria Present in Commercial Preparations. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:1108. [PMID: 33513771 PMCID: PMC7908519 DOI: 10.3390/ijerph18031108] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/12/2022]
Abstract
Probiotics are recommended, among others, in the diet of children who are under antibiotic therapy, or that suffer from food allergies or travel diarrhea, etc. In the case of toddlers taking probiotic preparations, it is highly recommended to first remove the special capsule, which normally protects probiotic strains against hard conditions in the gastrointestinal tract. Otherwise, the toddler may choke. This removal can impair probiotic survival and reduce its efficacy in a toddler's organism. The aim of this study was to evaluate the survivability of five strains of lactic acid bacteria from the commercial probiotics available on the Polish market under simulated conditions of the gastrointestinal tract. Five probiotics (each including one of these strains: Bifidobacterium BB-12, Lactobacillus (Lb.) rhamnosus GG, Lb. casei, Lb. acidophilus, Lb. plantarum) were protective capsule deprived, added in a food matrix (chicken-vegetable soup) and subjected under simulated conditions of the gastric and gastrointestinal passage. Strain survivability and possibility to growth were evaluated. Obtained results showed that, among all analyzed commercial probiotic strains, the Lb. plantarum was the most resistant to the applied conditions of the culture medium. They showed a noticeable growth under both in vitro gastric conditions at pH 4.0 and 5.0, as well as in vitro intestinal conditions at all tested concentrations of bile salts.
Collapse
Affiliation(s)
- Lidia Stasiak-Różańska
- Department of Food Technology and Assessment, Institute of Food Sciences, Warsaw University of Life Sciences-SGGW, Nowoursynowska St. 166, 02-787 Warsaw, Poland; (A.B.-P.); (A.S.P.); (K.D.); (M.G.)
| | | | | | | | | |
Collapse
|
15
|
Borrell García C, Ribelles Llop M, García Esparza MÁ, Flichy-Fernández AJ, Marqués Martínez L, Izquierdo Fort R. The use of Lactobacillus reuteri DSM 17938 and ATCC PTA 5289 on oral health indexes in a school population: A pilot randomized clinical trial. Int J Immunopathol Pharmacol 2021; 35:20587384211031107. [PMID: 34325540 PMCID: PMC8327006 DOI: 10.1177/20587384211031107] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 06/16/2021] [Indexed: 11/23/2022] Open
Abstract
To assess the effects of a probiotic upon oral health indices in adolescents and to establish relationships between these indices and dietary habits and oral hygiene. Twenty-seven adolescents between 12 and 18 years of age were randomized into two groups. The study group received tablets containing Lactobacillus reuteri DSM 17938/ATCC 5289 for 28 days, while the control group received tablets without any bacteria. Streptococcus mutans, Lactobacillus sp., and salivary pH were assessed at baseline and at 7, 14, 21, 28, and 45 days. The plaque, gingivitis, and bleeding indices were recorded at baseline and at 14, 28, and 45 days. Dietary and oral hygiene habits were also evaluated by means of a questionnaire. A less marked rise in S. mutans was recorded in the study group. Improvements were observed in terms of plaque, gingivitis, and bleeding, though statistical significance was not reached. Oral pH increased in the study group, though not to a significant degree. Poorer eating habits were significantly correlated to increased plaque. The study parameters decreased with the two strains of L. reuteri DSM 17938 and ATCC PTA 5289, though the results failed to reach statistical.
Collapse
Affiliation(s)
- Carla Borrell García
- Department of Dentistry, Faculty of
Health Sciences, CEU Cardenal Herrera University, Valencia, Spain
| | - Marta Ribelles Llop
- Department of Dentistry, Faculty of
Health Sciences, CEU Cardenal Herrera University, Valencia, Spain
| | | | | | - Laura Marqués Martínez
- Department of Dentistry, Faculty of
Health Sciences, CEU Cardenal Herrera University, Valencia, Spain
| | - Regina Izquierdo Fort
- Department of Periodontics, Valencia
University Medical and Dental School, Valencia, Spain
| |
Collapse
|
16
|
Saettone V, Biasato I, Radice E, Schiavone A, Bergero D, Meineri G. State-of-the-Art of the Nutritional Alternatives to the Use of Antibiotics in Humans and Monogastric Animals. Animals (Basel) 2020; 10:ani10122199. [PMID: 33255356 PMCID: PMC7759783 DOI: 10.3390/ani10122199] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/19/2020] [Accepted: 11/20/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Antibiotic resistance represents a worldwide recognized issue affecting both human and veterinary medicine, with a particular focus being directed towards monogastric animals destined for human consumption. This scenario is the result of frequent utilization of the antibiotics either for therapeutic purposes (humans and animals) or as growth promoters (farmed animals). Therefore, the search for nutritional alternatives has progressively been the object of significant efforts by the scientific community. So far, probiotics, prebiotics and postbiotics are considered the most promising products, as they are capable of preventing or treating gastrointestinal diseases as well as restoring a eubiosis condition after antibiotic-induced dysbiosis development. This review provides an updated state-of-the-art of these nutritional alternatives in both humans and monogastric animals. Abstract In recent years, the indiscriminate use of antibiotics has been perpetrated across human medicine, animals destined for zootechnical productions and companion animals. Apart from increasing the resistance rate of numerous microorganisms and generating multi-drug resistance (MDR), the nonrational administration of antibiotics causes sudden changes in the structure of the intestinal microbiota such as dysbiotic phenomena that can have a great clinical significance for both humans and animals. The aim of this review is to describe the state-of-the-art of alternative therapies to the use of antibiotics and their effectiveness in humans and monogastric animals (poultry, pigs, fish, rabbits, dogs and cats). In particular, those molecules (probiotics, prebiotics and postbiotics) which have a direct function on the gastrointestinal health are herein critically analysed in the prevention or treatment of gastrointestinal diseases or dysbiosis induced by the consumption of antibiotics.
Collapse
Affiliation(s)
- Vittorio Saettone
- Department of Veterinary Sciences, School of Agriculture and Veterinary Medicine, University of Turin, Grugliasco, Largo Braccini 2, 10095 Torino, Italy; (V.S.); (A.S.); (D.B.); (G.M.)
| | - Ilaria Biasato
- Department of Agricultural, Forestry and Food Sciences, School of Agriculture and Veterinary Medicine, University of Turin, Grugliasco, Largo Braccini 2, 10095 Torino, Italy
- Correspondence:
| | - Elisabetta Radice
- Department of Surgical Sciences, Medical School, University of Turin, Corso Dogliotti 14, 10126 Torino, Italy;
| | - Achille Schiavone
- Department of Veterinary Sciences, School of Agriculture and Veterinary Medicine, University of Turin, Grugliasco, Largo Braccini 2, 10095 Torino, Italy; (V.S.); (A.S.); (D.B.); (G.M.)
| | - Domenico Bergero
- Department of Veterinary Sciences, School of Agriculture and Veterinary Medicine, University of Turin, Grugliasco, Largo Braccini 2, 10095 Torino, Italy; (V.S.); (A.S.); (D.B.); (G.M.)
| | - Giorgia Meineri
- Department of Veterinary Sciences, School of Agriculture and Veterinary Medicine, University of Turin, Grugliasco, Largo Braccini 2, 10095 Torino, Italy; (V.S.); (A.S.); (D.B.); (G.M.)
| |
Collapse
|
17
|
Chen K, Xin J, Zhang G, Xie H, Luo L, Yuan S, Bu Y, Yang X, Ge Y, Liu C. A combination of three probiotic strains for treatment of acute diarrhoea in hospitalised children: an open label, randomised controlled trial. Benef Microbes 2020; 11:339-346. [PMID: 32720832 DOI: 10.3920/bm2020.0046] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Acute diarrhoea continues to be a leading cause of morbidity, hospitalisation, and mortality worldwide, and probiotics have been proposed as a complementary therapy in the treatment of acute diarrhoea. The goal of this study is to assess the efficacy and safety of three combined probiotic strains, Bifidobacterium lactis Bi-07, Lactobacillus rhamnosus HN001, and Lactobacillus acidophilus NCFM, as an adjunct to rehydration therapy in treatment of acute watery diarrhoea in hospitalised children. Eligible diarrheal children were randomised into intervention group (IG, n=96, conventional treatment for diarrhoea in combination with probiotics) and control group (CG, n=98, conventional treatment for diarrhoea without probiotics). The primary assessments of this study were duration of diarrhoea and hospital stay and improvement in diarrhoea symptoms. Significantly more children in the IG showed improvements in diarrhoea (defined as a decrease of stool frequency to no more than four times per day and an improved stool consistency within 24-48 h after the treatment) than those in the CG (96.9 vs 79.6%, P<0.05). Children supplemented with the mixed strains had a 22.5 h shorter (121.4±13.7 h vs 143.9±19.8 h) mean duration of diarrhoea and 1.2 d shorter hospital stays (5.1±1.2 d vs 6.3±1.4 d) than children only receiving the rehydration therapy (P<0.05). The prevalence of constipation of children in the IG (3.1%) was markedly lower (P<0.05) than that of children in the CG (13.3%) after treatment. In conclusion, the mixture of three probiotic strains given to children aged 1-3 years resulted in shorter durations of diarrhoea and hospitalisation and a higher percentage of improved children.
