1
|
Chaudhary S, Kaur P, Singh TA, Bano KS, Vyas A, Mishra AK, Singh P, Mehdi MM. The dynamic crosslinking between gut microbiota and inflammation during aging: reviewing the nutritional and hormetic approaches against dysbiosis and inflammaging. Biogerontology 2024; 26:1. [PMID: 39441393 DOI: 10.1007/s10522-024-10146-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 10/01/2024] [Indexed: 10/25/2024]
Abstract
The early-life gut microbiota (GM) is increasingly recognized for its contributions to human health and disease over time. Microbiota composition, influenced by factors like race, geography, lifestyle, and individual differences, is subject to change. The GM serves dual roles, defending against pathogens and shaping the host immune system. Disruptions in microbial composition can lead to immune dysregulation, impacting defense mechanisms. Additionally, GM aids digestion, releasing nutrients and influencing physiological systems like the liver, brain, and endocrine system through microbial metabolites. Dysbiosis disrupts intestinal homeostasis, contributing to age-related diseases. Recent studies are elucidating the bacterial species that characterize a healthy microbiota, defining what constitutes a 'healthy' colonic microbiota. The present review article focuses on the importance of microbiome composition for the development of homeostasis and the roles of GM during aging and the age-related diseases caused by the alteration in gut microbial communities. This article might also help the readers to find treatments targeting GM for the prevention of various diseases linked to it effectively.
Collapse
Affiliation(s)
- Sakshi Chaudhary
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Pardeep Kaur
- Department of Biotechnology, Chandigarh University, Mohali, Punjab, 140413, India
| | - Thokchom Arjun Singh
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Kaniz Shahar Bano
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Ashish Vyas
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Alok Kumar Mishra
- Department of Microbiology, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Prabhakar Singh
- Department of Biotechnology, School of Bio and Chemical Engineering, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, 600119, India
| | - Mohammad Murtaza Mehdi
- Department of Biochemistry, School of Bioengineering and Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India.
| |
Collapse
|
2
|
Chu VT, Tsitsiklis A, Mick E, Ambroggio L, Kalantar KL, Glascock A, Osborne CM, Wagner BD, Matthay MA, DeRisi JL, Calfee CS, Mourani PM, Langelier CR. The antibiotic resistance reservoir of the lung microbiome expands with age in a population of critically ill patients. Nat Commun 2024; 15:92. [PMID: 38168095 PMCID: PMC10762195 DOI: 10.1038/s41467-023-44353-1] [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: 08/30/2023] [Accepted: 12/11/2023] [Indexed: 01/05/2024] Open
Abstract
Antimicrobial resistant lower respiratory tract infections are an increasing public health threat and an important cause of global mortality. The lung microbiome can influence susceptibility of respiratory tract infections and represents an important reservoir for exchange of antimicrobial resistance genes. Studies of the gut microbiome have found an association between age and increasing antimicrobial resistance gene burden, however, corollary studies in the lung microbiome remain absent. We performed an observational study of children and adults with acute respiratory failure admitted to the intensive care unit. From tracheal aspirate RNA sequencing data, we evaluated age-related differences in detectable antimicrobial resistance gene expression in the lung microbiome. Using a multivariable logistic regression model, we find that detection of antimicrobial resistance gene expression was significantly higher in adults compared with children after adjusting for demographic and clinical characteristics. This association remained significant after additionally adjusting for lung bacterial microbiome characteristics, and when modeling age as a continuous variable. The proportion of adults expressing beta-lactam, aminoglycoside, and tetracycline antimicrobial resistance genes was higher compared to children. Together, these findings shape our understanding of the lung resistome in critically ill patients across the lifespan, which may have implications for clinical management and global public health.
Collapse
Affiliation(s)
- Victoria T Chu
- Division of Infectious Diseases & Global Health, University of California, San Francisco, CA, USA
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
| | - Alexandra Tsitsiklis
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
| | - Eran Mick
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Division of Pulmonary and Critical Care Medicine, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Lilliam Ambroggio
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO, USA
| | | | | | - Christina M Osborne
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO, USA
| | - Brandie D Wagner
- Department of Pediatrics, University of Colorado and Children's Hospital Colorado, Aurora, CO, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado, Aurora, CO, USA
| | - Michael A Matthay
- Division of Pulmonary and Critical Care Medicine, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Joseph L DeRisi
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Carolyn S Calfee
- Division of Pulmonary and Critical Care Medicine, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Peter M Mourani
- Arkansas Children's Research Institute, Arkansas Children's Hospital, Little Rock, AR, USA
| | - Charles R Langelier
- Division of Infectious Diseases, University of California, San Francisco, CA, USA.
- Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
3
|
Fu Y, Dou Q, Smalla K, Wang Y, Johnson TA, Brandt KK, Mei Z, Liao M, Hashsham SA, Schäffer A, Smidt H, Zhang T, Li H, Stedtfeld R, Sheng H, Chai B, Virta M, Jiang X, Wang F, Zhu Y, Tiedje JM. Gut microbiota research nexus: One Health relationship between human, animal, and environmental resistomes. MLIFE 2023; 2:350-364. [PMID: 38818274 PMCID: PMC10989101 DOI: 10.1002/mlf2.12101] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 06/01/2024]
Abstract
The emergence and rapid spread of antimicrobial resistance is of global public health concern. The gut microbiota harboring diverse commensal and opportunistic bacteria that can acquire resistance via horizontal and vertical gene transfers is considered an important reservoir and sink of antibiotic resistance genes (ARGs). In this review, we describe the reservoirs of gut ARGs and their dynamics in both animals and humans, use the One Health perspective to track the transmission of ARG-containing bacteria between humans, animals, and the environment, and assess the impact of antimicrobial resistance on human health and socioeconomic development. The gut resistome can evolve in an environment subject to various selective pressures, including antibiotic administration and environmental and lifestyle factors (e.g., diet, age, gender, and living conditions), and interventions through probiotics. Strategies to reduce the abundance of clinically relevant antibiotic-resistant bacteria and their resistance determinants in various environmental niches are needed to ensure the mitigation of acquired antibiotic resistance. With the help of effective measures taken at the national, local, personal, and intestinal management, it will also result in preventing or minimizing the spread of infectious diseases. This review aims to improve our understanding of the correlations between intestinal microbiota and antimicrobial resistance and provide a basis for the development of management strategies to mitigate the antimicrobial resistance crisis.
