1
|
Rybicka I, Kaźmierczak Z. The human phageome: niche-specific distribution of bacteriophages and their clinical implications. Appl Environ Microbiol 2025; 91:e0178824. [PMID: 40237489 DOI: 10.1128/aem.01788-24] [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] [Indexed: 04/18/2025] Open
Abstract
Bacteriophages (phages) play a crucial role in shaping the composition and diversity of the human microbiome across various body niches. Recent advancements in high-throughput sequencing technologies have enabled comprehensive analysis of the human phageome in different body sites. This review comprehensively analyzes phage populations across major human body niches, examining their distribution and dynamics through recent metagenomic discoveries. We explore how phage-bacteria interactions within different body sites contribute to homeostasis and disease development. Emerging evidence demonstrates that phageome perturbations can serve as early indicators of various disorders, particularly in the gut microbiome. Understanding these complex microbial interactions offers promising opportunities for developing novel diagnostic markers and therapeutic approaches. However, the causal relationship between phages, bacteria, and disease development remains unclear. Further research is needed to elucidate the role of phages in human health and disease and to explore their potential as diagnostic or therapeutic tools. Understanding the intricate interactions between phages, bacteria, and the human host is crucial for unraveling the complexities of the human microbiome and its impact on health and disease.
Collapse
Affiliation(s)
- Izabela Rybicka
- Laboratory of Phage Molecular Biology, Hirszfeld Institute of Immunology and Experimental Therapy, Wrocław, Poland
| | - Zuzanna Kaźmierczak
- Laboratory of Phage Molecular Biology, Hirszfeld Institute of Immunology and Experimental Therapy, Wrocław, Poland
- Research and Development Center, Regional Specialist Hospital in Wrocław, Wrocław, Poland
- Faculty of Medicine, Department of Preclinical Sciences, Pharmacology and Medical Diagnostics, Wrocław University of Science and Technology, Wrocław, Poland
| |
Collapse
|
2
|
Pai VV, Sarath AP, Kerkar Z. Gut microbiome in dermatology - A narrative review. Indian J Dermatol Venereol Leprol 2025; 0:1-11. [PMID: 40357977 DOI: 10.25259/ijdvl_1094_2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Accepted: 12/08/2024] [Indexed: 05/15/2025]
Abstract
The gut microbiome and human body have co-evolved in a synergistic host-microbial relationship. The ideal composition of human gut microbiota is an elusive concept, but every individual has a unique gut microbiota profile with regional differences. Newer diagnostic techniques have helped identify different bacteria and their roles in health and disease. The gut microbiome composition is affected by various factors like age, diet, immune system, environmental factors, exercise, and drugs. The microbiome has varied roles in metabolism, immune response, immune tolerance and antimicrobial protection. Diet plays an important role in maintaining the gut microbial diversity. Loss of homoeostasis in the microbiome results in dysbiosis. Dysbiosis plays a role in many dermatological diseases like atopic dermatitis, psoriasis, acne, rosacea, hidradenitis suppurativa, connective tissue disorders and many other systemic conditions like obesity, diabetes, neurological disease and malignancy. Reconstitution of the gut microbiome ecology in the form of bacteriotherapy with the reintegration of certain strains of microbiota has a beneficial role in many of these disorders.
Collapse
Affiliation(s)
| | | | - Zenia Kerkar
- Department of Dermatology, Goa Medical College, Bambolim, India
| |
Collapse
|
3
|
Liu H, Yin J, Huang X, Zang C, Zhang Y, Cao J, Gong M. Mosquito Gut Microbiota: A Review. Pathogens 2024; 13:691. [PMID: 39204291 PMCID: PMC11357333 DOI: 10.3390/pathogens13080691] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2024] [Revised: 07/29/2024] [Accepted: 08/06/2024] [Indexed: 09/03/2024] Open
Abstract
Mosquitoes are vectors of many important human diseases. The prolonged and widespread use of insecticides has led to the development of mosquito resistance to these insecticides. The gut microbiota is considered the master of host development and physiology; it influences mosquito biology, disease pathogen transmission, and resistance to insecticides. Understanding the role and mechanisms of mosquito gut microbiota in mosquito insecticide resistance is useful for developing new strategies for tackling mosquito insecticide resistance. We searched online databases, including PubMed, MEDLINE, SciELO, Web of Science, and the Chinese Science Citation Database. We searched all terms, including microbiota and mosquitoes, or any specific genera or species of mosquitoes. We reviewed the relationships between microbiota and mosquito growth, development, survival, reproduction, and disease pathogen transmission, as well as the interactions between microbiota and mosquito insecticide resistance. Overall, 429 studies were included in this review after filtering 8139 search results. Mosquito gut microbiota show a complex community structure with rich species diversity, dynamic changes in the species composition over time (season) and across space (environmental setting), and variation among mosquito species and mosquito developmental stages (larval vs. adult). The community composition of the microbiota plays profound roles in mosquito development, survival, and reproduction. There was a reciprocal interaction between the mosquito midgut microbiota and virus infection in mosquitoes. Wolbachia, Asaia, and Serratia are the three most studied bacteria that influence disease pathogen transmission. The insecticide resistance or exposure led to the enrichment or reduction in certain microorganisms in the resistant mosquitoes while enhancing the abundance of other microorganisms in insect-susceptible mosquitoes, and they involved many different species/genera/families of microorganisms. Conversely, microbiota can promote insecticide resistance in their hosts by isolating and degrading insecticidal compounds or altering the expression of host genes and metabolic detoxification enzymes. Currently, knowledge is scarce about the community structure of mosquito gut microbiota and its functionality in relation to mosquito pathogen transmission and insecticide resistance. The new multi-omics techniques should be adopted to find the links among environment, mosquito, and host and bring mosquito microbiota studies to the next level.
Collapse
Affiliation(s)
- Hongmei Liu
- Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai 200025, China;
- Digestive Disease Hospital of Shandong First Medical University, Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining 272000, China; (X.H.); (C.Z.); (Y.Z.)
- World Health Organization Collaborating Centre for Tropical Diseases, Shanghai 200025, China
| | - Jianhai Yin
- Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai 200025, China;
- World Health Organization Collaborating Centre for Tropical Diseases, Shanghai 200025, China
| | - Xiaodan Huang
- Digestive Disease Hospital of Shandong First Medical University, Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining 272000, China; (X.H.); (C.Z.); (Y.Z.)
| | - Chuanhui Zang
- Digestive Disease Hospital of Shandong First Medical University, Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining 272000, China; (X.H.); (C.Z.); (Y.Z.)
| | - Ye Zhang
- Digestive Disease Hospital of Shandong First Medical University, Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining 272000, China; (X.H.); (C.Z.); (Y.Z.)
| | - Jianping Cao
- Key Laboratory of Parasite and Vector Biology, National Health Commission of People’s Republic of China, National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai 200025, China;
- World Health Organization Collaborating Centre for Tropical Diseases, Shanghai 200025, China
| | - Maoqing Gong
- Digestive Disease Hospital of Shandong First Medical University, Shandong Institute of Parasitic Diseases, Shandong First Medical University & Shandong Academy of Medical Sciences, Jining 272000, China; (X.H.); (C.Z.); (Y.Z.)
| |
Collapse
|
4
|
Kulecka M, Czarnowski P, Bałabas A, Turkot M, Kruczkowska-Tarantowicz K, Żeber-Lubecka N, Dąbrowska M, Paszkiewicz-Kozik E, Walewski J, Ługowska I, Koseła-Paterczyk H, Rutkowski P, Kluska A, Piątkowska M, Jagiełło-Gruszfeld A, Tenderenda M, Gawiński C, Wyrwicz L, Borucka M, Krzakowski M, Zając L, Kamiński M, Mikula M, Ostrowski J. Microbial and Metabolic Gut Profiling across Seven Malignancies Identifies Fecal Faecalibacillus intestinalis and Formic Acid as Commonly Altered in Cancer Patients. Int J Mol Sci 2024; 25:8026. [PMID: 39125593 PMCID: PMC11311272 DOI: 10.3390/ijms25158026] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 07/18/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
The key association between gut dysbiosis and cancer is already known. Here, we used whole-genome shotgun sequencing (WGS) and gas chromatography/mass spectrometry (GC/MS) to conduct metagenomic and metabolomic analyses to identify common and distinct taxonomic configurations among 40, 45, 71, 34, 50, 60, and 40 patients with colorectal cancer, stomach cancer, breast cancer, lung cancer, melanoma, lymphoid neoplasms and acute myeloid leukemia (AML), respectively, and compared the data with those from sex- and age-matched healthy controls (HC). α-diversity differed only between the lymphoid neoplasm and AML groups and their respective HC, while β-diversity differed between all groups and their HC. Of 203 unique species, 179 and 24 were under- and over-represented, respectively, in the case groups compared with HC. Of these, Faecalibacillus intestinalis was under-represented in each of the seven groups studied, Anaerostipes hadrus was under-represented in all but the stomach cancer group, and 22 species were under-represented in the remaining five case groups. There was a marked reduction in the gut microbiome cancer index in all case groups except the AML group. Of the short-chain fatty acids and amino acids tested, the relative concentration of formic acid was significantly higher in each of the case groups than in HC, and the abundance of seven species of Faecalibacterium correlated negatively with most amino acids and formic acid, and positively with the levels of acetic, propanoic, and butanoic acid. We found more differences than similarities between the studied malignancy groups, with large variations in diversity, taxonomic/metabolomic profiles, and functional assignments. While the results obtained may demonstrate trends rather than objective differences that correlate with different types of malignancy, the newly developed gut microbiota cancer index did distinguish most of the cancer cases from HC. We believe that these data are a promising step forward in the search for new diagnostic and predictive tests to assess intestinal dysbiosis among cancer patients.
Collapse
Affiliation(s)
- Maria Kulecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Paweł Czarnowski
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Aneta Bałabas
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Maryla Turkot
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
- Department of Cancer Prevention, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Kamila Kruczkowska-Tarantowicz
- Department of Internal Medicine and Hematology, Military Institute of Medicine—National Research Institute, 04-141 Warsaw, Poland
| | - Natalia Żeber-Lubecka
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Michalina Dąbrowska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Ewa Paszkiewicz-Kozik
- Department of Lymphoid Malignancies, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Jan Walewski
- Department of Lymphoid Malignancies, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Iwona Ługowska
- Early Phase Clinical Trials Unit, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Hanna Koseła-Paterczyk
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Piotr Rutkowski
- Department of Soft Tissue/Bone Sarcoma and Melanoma, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Anna Kluska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Magdalena Piątkowska
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Agnieszka Jagiełło-Gruszfeld
- Department of Breast Cancer & Reconstructive Surgery, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Michał Tenderenda
- Department of Oncological Surgery and Neuroendocrine Tumors, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Cieszymierz Gawiński
- Department of Oncology and Radiotherapy, Maria Sklodowska-Curie National Cancer Research Institute, 02-781 Warsaw, Poland
| | - Lucjan Wyrwicz
- Department of Oncology and Radiotherapy, Maria Sklodowska-Curie National Cancer Research Institute, 02-781 Warsaw, Poland
| | - Magdalena Borucka
- Department of Lung and Chest Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Maciej Krzakowski
- Department of Lung and Chest Cancer, Maria Skłodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Leszek Zając
- Department of Gastrointestinal Surgical Oncology, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Michał Kamiński
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
- Department of Cancer Prevention, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Michał Mikula
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| | - Jerzy Ostrowski
- Department of Gastroenterology, Hepatology and Clinical Oncology, Centre of Postgraduate Medical Education, 02-781 Warsaw, Poland
- Department of Genetics, Maria Sklodowska-Curie National Research Institute of Oncology, 02-781 Warsaw, Poland
| |
Collapse
|
5
|
Suswał K, Tomaszewski M, Romaniuk A, Świechowska-Starek P, Zygmunt W, Styczeń A, Romaniuk-Suswał M. Gut-Lung Axis in Focus: Deciphering the Impact of Gut Microbiota on Pulmonary Arterial Hypertension. J Pers Med 2023; 14:8. [PMID: 38276223 PMCID: PMC10817474 DOI: 10.3390/jpm14010008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 12/11/2023] [Accepted: 12/14/2023] [Indexed: 01/27/2024] Open
Abstract
Recent advancements in the understanding of pulmonary arterial hypertension (PAH) have highlighted the significant role of the gut microbiota (GM) in its pathogenesis. This comprehensive review delves into the intricate relationship between the GM and PAH, emphasizing the influence of gut microbial composition and the critical metabolites produced. We particularly focus on the dynamic interaction between the gut and lung, examining how microbial dysbiosis contributes to PAH development through inflammation, altered immune responses, and changes in the gut-lung axis. Noteworthy findings include variations in the ratios of key bacterial groups such as Firmicutes and Bacteroidetes in PAH and the pivotal roles of metabolites like trimethylamine N-oxide (TMAO), short-chain fatty acids (SCFAs), and serotonin in the disease's progression. Additionally, the review elucidates potential diagnostic biomarkers and novel therapeutic approaches, including the use of probiotics and fecal microbiota transplantation, which leverage the gut microbiota for managing PAH. This review encapsulates the current state of research in this field, offering insights into the potential of gut microbiota modulation as a promising strategy in PAH diagnosing and treatment.