Collapse
Affiliation(s)
- K Chen
- Department of Nutrition, Chengdu Women's & Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, Sichuan 6100131, China P.R
| | - J Xin
- Infinitus (China) Company Ltd., Hangzhou, Zhejiang, China P.R
| | - G Zhang
- Department of Pediatric Intensive Care Unit, Chengdu Women's & Children's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, Sichuan, China P.R
| | - H Xie
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - L Luo
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - S Yuan
- Department of Nutrition, Chengdu Women's & Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, No. 1617, Riyue Avenue, Qingyang District, Chengdu, Sichuan 6100131, China P.R
| | - Y Bu
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - X Yang
- Department of Pediatrics, Dayi Maternal and Child Health Care Hospital, Chengdu, Sichuan, China P.R
| | - Y Ge
- Infinitus (China) Company Ltd., Hangzhou, Zhejiang, China P.R
| | - C Liu
- School of Exercise and Nutritional Sciences, San Diego State University, 5500 Campanile Dr, San Diego, CA 92182-7251, USA
| |
Collapse
|
18
|
Changes in the Intestinal Microbiome during a Multispecies Probiotic Intervention in Compensated Cirrhosis. Nutrients 2020; 12:nu12061874. [PMID: 32585997 PMCID: PMC7353185 DOI: 10.3390/nu12061874] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 06/17/2020] [Accepted: 06/19/2020] [Indexed: 02/07/2023] Open
Abstract
Probiotics have been used in trials to therapeutically modulate the gut microbiome and have shown beneficial effects in cirrhosis. However, their effect on the microbiome of cirrhosis patients is not fully understood yet. Here, we tested the effects of a multispecies probiotic on microbiome composition in compensated cirrhosis. The gut microbiome composition of 58 patients with compensated cirrhosis from a randomized controlled trial who received a daily dose of multispecies probiotics or placebo for six months was analysed by 16S rRNA gene sequencing. Microbiome composition of patients who received probiotics was enriched with probiotic strains and the abundance of Faecalibacterium prausnitzii, Syntrophococcus sucromutans, Bacteroides vulgatus, Alistipes shahii and a Prevotella species was increased in the probiotic group compared to the placebo group. Patients who had microbiome changes in response to probiotic treatment also showed a significant increase in neopterin and a significant decrease in faecal zonulin levels after intervention, which was not observed in placebo-treated patients or patients with unchanged microbiome compositions. In conclusion, multispecies probiotics may enrich the microbiome of compensated cirrhotic patients with probiotic bacteria during a six-month intervention and beneficially change the residential microbiome and gut barrier function.
Collapse
|
19
|
Behrouzi A, Mazaheri H, Falsafi S, Tavassol ZH, Moshiri A, Siadat SD. Intestinal effect of the probiotic Escherichia coli strain Nissle 1917 and its OMV. J Diabetes Metab Disord 2020; 19:597-604. [PMID: 32550212 DOI: 10.1007/s40200-020-00511-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2019] [Accepted: 02/26/2020] [Indexed: 12/15/2022]
Abstract
Several investigations have been conducted during the past years to examine the correlation between dysbiosis and both intestinal and extra-intestinal diseases such as inflammatory bowel disease (IBD) and ulcerative colitis (UC). E. coli Nissle 1917 (EcN) is a nonpathogenic gram-negative strain utilized in numerous gastrointestinal issues, consisting of diarrhea, uncomplicated diverticular malady, IBD and specifically UC. Many investigations have been done to examine the capability of assertive bacteria, inclusive of commensal and probiotic strains to enhance IBD in clinical testing. Bacterial secreted factors have been investigated to detect the EcN agents that facilitate the regulation of tight junction. These agents candiffuse smoothly through the mucin layer before reaching intestinal epithelial cells. Outer membrane vesicles (OMVs) are known as intercellular communicasomes as they facilitate the distal transfer of active compounds between cells. A few investigations have detailed immune-modulatory attributes for EcN through various systems that could be liable for its clinical viability in IBD. Today, the function of gut microbiota extracellular vesicles in health and disease has become a focus of attention as they serve as vehicles for the transmission of microorganisms to distal tissues of many bacterial effectors.
Collapse
Affiliation(s)
- Ava Behrouzi
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Hoora Mazaheri
- Department of Molecular Biology, Pasteur Institute of Iran , Tehran, Iran
| | - Sarvenaz Falsafi
- Department of Microbiology, Faculty of Advanced Science and Technology, Tehran Medical Science, Islamic Azad University, Tehran, Iran
| | - Zahra Hoseini Tavassol
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran
| | - Arfa Moshiri
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Pasteur Institute of Iran, Tehran, Iran.,Microbiology Research Center (MRC), Pasteur Institute of Iran, Tehran, Iran.,Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
20
|
Niu JW, Zhou L, Liu ZZ, Pei DP, Fan WQ, Ning W. A Systematic Review and Meta-Analysis of the Effects of Perioperative Immunonutrition in Gastrointestinal Cancer Patients. Nutr Cancer 2020; 73:252-261. [PMID: 32285694 DOI: 10.1080/01635581.2020.1749291] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Jin-Wei Niu
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Lei Zhou
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Zhi-Ze Liu
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Dong-Po Pei
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Wen-Qiang Fan
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Wu Ning
- Department of General Surgery, China-Japan Friendship Hospital, Beijing, China
| |
Collapse
|
21
|
Nam B, Kim SA, Park SD, Kim HJ, Kim JS, Bae CH, Kim JY, Nam W, Lee JL, Sim JH. Regulatory effects of Lactobacillus plantarum HY7714 on skin health by improving intestinal condition. PLoS One 2020; 15:e0231268. [PMID: 32275691 PMCID: PMC7147770 DOI: 10.1371/journal.pone.0231268] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 03/19/2020] [Indexed: 01/01/2023] Open
Abstract
Despite increasing research on the gut-skin axis, there is a lack of comprehensive studies on the improvement of skin health through the regulation of the intestinal condition in humans. In this study, we investigated the benefits of Lactobacillus plantarum HY7714 (HY7714) consumption on skin health through its modulatory effects on the intestine and ensuing immune responses. HY7714 consumption led to differences in bacterial abundances from phylum to genus level, including increases in Actinobacteria followed by Bifidobacterium and a decrease in Proteobacteria. Additionally, HY7714 significantly ameliorated inflammation by reducing matrix metallopeptidases (MMP-2 and MMP-9), zonulin, and calprotectin in plasma, all of which are related to skin and intestinal permeability. Furthermore, RNA-seq analysis revealed its efficacy at restoring the integrity of the gut barrier by regulating gene expression associated with the extracellular matrix and immunity. This was evident by the upregulation of IGFBP5, SERPINE1, EFEMP1, COL6A3, and SEMA3B and downregulation of MT2A, MT1E, MT1X, MT1G, and MT1F between TNF- α and TNF- α plus HY7714 treated Caco-2 cells. These results propose the potential mechanistic role of HY7714 on skin health by the regulation of the gut condition.