Collapse
Affiliation(s)
- Yuhao Fu
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil ScienceChinese Academy of SciencesNanjingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Qingyuan Dou
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil ScienceChinese Academy of SciencesNanjingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Kornelia Smalla
- Julius Kühn Institute (JKI) Federal Research Centre for Cultivated PlantsBraunschweigGermany
| | - Yu Wang
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil ScienceChinese Academy of SciencesNanjingChina
- University of Chinese Academy of SciencesBeijingChina
| | | | - Kristian K. Brandt
- Section for Microbial Ecology and Biotechnology, Department of Plant and Environmental SciencesUniversity of CopenhagenFrederiksberg CDenmark
- Sino‐Danish Center (SDC)BeijingChina
| | - Zhi Mei
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil ScienceChinese Academy of SciencesNanjingChina
- University of Chinese Academy of SciencesBeijingChina
- Department of MicrobiologyUniversity of HelsinkiHelsinkiFinland
| | - Maoyuan Liao
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil ScienceChinese Academy of SciencesNanjingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Syed A. Hashsham
- Department of Plant, Soil and Microbial Sciences, Center for Microbial EcologyMichigan State UniversityMichiganUSA
- Department of Civil and Environmental EngineeringMichigan State UniversityMichiganUSA
| | - Andreas Schäffer
- Institute for Environmental ResearchRWTH Aachen UniversityAachenGermany
| | - Hauke Smidt
- Laboratory of MicrobiologyWageningen University & ResearchWageningenThe Netherlands
| | - Tong Zhang
- Environmental Microbiome Engineering and Biotechnology Laboratory, Center for Environmental Engineering Research, Department of Civil EngineeringThe University of Hong KongPokfulamHong KongChina
| | - Hui Li
- Department of Plant, Soil and Microbial Sciences, Center for Microbial EcologyMichigan State UniversityMichiganUSA
| | - Robert Stedtfeld
- Department of Civil and Environmental EngineeringMichigan State UniversityMichiganUSA
| | - Hongjie Sheng
- Institute of Agricultural Resources and EnvironmentJiangsu Academy of Agricultural SciencesNanjingChina
| | - Benli Chai
- Department of Plant, Soil and Microbial Sciences, Center for Microbial EcologyMichigan State UniversityMichiganUSA
| | - Marko Virta
- Department of MicrobiologyUniversity of HelsinkiHelsinkiFinland
| | - Xin Jiang
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil ScienceChinese Academy of SciencesNanjingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Fang Wang
- State Key Laboratory of Soil and Sustainable Agriculture, Institute of Soil ScienceChinese Academy of SciencesNanjingChina
- University of Chinese Academy of SciencesBeijingChina
| | - Yong‐Guan Zhu
- University of Chinese Academy of SciencesBeijingChina
- Key Laboratory of Urban Environment and Health, Institute of Urban EnvironmentChinese Academy of SciencesXiamenChina
- State Key Laboratory of Urban and Regional EcologyChinese Academy of SciencesBeijingChina
| | - James M. Tiedje
- Department of Plant, Soil and Microbial Sciences, Center for Microbial EcologyMichigan State UniversityMichiganUSA
| |
Collapse
|
4
|
Mollova D, Gozmanova M, Apostolova E, Yahubyan G, Iliev I, Baev V. Illuminating the Genomic Landscape of Lactiplantibacillus plantarum PU3-A Novel Probiotic Strain Isolated from Human Breast Milk, Explored through Nanopore Sequencing. Microorganisms 2023; 11:2440. [PMID: 37894099 PMCID: PMC10609609 DOI: 10.3390/microorganisms11102440] [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: 08/17/2023] [Revised: 09/11/2023] [Accepted: 09/25/2023] [Indexed: 10/29/2023] Open
Abstract
Lactiplantibacillus plantarum stands out as a remarkably diverse species of lactic acid bacteria, occupying a myriad of ecological niches. Particularly noteworthy is its presence in human breast milk, which can serve as a reservoir of probiotic bacteria, contributing significantly to the establishment and constitution of infant gut microbiota. In light of this, our study attempted to conduct an initial investigation encompassing both genomic and phenotypic aspects of the L. plantarum PU3 strain, that holds potential as a probiotic agent. By employing the cutting-edge third-generation Nanopore sequencing technology, L. plantarum PU3 revealed a circular chromosome of 3,180,940 bp and nine plasmids of various lengths. The L. plantarum PU3 genome has a total of 2962 protein-coding and non-coding genes. Our in-depth investigations revealed more than 150 probiotic gene markers that unfold the genetic determinants for acid tolerance, bile resistance, adhesion, and oxidative and osmotic stress. The in vivo analysis showed the strain's proficiency in utilizing various carbohydrates as growth substrates, complementing the in silico analysis of the genes involved in metabolic pathways. Notably, the strain demonstrated a pronounced affinity for D-sorbitol, D-mannitol, and D-Gluconic acid, among other carbohydrate sources. The in vitro experimental verification of acid, osmotic and bile tolerance validated the robustness of the strain in challenging environments. Encouragingly, no virulence factors were detected in the genome of PU3, suggesting its safety profile. In search of beneficial properties, we found potential bacteriocin biosynthesis clusters, suggesting its capability for antimicrobial activity. The characteristics exhibited by L. plantarum PU3 pave the way for promising strain potential, warranting further investigations to unlock its full capacity and contributions to probiotic and therapeutic avenues.
Collapse
Affiliation(s)
- Daniela Mollova
- Faculty of Biology, Department of Biochemistry and Microbiology, University of Plovdiv, Tzar Assen 24, 4000 Plovdiv, Bulgaria; (D.M.); (I.I.)
| | - Mariyana Gozmanova
- Faculty of Biology, Department of Plant Physiology and Molecular Biology, University of Plovdiv, Tzar Assen 24, 4000 Plovdiv, Bulgaria; (M.G.); (E.A.); (G.Y.)
| | - Elena Apostolova
- Faculty of Biology, Department of Plant Physiology and Molecular Biology, University of Plovdiv, Tzar Assen 24, 4000 Plovdiv, Bulgaria; (M.G.); (E.A.); (G.Y.)
| | - Galina Yahubyan
- Faculty of Biology, Department of Plant Physiology and Molecular Biology, University of Plovdiv, Tzar Assen 24, 4000 Plovdiv, Bulgaria; (M.G.); (E.A.); (G.Y.)
| | - Ilia Iliev
- Faculty of Biology, Department of Biochemistry and Microbiology, University of Plovdiv, Tzar Assen 24, 4000 Plovdiv, Bulgaria; (D.M.); (I.I.)
| | - Vesselin Baev
- Faculty of Biology, Department of Plant Physiology and Molecular Biology, University of Plovdiv, Tzar Assen 24, 4000 Plovdiv, Bulgaria; (M.G.); (E.A.); (G.Y.)
| |
Collapse
|
5
|
Chu VT, Tsitsiklis A, Mick E, Ambroggio L, Kalantar KL, Glascock A, Osborne CM, Wagner BD, Matthay MA, DeRisi JL, Calfee CS, Mourani PM, Langelier CR. The antibiotic resistance reservoir of the lung microbiome expands with age. RESEARCH SQUARE 2023:rs.3.rs-3283415. [PMID: 37790384 PMCID: PMC10543260 DOI: 10.21203/rs.3.rs-3283415/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Antimicrobial resistant lower respiratory tract infections (LRTI) are an increasing public health threat, and an important cause of global mortality. The lung microbiome influences LRTI susceptibility and represents an important reservoir for exchange of antimicrobial resistance genes (ARGs). Studies of the gut microbiome have found an association between age and increasing antimicrobial resistance gene (ARG) burden, however corollary studies in the lung microbiome remain absent, despite the respiratory tract representing one of the most clinically significant sites for drug resistant infections. We performed a prospective, multicenter observational study of 261 children and 88 adults with acute respiratory failure, ranging in age from 31 days to ≥ 89 years, admitted to intensive care units in the United States. We performed RNA sequencing on tracheal aspirates collected within 72 hours of intubation, and evaluated age-related differences in detectable ARG expression in the lung microbiome as a primary outcome. Secondary outcomes included number and classes of ARGs detected, proportion of patients with an ARG class, and composition of the lung microbiome. Multivariable logistic regression models (adults vs children) or continuous age (years) were adjusted for sex, race/ethnicity, LRTI status, and days from intubation to specimen collection. Detection of ARGs was significantly higher in adults compared with children after adjusting for sex, race/ethnicity, LRTI diagnosis, and days from intubation to specimen collection (adjusted odds ratio (aOR): 2.16, 95% confidence interval (CI): 1.10-4.22). A greater proportion of adults compared with children had beta-lactam ARGs (31% (CI: 21-41%) vs 13% (CI: 10-18%)), aminoglycoside ARGs (20% (CI: 13-30%) vs 2% (CI: 0.6-4%)), and tetracycline ARGs (14% (CI: 7-23%) vs 3% (CI: 1-5%)). Adults ≥70 years old had the highest proportion of these three ARG classes. The total bacterial abundance of the lung microbiome increased with age, and microbiome alpha diversity varied with age. Taxonomic composition of the lung microbiome, measured by Bray Curtis dissimilarity index, differed between adults and children (p = 0.003). The association between age and increased ARG detection remained significant after additionally including lung microbiome total bacterial abundance and alpha diversity in the multivariable logistic regression model (aOR: 2.38, (CI: 1.25-4.54)). Furthermore, this association remained robust when modeling age as a continuous variable (aOR: 1.02, (CI: 1.01-1.03) per year of age). Taken together, our results demonstrate that age is an independent risk factor for ARG detection in the lower respiratory tract microbiome. These data shape our understanding of the lung resistome in critically ill patients across the lifespan, which may have implications for clinical management and global public health.