Collapse
Affiliation(s)
- Konrad Suswał
- Department of Pulmonology, Alergollogy and Oncology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Michał Tomaszewski
- Department of Cardiology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Aleksandra Romaniuk
- Cardiology Student Scientific Circle, Academy of Silesia, 40-555 Katowice, Poland;
| | | | - Wojciech Zygmunt
- Department of Pulmonology, Alergollogy and Oncology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Agnieszka Styczeń
- Department of Cardiology, Medical University of Lublin, 20-954 Lublin, Poland;
| | - Małgorzata Romaniuk-Suswał
- Department of Psychiatry, Psychotheraphy and Early Intervention, Medical University of Lublin, 20-954 Lublin, Poland
| |
Collapse
|
6
|
Xue H, Mei C, Wang F, Tang X. Relationship among Chinese herb polysaccharide (CHP), gut microbiota, and chronic diarrhea and impact of CHP on chronic diarrhea. Food Sci Nutr 2023; 11:5837-5855. [PMID: 37823142 PMCID: PMC10563694 DOI: 10.1002/fsn3.3596] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 07/13/2023] [Accepted: 07/22/2023] [Indexed: 10/13/2023] Open
Abstract
Chronic diarrhea, including diarrhea-predominant irritable bowel syndrome (IBS-D), osmotic diarrhea, bile acid diarrhea, and antibiotic-associated diarrhea, is a common problem which is highly associated with disorders of the gut microbiota composition such as small intestinal bacterial overgrowth (SIBO) and so on. A growing number of studies have supported the view that Chinese herbal formula alleviates the symptoms of diarrhea by modulating the fecal microbiota. Chinese herbal polysaccharides (CHPs) are natural polymers composed of monosaccharides that are widely found in Chinese herbs and function as important active ingredients. Commensal gut microbiota has an extensive capacity to utilize CHPs and play a vital role in degrading polysaccharides into short-chain fatty acids (SCFAs). Many CHPs, as prebiotics, have an antidiarrheal role to promote the growth of beneficial bacteria and inhibit the colonization of pathogenic bacteria. This review systematically summarizes the relationship among gut microbiota, chronic diarrhea, and CHPs as well as recent progress on the impacts of CHPs on the gut microbiota and recent advances on the possible role of CHPs in chronic diarrhea.
Collapse
Affiliation(s)
- Hong Xue
- Digestive Laboratory of Traditional Chinese Medicine Research Institute of Spleen and Stomach DiseasesXiyuan Hospital, China Academy of Chinese Medical SciencesBeijingChina
| | - Chun‐Feng Mei
- Digestive Laboratory of Traditional Chinese Medicine Research Institute of Spleen and Stomach DiseasesXiyuan Hospital, China Academy of Chinese Medical SciencesBeijingChina
| | - Feng‐Yun Wang
- Digestive Laboratory of Traditional Chinese Medicine Research Institute of Spleen and Stomach DiseasesXiyuan Hospital, China Academy of Chinese Medical SciencesBeijingChina
| | - Xu‐Dong Tang
- Digestive Laboratory of Traditional Chinese Medicine Research Institute of Spleen and Stomach DiseasesXiyuan Hospital, China Academy of Chinese Medical SciencesBeijingChina
| |
Collapse
|
7
|
Costa SK, Antosca K, Beekman CN, Peterson RL, Penumutchu S, Belenky P. Short-Term Dietary Intervention with Whole Oats Protects from Antibiotic-Induced Dysbiosis. Microbiol Spectr 2023; 11:e0237623. [PMID: 37439681 PMCID: PMC10434222 DOI: 10.1128/spectrum.02376-23] [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: 06/06/2023] [Accepted: 06/22/2023] [Indexed: 07/14/2023] Open
Abstract
Antibiotic-induced gut microbiome dysbiosis (AID) is known to be influenced by host dietary composition. However, how and when diet modulates gut dysbiosis remains poorly characterized. Thus, here, we utilize a multi-omics approach to characterize how a diet supplemented with oats, a rich source of microbiota-accessible carbohydrates, or dextrose impacts amoxicillin-induced changes to gut microbiome structure and transcriptional activity. We demonstrate that oat administration during amoxicillin challenge provides greater protection from AID than the always oats or recovery oats diet groups. In particular, the group in which oats were provided at the time of antibiotic exposure induced the greatest protection against AID while the other oat diets saw greater effects after amoxicillin challenge. The oat diets likewise reduced amoxicillin-driven elimination of Firmicutes compared to the dextrose diet. Functionally, gut communities fed dextrose were carbohydrate starved and favored respiratory metabolism and consequent metabolic stress management while oat-fed communities shifted their transcriptomic profile and emphasized antibiotic stress management. The metabolic trends were exemplified when assessing transcriptional activity of the following two common gut commensal bacteria: Akkermansia muciniphila and Bacteroides thetaiotaomicron. These findings demonstrate that while host diet is important in shaping how antibiotics effect the gut microbiome composition and function, diet timing may play an even greater role in dietary intervention-based therapeutics. IMPORTANCE We utilize a multi-omics approach to demonstrate that diets supplemented with oats, a rich source of microbiota-accessible carbohydrates, are able to confer protection against antibiotic-induced dysbiosis (AID). Our findings affirm that not only is host diet important in shaping antibiotics effects on gut microbiome composition and function but also that the timing of these diets may play an even greater role in managing AID. This work provides a nuanced perspective on dietary intervention against AID and may be informative on preventing AID during routine antibiotic treatment.
Collapse
Affiliation(s)
- Stephen K. Costa
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Katherine Antosca
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Chapman N. Beekman
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Rachel L. Peterson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Swathi Penumutchu
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island, USA
| |
Collapse
|
8
|
Latif AS, Saparbekova AA, Akhmedova ZR, Kaldybekova G, Daugaliyeva ST. Probiotic yeast Saccharomyces cerevisiae Az-12 isolated from pomegranate juice presented inhibitory effects against pathogenic bacteria. BRAZ J BIOL 2023; 83:e271997. [PMID: 37585928 DOI: 10.1590/1519-6984.271997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Accepted: 06/04/2023] [Indexed: 08/18/2023] Open
Abstract
The potential probiotic yeast was isolated from the Kyzyl Anor pomegranate variety growing in the Turkestan region (Kazakhstan). The yeast strain was identified as Saccharomyces cerevisiae Az-12. Molecular genetic identification was carried out using the Sanger sequencing method. The degree of homology of the S. cerevisiae Az-12 strain with the strain MH608341.1 Saccharomyces cerevisiae isolate extr03 was 99.65%. Antagonistic effect of the yeast against pathogenic bacteria was confirmed according inhibition zones for Staphylococcus aureus 13.5 ± 0.05 mm; the inhibition zones for Escherichia coli 12.8 ± 0.05 mm; and 10.7 ± 0.05 mm for Pseudomonas aeruginosa. Scanning microscopy of S. cerevisiae Az-12 and S. aureus confirmed the adhesive ability of the yeast cell surface to S. aureus. S. cerevisiae Az-12 were chosen as the most promising, as they are able to quickly ferment juices. Functional drinks containing pomegranate juice and yeast with a probiotic effect can be considered as a useful synbiotic product formulation.
Collapse
Affiliation(s)
- A S Latif
- M. Auezov South Kazakhstan University, Department of Biotechnology, Shymkent, Kazakhstan
| | - A A Saparbekova
- M. Auezov South Kazakhstan University, Department of Biotechnology, Shymkent, Kazakhstan
| | - Z R Akhmedova
- Institute of Microbiology of the Academy of Sciences of the Republic of Uzbekistan, Department of Environmental Biotechnology, Tashkent, Uzbekistan
| | - G Kaldybekova
- M. Auezov South Kazakhstan University, Department of Biotechnology, Shymkent, Kazakhstan
| | - S T Daugaliyeva
- Institute of Microbiology and Virology, Laboratory of Molecular Genetics, Almaty, Kazakhstan
| |
Collapse
|
9
|
Ji L, Deng H, Xue H, Wang J, Hong K, Gao Y, Kang X, Fan G, Huang W, Zhan J, You Y. Research progress regarding the effect and mechanism of dietary phenolic acids for improving nonalcoholic fatty liver disease via gut microbiota. Compr Rev Food Sci Food Saf 2023; 22:1128-1147. [PMID: 36717374 DOI: 10.1111/1541-4337.13106] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 02/01/2023]
Abstract
Phenolic acids (PAs), a class of small bioactive molecules widely distributed in food and mainly found as secondary plant metabolites, present significant advantages such as antioxidant activity and other health benefits. The global epidemic of nonalcoholic fatty liver disease (NAFLD) is becoming a serious public health problem. Existing studies showed that gut microbiota (GM) dysbiosis is highly associated with the occurrence and development of NAFLD. In recent years, progress has been made in the study of the relationship among PA compounds, GM, and NAFLD. PAs can regulate the composition and functions of the GM to promote human health, while GM can increase the dietary sources of PAs and improve its bioavailability. This paper discussed PAs, GM, and their interrelationship while introducing several representative dietary PA sources and examining the absorption and metabolism of PAs mediated by GM. It also summarizes the effect and mechanisms of PAs in improving and regulating NAFLD via GM and their metabolites. This helps to better evaluate the potential preventive effect of PAs on NAFLD via the regulation of GM and expands the utilization of PAs and PA-rich food resources.
Collapse
Affiliation(s)
- Lin Ji
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Huan Deng
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Huimin Xue
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Jiting Wang
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Kexin Hong
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Yunxiao Gao
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Xiping Kang
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Guanghe Fan
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Weidong Huang
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Jicheng Zhan
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| | - Yilin You
- College of Food Science and Nutritional Engineering, Beijing Key Laboratory of Viticulture and Enology, China Agricultural University, Beijing, China
| |
Collapse
|
10
|
Maev IV, Andreev DN, Sokolov PS, Fomenko AК, Devkota MK, Andreev NG, Zaborovsky AV. [Efficacy of Saccharomyces boulardii CNCM I-745 probiotic drug in the prevention and treatment of diarrhea in hospitalized patients with new coronavirus infection COVID-19]. TERAPEVT ARKH 2022; 94:1163-1170. [PMID: 36468990 DOI: 10.26442/00403660.2022.10.201881] [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: 11/22/2022] [Accepted: 11/22/2022] [Indexed: 11/23/2022]
Abstract
AIM To evaluate the efficacy of Saccharomyces boulardii (S. boulardii) CNCM I-745 probiotic drug in preventing and treating diarrhea in hospitalized patients with COVID-19. MATERIALS AND METHODS A prospective comparative study was conducted in two parallel groups. The study included males and females aged 18 to 60 with the following diagnosis confirmed by polymerase chain reaction: U07.2 Coronavirus infection COVID-19, caused by SARS-CoV-2 virus (grade 1-3 pneumonia according to CT scan). All patients received antibiotic therapy. The patients were subdivided into two equal groups (n=60) depending on the administration of S. boulardii CNCM I-745 probiotic drug in addition to standard treatment. The probiotic was prescribed by the attending physician; the dose was 2 capsules per day (500 mg/day) 30 min before the meal for 10 days. All patients were monitored for main clinical, laboratory, and instrumental parameters during the study. In addition, the symptom of diarrhea (stool with a frequency of more than 3 times a day of type 6 and 7 according to the Bristol stool scale), including its frequency, duration, and the number of bowel movements of loose stool per day were precisely evaluated in both groups. RESULTS In the overall patient pool, diarrhea was reported in 21.7% of in-patients during the observation period (95% confidence interval [CI] 14.2-29.1) with a mean duration of 4.6154 days (95% CI 3.7910-5.4398). The incidence of diarrhea in group 1 was 13.3% (95% CI 4.5-22.2), and in group 2, it was 30.0% (95% CI 18.1-41.9). Relative risk showed that the use of the S. boulardii CNCM I-745 probiotic drug leads to a significant reduction in the risk of diarrhea in hospitalized patients with COVID-19 infection receiving antibiotic therapy (odds ratio [OR] 0.3590, 95% CI 0.1421-0.9069; p=0.0303). In group 1, the duration of diarrhea was 3.1250 days (95% CI 2.5892-3.6608) versus 5.2778 days (95% CI 4.2290-6.3265) in group 2, p=0.0112. The mean daily frequency of loose stools in patients with diarrhea in group 1 was 3.2500 (95% CI 2.6588-3.8412) versus 4.3889 (95% CI 3.7252-5.0525) in group 2, p=0.0272. The secondary endpoint, duration of hospital stay, was also significantly shorter in group 1 patients - 11.6833 days (95% CI 11.2042-12.1625) versus 12.7333 days (95% CI 12.1357-13.3309) in group 2, p=0.0120. CONCLUSION The present prospective comparative study demonstrated that adding S. boulardii CNCM I-745 probiotic drug into the standard treatment regimen of patients with new coronavirus infection COVID-19 receiving antibiotic therapy helps reduce the incidence of diarrhea and its severity during hospitalization, as well as the duration of hospital stay.
Collapse
Affiliation(s)
- I V Maev
- Yevdokimov Moscow State University of Medicine and Dentistry
| | - D N Andreev
- Yevdokimov Moscow State University of Medicine and Dentistry
| | - P S Sokolov
- Yevdokimov Moscow State University of Medicine and Dentistry
| | - A К Fomenko
- Yevdokimov Moscow State University of Medicine and Dentistry
| | - M K Devkota
- Yevdokimov Moscow State University of Medicine and Dentistry
| | - N G Andreev
- Yevdokimov Moscow State University of Medicine and Dentistry
| | - A V Zaborovsky
- Yevdokimov Moscow State University of Medicine and Dentistry
| |
Collapse
|
11
|
Ali K, Zaidi S, Khan AA, Khan AU. Orally fed EGCG coronate food released TiO 2 and enhanced penetrability into body organs via gut. BIOMATERIALS ADVANCES 2022; 144:213205. [PMID: 36442452 DOI: 10.1016/j.bioadv.2022.213205] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 10/16/2022] [Accepted: 11/16/2022] [Indexed: 11/22/2022]
Abstract
Owing to unique nano-scale properties, TiO2-NPs (T-NPs) are employed as food-quality enhancers in >900 processed food products. Whereas, epigallocatechin-3-gallate (EGCG), a green tea polyphenol is consumed in traditional brewed tea, globally. Taken together, we aimed to investigate whether human gastric-acid digested T-NPs and complex tea catechins yield ionic species (Ti4+, Ti3+ etc.) and active EGCG forms to meet favourable conditions for in vivo bio-genesis of EGCG-coronated TiO2-NPs (ET-NPs) in human gut. Secondly, compared to bare-surface micro and nano-scale TiO2, i.e., T-MPs and T-NPs, respectively, how EGCG coronation on ET-NPs in the gut facilitates the modulation of intrinsic propensity of internalization of TiO2 species into bacteria, body-organs, and gut-microbiota (GM), and immune system. ET-NPs were synthesized in non-toxic aqueous solution at varied pH (3-10) and characterised by state-of-the-arts for crystallinity, surface-charge, EGCG-encapsulation, stability, size, composition and morphology. Besides, flow-cytometry (FCM), TEM, EDS, histopathology, RT-PCR, 16S-rRNA metagenomics and ELISA were also performed to assess the size and surface dependent activities of ET-NPs, T-NPs and T-MPs vis-a-vis planktonic bacteria, biofilm, GM bacterial communities and animal's organs. Electron-microscopic, NMR, FTIR, DLS, XRD and EDS confirmed the EGCG coronation, dispersity, size-stability of ET-NPs, crystallinity and elemental composition of ET-NPs-8 and T-NPs. Besides, FCM, RT-PCR, 16S-rRNA metagenomics, histopathology, SEM and EDS analyses exhibited that EGCG coronation in ET-NPs-8 enhanced the penetration into body organs (i.e., liver and kidney etc.) and metabolically active bacterial communities of GM.