Collapse
Affiliation(s)
- Bora Nam
- R&BD Center, Korea Yakult Co. Ltd., Yongin, Republic of Korea
| | - Soo A. Kim
- R&BD Center, Korea Yakult Co. Ltd., Yongin, Republic of Korea
| | - Soo Dong Park
- R&BD Center, Korea Yakult Co. Ltd., Yongin, Republic of Korea
| | - Hyeon Ji Kim
- R&BD Center, Korea Yakult Co. Ltd., Yongin, Republic of Korea
| | - Ji Soo Kim
- R&BD Center, Korea Yakult Co. Ltd., Yongin, Republic of Korea
| | - Chu Hyun Bae
- R&BD Center, Korea Yakult Co. Ltd., Yongin, Republic of Korea
| | - Joo Yun Kim
- R&BD Center, Korea Yakult Co. Ltd., Yongin, Republic of Korea
| | - Woo Nam
- R&BD Center, Korea Yakult Co. Ltd., Yongin, Republic of Korea
| | - Jung Lyoul Lee
- R&BD Center, Korea Yakult Co. Ltd., Yongin, Republic of Korea
| | - Jae Hun Sim
- R&BD Center, Korea Yakult Co. Ltd., Yongin, Republic of Korea
- * E-mail:
| |
Collapse
|
22
|
Li Y, Yin Z, Zhang Y, Liu J, Cheng Y, Wang J, Pi F, Zhang Y, Sun X. Perspective of Microbe-based Minerals Fortification in Nutrition Security. FOOD REVIEWS INTERNATIONAL 2020. [DOI: 10.1080/87559129.2020.1728308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Ying Li
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, People’s Republic of China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, People’s Republic of China
- Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, People’s Republic of China
| | - Ziye Yin
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, People’s Republic of China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, People’s Republic of China
- Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, People’s Republic of China
| | - Yuanyuan Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, People’s Republic of China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, People’s Republic of China
- Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, People’s Republic of China
| | - Jinghan Liu
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, People’s Republic of China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, People’s Republic of China
- Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, People’s Republic of China
| | - Yuliang Cheng
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, People’s Republic of China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, People’s Republic of China
- Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, People’s Republic of China
| | - Jiahua Wang
- College of Food Science and Engineering, Wuhan Polytechnic University, Wuhan, People’s Republic of China
| | - Fuwei Pi
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, People’s Republic of China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, People’s Republic of China
- Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, People’s Republic of China
| | - Yinzhi Zhang
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, People’s Republic of China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, People’s Republic of China
- Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, People’s Republic of China
| | - Xiulan Sun
- State Key Laboratory of Food Science and Technology, School of Food Science and Technology, Jiangnan University, Wuxi, People’s Republic of China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, People’s Republic of China
- Synergetic Innovation Center of Food Safety and Quality Control, Jiangnan University, Wuxi, People’s Republic of China
| |
Collapse
|
23
|
Kassaian N, Feizi A, Aminorroaya A, Amini M. Probiotic and synbiotic supplementation could improve metabolic syndrome in prediabetic adults: A randomized controlled trial. Diabetes Metab Syndr 2019; 13:2991-2996. [PMID: 30076087 DOI: 10.1016/j.dsx.2018.07.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2018] [Accepted: 07/29/2018] [Indexed: 02/07/2023]
Abstract
AIMS Modulation of the gastrointestinal microbiome is suggested to contribute to the progression of metabolic syndrome associated diseases. This study was designed to assess the effects of probiotics and synbiotics on metabolic syndrome in individuals with prediabetes. METHODS 120 adults with prediabetes were enrolled in a double-blind, placebo-controlled randomized parallel-group clinical trial. Participants were randomized to a multi-species probiotic or inulin-based synbiotic or placebo. Blood samples and anthropometric measures were collected at baseline, 12 and 24 weeks after treatment. The primary outcome measures were the changes between groups in metabolic syndrome and its components' prevalence. RESULTS A significant trend for a reduction in the prevalence of hyperglycemia in probiotic and synbiotic groups (p = 0.01 and 0.005 respectively), and hypertension in probiotic group (p = 0.04) was found. The decreases in metabolic syndrome prevalence were significant after taking probiotic and synbiotic supplementation as compared with placebo (p = 0.02). Also, the prevalence of low HDL-cholesterol level was decreased during the study in the probiotic group compared with placebo (p = 0.02). CONCLUSIONS The potential benefits of using probiotic and synbiotic for metabolic syndrome management in prediabetes have been supported by the results in the current study which might provide an important strategy to combat metabolic syndrome-associated diseases.
Collapse
Affiliation(s)
- Nazila Kassaian
- Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Awat Feizi
- Professor of Biostatistics. Isfahan Endocrine and Metabolism Research Center and Department of Biostatistics and Epidemiology, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Ashraf Aminorroaya
- Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Masoud Amini
- Isfahan Endocrine and Metabolism Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
24
|
Kamiński M, Skonieczna-Żydecka K, Łoniewski I, Koulaouzidis A, Marlicz W. Are probiotics useful in the treatment of chronic idiopathic constipation in adults? A review of existing systematic reviews, meta-analyses, and recommendations. PRZEGLAD GASTROENTEROLOGICZNY 2019; 15:103-118. [PMID: 32550942 PMCID: PMC7294971 DOI: 10.5114/pg.2019.86747] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 06/03/2019] [Indexed: 12/13/2022]
Abstract
Chronic idiopathic constipation (CIC) has emerged as common problem for contemporary gastroenterology and is one of the most frequent complaints in primary care. Chronic idiopathic constipation significantly affects patients' quality of life and has an impact on global health and economy. Functional gastrointestinal disorders and bowel disorders, according to Rome IV criteria, result from inappropriate gut-brain interactions. The pathophysiology is complex and poorly understood, with evidence accumulating that gut microbiota can be implicated in the development and function of the enteric nervous system. Gut bacteria modulate gut barrier function, short chain fatty acid synthesis, and bile acid metabolism, factors which play roles in the gut peristalsis regulation. The high prevalence of CIC, with poor treatment outcomes, warrants searches for new forms of therapy, including probiotic therapies. Probiotics are often recommended by medical practitioners, but evidence-based utility in adults with CIC is uncertain. Recommendations/guidelines are often based on results from individual studies, rather than meta-analyses or umbrella reviews. Additionally, meta-analyses often indicate a group of probiotics rather than individual strains, and they create difficulty for physicians in making therapeutic choices. More CIC patient randomised clinical studies utilising well-defined strains, or combinations, are necessary.
Collapse
Affiliation(s)
| | | | - Igor Łoniewski
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University, Szczecin, Poland
| | | | - Wojciech Marlicz
- Department of Gastroenterology, Pomeranian Medical University, Szczecin, Poland
| |
Collapse
|
25
|
GOZUYESIL E, ARIOZ DUZGUN A, SUCU M. WOMEN’S CONSUMPTION OF PROBIOTIC FOOD; THE EXAMPLE OF YOGURT AND KEFIR. CLINICAL AND EXPERIMENTAL HEALTH SCIENCES 2019. [DOI: 10.33808/clinexphealthsci.565866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
26
|
Jiang B, Li Z, Ou B, Duan Q, Zhu G. Targeting ideal oral vaccine vectors based on probiotics: a systematical view. Appl Microbiol Biotechnol 2019; 103:3941-3953. [PMID: 30915504 DOI: 10.1007/s00253-019-09770-7] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 12/29/2022]
Abstract
Probiotics have great potential to be engineered into oral vaccine delivery systems, which can facilitate elicitation of mucosal immunity without latent risks of pathogenicity. Combined with the progressive understanding of probiotics and the mucosal immune system as well as the advanced biotechniques of genetic engineering, the development of promising oral vaccine vectors based on probiotics is available while complicated and demanding. Therefore, a systematical view on the design of practical probiotic vectors is necessary, which will help to logically analyze and resolve the problems that might be neglected during our exploration. Here, we attempt to systematically summarize several fundamental issues vital to the effectiveness of the vector of probiotics, including the stability of the engineered vectors, the optimization of antigen expression, the improvement of colonization, and the enhancement of immunoreactivity. We also compared the existent strategies and some developing ones, attempting to figure out an optimal strategy that might deserve to be referred in the future development of oral vaccine vectors based on probiotics.