Collapse
Affiliation(s)
- Victoria T. Chu
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Alexandra Tsitsiklis
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
| | - Eran Mick
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Division of Pulmonary and Critical Care Medicine, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Lilliam Ambroggio
- Department of Pediatrics, University of Colorado and Children’s Hospital Colorado, Aurora, CO, USA
| | | | | | - Christina M. Osborne
- Department of Pediatrics, University of Colorado and Children’s Hospital Colorado, Aurora, CO, USA
| | - Brandie D. Wagner
- Department of Pediatrics, University of Colorado and Children’s Hospital Colorado, Aurora, CO, USA
- Department of Biostatistics and Informatics, Colorado School of Public Health, University of Colorado, Aurora, CO, USA
| | - Michael A. Matthay
- Division of Pulmonary and Critical Care Medicine, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Joseph L. DeRisi
- Chan Zuckerberg Biohub, San Francisco, CA, USA
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Carolyn S. Calfee
- Division of Pulmonary and Critical Care Medicine, Cardiovascular Research Institute, University of California, San Francisco, CA, USA
| | - Peter M. Mourani
- Arkansas Children’s Research Institute, Arkansas Children’s Hospital, Little Rock, AR, USA
| | - Charles R. Langelier
- Division of Infectious Diseases, University of California, San Francisco, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| |
Collapse
|
6
|
Todman H, Arya S, Baker M, Stekel DJ. A model of antibiotic resistance genes accumulation through lifetime exposure from food intake and antibiotic treatment. PLoS One 2023; 18:e0289941. [PMID: 37590256 PMCID: PMC10434901 DOI: 10.1371/journal.pone.0289941] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 07/30/2023] [Indexed: 08/19/2023] Open
Abstract
Antimicrobial resistant bacterial infections represent one of the most serious contemporary global healthcare crises. Acquisition and spread of resistant infections can occur through community, hospitals, food, water or endogenous bacteria. Global efforts to reduce resistance have typically focussed on antibiotic use, hygiene and sanitation and drug discovery. However, resistance in endogenous infections, e.g. many urinary tract infections, can result from life-long acquisition and persistence of resistance genes in commensal microbial flora of individual patients, which is not normally considered. Here, using individual based Monte Carlo models calibrated using antibiotic use data and human gut resistomes, we show that the long-term increase in resistance in human gut microbiomes can be substantially lowered by reducing exposure to resistance genes found food and water, alongside reduced medical antibiotic use. Reduced dietary exposure is especially important during patient antibiotic treatment because of increased selection for resistance gene retention; inappropriate use of antibiotics can be directly harmful to the patient being treated for the same reason. We conclude that a holistic approach to antimicrobial resistance that additionally incorporates food production and dietary considerations will be more effective in reducing resistant infections than a purely medical-based approach.
Collapse
Affiliation(s)
- Henry Todman
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Nottingham, United Kingdom
| | - Sankalp Arya
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Nottingham, United Kingdom
| | - Michelle Baker
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Nottingham, United Kingdom
| | - Dov Joseph Stekel
- School of Biosciences, University of Nottingham, Sutton Bonington Campus, Loughborough, Nottingham, United Kingdom
- Department of Mathematics and Applied Mathematics, University of Johannesburg, Rossmore, South Africa
| |
Collapse
|
7
|
Saturio S, Rey A, Samarra A, Collado MC, Suárez M, Mantecón L, Solís G, Gueimonde M, Arboleya S. Old Folks, Bad Boon: Antimicrobial Resistance in the Infant Gut Microbiome. Microorganisms 2023; 11:1907. [PMID: 37630467 PMCID: PMC10458625 DOI: 10.3390/microorganisms11081907] [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: 06/15/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/27/2023] Open
Abstract
The development of the intestinal microbiome in the neonate starts, mainly, at birth, when the infant receives its founding microbial inoculum from the mother. This microbiome contains genes conferring resistance to antibiotics since these are found in some of the microorganisms present in the intestine. Similarly to microbiota composition, the possession of antibiotic resistance genes is affected by different perinatal factors. Moreover, antibiotics are the most used drugs in early life, and the use of antibiotics in pediatrics covers a wide variety of possibilities and treatment options. The disruption in the early microbiota caused by antibiotics may be of great relevance, not just because it may limit colonization by beneficial microorganisms and increase that of potential pathogens, but also because it may increase the levels of antibiotic resistance genes. The increase in antibiotic-resistant microorganisms is one of the major public health threats that humanity has to face and, therefore, understanding the factors that determine the development of the resistome in early life is of relevance. Recent advancements in sequencing technologies have enabled the study of the microbiota and the resistome at unprecedent levels. These aspects are discussed in this review as well as some potential interventions aimed at reducing the possession of resistance genes.
Collapse
Affiliation(s)
- Silvia Saturio
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.R.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
| | - Alejandra Rey
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.R.)
| | - Anna Samarra
- Institute of Agrochemistry and Food Technology (IATA-CSIC), 46980 Paterna, Spain; (A.S.); (M.C.C.)
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology (IATA-CSIC), 46980 Paterna, Spain; (A.S.); (M.C.C.)
| | - Marta Suárez
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
| | - Laura Mantecón
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
| | - Gonzalo Solís
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
- Pediatrics Service, Central University Hospital of Asturias (HUCA-SESPA), 33011 Oviedo, Spain
| | - Miguel Gueimonde
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.R.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
| | - Silvia Arboleya
- Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Spain; (S.S.); (A.R.)
- Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), 33011 Oviedo, Spain; (M.S.); (L.M.); (G.S.)
| |
Collapse
|
8
|
Recent Approaches for Downplaying Antibiotic Resistance: Molecular Mechanisms. BIOMED RESEARCH INTERNATIONAL 2023; 2023:5250040. [PMID: 36726844 PMCID: PMC9886476 DOI: 10.1155/2023/5250040] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2022] [Revised: 10/06/2022] [Accepted: 10/12/2022] [Indexed: 01/25/2023]
Abstract
Antimicrobial resistance (AMR) is a ubiquitous public health menace. AMR emergence causes complications in treating infections contributing to an upsurge in the mortality rate. The epidemic of AMR in sync with a high utilization rate of antimicrobial drugs signifies an alarming situation for the fleet recovery of both animals and humans. The emergence of resistant species calls for new treatments and therapeutics. Current records propose that health drug dependency, veterinary medicine, agricultural application, and vaccination reluctance are the primary etymology of AMR gene emergence and spread. Recently, several encouraging avenues have been presented to contest resistance, such as antivirulent therapy, passive immunization, antimicrobial peptides, vaccines, phage therapy, and botanical and liposomal nanoparticles. Most of these therapies are used as cutting-edge methodologies to downplay antibacterial drugs to subdue the resistance pressure, which is a featured motive of discussion in this review article. AMR can fade away through the potential use of current cutting-edge therapeutics, advancement in antimicrobial susceptibility testing, new diagnostic testing, prompt clinical response, and probing of new pharmacodynamic properties of antimicrobials. It also needs to promote future research on contemporary methods to maintain host homeostasis after infections caused by AMR. Referable to the microbial ability to break resistance, there is a great ultimatum for using not only appropriate and advanced antimicrobial drugs but also other neoteric diverse cutting-edge therapeutics.