Collapse
Affiliation(s)
- Khursheed Ali
- Medical Microbiology and Molecular Biology, Laboratory Interdisciplinary, Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Sahar Zaidi
- Medical Microbiology and Molecular Biology, Laboratory Interdisciplinary, Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Aijaz A Khan
- Department of Anatomy, Jawaharlal Nehru Medical College & Hospital, Aligarh Muslim University, Aligarh 202002, UP, India
| | - Asad U Khan
- Medical Microbiology and Molecular Biology, Laboratory Interdisciplinary, Biotechnology Unit, Aligarh Muslim University, Aligarh 202002, UP, India.
| |
Collapse
|
12
|
Leitner E, Bozic M, Kienesberger S, Cosic A, Landt O, Högenauer C, Kessler HH. Improved diagnosis of antibiotic-associated haemorrhagic colitis (AAHC) in faecal specimens by a new qualitative real-time PCR assay detecting relevant toxin genes of Klebsiella oxytoca sensu lato. Clin Microbiol Infect 2022; 28:690-694. [PMID: 34582979 DOI: 10.1016/j.cmi.2021.09.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Revised: 08/27/2021] [Accepted: 09/15/2021] [Indexed: 02/04/2023]
Abstract
OBJECTIVE Toxin-producing Klebsiella oxytoca causes antibiotic-associated haemorrhagic colitis (AAHC). The disease-relevant cytotoxins tilivalline and tilimycine produced by certain K. oxytoca isolates are encoded by the non-ribosomal peptide synthetase genes A (npsA) and B (npsB). In this study, the new LightMix® Modular kit for the detection of relevant K. oxytoca sensu lato (s.l.) toxin genes was evaluated. METHODS DNA was extracted on the automated EMAG® platform. Amplification was done on the Light Cycler® 480 II instrument. In total, 130 residual faecal specimens collected from patients with antibiotic-associated diarrhoea were studied to determine the clinical sensitivity and specificity. Toxigenic culture served as reference method. RESULTS With the new kit, the limit of detection was 15 CFU/mL for all targets. For the pehX target specific to K. oxytoca s.l., 65 of 130 clinical specimens were positive, while toxin-specific targets (npsA/npsB) were positive in 47 of 130. The npsA/npsB PCR targets showed a clinical sensitivity of 100% (95%CI 80.5-100%) and a specificity of 73.5% (95%CI 64.3-81.3%) with a positive predictive value of 16.5% (95%CI 12.7-21.2%) and a negative predictive value of 100%. CONCLUSION Compared with culture, additional clinical specimens positive for K. oxytoca s.l. were detected with real-time PCR. The specificity of the toxin targets appears moderate due to the inferior sensitivity of the culture-based reference method. Since the developed assay is highly sensitive, it may be used as first-line method to improve the diagnosis of AAHC.
Collapse
Affiliation(s)
- Eva Leitner
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Austria.
| | - Michael Bozic
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Austria
| | - Sabine Kienesberger
- Institute of Molecular Biosciences, University of Graz, Austria; BioTechMed-Graz, Austria; Field of Excellence BioHealth, University of Graz, Austria
| | - Amar Cosic
- Institute of Molecular Biosciences, University of Graz, Austria
| | | | - Christoph Högenauer
- BioTechMed-Graz, Austria; Division of Gastroenterology and Hepatology, Department of Internal Medicine, Medical University of Graz, Austria
| | - Harald H Kessler
- Diagnostic and Research Institute of Hygiene, Microbiology and Environmental Medicine, Medical University of Graz, Austria
| |
Collapse
|
13
|
Tallei TE, Fatimawali, Yelnetty A, Kusumawaty D, Effendi Y, Park MN, Alhumaydhi FA, Emran TB, Kim B. Predictive Microbial Community and Functional Gene Expression Profiles in Pineapple Peel Fermentation Using 16S rRNA Gene Sequences. FERMENTATION-BASEL 2022; 8:194. [DOI: 10.3390/fermentation8050194] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
Abstract
Pineapple peel (PP) is a by-product with the potential to be used as a raw material for functional beverages. Traditional PP fermentation has so far paid little attention to the microbial community and its role in the fermentation process. As a result, the current research looked into the microbial communities and their roles during PP fermentation. A metagenomic approach based on the 16S rRNA sequencing data was used to assess the microbial communities. Subsequent analysis was performed using PICRUSt (phylogenetic investigation of communities by reconstruction of unobserved states) to analyze the microbial functions in the fermentation system. The microecology of the fermentation process in three samples was predominated by Firmicutes. Furthermore, the well-known probiotic genera Weissella, Lactobacillus, and Lactococcus were found to be predominating in the gumer, promic, and control samples, respectively. It was obvious that microenvironmental differences have an effect on the microbial composition of PP fermentation. Moreover, functional prediction revealed that carbohydrate metabolism was the most prevalent metabolic pathway during the fermentation process. Additionally, it was discovered that all of the bacteria found in the samples played significant roles in carbohydrate, amino acid, vitamin, and co-factor metabolism, which can be inferred to result in the production of beneficial metabolites.
Collapse
Affiliation(s)
- Trina Ekawati Tallei
- Department of Biology, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado 95115, North Sulawesi, Indonesia
| | - Fatimawali
- Pharmacy Study Program, Faculty of Mathematics and Natural Sciences, Sam Ratulangi University, Manado 95115, North Sulawesi, Indonesia
| | - Afriza Yelnetty
- Department of Animal Production, Faculty of Animal Husbandry, Sam Ratulangi University, Manado 95115, North Sulawesi, Indonesia
| | - Diah Kusumawaty
- Department of Biology, Faculty of Mathematics and Natural Sciences Education, Universitas Pendidikan Indonesia, Bandung 40154, Indonesia
| | - Yunus Effendi
- Department of Biology, Faculty of Science and Technology, Al-Azhar Indonesia University, Jakarta 12110, Indonesia
| | - Moon Nyeo Park
- Department of Pathology College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05254, Korea
| | - Fahad A. Alhumaydhi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah 52571, Saudi Arabia
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| | - Bonglee Kim
- Department of Pathology College of Korean Medicine, Kyung Hee University, Hoegidong Dongdaemungu, Seoul 05254, Korea
| |
Collapse
|
14
|
Gu X, Sim JX, Lee WL, Cui L, Chan YF, Chang ED, Teh YE, Zhang AN, Armas F, Chandra F, Chen H, Zhao S, Lee Z, Thompson JR, Ooi EE, Low JG, Alm EJ, Kalimuddin S. Gut Ruminococcaceae levels at baseline correlate with risk of antibiotic-associated diarrhea. iScience 2022; 25:103644. [PMID: 35005566 PMCID: PMC8718891 DOI: 10.1016/j.isci.2021.103644] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/19/2021] [Accepted: 12/13/2021] [Indexed: 12/11/2022] Open
Abstract
Antibiotic-associated diarrhea (AAD) affects a significant proportion of patients receiving antibiotics. We sought to understand if differences in the gut microbiome would influence the development of AAD. We administered a 3-day course of amoxicillin-clavulanate to 30 healthy adult volunteers, and analyzed their stool microbiome, using 16S rRNA gene sequencing, at baseline and up to 4 weeks post antibiotic administration. Lower levels of gut Ruminococcaceae were significantly and consistently observed from baseline until day 7 in participants who developed AAD. Overall, participants who developed AAD experienced a greater decrease in microbial diversity. The probability of AAD could be predicted based on qPCR-derived levels of Faecalibacterium prausnitzii at baseline. Our findings suggest that a lack of gut Ruminococcaceae influences development of AAD. Quantification of F. prausnitzii in stool prior to antibiotic administration may help identify patients at risk of AAD, and aid clinicians in devising individualized treatment regimens to minimize such adverse effects.
Collapse
Affiliation(s)
- Xiaoqiong Gu
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Jean X.Y. Sim
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - Wei Lin Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Liang Cui
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Yvonne F.Z. Chan
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - Ega Danu Chang
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Yii Ean Teh
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
| | - An-Ni Zhang
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
| | - Federica Armas
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Franciscus Chandra
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Hongjie Chen
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Shijie Zhao
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
| | - Zhanyi Lee
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
| | - Janelle R. Thompson
- Singapore Centre for Environmental Life Sciences Engineering, Nanyang Technological University, 60 Nanyang Drive, Singapore 637551, Singapore
- Asian School of the Environment, Nanyang Technological University, 62 Nanyang Drive, Singapore 637459, Singapore
| | - Eng Eong Ooi
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Centre (ViREMiCS), 20 College Road, Singapore 169856, Singapore
- Saw Swee Hock School of Public Health, National University of Singapore, 12 Science Drive 2, Singapore 117549, Singapore
| | - Jenny G. Low
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
- Viral Research and Experimental Medicine Center, SingHealth Duke-NUS Academic Medical Centre (ViREMiCS), 20 College Road, Singapore 169856, Singapore
| | - Eric J. Alm
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, 1 Create Way, Singapore 138602, Singapore
- Campus for Research Excellence and Technological Enterprise (CREATE), Singapore 138602, Singapore
- Department of Biological Engineering, Massachusetts Institute of Technology, 21 Ames Street, Cambridge, MA 02142, USA
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Building E25-321, Cambridge, MA 02139, USA
- Broad Institute of MIT and Harvard, 415 Main Street, Cambridge, MA 02142, USA
| | - Shirin Kalimuddin
- Department of Infectious Diseases, Singapore General Hospital, Academia Level 3, 20 College Road, Singapore 169856, Singapore
- Program in Emerging Infectious Diseases, Duke-NUS Medical School, 8 College Road, Singapore 169857, Singapore
| |
Collapse
|
15
|
Gut microbiota in gastrointestinal diseases. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 191:141-151. [DOI: 10.1016/bs.pmbts.2022.06.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
16
|
Tomczyk-Warunek A, Blicharski T, Muszyński S, Tomaszewska E, Dobrowolski P, Blicharski R, Jarecki J, Arczewska-Włosek A, Świątkiewicz S, Józefiak D. Structural Changes in Trabecular Bone, Cortical Bone and Hyaline Cartilage as Well as Disturbances in Bone Metabolism and Mineralization in an Animal Model of Secondary Osteoporosis in Clostridium perfringens Infection. J Clin Med 2021; 11:205. [PMID: 35011946 PMCID: PMC8746067 DOI: 10.3390/jcm11010205] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Revised: 12/15/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
There is no information regarding whether changes in the microbiological balance of the gastrointestinal tract as a result of an infection with Clostridium perfringens influence the development of metabolic bone disorders. The experiment was carried out on male broiler chickens divided into two groups: control (n = 10) and experimental (n = 10). The experimental animals were infected with Clostridium perfringens between 17 and 20 days of age. The animals were euthanized at 42 days of age. The structural parameters of the trabecular bone, cortical bone, and hyaline cartilage as well as the mineralization of the bone were determined. The metabolism of the skeletal system was assessed by determining the levels of bone turnover markers, hormones, and minerals in the blood serum. The results confirm that the disturbed composition of the gastrointestinal microflora has an impact on the mineralization and metabolism of bone tissue, leading to the structural changes in cortical bone, trabecular bone, and hyaline cartilage. On the basis of the obtained results, it can be concluded that changes in the microenvironment of the gastrointestinal tract by infection with C. perfringens may have an impact on the earlier development of osteoporosis.