Collapse
Affiliation(s)
- Boyu Jiang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, China
| | - Zhendong Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, China
| | - Bingming Ou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China.,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, China.,College of Life Science, Zhaoqing University, Zhaoqing, 526061, China
| | - Qiangde Duan
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, China.
| | - Guoqiang Zhu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China. .,Jiangsu Co-Innovation Center for Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety of Ministry of Education of China, Yangzhou, 225009, China.
| |
Collapse
|
27
|
Sudha MR, Jayanthi N, Pandey DC, Verma AK. Bacillus clausii UBBC-07 reduces severity of diarrhoea in children under 5 years of age: a double blind placebo controlled study. Benef Microbes 2019; 10:149-154. [PMID: 30638396 DOI: 10.3920/bm2018.0094] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Acute diarrhoea is one of the leading causes of mortality in infants and young children. Evidence suggests that probiotics can reduce diarrhoea duration. As the effects of probiotics are strain specific, the effect of Bacillus clausii UBBC-07, a safe probiotic strain in the treatment of acute diarrhoea in children was studied. The double blind, randomised, placebo-controlled, parallel group multicentric study was conducted at two outpatient facility sites in Lucknow, India. Children aged six months to five years suffering from acute diarrhoea, were randomly assigned to receive either probiotic (B. clausii UBBC-07) spore suspension or placebo suspension twice daily apart from oral rehydration solution (ORS). The duration of treatment was for five days with a follow -up until the 10th day. Outcomes evaluated were duration and frequency of diarrhoea, consistency of stool, fever and vomiting. The duration of diarrhoea was significantly shorter (P<0.05) in patients who received B. clausii suspension (75.66±13.23 h) than in placebo treated group (81.6±15.43 h). The average daily number of stools (frequency) was 8.67±3.42 at baseline in treatment group receiving B. clausii and 8.53±3.19 in placebo group. By day 4, there was a significant reduction (P<0.01) in frequency of stools in probiotic treated group (3.46±0.66) as compared to placebo group (4.57±1.59). Improvement in stool consistency was also observed in the probiotic treated group as compared to the placebo group. There was no effect on vomiting and duration of fever. B. clausii UBBC-07 significantly decreased the duration and frequency of diarrhoea as compared to placebo indicating effectiveness of strain in the treatment of acute diarrhoea in children and could be a safe alternative to antibiotics.
Collapse
Affiliation(s)
- M Ratna Sudha
- 1 Centre for Research & Development, Unique Biotech Ltd., Plot No. 2, Phase-II, Alexandria Knowledge Park, Hyderabad, Telangana 500078, India
| | - N Jayanthi
- 1 Centre for Research & Development, Unique Biotech Ltd., Plot No. 2, Phase-II, Alexandria Knowledge Park, Hyderabad, Telangana 500078, India
| | - D C Pandey
- 2 MV Hospital and Research Centre, Department of Pediatrics, Room No 01 314/30, Mirza Mandi Chowk, Lucknow, Uttar Pradesh, India
| | - A K Verma
- 3 K.R.M. hospital and Research Centre, Department of Pediatrics, Room No 01 3/92-93,Vijayant Khand Lucknow, Uttar Pradesh, India
| |
Collapse
|
28
|
Rostami FM, Mousavi H, Mousavi MRN, Shahsafi M. Efficacy of Probiotics in Prevention and Treatment of Infectious Diseases. ACTA ACUST UNITED AC 2018. [DOI: 10.1016/j.clinmicnews.2018.06.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
29
|
Mischke M, Arora T, Tims S, Engels E, Sommer N, van Limpt K, Baars A, Oozeer R, Oosting A, Bäckhed F, Knol J. Specific synbiotics in early life protect against diet-induced obesity in adult mice. Diabetes Obes Metab 2018; 20:1408-1418. [PMID: 29460474 PMCID: PMC5969090 DOI: 10.1111/dom.13240] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Revised: 01/17/2018] [Accepted: 01/26/2018] [Indexed: 12/26/2022]
Abstract
AIMS The metabolic state of human adults is associated with their gut microbiome. The symbiosis between host and microbiome is initiated at birth, and early life microbiome perturbation can disturb health throughout life. Here, we determined how beneficial microbiome interventions in early life affect metabolic health in adulthood. METHODS Postnatal diets were supplemented with either prebiotics (scGOS/lcFOS) or synbiotics (scGOS/lcFOS with Bifidobacterium breve M-16 V) until post-natal (PN) day 42 in a well-established rodent model for nutritional programming. Mice were subsequently challenged with a high-fat Western-style diet (WSD) for 8 weeks. Body weight and composition were monitored, as was gut microbiota composition at PN21, 42 and 98. Markers of glucose homeostasis, lipid metabolism and host transcriptomics of 6 target tissues were determined in adulthood (PN98). RESULTS Early life synbiotics protected mice against WSD-induced excessive fat accumulation throughout life, replicable in 2 independent European animal facilities. Adult insulin sensitivity and dyslipidaemia were improved and most pronounced changes in gene expression were observed in the ileum. We observed subtle changes in faecal microbiota composition, both in early life and in adulthood, including increased abundance of Bifidobacterium. Microbiota transplantation using samples collected from synbiotics-supplemented adolescent mice at PN42 to age-matched germ-free recipients did not transfer the beneficial phenotype, indicating that synbiotics-modified microbiota at PN42 is not sufficient to transfer long-lasting protection of metabolic health status. CONCLUSION Together, these findings show the potential and importance of timing of synbiotic interventions in early life during crucial microbiota development as a preventive measure to lower the risk of obesity and improve metabolic health throughout life.
Collapse
Affiliation(s)
| | - Tulika Arora
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Institute of MedicineUniversity of GothenburgGothenburgSweden
| | | | | | - Nina Sommer
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Institute of MedicineUniversity of GothenburgGothenburgSweden
| | | | | | | | | | - Fredrik Bäckhed
- Wallenberg Laboratory and Sahlgrenska Center for Cardiovascular and Metabolic Research, Institute of MedicineUniversity of GothenburgGothenburgSweden
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Jan Knol
- Nutricia ResearchUtrechtThe Netherlands
- Laboratory of MicrobiologyWageningen UniversityWageningenThe Netherlands
| |
Collapse
|
30
|
Ceccato-Antonini SR. Conventional and nonconventional strategies for controlling bacterial contamination in fuel ethanol fermentations. World J Microbiol Biotechnol 2018; 34:80. [PMID: 29802468 DOI: 10.1007/s11274-018-2463-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Accepted: 05/23/2018] [Indexed: 12/17/2022]
Abstract
Ethanol bio-production in Brazil has some unique characteristics that inevitably lead to bacterial contamination, which results in the production of organic acids and biofilms and flocculation that impair the fermentation yield by affecting yeast viability and diverting sugars to metabolites other than ethanol. The ethanol-producing units commonly give an acid treatment to the cells after each fermentative cycle to decrease the bacterial number, which is not always effective. An alternative strategy must be employed to avoid bacterial multiplication but must be compatible with economic, health and environmental aspects. This review analyzes the issue of bacterial contamination in sugarcane-based fuel ethanol fermentation, and the potential strategies that may be utilized to control bacterial growth besides acid treatment and antibiotics. We have emphasized the efficiency and suitability of chemical products other than acids and those derived from natural sources in industrial conditions. In addition, we have also presented bacteriocins, bacteriophages, and beneficial bacteria as non-conventional antimicrobial agents to mitigate bacterial contamination in the bioethanol industry.
Collapse
Affiliation(s)
- Sandra Regina Ceccato-Antonini
- Laboratory of Molecular and Agricultural Microbiology, Department Tecnologia Agroindustrial e Sócio-Economia Rural, Centro de Ciencias Agrárias, Universidade Federal de São Carlos, Via Anhanguera km 174, Araras, SP, 13600-970, Brazil.
| |
Collapse
|
31
|
Rodríguez-Nogales A, Algieri F, Garrido-Mesa J, Vezza T, Utrilla MP, Chueca N, Fernández-Caballero JA, García F, Rodríguez-Cabezas ME, Gálvez J. The Administration of Escherichia coli Nissle 1917 Ameliorates Development of DSS-Induced Colitis in Mice. Front Pharmacol 2018; 9:468. [PMID: 29867475 PMCID: PMC5958303 DOI: 10.3389/fphar.2018.00468] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2017] [Accepted: 04/23/2018] [Indexed: 12/17/2022] Open
Abstract
The beneficial effects of probiotics on immune-based pathologies such as inflammatory bowel disease (IBD) have been well reported. However, their exact mechanisms have not been fully elucidated. Few studies have focused on the impact of probiotics on the composition of the colonic microbiota. The aim of the present study was to correlate the intestinal anti-inflammatory activity of the probiotic Escherichia coli Nissle 1917 (EcN) in the dextran sodium sulfate (DSS) model of mouse colitis with the changes induced in colonic microbiota populations. EcN prevented the DSS-induced colonic damage, as evidenced by lower disease activity index (DAI) values and colonic weight/length ratio, when compared with untreated control mice. The beneficial effects were confirmed biochemically, since the probiotic treatment improved the colonic expression of different cytokines and proteins involved in epithelial integrity. In addition, it restored the expression of different micro-RNAs (miR-143, miR-150, miR-155, miR-223, and miR-375) involved in the inflammatory response that occurs in colitic mice. Finally, the characterization of the colonic microbiota by pyrosequencing showed that the probiotic administration was able to counteract the dysbiosis associated with the intestinal inflammatory process. This effect was evidenced by an increase in bacterial diversity in comparison with untreated colitic mice. The intestinal anti-inflammatory effects of the probiotic EcN were associated with an amelioration of the altered gut microbiome in mouse experimental colitis, especially when considering bacterial diversity, which is reduced in these intestinal conditions. Moreover, this probiotic has shown an ability to modulate expression levels of miRNAs and different mediators of the immune response involved in gut inflammation. This modulation could also be of great interest to understand the mechanism of action of this probiotic in the treatment of IBD.