Collapse
|
9
|
Leo S, Curtis N, Zimmermann P. The neonatal intestinal resistome and factors that influence it - a systematic review. Clin Microbiol Infect 2022; 28:1539-1546. [PMID: 35868586 DOI: 10.1016/j.cmi.2022.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/21/2022] [Accepted: 07/14/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND The intestinal microbiome provides a reservoir for antibiotic resistance genes (ARGs). The neonatal microbiome is more susceptible to disturbance from external factors than the established microbiome. OBJECTIVES In this review, we systematically summarise studies which investigated the intestinal resistome in neonates. DATA SOURCES MEDLINE and Embase databases were searched. STUDY ELIGIBILITY CRITERIA We included original studies which investigated ARGs in stool or rectal swabs in neonates using molecular diagnostics. METHODS OF DATA SYNTHESIS Two authors independently extracted data. Data was summarised in tables. RESULTS Our search identified 2,701 studies, of which 23 (22 cohorts) were included. The studies show that the neonatal intestine harbours a high abundance and variety of ARGs, even in the absence of direct antibiotic exposure. The most-commonly found ARGs confer resistance to aminoglycosides, beta-lactams, macrolides, tetracyclines or multi-drug resistance. There is evidence that ARGs can be transferred from mothers to neonates. Interestingly, however, compared to mothers, neonates are reported to have a higher abundance of ARGs. One likely reason for this is the bacterial phylogenetic composition with a high abundance of Gammaproteobacteria in neonatal stool. Factors that have been associated with a higher abundance of ARGs are intrapartum and neonatal antibiotic use. Breastfeeding and neonatal probiotic use have been associated with a lower abundance of ARGs. Antibiotics during pregnancy, delivery mode or sex are reported to have little effect. However, this might be because studies were underpowered and because it is difficult to account for effect modifiers. DISCUSSION The neonatal intestine seems to have a lower colonisation resistance, which could make it easier for antibiotic-resistant populations to establish themselves. Future studies will help in the development of evidence-based interventions to modulate the abundance of ARGs in neonates, for example, by the use of pre- and probiotics and bacteriophages.
Collapse
Affiliation(s)
- Stefano Leo
- Department for Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland; Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland
| | - Nigel Curtis
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia; Department of Paediatrics, The University of Melbourne, Parkville, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, Australia
| | - Petra Zimmermann
- Department for Community Health, Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland; Department of Paediatrics, Fribourg Hospital, Fribourg, Switzerland; Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, Australia.
| |
Collapse
|
10
|
Shuai M, Zhang G, Zeng F, Fu Y, Liang X, Yuan L, Xu F, Gou W, Miao Z, Jiang Z, Wang J, Zhuo L, Chen Y, Ju F, Zheng J. Human Gut Antibiotic Resistome and Progression of Diabetes. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104965. [PMID: 35142450 PMCID: PMC9008416 DOI: 10.1002/advs.202104965] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 01/13/2022] [Indexed: 05/02/2023]
Abstract
The antibiotic resistance crisis underlies globally increasing failures in treating deadly bacterial infections, largely due to the selection of antibiotic resistance genes (ARG) collection, known as the resistome, in human gut microbiota. So far, little is known about the relationship between gut antibiotic resistome and host metabolic disorders such as type 2 diabetes (T2D). Here, metagenomic landscape of gut antibiotic resistome is profiled in a large multiomics human cohort (n = 1210). There is a significant overall shift in gut antibiotic resistome structure among healthy, prediabetes, and T2D groups. It is found that larger ARG diversity is associated with a higher risk of T2D. The novel diabetes ARG score is positively associated with glycemic traits. Longitudinal validation analysis confirms that the ARG score is associated with T2D progression, characterized by the change of insulin resistance. Collectively, the data describe the profiles of gut antibiotic resistome and support its close relationship with T2D progression.
Collapse
Affiliation(s)
- Menglei Shuai
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhou310030China
- Westlake Intelligent Biomarker Discovery LabWestlake Laboratory of Life Sciences and BiomedicineHangzhou310024China
| | - Guoqing Zhang
- Key Laboratory of Coastal Environment and Resources of Zhejiang ProvinceSchool of EngineeringWestlake UniversityHangzhou310030China
- Institute of Advanced TechnologyWestlake Institute for Advanced StudyHangzhou310024China
| | - Fang‐fang Zeng
- Guangdong Provincial Key Laboratory of FoodNutrition and HealthDepartment of EpidemiologySchool of Public HealthSun Yat‐sen UniversityGuangzhou510275China
- Department of EpidemiologySchool of MedicineJinan UniversityGuangzhou510632China
| | - Yuanqing Fu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhou310030China
- Westlake Intelligent Biomarker Discovery LabWestlake Laboratory of Life Sciences and BiomedicineHangzhou310024China
- Institute of Basic Medical SciencesWestlake Institute for Advanced StudyHangzhou310024China
| | - Xinxiu Liang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhou310030China
- Westlake Intelligent Biomarker Discovery LabWestlake Laboratory of Life Sciences and BiomedicineHangzhou310024China
| | - Ling Yuan
- Key Laboratory of Coastal Environment and Resources of Zhejiang ProvinceSchool of EngineeringWestlake UniversityHangzhou310030China
- Institute of Advanced TechnologyWestlake Institute for Advanced StudyHangzhou310024China
| | - Fengzhe Xu
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhou310030China
- Westlake Intelligent Biomarker Discovery LabWestlake Laboratory of Life Sciences and BiomedicineHangzhou310024China
| | - Wanglong Gou
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhou310030China
- Westlake Intelligent Biomarker Discovery LabWestlake Laboratory of Life Sciences and BiomedicineHangzhou310024China
| | - Zelei Miao
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhou310030China
- Westlake Intelligent Biomarker Discovery LabWestlake Laboratory of Life Sciences and BiomedicineHangzhou310024China
| | - Zengliang Jiang
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhou310030China
- Westlake Intelligent Biomarker Discovery LabWestlake Laboratory of Life Sciences and BiomedicineHangzhou310024China
- Institute of Basic Medical SciencesWestlake Institute for Advanced StudyHangzhou310024China
| | - Jia‐ting Wang
- Guangdong Provincial Key Laboratory of FoodNutrition and HealthDepartment of EpidemiologySchool of Public HealthSun Yat‐sen UniversityGuangzhou510275China
| | - Lai‐bao Zhuo
- Guangdong Provincial Key Laboratory of FoodNutrition and HealthDepartment of EpidemiologySchool of Public HealthSun Yat‐sen UniversityGuangzhou510275China
| | - Yu‐ming Chen
- Guangdong Provincial Key Laboratory of FoodNutrition and HealthDepartment of EpidemiologySchool of Public HealthSun Yat‐sen UniversityGuangzhou510275China
| | - Feng Ju
- Westlake Intelligent Biomarker Discovery LabWestlake Laboratory of Life Sciences and BiomedicineHangzhou310024China
- Key Laboratory of Coastal Environment and Resources of Zhejiang ProvinceSchool of EngineeringWestlake UniversityHangzhou310030China
- Institute of Advanced TechnologyWestlake Institute for Advanced StudyHangzhou310024China
| | - Ju‐Sheng Zheng
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang ProvinceSchool of Life SciencesWestlake UniversityHangzhou310030China
- Westlake Intelligent Biomarker Discovery LabWestlake Laboratory of Life Sciences and BiomedicineHangzhou310024China
- Institute of Basic Medical SciencesWestlake Institute for Advanced StudyHangzhou310024China
| |
Collapse
|
11
|
Yu D, Meng X, de Vos WM, Wu H, Fang X, Maiti AK. Implications of Gut Microbiota in Complex Human Diseases. Int J Mol Sci 2021; 22:12661. [PMID: 34884466 PMCID: PMC8657718 DOI: 10.3390/ijms222312661] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 10/30/2021] [Accepted: 11/17/2021] [Indexed: 02/07/2023] Open
Abstract
Humans, throughout the life cycle, from birth to death, are accompanied by the presence of gut microbes. Environmental factors, lifestyle, age and other factors can affect the balance of intestinal microbiota and their impact on human health. A large amount of data show that dietary, prebiotics, antibiotics can regulate various diseases through gut microbes. In this review, we focus on the role of gut microbes in the development of metabolic, gastrointestinal, neurological, immune diseases and, cancer. We also discuss the interaction between gut microbes and the host with respect to their beneficial and harmful effects, including their metabolites, microbial enzymes, small molecules and inflammatory molecules. More specifically, we evaluate the potential ability of gut microbes to cure diseases through Fecal Microbial Transplantation (FMT), which is expected to become a new type of clinical strategy for the treatment of various diseases.