Collapse
Affiliation(s)
- Agnieszka Tomczyk-Warunek
- Chair and Department of Rehabilitation and Orthopaedics, Medical University in Lublin, 20-090 Lublin, Poland; (A.T.-W.); (R.B.); (J.J.)
| | - Tomasz Blicharski
- Chair and Department of Rehabilitation and Orthopaedics, Medical University in Lublin, 20-090 Lublin, Poland; (A.T.-W.); (R.B.); (J.J.)
| | - Siemowit Muszyński
- Department of Biophysics, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| | - Ewa Tomaszewska
- Department of Animal Physiology, University of Life Sciences in Lublin, 20-950 Lublin, Poland;
| | - Piotr Dobrowolski
- Department of Functional Anatomy and Cytobiology, Maria Curie-Skłodowska University, 20-033 Lublin, Poland;
| | - Rudolf Blicharski
- Chair and Department of Rehabilitation and Orthopaedics, Medical University in Lublin, 20-090 Lublin, Poland; (A.T.-W.); (R.B.); (J.J.)
| | - Jaromir Jarecki
- Chair and Department of Rehabilitation and Orthopaedics, Medical University in Lublin, 20-090 Lublin, Poland; (A.T.-W.); (R.B.); (J.J.)
| | - Anna Arczewska-Włosek
- Department of Animal Nutrition and Feed Science, National Research Institute of Animal Production, Krakowska St. 1, 32-083 Balice, Poland; (A.A.-W.); (S.Ś.)
| | - Sylwester Świątkiewicz
- Department of Animal Nutrition and Feed Science, National Research Institute of Animal Production, Krakowska St. 1, 32-083 Balice, Poland; (A.A.-W.); (S.Ś.)
| | - Damian Józefiak
- Department of Animal Nutrition, Faculty of Veterinary Medicine and Animal Science, Poznań University of Life Sciences, Wołyńska 33, 60-637 Poznań, Poland;
| |
Collapse
|
17
|
Pakhomovskaia NL, Tatyanina OF, Lazareva ТY. Approaches to prevention of antibiotic-associated diarrhea in children. MEDITSINSKIY SOVET = MEDICAL COUNCIL 2021:283-292. [DOI: 10.21518/2079-701x-2021-17-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The incidence of antibiotic-associated diarrhea, according to various authors, ranges from 5 to 39% and depends on the patient’s age and other contributing factors. Antibiotic-associated diarrhea can be caused by any antibiotic, regardless of dosage form or route of administration. In the pediatric population, the prevalence of antibiotic-associated diarrhea ranges from 6 to 70%. An urgent problem is the development of this disease against the background of a course of H. pylori eradication therapy, which significantly complicates tolerance and adherence to therapy. This article presents current data on the pathogenesis and risk factors of antibiotic-associated diarrhea in children. The clinical picture ranges from idiopathic enteritis to antibiotic-associated diarrhea caused by Cl. difficile - pseudomembranous colitis.The main principle of antibiotic-associated diarrhea treatment is cancellation of the antibacterial medicine that caused the diarrhea, or reducing its dose (if the course of the disease allows it). In complex treatment sorbents are used, correction of water-electrolyte balance is carried out. The use of probiotics seems quite logical for the treatment and prevention of antibiotic-associated diarrhea in terms of the pathogenesis of this condition. To correct dysbiosis, drugs are used to maintain and restore the quantitative and qualitative composition of the intestinal microbiota.Taking into account modern recommendations the main groups of drugs (probiotics, prebiotics, synbiotics) used for correction of intestinal microbiocenosis are presented. The mechanism of action of probiotics and mechanisms of their effect on intestinal microflora are considered. The basic requirements for bacterial strains that are part of the probiotic drugs are presented.The results of various randomized clinical trials and meta-analyses confirming the necessity of including probiotic complexes in antibiotic-associated diarrhea treatment regimens are presented from an evidence-based medicine perspective. The clinical effects of strains of Lactobacillusspp., Bifidobacterium spp.,Streptococcusspp. and Lactococcusspp. on the digestive tract microbiota are considered. The role of a synbiotic containing 9 probiotic strains of 4.5 * 109 CFU in one capsule and the prebiotic component fructooligosac-charides in the prevention of antibiotic-associated diarrhea in children is discussed separately. The results of microbiological studies confirmed the presence of microorganisms of genera Bifidobacterium, Lactobacillus, Streptococcus in the product, and the content of bacteria in one dose of the product was not less than 2 x 1010 CFU.
Collapse
|
18
|
Ianiro G, Giambò F, Cammarota G. Residual Gastrointestinal Symptoms after Fecal Microbiota Transplantation for Clostridioides difficile Infection: A Matter of Efficacy Rather Than Safety? Gastroenterology 2021; 161:1344. [PMID: 34090885 DOI: 10.1053/j.gastro.2021.06.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 06/01/2021] [Indexed: 12/02/2022]
Affiliation(s)
- Gianluca Ianiro
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS and, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Federica Giambò
- Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Giovanni Cammarota
- Digestive Disease Center, Fondazione Policlinico Universitario Agostino Gemelli IRCCS and, Dipartimento Universitario di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| |
Collapse
|
19
|
di Michele F. Why use nutraceutical strategies for the Irritable Bowel Syndrome. Curr Med Chem 2021; 29:2075-2092. [PMID: 34533437 DOI: 10.2174/0929867328666210917115255] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 07/30/2021] [Accepted: 08/11/2021] [Indexed: 11/22/2022]
Abstract
Irritable bowel syndrome (IBS) is a chronic functional bowel disorder, often stress-related, identified by many abdominal symptoms, the most important of which is the chronic visceral abdominal pain. Therefore, IBS commonly impairs the quality of life of patients, moreover it is frequently linked to depressive and anxiety symptoms. The treatment of IBS primarily focuses on symptoms relief. Unfortunately, up to now, no efficacious therapies have been found. Therefore, it would be important to develop new anti-IBS interventions. The aim of this brief review is to summarize the current evidence of nutraceutical supplementation in IBS treatment, with probiotics, prebiotics, synbiotics, butyrate, palmitoylethanolamide and colostrum. Since nutraceutics are over the counter products, the review has the purpose to better inform the medicinal chemist and the practitioner about the possible benefit mechanisms and the many advantages that these therapies offer. All of these compounds present multiple mechanisms of action, such as restoring the physiological microbiota, potentiating gastrointestinal barrier's function, immunomodulatory, anti-inflammatory and antinociceptive activities. From the literature data it results that these compounds are not only capable to improve IBS symptomatology, but mainly display an optimal safety and tolerability profile. Although extensive studies must be carried out to reinforce the evidences from the so far limited clinical trials, the supplementation with these compounds may be useful considering the warnings of prescription medicines for special populations of patients, such as elders, youngsters, or patients who need a combination therapy. Finally, the nutraceutical approach may improve adherence to treatment, given its better acceptance by the patients compared to pharmacological therapy.
Collapse
Affiliation(s)
- Flavia di Michele
- Dept Systems Medicine, Policlinico Tor Vergata (PTV) Foundation, Rome, Italy
| |
Collapse
|
20
|
Mateen BA, Samanta S, Tullie S, O'Neill S, Cargill Z, Kelly G, Brennan E, Patel M, Al-Agil M, Galloway J, Teo J, Shawcross DL, Kent AJ, Hayee B. Diarrhoea and preadmission antibiotic exposure in COVID-19: a retrospective cohort study of 1153 hospitalised patients. BMJ Open Gastroenterol 2021; 8:e000593. [PMID: 34489301 PMCID: PMC8423520 DOI: 10.1136/bmjgast-2020-000593] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Accepted: 03/14/2021] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVE The aims of this study were to describe community antibiotic prescribing patterns in individuals hospitalised with COVID-19, and to determine the association between experiencing diarrhoea, stratified by preadmission exposure to antibiotics, and mortality risk in this cohort. DESIGN/METHODS Retrospective study of the index presentations of 1153 adult patients with COVID-19, admitted between 1 March 2020 and 29 June 2020 in a South London NHS Trust. Data on patients' medical history (presence of diarrhoea, antibiotic use in the previous 14 days, comorbidities); demographics (age, ethnicity, and body mass index); and blood test results were extracted. Time to event modelling was used to determine the risk of mortality for patients with diarrhoea and/or exposure to antibiotics. RESULTS 19.2% of the cohort reported diarrhoea on presentation; these patients tended to be younger, and were less likely to have recent exposure to antibiotics (unadjusted OR 0.64, 95% CI 0.42 to 0.97). 19.1% of the cohort had a course of antibiotics in the 2 weeks preceding admission; this was associated with dementia (unadjusted OR 2.92, 95% CI 1.14 to 7.49). After adjusting for confounders, neither diarrhoea nor recent antibiotic exposure was associated with increased mortality risk. However, the absence of diarrhoea in the presence of recent antibiotic exposure was associated with a 30% increased risk of mortality. CONCLUSION Community antibiotic use in patients with COVID-19, prior to hospitalisation, is relatively common, and absence of diarrhoea in antibiotic-exposed patients may be associated with increased risk of mortality. However, it is unclear whether this represents a causal physiological relationship or residual confounding.
Collapse
Affiliation(s)
- Bilal Akhter Mateen
- Institute of Health Informatics, University College London, London, UK
- King's College Hospital NHS Foundation Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Sandip Samanta
- King's College Hospital NHS Foundation Trust, London, UK
| | | | - Sarah O'Neill
- King's College Hospital NHS Foundation Trust, London, UK
| | - Zillah Cargill
- King's College Hospital NHS Foundation Trust, London, UK
| | - Gillian Kelly
- King's College Hospital NHS Foundation Trust, London, UK
| | - Ewen Brennan
- King's College Hospital NHS Foundation Trust, London, UK
| | - Mehul Patel
- King's College Hospital NHS Foundation Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| | | | - James Galloway
- King's College Hospital NHS Foundation Trust, London, UK
| | - James Teo
- King's College Hospital NHS Foundation Trust, London, UK
| | - Debbie L Shawcross
- King's College Hospital NHS Foundation Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Alexandra J Kent
- King's College Hospital NHS Foundation Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| | - Bu'Hussain Hayee
- King's College Hospital NHS Foundation Trust, London, UK
- School of Immunology & Microbial Sciences, King's College London, London, UK
| |
Collapse
|
21
|
Liao W, Chen C, Wen T, Zhao Q. Probiotics for the Prevention of Antibiotic-associated Diarrhea in Adults: A Meta-Analysis of Randomized Placebo-Controlled Trials. J Clin Gastroenterol 2021; 55:469-480. [PMID: 33234881 PMCID: PMC8183490 DOI: 10.1097/mcg.0000000000001464] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 10/06/2020] [Indexed: 12/12/2022]
Abstract
OBJECTIVE This meta-analysis aims to combine the latest research evidence to assess the effect of probiotics on preventing antibiotic-associated diarrhea (AAD) in adults. METHODS PubMed, Cochrane Library, EMBASE, and Web of Science were searched for randomized placebo-controlled trials on probiotics preventing AAD. A random or fixed effect model was used to combine the incidence of AAD (primary outcome) and the adverse event rates. The authors performed subgroup analyses to explore the effects of different participants population, probiotics species, and dosage. RESULTS Thirty-six studies were included with 9312 participants. Probiotics reduced the incidence of AAD by 38% (pooled relative risk, 0.62; 95% confidence interval, 0.51-0.74). The protective effect of probiotics was still significant when grouped by reasons for antibiotics treatment, probiotic duration, probiotic dosage, and time from antibiotic to probiotic. However, there were no statistically significant increased adverse events in the probiotics group (relative risk, 1.00; 95% confidence interval, 0.87-1.14). CONCLUSIONS This updated meta-analysis suggested that using probiotics as early as possible during antibiotic therapy has a positive and safe effect on preventing AAD in adults. Further studies should focus on the optimal dosage and duration of probiotics to develop a specific recommendation.
Collapse
Affiliation(s)
- Wanqian Liao
- Department of Intensive Care Unit
- Infection Control Department, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Chongxiang Chen
- Department of Intensive Care Unit
- Infection Control Department, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou
| | - Tianmeng Wen
- School of Public Health, Sun Yat-Sen University, Guangzhou, Guangdong Province, People’s Republic of China
| | - Qingyu Zhao
- Department of Intensive Care Unit
- Infection Control Department, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou
| |
Collapse
|
22
|
Dingeo G, Brito A, Samouda H, Iddir M, La Frano MR, Bohn T. Phytochemicals as modifiers of gut microbial communities. Food Funct 2021; 11:8444-8471. [PMID: 32996966 DOI: 10.1039/d0fo01483d] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A healthy gut microbiota (GM) is paramount for a healthy lifestyle. Alterations of the GM have been involved in the aetiology of several chronic diseases, including obesity and type 2 diabetes, as well as cardiovascular and neurodegenerative diseases. In pathological conditions, the diversity of the GM is commonly reduced or altered, often toward an increased Firmicutes/Bacteroidetes ratio. The colonic fermentation of dietary fiber has shown to stimulate the fraction of bacteria purported to have beneficial health effects, acting as prebiotics, and to increase the production of short chain fatty acids, e.g. propionate and butyrate, while also improving gut epithelium integrity such as tight junction functionality. However, a variety of phytochemicals, often associated with dietary fiber, have also been proposed to modulate the GM. Many phytochemicals possess antioxidant and anti-inflammatory properties that may positively affect the GM, including polyphenols, carotenoids, phytosterols/phytostanols, lignans, alkaloids, glucosinolates and terpenes. Some polyphenols may act as prebiotics, while carotenoids have been shown to alter immunoglobulin A expression, an important factor for bacteria colonization. Other phytochemicals may interact with the mucosa, another important factor for colonization, and prevent its degradation. Certain polyphenols have shown to influence bacterial communication, interacting with quorum sensing. Finally, phytochemicals can be metabolized in the gut into bioactive constituents, e.g. equol from daidzein and enterolactone from secoisolariciresinol, while bacteria can use glycosides for energy. In this review, we strive to highlight the potential interactions between prominent phytochemicals and health benefits related to the GM, emphasizing their potential as adjuvant strategies for GM-related diseases.