Collapse
Affiliation(s)
- Alba Rodríguez-Nogales
- CIBEREHD, Department of Pharmacology, Instituto de Investigación Biosanitaria de Granada, Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Francesca Algieri
- CIBEREHD, Department of Pharmacology, Instituto de Investigación Biosanitaria de Granada, Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - José Garrido-Mesa
- CIBEREHD, Department of Pharmacology, Instituto de Investigación Biosanitaria de Granada, Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Teresa Vezza
- CIBEREHD, Department of Pharmacology, Instituto de Investigación Biosanitaria de Granada, Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Maria P Utrilla
- CIBEREHD, Department of Pharmacology, Instituto de Investigación Biosanitaria de Granada, Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Natalia Chueca
- Department of Microbiology, Complejo Hospitalario Universitario de Granada, Instituto de Investigación Biosanitaria de Granada, Granada, Spain
| | - Jose A Fernández-Caballero
- Department of Microbiology, Complejo Hospitalario Universitario de Granada, Instituto de Investigación Biosanitaria de Granada, Granada, Spain
| | - Federico García
- Department of Microbiology, Complejo Hospitalario Universitario de Granada, Instituto de Investigación Biosanitaria de Granada, Granada, Spain
| | - Maria E Rodríguez-Cabezas
- CIBEREHD, Department of Pharmacology, Instituto de Investigación Biosanitaria de Granada, Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
| | - Julio Gálvez
- CIBEREHD, Department of Pharmacology, Instituto de Investigación Biosanitaria de Granada, Centre for Biomedical Research (CIBM), University of Granada, Granada, Spain
| |
Collapse
|
32
|
Abstract
The three main oral diseases of humans, that is, caries, periodontal diseases, and oral candidiasis, are associated with microbiome shifts initiated by changes in the oral environment and/or decreased effectiveness of mucosal immune surveillance. In this review, we discuss the role that microbial-based therapies may have in the control of these conditions. Most investigations on the use of microorganisms for management of oral disease have been conducted with probiotic strains with some positive but very discrete clinical outcomes. Other strategies such as whole oral microbiome transplantation or modification of community function by enrichment with health-promoting indigenous oral strains may offer more promise, but research in this field is still in its infancy. Any microbial-based therapeutics for oral conditions, however, are likely to be only one component within a holistic preventive strategy that should also aim at modification of the environmental influences responsible for the initiation and perpetuation of microbiome shifts associated with oral dysbiosis.
Collapse
|
33
|
Abstract
With the advent of the scientific realization that the microbiota of the gastrointestinal tract was more than the cells that exist in the body, the full importance of prebiotics and probiotics has come forth. The importance has been stressed and is available in the new textbook entitled, "The Microbiota in Gastrointestinal Pathophysiology: Implication for Human Health, Prebiotics, Probiotics and Dysbiosis." There is enough evidence now published in the literature so that the scientific world now believes that prebiotics and probiotics are important in gastrointestinal disease.
Collapse
Affiliation(s)
- Martin H Floch
- Section of Digestive Diseases, Yale University School of Medicine, 333 Cedar Street, 1089 LMP, New Haven, CT 06850, USA.
| |
Collapse
|
34
|
Wasfi R, Abd El‐Rahman OA, Zafer MM, Ashour HM. Probiotic Lactobacillus sp. inhibit growth, biofilm formation and gene expression of caries-inducing Streptococcus mutans. J Cell Mol Med 2018; 22:1972-1983. [PMID: 29316223 PMCID: PMC5824418 DOI: 10.1111/jcmm.13496] [Citation(s) in RCA: 154] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2017] [Accepted: 11/16/2017] [Indexed: 01/01/2023] Open
Abstract
Streptococcus mutans contributes significantly to dental caries, which arises from homoeostasic imbalance between host and microbiota. We hypothesized that Lactobacillus sp. inhibits growth, biofilm formation and gene expression of Streptococcus mutans. Antibacterial (agar diffusion method) and antibiofilm (crystal violet assay) characteristics of probiotic Lactobacillus sp. against Streptococcus mutans (ATCC 25175) were evaluated. We investigated whether Lactobacillus casei (ATCC 393), Lactobacillus reuteri (ATCC 23272), Lactobacillus plantarum (ATCC 14917) or Lactobacillus salivarius (ATCC 11741) inhibit expression of Streptococcus mutans genes involved in biofilm formation, quorum sensing or stress survival using quantitative real-time polymerase chain reaction (qPCR). Growth changes (OD600) in the presence of pH-neutralized, catalase-treated or trypsin-treated Lactobacillus sp. supernatants were assessed to identify roles of organic acids, peroxides and bacteriocin. Susceptibility testing indicated antibacterial (pH-dependent) and antibiofilm activities of Lactobacillus sp. against Streptococcus mutans. Scanning electron microscopy revealed reduction in microcolony formation and exopolysaccharide structural changes. Of the oral normal flora, L. salivarius exhibited the highest antibiofilm and peroxide-dependent antimicrobial activities. All biofilm-forming cells treated with Lactobacillus sp. supernatants showed reduced expression of genes involved in exopolysaccharide production, acid tolerance and quorum sensing. Thus, Lactobacillus sp. can inhibit tooth decay by limiting growth and virulence properties of Streptococcus mutans.
Collapse
Affiliation(s)
- Reham Wasfi
- Department of Microbiology and ImmunologyFaculty of PharmacyOctober University for Modern Sciences and Arts (MSA)GizaEgypt
| | - Ola A. Abd El‐Rahman
- Department of Microbiology and ImmunologyFaculty of PharmacyAl‐Azhar University (Girls)CairoEgypt
| | - Mai M. Zafer
- Department of Microbiology and ImmunologyFaculty of PharmacyAhram Canadian University (ACU)GizaEgypt
| | - Hossam M. Ashour
- Department of Biological SciencesCollege of Arts and SciencesUniversity of South Florida St. PetersburgSt. PetersburgFLUSA
- Department of Microbiology and ImmunologyFaculty of PharmacyCairo UniversityCairoEgypt
| |
Collapse
|
35
|
|
36
|
Holleran G, Lopetuso L, Petito V, Graziani C, Ianiro G, McNamara D, Gasbarrini A, Scaldaferri F. The Innate and Adaptive Immune System as Targets for Biologic Therapies in Inflammatory Bowel Disease. Int J Mol Sci 2017; 18:E2020. [PMID: 28934123 PMCID: PMC5666702 DOI: 10.3390/ijms18102020] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2017] [Revised: 09/10/2017] [Accepted: 09/14/2017] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel disease (IBD) is an immune-mediated inflammatory condition causing inflammation of gastrointestinal and systemic cells, with an increasing prevalence worldwide. Many factors are known to trigger and maintain inflammation in IBD including the innate and adaptive immune systems, genetics, the gastrointestinal microbiome and several environmental factors. Our knowledge of the involvement of the immune system in the pathophysiology of IBD has advanced rapidly over the last two decades, leading to the development of several immune-targeted treatments with a biological source, known as biologic agents. The initial focus of these agents was directed against the pro-inflammatory cytokine tumor necrosis factor-α (TNF-α) leading to dramatic changes in the disease course for a proportion of patients with IBD. However, more recently, it has been shown that a significant proportion of patients do not respond to anti-TNF-α directed therapies, leading a shift to other inflammatory pathways and targets, including those of both the innate and adaptive immune systems, and targets linking both systems including anti-leukocyte trafficking agents-integrins and adhesion molecules. This review briefly describes the molecular basis of immune based gastrointestinal inflammation in IBD, and then describes how several current and future biologic agents work to manipulate these pathways, and their clinical success to date.