Collapse
Affiliation(s)
- Dahai Yu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China; (X.M.); (X.F.)
| | - Xin Meng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China; (X.M.); (X.F.)
| | - Willem M. de Vos
- Laboratory of Microbiology, Wageningen University, Dreijenplein 10, 6703 HB Wageningen, The Netherlands;
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland
| | - Hao Wu
- Vascular Biology Program, Department of Surgery, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02115, USA;
| | - Xuexun Fang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, School of Life Sciences, Jilin University, 2699 Qianjin Street, Changchun 130012, China; (X.M.); (X.F.)
| | - Amit K. Maiti
- Department of Genetics and Genomics, Mydnavar, 2645 Somerset Boulevard, Troy, MI 48084, USA
| |
Collapse
|
12
|
Wu L, Xie X, Li Y, Liang T, Zhong H, Ma J, Yang L, Yang J, Li L, Xi Y, Li H, Zhang J, Chen X, Ding Y, Wu Q. Metagenomics-Based Analysis of the Age-Related Cumulative Effect of Antibiotic Resistance Genes in Gut Microbiota. Antibiotics (Basel) 2021; 10:1006. [PMID: 34439056 PMCID: PMC8388928 DOI: 10.3390/antibiotics10081006] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 08/10/2021] [Accepted: 08/13/2021] [Indexed: 12/21/2022] Open
Abstract
Antibiotic resistance in bacteria has become a major global health problem. One of the main reservoirs of antibiotic resistance genes is the human gut microbiota. To characterise these genes, a metagenomic approach was used. In this study, a comprehensive antibiotic resistome catalog was established using fecal samples from 246 healthy individuals from world's longevity township in Jiaoling, China. In total, 606 antibiotic resistance genes were detected. Our results indicated that antibiotic resistance genes in the human gut microbiota accumulate and become more complex with age as older groups harbour the highest abundance of these genes. Tetracycline resistance gene type tetQ was the most abundant group of antibiotic resistance genes in gut microbiota, and the main carrier of antibiotic resistance genes was Bacteroides. Antibiotic efflux, inactivation, and target alteration were found to be the dominant antimicrobial resistance mechanisms. This research may help to establish a comprehensive antibiotic resistance catalog that includes extremely long-lived healthy people such as centenarians, and may provide potential recommendations for controlling the use of antibiotics.
Collapse
Affiliation(s)
- Lei Wu
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (L.W.); (T.L.); (J.M.); (J.Y.); (X.C.)
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Xinqiang Xie
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Ying Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Tingting Liang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (L.W.); (T.L.); (J.M.); (J.Y.); (X.C.)
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Haojie Zhong
- The First Affiliated Hospital, School of Clinical Medicine of Guangdong Pharmaceutical University, Guangzhou 510080, China;
| | - Jun Ma
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (L.W.); (T.L.); (J.M.); (J.Y.); (X.C.)
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Lingshuang Yang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Juan Yang
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (L.W.); (T.L.); (J.M.); (J.Y.); (X.C.)
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Longyan Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Yu Xi
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Haixin Li
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Jumei Zhang
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| | - Xuefeng Chen
- School of Food and Biological Engineering, Shaanxi University of Science and Technology, Xi’an 710021, China; (L.W.); (T.L.); (J.M.); (J.Y.); (X.C.)
| | - Yu Ding
- Department of Food Science and Technology, Institute of Food Safety and Nutrition, Jinan University, Guangzhou 510632, China
| | - Qingping Wu
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, Guangzhou 510070, China; (X.X.); (Y.L.); (L.Y.); (L.L.); (Y.X.); (H.L.); (J.Z.)
| |
Collapse
|
13
|
Li Z, Xia J, Jiang L, Tan Y, An Y, Zhu X, Ruan J, Chen Z, Zhen H, Ma Y, Jie Z, Xiao L, Yang H, Wang J, Kristiansen K, Xu X, Jin L, Nie C, Krutmann J, Liu X, Wang J. Characterization of the human skin resistome and identification of two microbiota cutotypes. MICROBIOME 2021; 9:47. [PMID: 33597039 PMCID: PMC7890624 DOI: 10.1186/s40168-020-00995-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Accepted: 12/29/2020] [Indexed: 05/04/2023]
Abstract
BACKGROUND The human skin microbiota is considered to be essential for skin homeostasis and barrier function. Comprehensive analyses of its function would substantially benefit from a catalog of reference genes derived from metagenomic sequencing. The existing catalog for the human skin microbiome is based on samples from limited individuals from a single cohort on reference genomes, which limits the coverage of global skin microbiome diversity. RESULTS In the present study, we have used shotgun metagenomics to newly sequence 822 skin samples from Han Chinese, which were subsequently combined with 538 previously sequenced North American samples to construct an integrated Human Skin Microbial Gene Catalog (iHSMGC). The iHSMGC comprised 10,930,638 genes with the detection of 4,879,024 new genes. Characterization of the human skin resistome based on iHSMGC confirmed that skin commensals, such as Staphylococcus spp, are an important reservoir of antibiotic resistance genes (ARGs). Further analyses of skin microbial ARGs detected microbe-specific and skin site-specific ARG signatures. Of note, the abundance of ARGs was significantly higher in Chinese than Americans, while multidrug-resistant bacteria ("superbugs") existed on the skin of both Americans and Chinese. A detailed analysis of microbial signatures identified Moraxella osloensis as a species specific for Chinese skin. Importantly, Moraxella osloensis proved to be a signature species for one of two robust patterns of microbial networks present on Chinese skin, with Cutibacterium acnes indicating the second one. Each of such "cutotypes" was associated with distinct patterns of data-driven marker genes, functional modules, and host skin properties. The two cutotypes markedly differed in functional modules related to their metabolic characteristics, indicating that host-dependent trophic chains might underlie their development. CONCLUSIONS The development of the iHSMGC will facilitate further studies on the human skin microbiome. In the present study, it was used to further characterize the human skin resistome. It also allowed to discover the existence of two cutotypes on the human skin. The latter finding will contribute to a better understanding of the interpersonal complexity of the skin microbiome. Video abstract.
Collapse
Affiliation(s)
- Zhiming Li
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
| | - Jingjing Xia
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
| | - Liuyiqi Jiang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Yimei Tan
- Human Phenome Institute, Fudan University, Shanghai, China
- Department of Skin & Cosmetic Research, Shanghai Skin Disease Hospital, Shanghai, China
| | - Yitai An
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
| | - Xingyu Zhu
- Human Phenome Institute, Fudan University, Shanghai, China
- Institute for Six-sector Economy, Fudan University, Shanghai, China
| | - Jie Ruan
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
| | - Zhihua Chen
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
| | - Hefu Zhen
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
| | - Yanyun Ma
- Human Phenome Institute, Fudan University, Shanghai, China
- Institute for Six-sector Economy, Fudan University, Shanghai, China
| | - Zhuye Jie
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
| | - Liang Xiao
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
| | - Huanming Yang
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
| | - Jian Wang
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
| | - Karsten Kristiansen
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
- Laboratory of Genomics and Molecular Biomedicine, Department of Biology, University of Copenhagen, Copenhagen, Denmark
| | - Xun Xu
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, Shenzhen, China
| | - Li Jin
- Human Phenome Institute, Fudan University, Shanghai, China
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai, China
| | - Chao Nie
- BGI-Shenzhen, Shenzhen, China
- China National Genebank, Shenzhen, China
| | - Jean Krutmann
- Human Phenome Institute, Fudan University, Shanghai, China
- IUF-Leibniz Research Institute for Environmental Medicine, Düsseldorf, Germany
- Medical Faculty, Heinrich-Heine University, Düsseldorf, Germany
| | - Xiao Liu
- BGI-Shenzhen, Shenzhen, China
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
- BGI Education Center, University of Chinese Academy of Sciences, Shenzhen, China
| | - Jiucun Wang
- State Key Laboratory of Genetic Engineering, Collaborative Innovation Center for Genetics and Development, School of Life Sciences, and Department of Dermatology, Huashan Hospital, Fudan University, Shanghai, China
- Human Phenome Institute, Fudan University, Shanghai, China
- Research Unit of Dissecting the Population Genetics and Developing New Technologies for Treatment and Prevention of Dermatological Diseases (2019RU058), Chinese Academy of Medical Sciences, Shanghai, China
- Institute of Rheumatology, Immunology and Allergy, Fudan University, Shanghai, China
| |
Collapse
|
14
|
Li J, Si H, Du H, Guo H, Dai H, Xu S, Wan J. Comparison of gut microbiota structure and Actinobacteria abundances in healthy young adults and elderly subjects: a pilot study. BMC Microbiol 2021; 21:13. [PMID: 33407122 PMCID: PMC7788997 DOI: 10.1186/s12866-020-02068-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 12/10/2020] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND The aim was to determine the potential association of the gut microbiota composition, especially the abundance of Actinobacteria, as well as the differentiation of functional and resistance genes with age (young adults vs elderly subjects) in China. RESULTS The patterns of relative abundance of all bacteria isolated from fecal samples differed between young adults and elderly subjects, but the alpha diversity (Chao1 P = 0.370, Shannon P = 0.560 and Simpson P = 0.270) and beta diversity (ANOSIM R = 0.031, P = 0.226) were not significantly different. There were 3 Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolic pathways (carbon metabolism, inositol phosphate metabolism, and sesquiterpenoid and triterpenoid biosynthesis) and 7 antibiotic resistant genes (ARGs) (macrolide lincosamide-streptogramin B (MLSB), tetracycline, aminoglycoside, sulfonamide, fosmidomycin, lincomycin, and vancomycin) that showed significant differences between the 2 groups (all P < 0.05). The abundance of Actinomycetes was enriched (about 2.4-fold) in young adults. Bifidobacteria dominated in both young adults and elderly subjects, with overall higher abundances in young adults (P > 0.05). Only the Bifidobacterium_dentium species showed significant differences between the 2 groups (P = 0.013), with a higher abundance in elderly subjects but absent in young adults. CONCLUSIONS The present study revealed that there were 3 KEGG metabolic pathways and 7 ARGs as well as enhanced Bifidobacterium_dentium species abundance in elderly compared to young subjects.