Collapse
Affiliation(s)
| | - Alex Brito
- Luxembourg Institute of Health, Population Health Department, Nutrition and Health Research Group, 1A-B, rue Thomas Edison, Strassen L-1445, Luxembourg. and Laboratory of Pharmacokinetics and Metabolomic Analysis, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow Medical University, Moscow, Russia.
| | - Hanen Samouda
- Luxembourg Institute of Health, Population Health Department, Nutrition and Health Research Group, 1A-B, rue Thomas Edison, Strassen L-1445, Luxembourg.
| | - Mohammed Iddir
- Luxembourg Institute of Health, Population Health Department, Nutrition and Health Research Group, 1A-B, rue Thomas Edison, Strassen L-1445, Luxembourg.
| | - Michael R La Frano
- Department of Food Science and Nutrition, California Polytechnic State University, San Luis Obispo, CA, USA. and Center for Health Research, California Polytechnic State University, San Luis Obispo, CA, USA.
| | - Torsten Bohn
- Luxembourg Institute of Health, Population Health Department, Nutrition and Health Research Group, 1A-B, rue Thomas Edison, Strassen L-1445, Luxembourg.
| |
Collapse
|
23
|
Wu ZC, Wu L, Zhang M, Zhou W. Genome sequence and annotation of Bacteroides sp aff. Thetaiotaomicron strain isolated from blood. INFECTION GENETICS AND EVOLUTION 2021; 91:104816. [PMID: 33771725 DOI: 10.1016/j.meegid.2021.104816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 01/13/2021] [Accepted: 03/21/2021] [Indexed: 11/27/2022]
Abstract
This study is focused on genome sequence and annotation of the Bacteroides strain isolated from the blood of a patient with descending colon cancer. According to a comparison of the 16S ribosomal RNA sequence with the National Center for Biotechnology Information database, this strain was identified as Bacteroides sp. aff. Thetaiotaomicron. The next-generation sequencing of the strain was performed in a GENEWIZ laboratory (Jiangsu, China) on Illumina HiSeq device. According to CAZy classification, metabolic pathways related to carbohydrate metabolism of this strain engage the following enzymes: 427 glycosylhydrolases, 277 glycosyltransferases, 137 carbohydrate-binding modules, 48 carbohydrate esterases, and 24 polysaccharide lyases. According to the KEGG pathway database, Bacteroides sp. aff thetaiotaomicron strain is reported to incorporate 199 pathway associated genes. Bacteroides sp. aff. Thetaiotaomicron exposes the capacity of metabolizing a variety of polysaccharides. Its genome is enriched with an expanded repertoire of enzymes for the hydrolysis of glycosidic bonds and, thus, likely to hydrolyze most of glycosidic bonds in biological polysaccharides. The advanced capabilities of the studied strain to recognize and respond to environmental signals are expressed in the rich representation of one- and two-component signal transduction systems.
Collapse
Affiliation(s)
- Zhi Cheng Wu
- Clinical Laboratory, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Haikou, Hainan Province 570102, China.
| | - Lin Wu
- School of Tropical and Laboratory Medicine, Hainan Medical University, 31 Longhua Road, Haikou City, Hainan Province 571199, China; Faculty of Biotechnology and Biotechnics, National Technical University of Ukraine, Kyiv, Ukraine; Key Laboratory of Tropical Translational Medicine, Ministry of Education, Hainan Medical University, Haikou City, Hainan Province, China.
| | - Meng Zhang
- Sanya People's Hospital, Jiefang Third Road, 558, Sanya City, Hainan Province 572000, China
| | - WeiLan Zhou
- Clinical Laboratory, The First Affiliated Hospital of Hainan Medical University, 31 Longhua Road, Haikou, Hainan Province 570102, China
| |
Collapse
|
24
|
Yu H, Fang C, Li P, Wu M, Shen S. The relevance of DHA with modulating of host-gut microbiome signatures alterations and repairing of lipids metabolism shifts. Eur J Pharmacol 2021; 895:173885. [PMID: 33482183 DOI: 10.1016/j.ejphar.2021.173885] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 01/12/2021] [Accepted: 01/14/2021] [Indexed: 12/12/2022]
Abstract
Huge of previous reports recommended that gut microbiome have a crucial role in the human health and its change was profound impact for the metabolic improvements associated with lipids metabolism. In order to explore the relevance of a direct dysbiosis effect of gut microbiome on lipids metabolism shifts and repaired position of DHA, we built the animal model for the study with gut microbiome dysbiosis administrated by i.g. with CRO and intervened by DHA in the present work. Gut microbiome was analyzed by high throughput sequencing and bioinformatics analyses of bacteria. The composition of fatty acids and short chain fatty acids (SCFAs) were determined by gas chromatography. Blood lipids and bile acids were assayed by kit and UPLC-MS/MS, respectively. The expressions of enzymes of long chain fatty acid metabolism were analyzed by qRT-PCR. The results showed that gut microbiome dysbiosis caused lipid metabolism abnormal, and DHA was able to repair the lipids metabolism shifts resulted from gut microbiome dysbiosis. DHA could modulate host-gut microbiome signatures, improve concentrations of SCFAs, regulate fatty acids metabolism but modify bile acid profiles. In conclusion, we considered that DHA repaired lipid metabolism by modulating gut microbiome and regulating fatty acids metabolism pathway.
Collapse
Affiliation(s)
- Haining Yu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China.
| | - Chengjie Fang
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | - Peng Li
- Department of Geratoloy, The Third People's Hospital of Hangzhou, Hangzhou, China
| | - Manman Wu
- College of Pharmaceutical Science, Zhejiang University of Technology, Hangzhou, China
| | | |
Collapse
|
25
|
Levast B, Benech N, Gasc C, Batailler C, Senneville E, Lustig S, Pouderoux C, Boutoille D, Boucinha L, Dauchy FA, Zeller V, Maynard M, Cazanave C, Le Thi TT, Josse J, Doré J, Laurent F, Ferry T. Impact on the Gut Microbiota of Intensive and Prolonged Antimicrobial Therapy in Patients With Bone and Joint Infection. Front Med (Lausanne) 2021; 8:586875. [PMID: 33748154 PMCID: PMC7977441 DOI: 10.3389/fmed.2021.586875] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 01/25/2021] [Indexed: 01/02/2023] Open
Abstract
There is a growing interest in the potentially deleterious impact of antibiotics on gut microbiota. Patients with bone and joint infection (BJI) require prolonged treatment that may impact significantly the gut microbiota. We collected samples from patients with BJI at baseline, end of antibiotics (EOT), and 2 weeks after antibiotic withdrawal (follow-up, FU) in a multicenter prospective cohort in France. Microbiota composition was determined by shotgun metagenomic sequencing. Fecal markers of gut permeability and inflammation as well as multi-drug-resistant bacteria (MDRB) and Clostridioides difficile carriage were assessed at each time point. Sixty-two patients were enrolled: 27 native BJI, 14 osteosynthesis-related BJI, and 21 prosthetic joint infections (PJI). At EOT, there was a significant loss of alpha-diversity that recovered at FU in patients with native BJI and PJI, but not in patients with osteosynthesis-related BJI. At EOT, we observed an increase of Proteobacteria and Bacteroidetes that partially recovered at FU. The principal component analysis (PCoA) of the Bray–Curtis distance showed a significant change of the gut microbiota at the end of treatment compared to baseline that only partially recover at FU. Microbiota composition at FU does not differ significantly at the genus level when comparing patients treated for 6 weeks vs. those treated for 12 weeks. The use of fluoroquinolones was not associated with a lower Shannon index at the end of treatment; however, the PCoA of the Bray–Curtis distance showed a significant change at EOT, compared to baseline, that fully recovered at FU. Levels of fecal neopterin were negatively correlated with the Shannon index along with the follow-up (r2 = 0.17; p < 0.0001). The PCoA analysis of the Bray–Curtis distance shows that patients with an elevated plasma level of C-reactive protein (≥5 mg/L) at EOT had a distinct gut microbial composition compared to others. MDRB and C. difficile acquisition at EOT and FU represented 20% (7/35) and 37.1% (13/35) of all MDRB/C. difficile-free patients at the beginning of the study, respectively. In patients with BJI, antibiotics altered the gut microbiota diversity and composition with only partial recovery, mucosal inflammation, and permeability and acquisition of MDRB carriage. Microbiome interventions should be explored in patients with BJI to address these issues.
Collapse
Affiliation(s)
| | - Nicolas Benech
- Service des Maladies Infectieuses et Tropicales, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence Pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,TGF-ß Immune Evasion, Tumor Escape Resistance Immunity Department, Cancer Research Center of Lyon, Inserm 1052, CNRS 5286, Lyon, France
| | | | - Cécile Batailler
- Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence Pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Eric Senneville
- Service de Maladies Infectieuses et du Voyageur, Centre Hospitaliser Gustave Dron, Tourcoing, France.,Université de Lille, Lille, France.,Centre de Référence Pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lille-tourcoing), Tourcoing, France
| | - Sébastien Lustig
- Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence Pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Service de Chirurgie Orthopédique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Cécile Pouderoux
- Service des Maladies Infectieuses et Tropicales, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,Centre de Référence Pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France
| | - David Boutoille
- Service de Maladies Infectieuses et Tropicales, Hôpital de l'Hôtel-Dieu, CHU de Nantes, Nantes, France.,Université de Nantes, Nantes, France.,Centre de Référence des Infections Ostéo-Articulaires Grand-Ouest, Nantes, France
| | | | - Frederic-Antoine Dauchy
- Centre Hospitalier Universitaire de Bordeaux, Service des Maladies Infectieuses et Tropicales, Hôpital Pellegrin, CHU de Bordeaux, Centre de référence des Infections Ostéoarticulaires Complexes du Grand Sud-Ouest (CRIOAc GSO), Bordeaux, France
| | - Valérie Zeller
- Service de Médecine Interne et Rhumatologie, GH Diaconesses-Croix Saint-Simon, Paris, France.,Centre de Référence Infections Ostéoarticulaires Complexes de Paris (CRIOAc Paris), Paris, France
| | - Marianne Maynard
- Centre de Recherche Clinique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Charles Cazanave
- Centre Hospitalier Universitaire de Bordeaux, Service des Maladies Infectieuses et Tropicales, Hôpital Pellegrin, CHU de Bordeaux, Centre de référence des Infections Ostéoarticulaires Complexes du Grand Sud-Ouest (CRIOAc GSO), Bordeaux, France.,Univ. Bordeaux, USC EA 3671, Infections Humaines à Mycoplasmes et à Chlamydiae, Bordeaux, France
| | - Thanh-Thuy Le Thi
- Centre de Ressource Biologique, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Jérôme Josse
- Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence Pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Institut des Agents Infectieux, Laboratoire de Bactériologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, Lyon, France
| | - Joël Doré
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS, Jouy-en-Josas, France
| | - Frederic Laurent
- Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence Pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,Institut des Agents Infectieux, Laboratoire de Bactériologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, Lyon, France
| | - Tristan Ferry
- Service des Maladies Infectieuses et Tropicales, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France.,Université Claude Bernard Lyon 1, Lyon, France.,Centre de Référence Pour la Prise en Charge des Infections Ostéo-Articulaires Complexes (CRIOAc Lyon), Hospices Civils de Lyon, Lyon, France.,CIRI - Centre International de Recherche en Infectiologie, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, Ecole Normale Supérieure de Lyon, Univ Lyon, Lyon, France
| |
Collapse
|
26
|
Tyagi N, Kumar A. Evaluation of recreational risks due to exposure of antibiotic-resistance bacteria from environmental water: A proposed framework. JOURNAL OF ENVIRONMENTAL MANAGEMENT 2021; 279:111626. [PMID: 33243622 DOI: 10.1016/j.jenvman.2020.111626] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 06/11/2023]
Abstract
This research provides a framework for the human health risk assessment due to exposure of AR (antibiotic resistance) E. coli from recreational water (swimming activity). Literature-based epidemiological studies were used for f-value formulation (i.e., AR E. coli/total number of E. coli isolates) and the theoretical calculation of AR and non-AR E. coli concentrations. Risk was estimated using calculated values by considering four different dose-response (D-R) scenarios with known characteristics due to current lack of availability of D-R for AR bacteria. f-values ranged between 0.14 and 0.59 and the order of calculated theoretical values of maximum AR E. coli are as follows: ampicillin or amoxicillin (38 CFU/dip) > co-trimoxazole (19 CFU/dip) ~ tetracycline (18 CFU/dip) > ceftriaxone or cefotaxime or ceftazidime (10 CFU/dip) ~ ciprofloxacin or ofloxacin (9 CFU/dip). The risk of infection was considerably high for theoretical calculated concentration values regardless of the chosen D-R model (annual risk of infection (95th percentile) = 1, Spearman's rank correlation coefficient = -0.06 to 0.94), under the conditions studied. Further, AR levels of human gastrointestinal-tract were determined using literature-reported data in stool samples and indicated that the resistance level was very high in healthy human (range: 3.7 × 107-8.4 × 107 CFU/g of wet lumen content). The maximum allowable concentration values for AR E. coli and non-ARB (0.0075 CFU/dip and 2.56 CFU/dip) were found to be smaller than the USEPA recreational water quality guidelines (≤126 CFU/100 mL), which can help the USEPA and other regulatory bodies in revisiting the current guidelines. So based on the noted results, we can conclude that the maintenance of inventory of actual measured concentration of ARB in the recreational water sites is needed to prevent unwanted complication related to the treatment of infectious sustained by resistant microbes.
Collapse
Affiliation(s)
- Neha Tyagi
- Department of Civil Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India.
| | - Arun Kumar
- Department of Civil Engineering, Indian Institute of Technology Delhi, New Delhi, 110016, India.
| |
Collapse
|
27
|
Angurana S, Mehta A. Probiotics in critically ill children: An updated review. JOURNAL OF PEDIATRIC CRITICAL CARE 2021. [DOI: 10.4103/jpcc.jpcc_73_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
|
28
|
Javed I, Cui X, Wang X, Mortimer M, Andrikopoulos N, Li Y, Davis TP, Zhao Y, Ke PC, Chen C. Implications of the Human Gut-Brain and Gut-Cancer Axes for Future Nanomedicine. ACS NANO 2020; 14:14391-14416. [PMID: 33138351 DOI: 10.1021/acsnano.0c07258] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
Recent clinical and pathological evidence have implicated the gut microbiota as a nexus for modulating the homeostasis of the human body, impacting conditions from cancer and dementia to obesity and social behavior. The connections between microbiota and human diseases offer numerous opportunities in medicine, most of which have limited or no therapeutic solutions available. In light of this paradigm-setting trend in science, this review aims to provide a comprehensive and timely summary of the mechanistic pathways governing the gut microbiota and their implications for nanomedicines targeting cancer and neurodegenerative diseases. Specifically, we discuss in parallel the beneficial and pathogenic relationship of the gut microbiota along the gut-brain and gut-cancer axes, elaborate on the impact of dysbiosis and the gastrointestinal corona on the efficacy of nanomedicines, and highlight a molecular mimicry that manipulates the universal cross-β backbone of bacterial amyloid to accelerate neurological disorders. This review further offers a forward-looking section on the rational design of cancer and dementia nanomedicines exploiting the gut-brain and gut-cancer axes.