Collapse
Affiliation(s)
- Grainne Holleran
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
- Gastroenterology Department, Department of Clinical Medicine, Trinity College Dublin, Dublin 2, Ireland.
| | - Loris Lopetuso
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Valentina Petito
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Cristina Graziani
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Gianluca Ianiro
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Deirdre McNamara
- Gastroenterology Department, Department of Clinical Medicine, Trinity College Dublin, Dublin 2, Ireland.
| | - Antonio Gasbarrini
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| | - Franco Scaldaferri
- Internal Medicine, Gastroenterology and Liver Unit, Gastroenterology Area, Fondazione Policlinico Universitario Gemelli, Università Cattolica del Sacro Cuore, 00168 Rome, Italy.
| |
Collapse
|
37
|
Fábrega MJ, Rodríguez-Nogales A, Garrido-Mesa J, Algieri F, Badía J, Giménez R, Gálvez J, Baldomà L. Intestinal Anti-inflammatory Effects of Outer Membrane Vesicles from Escherichia coli Nissle 1917 in DSS-Experimental Colitis in Mice. Front Microbiol 2017; 8:1274. [PMID: 28744268 PMCID: PMC5504144 DOI: 10.3389/fmicb.2017.01274] [Citation(s) in RCA: 147] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/26/2017] [Indexed: 12/19/2022] Open
Abstract
Escherichia coli Nissle 1917 (EcN) is a probiotic strain with proven efficacy in inducing and maintaining remission of ulcerative colitis. However, the microbial factors that mediate these beneficial effects are not fully known. Gram-negative bacteria release outer membrane vesicles (OMVs) as a direct pathway for delivering selected bacterial proteins and active compounds to the host. In fact, vesicles released by gut microbiota are emerging as key players in signaling processes in the intestinal mucosa. In the present study, the dextran sodium sulfate (DSS)-induced colitis mouse model was used to investigate the potential of EcN OMVs to ameliorate mucosal injury and inflammation in the gut. The experimental protocol involved pre-treatment with OMVs for 10 days before DSS intake, and a 5-day recovery period. Oral administration of purified EcN OMVs (5 μg/day) significantly reduced DSS-induced weight loss and ameliorated clinical symptoms and histological scores. OMVs treatment counteracted altered expression of cytokines and markers of intestinal barrier function. This study shows for the first time that EcN OMVs can mediate the anti-inflammatory and barrier protection effects previously reported for this probiotic in experimental colitis. Remarkably, translation of probiotics to human healthcare requires knowledge of the molecular mechanisms involved in probiotic–host interactions. Thus, OMVs, as a non-replicative bacterial form, could be explored as a new probiotic-derived therapeutic approach, with even lower risk of adverse events than probiotic administration.
Collapse
Affiliation(s)
- María-José Fábrega
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Institut de Biomedicina de la, Universitat de BarcelonaBarcelona, Spain.,Microbiota Intestinal, Institut de Recerca Sant Joan de DéuEsplugues de Llobregat, Spain
| | - Alba Rodríguez-Nogales
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research, University of GranadaGranada, Spain
| | - José Garrido-Mesa
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research, University of GranadaGranada, Spain
| | - Francesca Algieri
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research, University of GranadaGranada, Spain
| | - Josefa Badía
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Institut de Biomedicina de la, Universitat de BarcelonaBarcelona, Spain.,Microbiota Intestinal, Institut de Recerca Sant Joan de DéuEsplugues de Llobregat, Spain
| | - Rosa Giménez
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Institut de Biomedicina de la, Universitat de BarcelonaBarcelona, Spain.,Microbiota Intestinal, Institut de Recerca Sant Joan de DéuEsplugues de Llobregat, Spain
| | - Julio Gálvez
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas, Department of Pharmacology, ibs.GRANADA, Center for Biomedical Research, University of GranadaGranada, Spain
| | - Laura Baldomà
- Departament de Bioquímica i Fisiologia, Facultat de Farmàcia i Ciències de l'Alimentació, Universitat de Barcelona, Institut de Biomedicina de la, Universitat de BarcelonaBarcelona, Spain.,Microbiota Intestinal, Institut de Recerca Sant Joan de DéuEsplugues de Llobregat, Spain
| |
Collapse
|
38
|
Abstract
Inflammatory bowel diseases (IBD) are chronic immune disorders of unclear aetiology. Dietary deficiencies may be a potential pathogenic factor in their development. Patients often take food supplements without knowledge of any evidence base. We have therefore assessed the evidence for food supplementation in the management of IBD. A PubMed search was performed for the terms Inflammatory bowel disease; nutritional deficiencies; dietary supplements; curcumin; green tea; vitamin D/other vitamins; folic acid; iron; zinc; probiotics; andrographis paniculata; and boswellia serrate. PubMed was used to search for all relevant articles published between January 1975 and September 2015. Curcumin supplementation has been reported to be effective in reducing the symptoms and the inflammatory indices in IBD patients. Similar results have been observed for green tea; however, pertinent studies are limited. Vitamin D supplementation may help to increase bone mineral density in IBD patients and to reduce disease activity. IBD patients with ileal resections higher than 20 cm may develop vitamin B12 deficiency that requires parenteral supplementation. There is no current evidence to support fat-soluble vitamin supplementation in IBD patients. Zinc and iron should be supplemented in selected cases. Probiotics (VSL#3) may reduce disease activity in IBD patients with pouchitis. Complementary and alternative medicines are used by IBD patients and some studies have shown promising results. In summary, attention to dietary factors such as curcumin, green tea and vitamins, including vitamins D and B12, appears to be beneficial and, if necessary, supplementation may be appropriate.
Collapse
|
39
|
Horvath A, Leber B, Schmerboeck B, Tawdrous M, Zettel G, Hartl A, Madl T, Stryeck S, Fuchs D, Lemesch S, Douschan P, Krones E, Spindelboeck W, Durchschein F, Rainer F, Zollner G, Stauber RE, Fickert P, Stiegler P, Stadlbauer V. Randomised clinical trial: the effects of a multispecies probiotic vs. placebo on innate immune function, bacterial translocation and gut permeability in patients with cirrhosis. Aliment Pharmacol Ther 2016; 44:926-935. [PMID: 27593544 PMCID: PMC5053220 DOI: 10.1111/apt.13788] [Citation(s) in RCA: 86] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 07/12/2016] [Accepted: 08/08/2016] [Indexed: 02/06/2023]
Abstract
BACKGROUND Probiotics may correct intestinal dysbiosis and proinflammatory conditions in patients with liver cirrhosis. AIM To test the effects of a multispecies probiotic on innate immune function, bacterial translocation and gut permeability. METHODS In a randomised, double blind, placebo-controlled study, stable cirrhotic out-patients either received a daily dose of a probiotic powder containing eight different bacterial strains (Ecologic Barrier, Winclove, Amsterdam, The Netherlands) (n = 44) or a placebo (n = 36) for 6 months and were followed up for another 6 months. RESULTS We found a significant but subclinical increase in neutrophil resting burst (2.6-3.2%, P = 0.0134) and neopterin levels (7.7-8.4 nmol/L, P = 0.001) with probiotics but not with placebo. Probiotic supplementation did not have a significant influence on neutrophil phagocytosis, endotoxin load, gut permeability or inflammatory markers. Ten severe infections occurred in total; one during intervention in the placebo group, and five and four after the intervention has ended in the probiotic and placebo group, respectively. Liver function showed some improvement with probiotics but not with placebo. CONCLUSIONS Probiotic supplementation significantly increased serum neopterin levels and the production of reactive oxygen species by neutrophils. These findings might explain the beneficial effects of probiotics on immune function. Furthermore, probiotic supplementation may be a well-tolerated method to maintain or even improve liver function in stable cirrhosis. However, its influence on gut barrier function and bacterial translocation in cirrhotic patients is minimal.