Collapse
Affiliation(s)
- Jun Li
- Department of Gastroenterology, 2nd Medical Center, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Haiyan Si
- Department of Oncology, First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Haitao Du
- Department of Gastroenterology, 2nd Medical Center, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing, 100853, China
| | - Hongxia Guo
- Department of Oncology, First Medical Center, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Huanqin Dai
- Chinese Academy Sciences Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing, China
| | - Shiping Xu
- Department of Gastroenterology, 2nd Medical Center, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing, 100853, China.
| | - Jun Wan
- Department of Gastroenterology, 2nd Medical Center, Chinese People's Liberation Army General Hospital, No. 28 Fuxing Road, Beijing, 100853, China.
| |
Collapse
|
15
|
Zhu Z, Cao M, Wang W, Zhang L, Ma T, Liu G, Zhang Y, Shang Z, Chen X, Shi Y, Zhang J. Exploring the Prevalence and Distribution Patterns of Antibiotic Resistance Genes in Bovine Gut Microbiota Using a Metagenomic Approach. Microb Drug Resist 2020; 27:980-990. [PMID: 33395552 DOI: 10.1089/mdr.2020.0271] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Antibiotic resistance genes (ARGs) have become recognized contaminants and pose a high public health risk. The animal gut microbiota is a reservoir of ARGs, but the knowledge of the origin and dissemination of ARGs remains unclear. In this study, we provide a comprehensive profile of ARGs and mobile genetic elements in the gut microbiota from 30 bovines to study the impact of modern antibiotics on resistance. A total of 42 ARG types were detected by annotating the metagenomic sequencing data from Comprehensive Antibiotic Resistance Database (CARD). We found that the diversity and abundance of ARGs in individual yaks were significantly lower than those in dairy and beef cattle (p < 0.0001). The results of heat map and single nucleotide polymorphism clustering suggest that ARGs from dairy and beef cattle are more similar, whereas those from yaks cluster separately. The long-term use of antibiotics may contribute to this difference, suggesting that antibiotic consumption is the main cause of ARG prevalence. Furthermore, abundant insertions were also found in this study, signifying a strong potential for horizontal transfer of ARGs among microbes, especially pathogens.
Collapse
Affiliation(s)
- Zhen Zhu
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of the Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China.,College of Life Science and Food Engineering, Hebei University of Engineering, Handan, China
| | - Mingze Cao
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan, China
| | - Weiwei Wang
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of the Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| | - Liwei Zhang
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan, China
| | - Tenghe Ma
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan, China
| | - Guanhui Liu
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan, China
| | - Yue Zhang
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan, China
| | - Zixuang Shang
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of the Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| | - Xu Chen
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan, China
| | - Yuxiang Shi
- College of Life Science and Food Engineering, Hebei University of Engineering, Handan, China
| | - Jiyu Zhang
- Key Laboratory of New Animal Drug Project of Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development of the Ministry of Agriculture, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of CAAS, Lanzhou, China
| |
Collapse
|
16
|
Qiu Q, Wang J, Yan Y, Roy B, Chen Y, Shang X, Dou T, Han L. Metagenomic Analysis Reveals the Distribution of Antibiotic Resistance Genes in a Large-Scale Population of Healthy Individuals and Patients With Varied Diseases. Front Mol Biosci 2020; 7:590018. [PMID: 33330625 PMCID: PMC7673455 DOI: 10.3389/fmolb.2020.590018] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 10/12/2020] [Indexed: 01/12/2023] Open
Abstract
The human gut microbiome is a reservoir for antibiotic resistance gene (ARG). Therefore, characterizing resistome distribution and potential disease markers can help manage antibiotics at the clinical level. While much population-level research has highlighted the strong effect of donor geographic origin on ARG prevalence in the human gut, little is known regarding the effects of other properties, such as age, sex, and disease. Here we employed 2,037 fecal metagenomes from 12 countries. By quantifying the known resistance genes for 24 types of antibiotics in each community, we showed that tetracycline, aminoglycoside, beta-lactam, macrolide-lincosamide-streptogramin (MLS), and vancomycin resistance genes were the dominant ARG types in the human gut. We then compared the ARG profiles of 1427 healthy individuals from the 2,037 samples and observed significant differences across countries. This was consistent with expectations that regional antibiotic usage and exposure in medical and food production contexts affect distribution. Although no specific uniform pattern of ARG was observed, a significant increase in resistance potential among multiple disease groups implied that the disease condition may be another source of ARG variance. In particular, the co-occurrence pattern of some enriched bacterial species and ARGs that were obtained in type 2 diabetes (T2D) and liver cirrhosis patients implied that some disease-associated species may be potential hosts of enriched ARGs, which could be potential biomarkers for the prediction and intervention of such diseases. Overall, our study identifies factors associated with the human gut resistome, including substantial effects of region and heterogeneous effects of disease status, and highlights the value of ARG analysis in disease research and clinical applications.
Collapse
Affiliation(s)
- Qinwei Qiu
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Jingjing Wang
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China.,Department of Scientific Research, KMHD, Shenzhen, China
| | - Yuhong Yan
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | | | - Yang Chen
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Xiaoxiao Shang
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Tongyi Dou
- School of Life and Pharmaceutical Sciences, Dalian University of Technology, Panjin, China
| | - Lijuan Han
- State Key Laboratory of Dampness Syndrome of Chinese Medicine, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China.,Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
17
|
Ho J, Yeoh YK, Barua N, Chen Z, Lui G, Wong SH, Yang X, Chan MCW, Chan PKS, Hawkey PM, Ip M. Systematic review of human gut resistome studies revealed variable definitions and approaches. Gut Microbes 2020; 12:1700755. [PMID: 31942825 PMCID: PMC7524153 DOI: 10.1080/19490976.2019.1700755] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
In this review, we highlight the variations of gut resistome studies, which may preclude comparisons and translational interpretations. Of 22 included studies, a range of 12 to 2000 antibiotic resistance (AR) genes were profiled. Overall, studies defined a healthy gut resistome as subjects who had not taken antibiotics in the last three to 12 months prior to sampling. In studies with de novo assembly, AR genes were identified based on variable nucleotide or amino acid sequence similarities. Different marker genes were used for defining resistance to a given antibiotic class. Validation of phenotypic resistance in the laboratory is frequently lacking. Cryptic resistance, collateral sensitivity and the interaction with repressors or promotors were not investigated. International consensus is needed for selecting marker genes to define resistance to a given antibiotic class in addition to uniformity in phenotypic validation and bioinformatics pipelines.