Collapse
Affiliation(s)
- Ibrahim Javed
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
| | - Xuejing Cui
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Xiaoyu Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Monika Mortimer
- Institute of Environmental and Health Sciences, College of Quality and Safety Engineering, China Jiliang University, Hangzhou, Zhejiang 310018, China
| | - Nikolaos Andrikopoulos
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
| | - Yuhuan Li
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
- Zhongshan Hospital, Fudan University, 111 Yixueyuan Rd, Xuhui District, Shanghai 200032, China
| | - Thomas P Davis
- Australian Institute for Bioengineering and Nanotechnology, University of Queensland, Brisbane, Queensland 4072, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
| | - Yuliang Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Pu Chun Ke
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash Institute of Pharmaceutical Sciences, Monash University, Melbourne, Victoria 3052, Australia
- Zhongshan Hospital, Fudan University, 111 Yixueyuan Rd, Xuhui District, Shanghai 200032, China
| | - Chunying Chen
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety and CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- GBA Research Innovation Institute for Nanotechnology, Guangdong 510700, China
| |
Collapse
|
29
|
Antibiotics Disturb Dentin Formation and Differentiation of Dental Pulp Stem Cells: The Role of Microbiota in Cellular Turnover of Mouse Incisor. Stem Cells Int 2020; 2020:5154707. [PMID: 33014068 PMCID: PMC7519450 DOI: 10.1155/2020/5154707] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Accepted: 03/30/2020] [Indexed: 01/09/2023] Open
Abstract
Dentin formation was dependent on osteo-/odontogenic differentiation of dental pulp stem cells (DPSCs). It was observed in previous studies that antibiotic treatment in a clinical and animal model resulted in impaired mineralization of dental tissues. We previously reported that microbiota maintained the function of bone marrow mesenchymal stem cells, while whether microbiota dysbiosis caused by antibiotic treatment contributed to DPSCs dysfunction and impaired dentin formation is still not known. In this study, we aimed to clarify the role of microbiota or its metabolic products on dental mineralization and the function of DPSCs. Mice were treated with antibiotics to disrupt microbiota; then, the growth rate and histological characteristics of incisors as well as the biological characteristics of DPSCs in vitro were compared with specific pathogen-free (SPF) mice. In antibiotic-treated mice (AbT), we found a diminished quantity of microbiota and reduced growth rate of mechanical injured incisor, as well as decreased colony-forming rate and impaired ability of osteo-/odontogenic differentiation of DPSCs, in comparison to SPF mice. Colonization of AbT mice with SPF mice replanted the microbiota by cohousing (conventionalized (ConvD)) and normalized the growth rate of injured incisors and colony-forming and osteo-/odontogenic differentiation ability of DPSCs. Giving short-chain fatty acids (SCFAs) by oral gavage after antibiotic treatment also rescued the growth rate of incisors and the differentiation ability of DPSCs and enhanced proliferation ability of DPSCs. Collectively, gut microbiota could make contribution to maintain continuous growth of injured rodent incisor and differentiation capacity of DPSCs; SCFAs might play a crucial role in this process.
Collapse
|
30
|
Drago L, Meroni G, Chiaretti A, Laforgia N, Cucchiara S, Baldassarre ME. Effect of Limosilactobacillus reuteri LRE02- Lacticaseibacillus rhamnosus LR04 Combination on Antibiotic-Associated Diarrhea in a Pediatric Population: A National Survey. J Clin Med 2020; 9:jcm9103080. [PMID: 32987822 PMCID: PMC7650601 DOI: 10.3390/jcm9103080] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/16/2020] [Accepted: 09/22/2020] [Indexed: 11/16/2022] Open
Abstract
Probiotics are living microorganisms, which, upon oral ingestion, may prevent antibiotic-associated diarrhea (AAD) through the normalization of an unbalanced gastrointestinal flora. The objective of this study was to evaluate the benefits of a probiotic combination (Limosilactibacillus reuteri LRE02-DSM 23878 and Lacticaseibacillus rhamnosus LR04-DSM 16605) on the prevention of AAD in an outpatient pediatric setting. Questionnaires were delivered to pediatricians by each patient/parent during the visits after antibiotics and probiotics treatment to monitor physiological parameters. The primary outcome of both groups (probiotics and no probiotics treated) was the evaluation of the prevalence of AAD between the two groups. Evaluation of stool consistency using the Bristol Stool Scale (BSS) score was performed, as well as the evaluation of AAD duration, frequencies of daily evacuation, and the beginning of diarrhea and weight loss during AAD in both groups and related to antibiotic categories. Results indicated that probiotics, at the recommended dosage of 1.2 × 109 CFU (Colony Forming Unit) per day for 30 days, are associated with lower rates of AAD and a decreased number of days with diarrhea, independent of the type of antibiotic used. Moreover, the use of probiotics resulted in a normal stool consistency in a shorter time period, as evaluated by the BSS.
Collapse
Affiliation(s)
- Lorenzo Drago
- Laboratory of Clinical Microbiology and Microbial Metagenomic Unit, Department of Biomedical Sciences, University of Milan, 20133 Milan, Italy;
- Pediatric Research Center Romeo and Enrica Invernizzi, University of Milan, 20122 Milan, Italy
- Correspondence:
| | - Gabriele Meroni
- Laboratory of Clinical Microbiology and Microbial Metagenomic Unit, Department of Biomedical Sciences, University of Milan, 20133 Milan, Italy;
| | - Antonio Chiaretti
- Pediatric Emergency Department, Fondazione Policlinico Universitario A. Gemelli, IRCCS–Rome, 00168 Rome, Italy;
| | - Nicola Laforgia
- Department of Biomedical Science and Human Oncology-Section of Neonatology and NICU, University “Aldo Moro” of Bari, 70124 Bari, Italy; (N.L.); (M.E.B.)
| | - Salvatore Cucchiara
- Pediatric Gastroenterology and Liver Unit, Department of Women’s and Children’s Health, Sapienza University of Rome, 00161 Rome, Italy;
| | - Maria Elisabetta Baldassarre
- Department of Biomedical Science and Human Oncology-Section of Neonatology and NICU, University “Aldo Moro” of Bari, 70124 Bari, Italy; (N.L.); (M.E.B.)
| |
Collapse
|
31
|
Rajkumar C, Wilks M, Islam J, Ali K, Raftery J, Davies K, Timeyin J, Cheek E, Cohen J, Wright J, Natarajan U, Nicholl C, Dewhurst G, Fonseka M, Slovick D, Maskell P, Mukherjee S, Ali K, Nari R, Qureshi A, Gertner D, Khan Z, Shinh N, Bodmer C, Martin-Marero C, Poullis A, Pollok R, Ala A, Chauhan A, Patel M, Roberts H, Conroy S, McGowan D, Pathansali R, Yau C, Vasileiadis E, Guleri A, Orr D, Aldulami D. Do probiotics prevent antibiotic-associated diarrhoea? Results of a multicentre randomized placebo-controlled trial. J Hosp Infect 2020; 105:280-288. [DOI: 10.1016/j.jhin.2020.01.018] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Accepted: 01/27/2020] [Indexed: 02/07/2023]
|
32
|
Peng M, Tabashsum Z, Patel P, Bernhardt C, Biswas C, Meng J, Biswas D. Prevention of enteric bacterial infections and modulation of gut microbiota with conjugated linoleic acids producing Lactobacillus in mice. Gut Microbes 2020; 11:433-452. [PMID: 31411526 PMCID: PMC7524329 DOI: 10.1080/19490976.2019.1638724] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Probiotics are recognized for outcompeting pathogenic bacteria by competitive receptor-mediated colonization and secretion of functional metabolites which are antimicrobial against certain microbes as well as improving host's gut health and immunity. Recently, we have constructed a bioactive Lactobacillus casei (LC) strain, LC+mcra , by inserting mcra (myosin cross-reactive antigen) gene, which stimulates the conversion of conjugated linoleic acids. In this study, we evaluated the modulation of gut microbiome and protective roles of LC+mcra against pathogenic Salmonella enterica serovar Typhimurium (ST) and enterohemorrhagic E. coli (EHEC) infections in BALB/cJ mice. We observed that LC+mcra colonized efficiently in mice gut intestine and competitively reduced the infection with ST and EHEC in various locations of small and large intestine, specifically cecum, jejunum, and ileum (p < 0.05). Positive modulation of the cecal microbiota, for example, higher relative abundances of Firmicutes, lower relative abundances of Proteobacteria, and increased bacterial species diversity/richness, was detected in ST-challenged mice pretreated with LC+mcra based on 16S metagenomic sequencing. Cytokine gene expression analysis indicated that mice pretreated with LC+mcra associated with attenuated bacterial pathogen-induced gut inflammation. Furthermore, mice fed daily with LC+mcra for one week could protect themselves from the impairments caused by enteric infections with ST or EHEC. These impairments include weight loss, negative hematological changes, intestinal histological alterations, and potential death. This in vivo study suggests that daily consumption of novel conjugated linoleic acids over-producing probiotic effectively improves intestinal microbiota composition and prevents/combats foodborne enteric bacterial infections with pathogenic Salmonella and diarrheagenic E. coli.
Collapse
Affiliation(s)
- Mengfei Peng
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA,Biological Sciences Program, University of Maryland, College Park, MD, USA
| | - Zajeba Tabashsum
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| | - Puja Patel
- Biological Sciences Program, University of Maryland, College Park, MD, USA
| | - Cassandra Bernhardt
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| | - Chitrine Biswas
- Biological Sciences Program, University of Maryland, College Park, MD, USA
| | - Jianghong Meng
- Center for Food Safety and Security Systems, University of Maryland, College Park, MD, USA,Department of Nutrition and Food Science, University of Maryland, College Park, MD, USA
| | - Debabrata Biswas
- Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA,Biological Sciences Program, University of Maryland, College Park, MD, USA,Center for Food Safety and Security Systems, University of Maryland, College Park, MD, USA,CONTACT Debabrata Biswas Department of Animal and Avian Sciences, University of Maryland, College Park, MD, USA
| |
Collapse
|
33
|
Tabasi M, Ashrafian F, Khezerloo JK, Eshghjoo S, Behrouzi A, Javadinia SA, Poursadegh F, Eybpoosh S, Ahmadi S, Radmanesh A, Soroush A, Siadat SD. Changes in Gut Microbiota and Hormones After Bariatric Surgery: a Bench-to-Bedside Review. Obes Surg 2020; 29:1663-1674. [PMID: 30793228 DOI: 10.1007/s11695-019-03779-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Overweight and obesity are among the most prevalent non-communicable diseases which are generally treated successfully by bariatric or sleeve surgery. There are evidences affirming that sleeve surgery can manipulate the pH of the stomach and interact with the metabolism of fatty acids, carbohydrates, and bile acid transfer, leading to the overgrowth of gut microbiota. Therefore, this study aims to review the changes in gut microbiota and hormones after bariatric surgery.
Collapse
Affiliation(s)
- Mohsen Tabasi
- Department of Molecular Biology, Pasteur Institute of Iran, Pasteur Ave., Tehran, 13164, Iran
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Fatemeh Ashrafian
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Jamil Kheirvari Khezerloo
- Young Researchers and Elite Club, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Sahar Eshghjoo
- Microbial Pathogenesis and Immunology Department, Texas A&M University, Bryan, TX, USA
| | - Ava Behrouzi
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Seyed Alireza Javadinia
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran
| | - Farid Poursadegh
- Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran
| | - Sana Eybpoosh
- Department of Epidemiology and Biostatistics, Research Centre for Emerging and Reemerging Infectious Diseases, Pasteur Institute of Iran, Tehran, Iran
| | - Sara Ahmadi
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Amin Radmanesh
- Legal Medicine Research Center, Legal Medicine Organization, Tehran, Iran
| | - Ahmadreza Soroush
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Seyed Davar Siadat
- Department of Mycobacteriology and Pulmonary Research, Microbiology Research Center, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
34
|
Ma G, Chen Y. Polyphenol supplementation benefits human health via gut microbiota: A systematic review via meta-analysis. J Funct Foods 2020. [DOI: 10.1016/j.jff.2020.103829] [Citation(s) in RCA: 114] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
35
|
Kessler C. The Florajen Digestion Balance Patient Experience Study. Health (London) 2020. [DOI: 10.4236/health.2020.1211107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
36
|
Smith CJ, Perfetti TA. Exposure to chemicals formed from natural processes is ubiquitous. TOXICOLOGY RESEARCH AND APPLICATION 2020. [DOI: 10.1177/2397847320922940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Exposure to chemicals produced by natural processes is ubiquitous. First, in addition to the products of normal metabolism produced in humans of normal body weight, adipose tissue produces a large number of chemicals, including estrogen, testosterone from the produced estrogen, thyroid-stimulating hormone, leptin and approximately 500 other molecules termed adipokines, and a large number of inflammatory mediators. Second, the gut biome contains approximately the same number of bacteria as cells found in the entire body and produces a large number of small molecules. Third, the overwhelming majority (99.9%) of pesticide exposure occurs during ingestion of natural plant pesticides from eating vegetables. Fourth, consumption of cooked muscles meats leads to significant exposure to mutagenic and carcinogenic heterocyclic amines, polycyclic aromatic amines, and nitropyrenes. Fifth, many common beverages, for example, beer, coffee, and tea contain organic chemicals that display mutagenic activity. As compared with man-made production levels, from 1945 to 2015, an estimated 5000-fold more organic compounds were produced by a variety of natural processes, including common wood-degrading and forest litter-degrading fungi, microorganisms in temperate and boreal forest soils, bacteria in marine sponges, marine macro-algae, volcanoes, and forest fires. Exposure to these naturally produced organic compounds occurs via inhalation of ambient air, ingestion of food and water, and contact with soil, freshwater, and seawater. Contact with several thousand different endogenous or exogenous chemicals per day is unavoidable. This understanding might assist in better allocating resources toward controlling exposures to agents of highest concern as determined by current concepts of chronic disease causation.