Collapse
Affiliation(s)
- A. Horvath
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - B. Leber
- Department of Transplantation SurgeryMedical University of GrazGrazAustria,Centre for Biomarker Research in Medicine (CBmed)GrazAustria
| | - B. Schmerboeck
- Department of Transplantation SurgeryMedical University of GrazGrazAustria
| | - M. Tawdrous
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - G. Zettel
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - A. Hartl
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - T. Madl
- Institute of Molecular Biology and BiochemistryMedical University of GrazGrazAustria
| | - S. Stryeck
- Institute of Molecular Biology and BiochemistryMedical University of GrazGrazAustria
| | - D. Fuchs
- Division of Biological ChemistryBiocentreMedical University of InnsbruckInnsbruckAustria
| | - S. Lemesch
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - P. Douschan
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - E. Krones
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - W. Spindelboeck
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - F. Durchschein
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - F. Rainer
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - G. Zollner
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - R. E. Stauber
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - P. Fickert
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| | - P. Stiegler
- Department of Transplantation SurgeryMedical University of GrazGrazAustria
| | - V. Stadlbauer
- Department of Gastroenterology and HepatologyMedical University of GrazGrazAustria
| |
Collapse
|
40
|
|
41
|
Stevens GA. Probiotics for Digestive Health: A Review of Information Resources. JOURNAL OF CONSUMER HEALTH ON THE INTERNET 2016. [DOI: 10.1080/15398285.2016.1202019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
42
|
O'Neill CA, Monteleone G, McLaughlin JT, Paus R. The gut-skin axis in health and disease: A paradigm with therapeutic implications. Bioessays 2016; 38:1167-1176. [PMID: 27554239 DOI: 10.1002/bies.201600008] [Citation(s) in RCA: 245] [Impact Index Per Article: 27.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
As crucial interface organs gut and skin have much in common. Therefore it is unsurprising that several gut pathologies have skin co-morbidities. Nevertheless, the reason for this remains ill explored, and neither mainstream gastroenterology nor dermatology research have systematically investigated the 'gut-skin axis'. Here, in reviewing the field, we propose several mechanistic levels on which gut and skin may interact under physiological and pathological circumstances. We focus on the gut microbiota, with its huge metabolic capacity, and the role of dietary components as potential principle actors along the gut-skin axis. We suggest that metabolites from either the diet or the microbiota are skin accessible. After defining open key questions around the nature of these metabolites, how they are sensed, and which cutaneous changes they can induce, we propose that understanding of these pathways will lead to novel therapeutic strategies based on targeting one organ to improve the health of the other.
Collapse
Affiliation(s)
- Catherine A O'Neill
- Dermatology Research Centre, Institute of Inflammation and Repair, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, UK.
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - John T McLaughlin
- Gastrointestinal Research Centre, Institute of Inflammation and Repair, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, UK
| | - Ralf Paus
- Dermatology Research Centre, Institute of Inflammation and Repair, University of Manchester and Manchester Academic Health Sciences Centre, Manchester, UK.,Department of Dermatology, University of Münster, Münster, Germany
| |
Collapse
|
43
|
Bifidobacterium animalis subsp. lactis in prevention of common infections in healthy children attending day care centers – Randomized, double blind, placebo-controlled study. Clin Nutr 2016; 35:587-91. [DOI: 10.1016/j.clnu.2015.05.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/01/2015] [Accepted: 05/12/2015] [Indexed: 11/17/2022]
|
44
|
Spinler JK, Ross CL, Savidge TC. Probiotics as adjunctive therapy for preventing Clostridium difficile infection - What are we waiting for? Anaerobe 2016; 41:51-57. [PMID: 27180657 DOI: 10.1016/j.anaerobe.2016.05.007] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Revised: 05/06/2016] [Accepted: 05/10/2016] [Indexed: 12/14/2022]
Abstract
With the end of the golden era of antibiotic discovery, the emergence of a new post-antibiotic age threatens to thrust global health and modern medicine back to the pre-antibiotic era. Antibiotic overuse has resulted in the natural evolution and selection of multi-drug resistant bacteria. One major public health threat, Clostridium difficile, is now the single leading cause of hospital-acquired bacterial infections and is by far the most deadly enteric pathogen for the U.S. POPULATION Due to the high morbidity and mortality and increasing incidence that coincides with antibiotic use, non-traditional therapeutics are ideal alternatives to current treatment methods and also provide an avenue towards prevention. Despite the need for alternative therapies to antibiotics and the safety of most probiotics on the market, researchers are inundated with regulatory issues that hinder the translational science required to push these therapies forward. This review discusses the regulatory challenges of probiotic research, expert opinion regarding the application of probiotics to C. difficile infection and the efficacy of probiotics in preventing this disease.
Collapse
Affiliation(s)
- Jennifer K Spinler
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, 1102 Bates Ave., Houston, TX, USA; Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA.
| | - Caná L Ross
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, 1102 Bates Ave., Houston, TX, USA; Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| | - Tor C Savidge
- Texas Children's Microbiome Center, Department of Pathology, Texas Children's Hospital, 1102 Bates Ave., Houston, TX, USA; Department of Pathology & Immunology, Baylor College of Medicine, One Baylor Plaza, Houston, TX, USA
| |
Collapse
|
45
|
Fotbolcu H, Zorlu E. Nonalcoholic fatty liver disease as a multi-systemic disease. World J Gastroenterol 2016; 22:4079-4090. [PMID: 27122660 PMCID: PMC4837427 DOI: 10.3748/wjg.v22.i16.4079] [Citation(s) in RCA: 76] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Revised: 02/02/2016] [Accepted: 03/02/2016] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most common cause of chronic liver disease. NAFLD includes a wide spectrum of liver conditions ranging from simple steatosis to nonalcoholic steatohepatitis and advanced hepatic fibrosis. NAFLD has been recognized as a hepatic manifestation of metabolic syndrome linked with insulin resistance. NAFLD should be considered not only a liver specific disease but also an early mediator of systemic diseases. Therefore, NAFLD is usually associated with cardiovascular disease, chronic kidney disease, type 2 diabetes, obesity, and dyslipidemia. NAFLD is highly prevalent in the general population and is associated with increased cardiovascular morbidity and mortality. The underlying mechanisms and pathogenesis of NAFLD with regard to other medical disorders are not yet fully understood. This review focuses on pathogenesis of NAFLD and its relation with other systemic diseases.
Collapse
|
46
|
Matijašić M, Meštrović T, Perić M, Čipčić Paljetak H, Panek M, Vranešić Bender D, Ljubas Kelečić D, Krznarić Ž, Verbanac D. Modulating Composition and Metabolic Activity of the Gut Microbiota in IBD Patients. Int J Mol Sci 2016; 17:ijms17040578. [PMID: 27104515 PMCID: PMC4849034 DOI: 10.3390/ijms17040578] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2016] [Revised: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 02/06/2023] Open
Abstract
The healthy intestine represents a remarkable interface where sterile host tissues come in contact with gut microbiota, in a balanced state of homeostasis. The imbalance of gut homeostasis is associated with the onset of many severe pathological conditions, such as inflammatory bowel disease (IBD), a chronic gastrointestinal disorder increasing in incidence and severely influencing affected individuals. Despite the recent development of next generation sequencing and bioinformatics, the current scientific knowledge of specific triggers and diagnostic markers to improve interventional approaches in IBD is still scarce. In this review we present and discuss currently available and emerging therapeutic options in modulating composition and metabolic activity of gut microbiota in patients affected by IBD. Therapeutic approaches at the microbiota level, such as dietary interventions alone or with probiotics, prebiotics and synbiotics, administration of antibiotics, performing fecal microbiota transplantation (FMT) and the use of nematodes, all represent a promising opportunities towards establishing and maintaining of well-being as well as improving underlying IBD symptoms.
Collapse
Affiliation(s)
- Mario Matijašić
- Center for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia.
| | - Tomislav Meštrović
- Clinical Microbiology and Parasitology Unit, Polyclinic "Dr. Zora Profozić", Bosutska 19, 10000 Zagreb, Croatia.
| | - Mihaela Perić
- Center for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia.
| | - Hana Čipčić Paljetak
- Center for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia.
| | - Marina Panek
- Center for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia.
| | - Darija Vranešić Bender
- Department of Internal Medicine, Division of Clinical Nutrition, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia.
| | - Dina Ljubas Kelečić
- Department of Internal Medicine, Division of Clinical Nutrition, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia.
| | - Željko Krznarić
- Department of Internal Medicine, Division of Clinical Nutrition, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia.
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Clinical Hospital Centre Zagreb, 10000 Zagreb, Croatia.