Collapse
Affiliation(s)
- Jeffery Ho
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China,Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China,Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yun Kit Yeoh
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China,Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China,Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Nilakshi Barua
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Zigui Chen
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China,Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China,Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Grace Lui
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China,Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China,Department of Medicine & Therapeutics, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Sunny H Wong
- Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China,Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China,Department of Medicine & Therapeutics, Faculty of Medicine, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao Yang
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Martin CW Chan
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Paul KS Chan
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China,Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China,Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Peter M Hawkey
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Margaret Ip
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong SAR, China,Centre for Gut Microbiota Research, The Chinese University of Hong Kong, Hong Kong SAR, China,Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China,CONTACT Margaret Ip Department of Microbiology, Chinese University of Hong Kong, Prince of Wales Hospital, Shatin, Hong Kong, China
| |
Collapse
|
18
|
Meng X, Zhang G, Cao H, Yu D, Fang X, de Vos WM, Wu H. Gut dysbacteriosis and intestinal disease: mechanism and treatment. J Appl Microbiol 2020; 129:787-805. [PMID: 32277534 PMCID: PMC11027427 DOI: 10.1111/jam.14661] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/14/2020] [Accepted: 03/30/2020] [Indexed: 12/13/2022]
Abstract
The gut microbiome functions like an endocrine organ, generating bioactive metabolites, enzymes or small molecules that can impact host physiology. Gut dysbacteriosis is associated with many intestinal diseases including (but not limited to) inflammatory bowel disease, primary sclerosing cholangitis-IBD, irritable bowel syndrome, chronic constipation, osmotic diarrhoea and colorectal cancer. The potential pathogenic mechanism of gut dysbacteriosis associated with intestinal diseases includes the alteration of composition of gut microbiota as well as the gut microbiota-derived signalling molecules. The many correlations between the latter and the susceptibility for intestinal diseases has placed a spotlight on the gut microbiome as a potential novel target for therapeutics. Currently, faecal microbial transplantation, dietary interventions, use of probiotics, prebiotics and drugs are the major therapeutic tools utilized to impact dysbacteriosis and associated intestinal diseases. In this review, we systematically summarized the role of intestinal microbiome in the occurrence and development of intestinal diseases. The potential mechanism of the complex interplay between gut dysbacteriosis and intestinal diseases, and the treatment methods are also highlighted.
Collapse
Affiliation(s)
- X Meng
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Science, Jilin University, Changchun, PR China
| | - G Zhang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Science, Jilin University, Changchun, PR China
| | - H Cao
- InnovHope Inc, Framingham, MA, USA
| | - D Yu
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Science, Jilin University, Changchun, PR China
| | - X Fang
- Key Laboratory for Molecular Enzymology and Engineering of Ministry of Education, College of Life Science, Jilin University, Changchun, PR China
| | - W M de Vos
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - H Wu
- Vascular Biology Program, Department of Surgery, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
19
|
Korry BJ, Cabral DJ, Belenky P. Metatranscriptomics Reveals Antibiotic-Induced Resistance Gene Expression in the Murine Gut Microbiota. Front Microbiol 2020; 11:322. [PMID: 32210932 PMCID: PMC7069102 DOI: 10.3389/fmicb.2020.00322] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/13/2020] [Indexed: 12/25/2022] Open
Abstract
Antibiotic resistance is a current and expanding threat to the practice of modern medicine. Antibiotic therapy has been shown to perturb the composition of the host microbiome with significant health consequences. In addition, the gut microbiome is known to be a reservoir of antibiotic resistance genes. Work has demonstrated that antibiotics can alter the collection of antibiotic resistance genes within the microbiome through selection and horizontal gene transfer. While antibiotics also have the potential to impact the expression of resistance genes, metagenomic-based pipelines currently lack the ability to detect these shifts. Here, we utilized a dual sequencing approach combining shotgun metagenomics and metatranscriptomics to profile how three antibiotics, amoxicillin, doxycycline, and ciprofloxacin, impact the murine gut resistome at the DNA and RNA level. We found that each antibiotic induced broad, but untargeted impacts on the gene content of the resistome. In contrast, changes in ARG transcript abundance were more targeted to the antibiotic treatment. Doxycycline and amoxicillin induced the expression of tetracycline and beta-lactamase resistance genes, respectively. Furthermore, the increased beta-lactamase resistance gene transcripts could contribute to an observed bloom of Bacteroides thetaiotaomicron during amoxicillin treatment. Based on these findings, we propose that the utilization of a dual sequencing methodology provides a unique capacity to fully understand the response of the resistome to antibiotic perturbation. In particular, the analysis of transcripts reveals that the expression and utilization of resistance genes is far narrower than their abundance at the genomic level would suggest.
Collapse
Affiliation(s)
- Benjamin J Korry
- Department of Molecular Microbiology and Immunology, Division of Biology and Medicine, Brown University, Providence, RI, United States
| | - Damien J Cabral
- Department of Molecular Microbiology and Immunology, Division of Biology and Medicine, Brown University, Providence, RI, United States
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Division of Biology and Medicine, Brown University, Providence, RI, United States
| |
Collapse
|
20
|
Duan Y, Chen Z, Tan L, Wang X, Xue Y, Wang S, Wang Q, Das R, Lin H, Hou J, Li L, Mao D, Luo Y. Gut resistomes, microbiota and antibiotic residues in Chinese patients undergoing antibiotic administration and healthy individuals. THE SCIENCE OF THE TOTAL ENVIRONMENT 2020; 705:135674. [PMID: 31785918 DOI: 10.1016/j.scitotenv.2019.135674] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2019] [Revised: 11/18/2019] [Accepted: 11/19/2019] [Indexed: 05/21/2023]
Abstract
Human gut microbiota is an important reservoir of antibiotic resistance genes (ARGs). Although dysbacteriosis after the antibiotic course has been previously observed in the patient guts, a comprehensive comparison of gut resistomes, microbiota and antibiotic residues in healthy individuals and patients undergoing antibiotic administration is little. Using high-throughput qPCR, 16S rRNA gene amplicon sequencing and UPLC-MS/MS, we systematically examined the antibiotic resistome, gut microbiota, and antibiotic residues in fecal samples from both Chinese healthy individuals and patients receiving antibiotic therapy. Compared with healthy individuals, patients' guts harbored lower diverse gut resistome and microbiota, but higher concentrations of antibiotics and ARGs. Antibiotic concentration in human guts was positively correlated with ARG total abundance, but was negatively related to the diversity of both ARGs and bacterial communities, which demonstrated that antibiotic administration could shape the antibiotic resistomes and bacterial communities in the patient guts. Gene cfxA was evaluated as a potential biomarker to distinguish the patients receiving antibiotic therapy from the healthy individuals in China since its wide detection and significant enrichment in the guts of the patients. The detection of some veterinary antibiotics in human guts illustrated the potential transmission of antibiotic from the external environment to human via the food chain. The obtained results could help to better understand the influence of antibiotic therapy in shaping antibiotic reistomes and bacterial communities in Chinese individuals.