Collapse
|
37
|
Effects of peanut meal extracts fermented by Bacillus natto on the growth performance, learning and memory skills and gut microbiota modulation in mice. Br J Nutr 2019; 123:383-393. [PMID: 31769373 DOI: 10.1017/s0007114519002988] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Recent studies have demonstrated that the nutritional properties of peanut meal (PM) can be improved after being fermented. The assessment of fermented PM has been reported to be limited to various physical and chemical evaluations in vitro. In the present study, PM was fermented by Bacillus natto to explore the effects of fermented PM extract (FE) on growth performance, learning and memory ability and intestinal microflora in mice. Ninety newly weaned male Kunming (KM) mice were randomly divided into seven groups: normal group (n 20), low-dose FE group (n 10), middle-dose FE group (MFE) (n 10), high-dose FE group (HFE) (n 20), unfermented extraction group (n 10), model group (10) and natural recovery group (10). Learning and memory skills were performed by the Morris water maze (MWM) test, and the variation in gut microbiota (GM) composition was assessed by 16S rDNA amplicon sequencing. The results show that HFE remarkably improved the growth performance in mice. In the MWM test, escape latency was shortened in both MFE and HFE groups, while the percentage of time, distance in target quadrant and the number crossing over the platform were significantly increased in the HFE group. Moreover, the FE played a preventive role in the dysbacteriosis of mice induced by antibiotic and increased the richness and species evenness of GM in mice.
Collapse
|
38
|
Nkenfou CN, Abange WB, Gonsu HK, Kamgaing N, Lyonga EM, Anoubissi JDD, Ndjolo A, Koki P. Evaluation of the effect of HIV virus on the digestive flora of infected versus non infected infants. Pan Afr Med J 2019; 34:24. [PMID: 31762893 PMCID: PMC6859050 DOI: 10.11604/pamj.2019.34.24.15039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 04/17/2019] [Indexed: 12/31/2022] Open
Abstract
Introduction HIV infection is characterized by changes in the composition and functions of gut microbiota. We carried out a study aiming at comparing the compositional changes of the digestive flora of HIV infected infants versus that of non infected infants in Cameroon. Methods A case-control study was carried out during which stool sample was collected from each participant after obtaining the proxy consent. Stools were cultured using aerobic, strict anaerobic, 10% CO2 and micro-aerophilic conditions and specific culture media and bacteria were identified biochemically. Fisher's exact test was used for data analyses. Results From the 80 infants enrolled for the study, 33 (41.3%) were HIV positive. A statistically significant difference was observed between the number of infected versus non infected infants harboring the following bacteria: Clostridium spp. (P=0.009); Enterococcus spp. (p<0.001); Klebsiella (p<0.001); Shigella (<0.001); Staphylococcus aureus (p=0.006) and Streptococcus spp. (P=0.015). Among infected infants, WHO-stage 3 and 4 infants harbored more opportunistic bacteria than stage 1 and stage 2 and Bacteriodes spp. population was depleted as the disease progresses, although not significantly. There was an imbalance in bacteria flora in HIV infected infants harboring qualitatively more bacteria including more opportunistic and pathogenic bacteria than in HIV non-infected infants. Conclusion HIV infected infants presented a qualitatively different flora from HIV non infected infants. They habored more pathogenic bacteria Than non infected infants. Systematic stool culture could benefit for follow-up of HIV infected infants to reduce the risk of gastrointestinal disorders and thus the risk of high morbidity or high mortability.
Collapse
Affiliation(s)
- Celine Nguefeu Nkenfou
- Chantal Biya's International Reference Centre (CBIRC), Centre for HIV/AIDS Prevention and Management, Yaounde, Cameroon.,Department of Biological Sciences, Higher Teachers Training College, University of Yaounde I, Cameroon
| | - William Baiye Abange
- University Teaching Hospital, Yaounde, Cameroon.,Catholic University of Central Africa, Yaounde, Cameroon
| | - Hortense Kamga Gonsu
- University Teaching Hospital, Yaounde, Cameroon.,Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Nelly Kamgaing
- Chantal Biya's International Reference Centre (CBIRC), Centre for HIV/AIDS Prevention and Management, Yaounde, Cameroon.,University Teaching Hospital, Yaounde, Cameroon.,Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Emilia Mbamyah Lyonga
- Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Yaounde, Cameroon
| | | | - Alexis Ndjolo
- Chantal Biya's International Reference Centre (CBIRC), Centre for HIV/AIDS Prevention and Management, Yaounde, Cameroon.,Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Yaounde, Cameroon
| | - Paul Koki
- Faculty of Medicine and Biomedical Sciences, University of Yaounde I, Yaounde, Cameroon.,Centre Mère Enfant, Fondation Chantal Biya, Yaounde, Cameroon
| |
Collapse
|
39
|
Tan X, Li J, Li Y, Li J, Wang Q, Fang L, Ding X, Huang P, Yang H, Yin Y. Effect of chicken egg yolk immunoglobulins on serum biochemical profiles and intestinal bacterial populations in early-weaned piglets. J Anim Physiol Anim Nutr (Berl) 2019; 103:1503-1511. [PMID: 31144409 PMCID: PMC7166376 DOI: 10.1111/jpn.13129] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Revised: 04/23/2019] [Accepted: 05/04/2019] [Indexed: 01/24/2023]
Abstract
This study was conducted to test the hypothesis that dietary supplementation with anti-E. coli, chicken egg yolk immunoglobulins (IgY), may affect early weaned piglet (EWP) intestinal functions and enteric micro-organisms. One hundred and forty-eight ([Landrace × Yorkshire] × Duroc) piglets, weaned at age day 21, were randomly assigned to receive one of three diets for 14 days. Treatment group one (control group) was fed the base diet. Treatment group two (antibiotics group) was fed the base diet which was supplemented with 100 ppm colistin sulphate and 15 ppm enramycin; treatment group three (IgY group) was fed the base diet which was supplemented with 500 mg/kg anti-E. coli IgY. The study evaluated the effects on EWPs of IgY on growth, serum biochemical, inflammatory profiles and also digestion content intestinal bacterial populations. Results showed no significant difference in diarrhoea rates between IgY-fed EWPs and antibiotic-treated EWPs. Serum biochemical analysis showed that EWPs fed an IgY-containing diet had both lower (p < 0.05) cholesterol and low-density lipoprotein compared to antibiotic-treated EWPs. Escherichia coli populations measured in IgY-fed EWP ileal contents, compared to the control group, were significantly reduced (p < 0.05). Enterococcus, Lactobacillus, Clostridium and Bifidobacterium populations were unaffected by the IgY treatment. Larger (p < 0.05) Enterococcus populations and lower (p < 0.05) expression levels of heat-stable enterotoxin b (STb) were observed in IgY-fed EWP caecal digesta compared to the control group. Enteric Lactobacillus significantly decreased (p < 0.05) in EWPs fed antibiotics while it was unaffected by IgY treatment. Dietary supplementation with anti-E. coli IgY has the potential to suppress enteric E. coli growth, but not Lactobacillus, Clostridium and Bifidobacterium. This promotes and maintains a healthy EWP intestinal environment. These findings suggest that IgY may be used as an alternative to antibiotics in EWP diets.
Collapse
Affiliation(s)
- Xian Tan
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life SciencesHunan Normal UniversityChangshaChina
| | - Jia Li
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life SciencesHunan Normal UniversityChangshaChina
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro‐ecological Processes in Subtropical Region, Institute of Subtropical AgricultureChinese Academy of SciencesChangshaChina
| | - Yali Li
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life SciencesHunan Normal UniversityChangshaChina
| | - Jianzhong Li
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life SciencesHunan Normal UniversityChangshaChina
| | - Qingping Wang
- Zyme Fast (Changsha) Biotechnology Co., Ltd.ChangshaChina
| | - Lin Fang
- Zyme Fast (Changsha) Biotechnology Co., Ltd.ChangshaChina
| | - Xueqin Ding
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life SciencesHunan Normal UniversityChangshaChina
| | - Pengfei Huang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life SciencesHunan Normal UniversityChangshaChina
| | - Huansheng Yang
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life SciencesHunan Normal UniversityChangshaChina
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro‐ecological Processes in Subtropical Region, Institute of Subtropical AgricultureChinese Academy of SciencesChangshaChina
| | - Yulong Yin
- Hunan International Joint Laboratory of Animal Intestinal Ecology and Health, Animal Nutrition and Human Health Laboratory, School of Life SciencesHunan Normal UniversityChangshaChina
- Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, Scientific Observing and Experimental Station of Animal Nutrition and Feed Science in South‐Central, Ministry of Agriculture, Hunan Provincial Engineering Research Center for Healthy Livestock and Poultry Production, Key Laboratory of Agro‐ecological Processes in Subtropical Region, Institute of Subtropical AgricultureChinese Academy of SciencesChangshaChina
| |
Collapse
|
40
|
Stokes JM, Lopatkin AJ, Lobritz MA, Collins JJ. Bacterial Metabolism and Antibiotic Efficacy. Cell Metab 2019; 30:251-259. [PMID: 31279676 PMCID: PMC6990394 DOI: 10.1016/j.cmet.2019.06.009] [Citation(s) in RCA: 374] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 06/06/2019] [Accepted: 06/10/2019] [Indexed: 02/07/2023]
Abstract
Antibiotics target energy-consuming processes. As such, perturbations to bacterial metabolic homeostasis are significant consequences of treatment. Here, we describe three postulates that collectively define antibiotic efficacy in the context of bacterial metabolism: (1) antibiotics alter the metabolic state of bacteria, which contributes to the resulting death or stasis; (2) the metabolic state of bacteria influences their susceptibility to antibiotics; and (3) antibiotic efficacy can be enhanced by altering the metabolic state of bacteria. Altogether, we aim to emphasize the close relationship between bacterial metabolism and antibiotic efficacy as well as propose areas of exploration to develop novel antibiotics that optimally exploit bacterial metabolic networks.
Collapse
Affiliation(s)
- Jonathan M Stokes
- Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Infectious Disease & Microbiome Program, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA; Machine Learning for Pharmaceutical Discovery and Synthesis Consortium, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Allison J Lopatkin
- Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Infectious Disease & Microbiome Program, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA
| | - Michael A Lobritz
- Roche Pharma Research and Early Development, Roche Innovation Center Basel, 4070 Basel, Switzerland
| | - James J Collins
- Institute for Medical Engineering & Science, Department of Biological Engineering, and Synthetic Biology Center, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Infectious Disease & Microbiome Program, Broad Institute of MIT & Harvard, Cambridge, MA 02142, USA; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA 02115, USA; Harvard-MIT Program in Health Sciences and Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
41
|
Potential Pathogenic Bacterial Contaminants of Doors Handles and Computers Keyboards in the Faculty Environment. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2019. [DOI: 10.22207/jpam.13.2.34] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
42
|
Belguesmia Y, Alard J, Mendil R, Ravallec R, Grangette C, Drider D, Cudennec B. In vitro probiotic properties of selected lactobacilli and multi-strain consortium on immune function, gut barrier strengthening and gut hormone secretion. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.04.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
|
43
|
Khanpour Ardestani S, Robinson JL, Dieleman LA, Huynh HQ, Jou H, Vohra S. Surveys of parents and clinicians concerning the minimally important difference of probiotic therapy for prevention of paediatric antibiotic-associated diarrhoea. BMJ Open 2019; 9:e024651. [PMID: 30944130 PMCID: PMC6500342 DOI: 10.1136/bmjopen-2018-024651] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
OBJECTIVES To establish the minimally important difference (MID) that would prompt parents and clinicians to use probiotics for prevention of paediatric antibiotic-associated diarrhoea (AAD) and to obtain parent and clinician opinion about the most important outcomes in clinical trials of AAD. METHODS In this survey, parents of children presenting to the emergency department of a Canadian tertiary care children's hospital and paediatricians working in that hospital were approached. A range of potential MIDs were presented and participants selected one that they would require to use probiotics for AAD prevention. In addition, participants were asked to rate a list of outcomes they would consider to be important in clinical trials of AAD. RESULTS In total, 127 parents and 45 paediatricians participated. About 51% (64/125) of parents and 51% (21/41) of clinicians responding to the MID question reported they would use probiotics if it reduced the risk of AAD by 39% (ie, reduce the risk of AAD from 19% to 12%). The most important outcomes to parents, in descending order, were need for hospitalisation, prevention of dehydration, disruption of normal daily activities, diarrhoea duration and physician revisit. Paediatricians considered need for hospitalisation along with physician revisit as the most important outcomes. They rated prevention of dehydration, diarrhoea duration and stool frequency as important outcomes as well. CONCLUSION There is good agreement between parents and clinicians regarding how effective probiotics would need to be in preventing AAD in order to warrant use. This information, along with outcomes perceived to be most important, will help in the design of future clinical trials.