- Department of Internal Medicine, University of Zagreb School of Medicine, 10000 Zagreb, Croatia.
| | - Donatella Verbanac
- Center for Translational and Clinical Research, University of Zagreb School of Medicine, 10000 Zagreb, Croatia.
| |
Collapse
|
47
|
Priyamvada S, Anbazhagan AN, Kumar A, Soni V, Alrefai WA, Gill RK, Dudeja PK, Saksena S. Lactobacillus acidophilus stimulates intestinal P-glycoprotein expression via a c-Fos/c-Jun-dependent mechanism in intestinal epithelial cells. Am J Physiol Gastrointest Liver Physiol 2016; 310:G599-608. [PMID: 26867563 PMCID: PMC4836133 DOI: 10.1152/ajpgi.00210.2015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 02/05/2016] [Indexed: 02/08/2023]
Abstract
Our previous studies showed that Lactobacillus acidophilus (LA) culture supernatant (CS) increased P-glycoprotein [Pgp/multidrug resistance 1 (MDR1)] function, expression, and promoter activity in Caco-2 cells. The current studies were designed to elucidate the molecular mechanisms mediating the stimulatory effects of LA CS on Pgp promoter activity. Deletion analysis indicated that the LA CS response element(s) is located in the -172/+428-bp region, and sequence analysis of this region revealed three potential binding sites for c-Fos or c-Jun: proximal activating protein (AP) 1a (-119/-98 bp), distal AP1b (-99/-78 bp), and AP1c (+175/+196 bp). LA CS (24 h) showed an approximately twofold increase in the protein expression of c-Fos and c-Jun in Caco-2 cells. Electrophoretic mobility shift assay showed that LA CS markedly increased the binding of Caco-2 nuclear proteins to AP1a and AP1b, but not AP1c. The DNA-protein complex was completely eliminated by c-Fos antibody, while c-Jun antibody partially eliminated the complex. Chromatin immunoprecipitation analysis also showed that LA CS enhanced the association of c-Fos and c-Jun (by ∼4- and 1.5-fold, respectively) with endogenous Pgp promoter in Caco-2 cells (p-172/+1). Interestingly, overexpression of c-Fos or c-Jun activated Pgp promoter by nearly twofold each. This increase was further enhanced (∼14-fold) when c-Fos and c-Jun were simultaneously overexpressed, suggesting that the presence of one of these transcription factors potentiates the effect of the other. These studies, for the first time, provide evidence for the involvement of c-Fos/c-Jun in stimulation of Pgp gene expression by LA CS in the human intestine.
Collapse
Affiliation(s)
- Shubha Priyamvada
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Arivarasu N. Anbazhagan
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Anoop Kumar
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Vikas Soni
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Waddah A. Alrefai
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and ,2Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Ravinder K. Gill
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| | - Pradeep K. Dudeja
- 1Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and ,2Jesse Brown Veterans Affairs Medical Center, Chicago, Illinois
| | - Seema Saksena
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, Illinois; and
| |
Collapse
|
48
|
Liu S, Li Y, Deng B, Xu Z. Recombinant Lactococcus lactis expressing porcine insulin-like growth factor I ameliorates DSS-induced colitis in mice. BMC Biotechnol 2016; 16:25. [PMID: 26932768 PMCID: PMC4774141 DOI: 10.1186/s12896-016-0255-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 02/22/2016] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Insulin-like growth factor I (IGF-I) is one important family of growth factors, which plays key role in intestinal growth, regeneration, and damage repair. However, the low natural abundance of IGF-I limits its research opportunities and practical application in the fields of medicine and animal husbandry. In this study, a tandem repeat strategy was used to express three copies of the same pIGF-I3 protein in L. lactis. The activity of recombinant pIGF-I3 (rpIGF-I3) was further examined by a mouse model of dextran sulfate sodium (DSS)-induced colitis. In addition, the potential of recombinant L. lactis expressing pIGF-I3 to reduce inflammatory disease was evaluated. RESULTS pIGF-I3 could be expressed in L. lactis by the detection of SDS-PAGE and Western blot. Experimental colitis was induced in BALB/c mice by administration of 5 % DSS in drinking water, and the clinical symptoms were observed in DSS-treated mice. Oral administration of recombinant L. lactis expressing pIGF-I3 improved the colonic architecture, and significantly reduced the increase of colonic damage score (P < 0.05). Furthermore, recombinant L. lactis expressing pIGF-I3 treatment significantly reduced serum DAO activity and colonic MPO level, and elevated colonic occludin level compared to the DSS group (P < 0.05). CONCLUSIONS The pIGF-I3 expressed in L. lactis has good biological activity, and oral administration of recombinant L. lactis expressing pIGF-I3 attenuated the symptoms and development of DSS-induced colitis in mice. These suggested that L. lactis could be a potential host bacterium for production and delivery of IGF-I against intestinal diseases.
Collapse
Affiliation(s)
- Shujie Liu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou, 310021, Zhejiang, China.
| | - Yongming Li
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou, 310021, Zhejiang, China.
| | - Bo Deng
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou, 310021, Zhejiang, China.
| | - Ziwei Xu
- Institute of Animal Husbandry and Veterinary Science, Zhejiang Academy of Agricultural Sciences, 145 Shiqiao Road, Hangzhou, 310021, Zhejiang, China.
| |
Collapse
|
49
|
Goulet O. Potential role of the intestinal microbiota in programming health and disease. Nutr Rev 2016; 73 Suppl 1:32-40. [PMID: 26175488 DOI: 10.1093/nutrit/nuv039] [Citation(s) in RCA: 161] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The composition of the microbiota varies according to prenatal events, delivery methods, infant feeding, infant care environment, and antibiotic use. Postnatal gut function and immune development are largely influenced by the intestinal microbiota. Emerging evidence has shown that early microbiota colonization may influence the occurrence of later diseases (microbial programming). The vast majority of microbial species (commensals) give rise to symbiotic host-bacterial interactions that are fundamental for human health. However, changes in the composition of the gut microbiota (dysbiosis) may be associated with several clinical conditions, including obesity and metabolic diseases, autoimmune diseases and allergy, acute and chronic intestinal inflammation, irritable bowel syndrome (IBS), allergic gastroenteritis (e.g., eosinophilic gastroenteritis and allergic IBS), and necrotizing enterocolitis. Based on recent advances, modulation of gut microbiota with probiotics, prebiotics, or fermented dairy products has been suggested as a treatment of, or prevention for, different disorders such as IBS, infectious diarrhea, allergic disease, and necrotizing enterocolitis.
Collapse
Affiliation(s)
- Olivier Goulet
- O. Goulet is with the Department of Pediatric Gastroenterology-Hepatology-Nutrition, National Reference Center for Rare Digestive Disease, Hôpital Necker-EnfantsMalades, University of Paris Descartes, Paris, France.
| |
Collapse
|
50
|
Goldstein EJC, Johnson S, Maziade PJ, McFarland LV, Trick W, Dresser L, Millette M, Mazloum H, Low DE. Pathway to Prevention of Nosocomial Clostridium difficile Infection. Clin Infect Dis 2016; 60 Suppl 2:S148-58. [PMID: 25922401 DOI: 10.1093/cid/civ142] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND To address the significant morbidity and mortality rates associated with nosocomial Clostridium difficile-associated diarrhea (CDAD), a series of recommendations and a pathway to prevention were developed. METHODS An expert panel of infectious disease (ID) specialists participated in a modified Delphi process with specific objectives: (1) conduct a review for CDAD and prevention; (2) develop statements based upon panel members' opinions; (3) hold a panel meeting during the 2012 IDWeek; and (4) review the final recommendations and prevention pathway prior to submission for publication. RESULTS The panel voted on (1) antibiotic stewardship (7 of 8 panelists); (2) reduction of other potentially modifiable risk factors (variable); (3) utilization of specific probiotics to prevent C. difficile overgrowth (8/8); (4) staff education regarding CDAD preventive measures (8/8); (5) appropriate hand hygiene for everyone (7/8); (6) environmental cleaning (8/8); (7) medical equipment disinfection (7/8); (8) early detection of CDAD in symptomatic patients (7/8); (9) usage of protective clothing/gloves (8/8); (10) proper measures during outbreak (8/8); and (11) surveillance to monitor efficacy data of preventive measures (8/8). CONCLUSIONS The panel members agreed with 11 of 17 recommendations presented. The additional recommendations by the panel were proton pump inhibitor use as a risk factor and the use of adjunctive therapy with specific probiotic, as it was approved by Health Canada for the risk reduction of CDAD in hospitalized patients.
Collapse
Affiliation(s)
- Ellie J C Goldstein
- RM Alden Research Laboratory and the David Geffen School of Medicine at UCLA, Los Angeles, California
| | - Stuart Johnson
- Hines VA Hospital and Loyola University Medical Center, Chicago, Illinois
| | | | - Lynne V McFarland
- Department of Medicinal Chemistry, University of Washington, Seattle
| | | | - Linda Dresser
- Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario
| | | | | | - Donald E Low
- Mount Sinai Hospital, University of Toronto, Ontario, Canada
| |
Collapse
|