Collapse
Affiliation(s)
- Samaneh Khanpour Ardestani
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Joan L Robinson
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Levinus A Dieleman
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Hien Q Huynh
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Hsing Jou
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Sunita Vohra
- Department of Pediatrics, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
- Department of Medicine, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| |
Collapse
|
44
|
Litou C, Effinger A, Kostewicz ES, Box KJ, Fotaki N, Dressman JB. Effects of medicines used to treat gastrointestinal diseases on the pharmacokinetics of coadministered drugs: a PEARRL Review. J Pharm Pharmacol 2019; 71:643-673. [PMID: 30062750 DOI: 10.1111/jphp.12983] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Accepted: 06/27/2018] [Indexed: 01/06/2025]
Abstract
OBJECTIVES Drugs used to treat gastrointestinal diseases (GI drugs) are widely used either as prescription or over-the-counter (OTC) medications and belong to both the 10 most prescribed and 10 most sold OTC medications worldwide. The objective of this review article is to discuss the most frequent interactions between GI and other drugs, including identification of the mechanisms behind these interactions, where possible. KEY FINDINGS Current clinical practice shows that in many cases, these drugs are administered concomitantly with other drug products. Due to their metabolic properties and mechanisms of action, the drugs used to treat gastrointestinal diseases can change the pharmacokinetics of some coadministered drugs. In certain cases, these interactions can lead to failure of treatment or to the occurrence of serious adverse events. The mechanism of interaction depends highly on drug properties and differs among therapeutic categories. Understanding these interactions is essential to providing recommendations for optimal drug therapy. SUMMARY Interactions with GI drugs are numerous and can be highly significant clinically in some cases. While alterations in bioavailability due to changes in solubility, dissolution rate, GI transit and metabolic interactions can be (for the most part) easily identified, interactions that are mediated through other mechanisms, such as permeability or microbiota, are less well-understood. Future work should focus on characterising these aspects.
Collapse
Affiliation(s)
- Chara Litou
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - Angela Effinger
- Department of Pharmacy and Pharmacology, Faculty of Science, University of Bath, Bath, UK
| | - Edmund S Kostewicz
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| | - Karl J Box
- Pion Inc. (UK) Ltd., Forest Row, East Sussex, UK
| | - Nikoletta Fotaki
- Department of Pharmacy and Pharmacology, Faculty of Science, University of Bath, Bath, UK
| | - Jennifer B Dressman
- Institute of Pharmaceutical Technology, Goethe University, Frankfurt am Main, Germany
| |
Collapse
|
45
|
Hossen I, Hua W, Ting L, Mehmood A, Jingyi S, Duoxia X, Yanping C, Hongqing W, Zhipeng G, Kaiqi Z, Fang Y, Junsong X. Phytochemicals and inflammatory bowel disease: a review. Crit Rev Food Sci Nutr 2019; 60:1321-1345. [PMID: 30729797 DOI: 10.1080/10408398.2019.1570913] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Gastrointestinal tract is the second largest organ in the body that mainly functions in nutrients and minerals intake through the intestinal barrier. Intestinal permeability maintains the circulation of minerals and nutrients from digested foods. Life and all the metabolic processes depend either directly or indirectly on proper functioning of GI tract. Compromised intestinal permeability and related disorders are common among all the patients with inflammatory bowel disease (IBD), which is a collective term of inflammatory diseases including Crohn's disease and ulcerative colitis. Many synthetic drugs are currently in use to treat IBD such as 5-aminosalicylic acid corticosteroids. However, they all have some drawbacks as long-term use result in many complications. These problems encourage us to look out for alternative medicine. Numerous in vitro and in vivo experiments showed that the plant-derived secondary metabolites including phenolic compounds, glucosinolates, alkaloids, terpenoids, oligosaccharides, and quinones could reduce permeability, ameliorate-related dysfunctions with promising results. In addition, many of them could modulate enzymatic activity, suppress the inflammatory transcriptional factors, ease oxidative stress, and reduce pro-inflammatory cytokines secretion. In this review, we summarized the phytochemicals, which were proven potent in treating increased intestinal permeability and related complication along with their mechanism of action.
Collapse
Affiliation(s)
- Imam Hossen
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China.,Beijing Key Lab of Plant Resource Research and Development, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing, China
| | - Wu Hua
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China.,Beijing Key Lab of Plant Resource Research and Development, Beijing, China
| | - Luo Ting
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Arshad Mehmood
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China.,Beijing Key Lab of Plant Resource Research and Development, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing, China
| | - Song Jingyi
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China.,Beijing Key Lab of Plant Resource Research and Development, Beijing, China
| | - Xu Duoxia
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China.,Beijing Key Lab of Plant Resource Research and Development, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing, China
| | - Cao Yanping
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China.,Beijing Key Lab of Plant Resource Research and Development, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing, China
| | - Wu Hongqing
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China.,Beijing Key Lab of Plant Resource Research and Development, Beijing, China
| | - Gao Zhipeng
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China.,Beijing Key Lab of Plant Resource Research and Development, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing, China
| | - Zhang Kaiqi
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China.,Beijing Key Lab of Plant Resource Research and Development, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing, China
| | - Yang Fang
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China.,Beijing Key Lab of Plant Resource Research and Development, Beijing, China
| | - Xiao Junsong
- School of Food and Chemical Engineering, Beijing Technology and Business University, Beijing, China.,Beijing Key Lab of Plant Resource Research and Development, Beijing, China.,Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing, China.,Beijing Engineering and Technology Research Center of Food Additives, Beijing, China
| |
Collapse
|
46
|
Nogueira T, David PHC, Pothier J. Antibiotics as both friends and foes of the human gut microbiome: The microbial community approach. Drug Dev Res 2018; 80:86-97. [PMID: 30370682 DOI: 10.1002/ddr.21466] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/14/2018] [Accepted: 08/15/2018] [Indexed: 12/28/2022]
Abstract
The exposure of the human gut to antibiotics can have a great impact on human health. Antibiotics pertain to the preservation of human health and are useful tools for fighting bacterial infections. They can be used for curing infections and can play a critical role in immunocompromised or chronic patients, or in fighting childhood severe malnutrition. Yet, the genomic and phylogenetic diversity of the human gut changes under antibiotic exposure. Antibiotics can also have severe side effects on human gut health, due to the spreading of potential antibiotic resistance genetic traits and to their correlation with virulence of some bacterial pathogens. They can shape, and even disrupt, the composition and functioning diversity of the human gut microbiome. Traditionally bacterial antibiotic resistances have been evaluated at clone or population level. However, the understanding of these two apparently disparate perspectives as both friends and foes may come from the study of microbiomes as a whole and from the evaluation of both positive and negative effects of antibiotics on microbial community dynamics and diversity. In this review we present some metagenomic tools and databases that enable the studying of antibiotic resistance in human gut metagenomes, promoting the development of personalized medicine strategies, new antimicrobial therapy protocols and patient follow-up.
Collapse
Affiliation(s)
- Teresa Nogueira
- cE3c - Centro de Ecologia, Evolução e Alterações Ambientais, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Pedro H C David
- cE3c - Centro de Ecologia, Evolução e Alterações Ambientais, Faculdade de Ciências, Universidade de Lisboa, Lisbon, Portugal
| | - Joël Pothier
- Institut de Systématique, Evolution, Biodiversité (ISYEB), Sorbonne Université, Muséum National d'Histoire naturelle, CNRS, EPHE, CP, Paris, France
| |
Collapse
|
47
|
Bacteriophages Synergize with the Gut Microbial Community To Combat Salmonella. mSystems 2018; 3:mSystems00119-18. [PMID: 30320220 PMCID: PMC6172775 DOI: 10.1128/msystems.00119-18] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2018] [Accepted: 08/31/2018] [Indexed: 12/14/2022] Open
Abstract
Antibiotic-resistant bacteria are a global threat. Therefore, alternative approaches for combatting bacteria, especially antibiotic-resistant bacteria, are urgently needed. Using a human gut microbiota model, we demonstrate that bacteriophages (phages) are able to substantially decrease pathogenic Salmonella without perturbing the microbiota. Conversely, antibiotic treatment leads to the eradication of close to all commensal bacteria, leaving only antibiotic-resistant bacteria. An unbalanced microbiota has been linked to many diseases both in the gastrointestinal tract or “nonintestinal” diseases. In our study, we show that the microbiota provides a protective effect against Salmonella. Since phage treatment preserves the healthy gut microbiota, it is a feasible superior alternative to antibiotic treatment. Furthermore, when combating infections caused by pathogenic bacteria, gut microbiota should be considered. Salmonella infection is one of the main causes of food-borne diarrheal diseases worldwide. Although most Salmonella infections can be cleared without treatment, some cause serious illnesses that require antibiotic treatment. In view of the growing emergence of antibiotic-resistant Salmonella strains, novel treatments are increasingly required. Furthermore, there is a striking paucity of data on how a balanced human gut microbiota responds to Salmonella infection. This study aimed to evaluate whether a balanced gut microbiota protects against Salmonella growth and to compare two antimicrobial approaches for managing Salmonella infection: bacteriophage (phage) treatment and antibiotic treatment. Anaerobically cultivated human intestinal microflora (ACHIM) is a feasible model for the human gut microbiota and naturally inhibits Salmonella infection. By mimicking Salmonella infection in vitro using ACHIM, we observed a large reduction of Salmonella growth by the ACHIM itself. Treatments with phage and antibiotic further inhibited Salmonella growth. However, phage treatment had less impact on the nontargeted bacteria in ACHIM than the antibiotic treatment did. Phage treatment has high specificity when combating Salmonella infection and offers a noninvasive alternative to antibiotic treatment. IMPORTANCE Antibiotic-resistant bacteria are a global threat. Therefore, alternative approaches for combatting bacteria, especially antibiotic-resistant bacteria, are urgently needed. Using a human gut microbiota model, we demonstrate that bacteriophages (phages) are able to substantially decrease pathogenic Salmonella without perturbing the microbiota. Conversely, antibiotic treatment leads to the eradication of close to all commensal bacteria, leaving only antibiotic-resistant bacteria. An unbalanced microbiota has been linked to many diseases both in the gastrointestinal tract or “nonintestinal” diseases. In our study, we show that the microbiota provides a protective effect against Salmonella. Since phage treatment preserves the healthy gut microbiota, it is a feasible superior alternative to antibiotic treatment. Furthermore, when combating infections caused by pathogenic bacteria, gut microbiota should be considered. Author Video: An author video summary of this article is available.
Collapse
|
48
|
Clostridium difficile, the Difficult "Kloster" Fuelled by Antibiotics. Curr Microbiol 2018; 76:774-782. [PMID: 30084095 DOI: 10.1007/s00284-018-1543-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Accepted: 07/17/2018] [Indexed: 02/07/2023]
Abstract
Clostridium difficile is normally present in low numbers in a healthy adult gastro-intestinal tract (GIT). Drastic changes in the microbial population, e.g., dysbiosis caused by extensive treatment with antibiotics, stimulates the growth of resistant strains and the onset of C. difficile infection (CDI). Symptoms of infection varies from mild diarrhea to colitis (associated with dehydration and bleeding), pseudomembranous colitis with yellow ulcerations in the mucosa of the colon, to fulminant colitis (perforation of the gut membrane), and multiple organ failure. Inflamed epithelial cells and damaged mucosal tissue predisposes the colon to other opportunistic pathogens such as Clostridium perfringens, Staphylococcus aureus, Klebsiella oxytoca, Candida spp., and Salmonella spp. This may lead to small intestinal bacterial overgrowth (SIBO), sepsis, toxic megacolon, and even colorectal cancer. Many stains of C. difficile are resistant to metronidazole and vancomycin. Vaccination may be an answer to CDI, but requires more research. Success in treatment with probiotics depends on the strains used. Oral or rectal fecal transplants are partly effective, as spores in the small intestine may germinate and colonize the colon. The effect of antibiotics on C. difficile and commensal gut microbiota is summarized and changes in gut physiology are discussed. The need to search for non-antibiotic methods in the treatment of CDI and C. difficile-associated disease (CDAD) is emphasized.
Collapse
|
49
|
Wu XD, Xu W, Liu MM, Hu KJ, Sun YY, Yang XF, Zhu GQ, Wang ZW, Huang W. Efficacy of prophylactic probiotics in combination with antibiotics versus antibiotics alone for colorectal surgery: A meta-analysis of randomized controlled trials. J Surg Oncol 2018; 117:1394-1404. [PMID: 29572838 DOI: 10.1002/jso.25038] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2017] [Accepted: 02/12/2018] [Indexed: 02/05/2023]
Abstract
This meta-analysis aimed to determine whether prophylactic probiotics in combination with antibiotics are superior to antibiotics alone in the prevention of surgical site infection (SSI) after colorectal surgery. Fourteen trials involving 1524 participants were included. Compared with antibiotics alone, prophylactic probiotics in combination with antibiotics reduced the risk of SSI as well as other complications, shortened the cumulative duration of antibiotic therapy. Current evidence suggested that probiotics in combination with antibiotics could be recommended.
Collapse
Affiliation(s)
- Xiang-Dong Wu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Xu
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Meng-Meng Liu
- Department of Pathology, Anhui Medical University, Hefei, Anhui Province, China
| | - Ke-Jia Hu
- Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
- Harvard-MIT Health Sciences and Technology, Cambridge, Massachusetts
- Department of Microsurgery, Huashan Hospital, Fudan University, Shanghai, China
| | - Ya-Ying Sun
- Department of Sports Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Xue-Fei Yang
- Department of Endocrinology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Gui-Qi Zhu
- Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Labolatory of Carcinogenesis and Cancer Invasion, Fudan University, Ministry of Education, Shanghai, China
| | - Zi-Wei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wei Huang
- Department of Orthopaedic Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
50
|
Probiotics and antibiotic-associated diarrhea in children: A review and new evidence on Lactobacillus rhamnosus GG during and after antibiotic treatment. Pharmacol Res 2018; 128:63-72. [DOI: 10.1016/j.phrs.2017.08.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 08/03/2017] [Accepted: 08/03/2017] [Indexed: 12/13/2022]
|