Collapse
Affiliation(s)
- Yujing Duan
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Zeyou Chen
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Lu Tan
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Xiaolong Wang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Yingang Xue
- Key Laboratory of Environmental Protection of Water Environment Biological Monitoring of Jiangsu Province, Changzhou Environmental Monitoring Center, Changzhou 213001, China
| | - Shaopeng Wang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Qing Wang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Ranjit Das
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Huai Lin
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Jie Hou
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Linyun Li
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Daqing Mao
- School of Medicine, Nankai University, Tianjin 300071, China.
| | - Yi Luo
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China.
| |
Collapse
|
21
|
Selvin J, Maity D, Sajayan A, Kiran GS. Revealing antibiotic resistance in therapeutic and dietary probiotic supplements. J Glob Antimicrob Resist 2020; 22:202-205. [PMID: 32084605 DOI: 10.1016/j.jgar.2020.02.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 02/03/2020] [Accepted: 02/10/2020] [Indexed: 11/30/2022] Open
Abstract
OBJECTIVES The aim of this study was to evaluate antibiotic susceptibility patterns in commercially available dietary and probiotic supplements. METHODS Probiotic strains were isolated from the dietary supplements (designated as sample B, D and V), and multidrug resistance profiles were tested using the Kirby-Bauer test. Minimum inhibitory concentrations and double-disk synergy tests were performed to detect the mechanism of action of the resistance, and the presence of extended spectrum β-lactamase activity (ESBL) was confirmed. RESULTS The isolates Streptococcus faecalis and Bacillus mesentericus (both from sample B) were found to be resistant to penicillin G, Lactobacillus acidophilus (sample D) was resistant to ampicillin, and all the isolates from samples B, D and V were resistant to ceftazidime. The isolates Lactobacillus sporogenes, S. faecalis, B. mesentericus from sample B, Lactobacillus. rhamnosus, Saccharomyces boulardii from sample D, and L. sporogenes (sample V) were resistant to erythromycin. CONCLUSIONS The findings showed the presence of antibiotic resistance in probiotic bacteria isolated from commercially available dietary supplements. Because multidrug resistance is a serious emerging issue, and the risk of drug-resistant gene transfer to commensals or pathogens of the gut is inevitable, the safety of probiotics has become a major criterion of interest. The findings of this study would serve as a platform for further screening and characterization of the determinants of antibiotic resistance and the genetic mechanisms of resistance.
Collapse
Affiliation(s)
- Joseph Selvin
- Department of Microbiology, Pondicherry University, Puducherry, India
| | - Debanjana Maity
- KIIT school of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, India
| | - Arya Sajayan
- Department of Food Science and Technology, Pondicherry University, Puducherry, India
| | - George Seghal Kiran
- Department of Food Science and Technology, Pondicherry University, Puducherry, India.
| |
Collapse
|
22
|
Wang Q, Duan YJ, Wang SP, Wang LT, Hou ZL, Cui YX, Hou J, Das R, Mao DQ, Luo Y. Occurrence and distribution of clinical and veterinary antibiotics in the faeces of a Chinese population. JOURNAL OF HAZARDOUS MATERIALS 2020; 383:121129. [PMID: 31546217 DOI: 10.1016/j.jhazmat.2019.121129] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/26/2019] [Accepted: 08/30/2019] [Indexed: 05/21/2023]
Abstract
Antibiotics ingested in the human gut may create selective pressure to change the composition of the gut microbiota, which could adversely effect the immune system of the host. However, the occurrence and distribution of antibiotics in the human gut remains unclear. A total population of 180 individuals, across three Chinses regions with different economic development levels, including children, adults, and elders, were sampled in 2017. A total of 19 representative antibiotics, including both clinical and veterinary antibiotics, were investigated in human faeces. While clinical use and prescriptions were the main exposure pathways for children, environmental media were the exposure pathway to adults. In addition, significant differences (P < 0.05) in antibiotic residues in human faeces were observed amongst various economic development levels, where human faeces from underdeveloped areas were mostly associated with higher levels of antibiotics. This study first to investigate the occurrence and distribution of typical antibiotics in the faeces of a Chinese population and thereby provide a reference for the intensive study of the effects and mechanisms of antibiotics on human gut microbiota.
Collapse
Affiliation(s)
- Qing Wang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, 300071, China; Hebei Key Laboratory of Air Pollution Cause and Impact (preparatory), College of Energy and Environmental Engineering, Hebei University of Engineering, Handan, 056038, China
| | - Yu-Jing Duan
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Shao-Peng Wang
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Li-Tao Wang
- Hebei Key Laboratory of Air Pollution Cause and Impact (preparatory), College of Energy and Environmental Engineering, Hebei University of Engineering, Handan, 056038, China
| | - Ze-Lin Hou
- Hebei Key Laboratory of Air Pollution Cause and Impact (preparatory), College of Energy and Environmental Engineering, Hebei University of Engineering, Handan, 056038, China
| | - Yu-Xiao Cui
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Jie Hou
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Ranjit Das
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, 300071, China
| | - Da-Qing Mao
- School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yi Luo
- Ministry of Education Key Laboratory of Pollution Processes and Environmental Criteria, College of Environmental Science and Engineering, Nankai University, Tianjin, 300071, China.
| |
Collapse
|
23
|
Fan P, Nelson CD, Driver JD, Elzo MA, Jeong KC. Animal Breed Composition Is Associated With the Hindgut Microbiota Structure and β-Lactam Resistance in the Multibreed Angus-Brahman Herd. Front Microbiol 2019; 10:1846. [PMID: 31456774 PMCID: PMC6700273 DOI: 10.3389/fmicb.2019.01846] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Accepted: 07/26/2019] [Indexed: 01/03/2023] Open
Abstract
Antibiotics have been widely used in livestock to treat and prevent bacterial diseases. However, use of antibiotics has led to the emergence of antibiotic resistant microorganisms (ARMs) in food animals. Due to the decreased efficacy of antibiotics, alternatives to antibiotics that can reduce infectious diseases in food animals to enhance animal health and growth performance are urgently required. Here, we show that animal genetics is associated with the hindgut microbiome, which is related to fat deposition and beta-lactam resistance in the gastrointestinal tract. We investigated the hindgut microbiota structure in 95 postweaning heifers belonging to the unique multibreed Angus-Brahman herd with breed composition ranging from 100% Angus to 100% Brahman. The hindgut microbial composition of postweaning heifers differed among breed groups. The mucin-degrading bacterium Akkermansia known for promoting energy expenditure was enriched in Brahman calves that contained less intramuscular fat content, while butyrate-producing bacterium Faecalibacterium was linearly positively correlated with Angus proportion. Moreover, the higher relative abundance of beta-lactam resistant genes including ampC gene and arcA gene was associated with the greater Brahman proportion. As the first study aimed at understanding changes in hindgut microbiota among beef cattle with linear gradient of breed composition and its association with marbling in meat, our results suggest that the effects of animal genetics on the gut microbiota structure is associated with fat deposition and potentially a factor affecting the gut antimicrobial resistance.
Collapse
Affiliation(s)
- Peixin Fan
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Corwin D Nelson
- Department of Animal Sciences, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - J Danny Driver
- Department of Animal Sciences, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Mauricio A Elzo
- Department of Animal Sciences, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| | - Kwangcheol Casey Jeong
- Emerging Pathogens Institute, University of Florida, Gainesville, FL, United States.,Department of Animal Sciences, Institute of Food and Agricultural Sciences, University of Florida, Gainesville, FL, United States
| |
Collapse
|
24
|
McConnell MJ. Where are we with monoclonal antibodies for multidrug-resistant infections? Drug Discov Today 2019; 24:1132-1138. [PMID: 30853568 DOI: 10.1016/j.drudis.2019.03.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 01/21/2019] [Accepted: 03/01/2019] [Indexed: 01/17/2023]
Abstract
Widespread antibiotic resistance threatens the continued efficacy of antimicrobial therapy based on small-molecule antibiotics. Infections caused by multidrug-resistant Gram-negative bacteria are particularly worrisome owing to the lack of antimicrobials retaining sufficient activity against these microorganisms. Despite the explosion in monoclonal antibody therapies that have been developed for oncologic and rheumatic indications, only three antibacterial monoclonal antibodies have been approved for clinical use. In the present review, the therapeutic potential of this drug class for treating multidrug-resistant infections is discussed, and considerations for the development of antibacterial monoclonal antibodies are presented. Finally, the state of development of monoclonal antibody therapies for some of the most problematic multidrug-resistant Gram-negative infections is summarized.
Collapse
Affiliation(s)
- Michael J McConnell
- Antimicrobial Resistance and Hospital Acquired Infections Reference Laboratory, National Centre for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain.
| |
Collapse
|
25
|
Microbiome as a tool and a target in the effort to address antimicrobial resistance. Proc Natl Acad Sci U S A 2019; 115:12902-12910. [PMID: 30559176 DOI: 10.1073/pnas.1717163115] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Reciprocal, intimate relationships between the human microbiome and the host immune system are shaped by past microbial encounters and prepare the host for future ones. Antibiotics and other antimicrobials leave their mark on both the microbiome and host immunity. Antimicrobials alter the structure of the microbiota, expand the host-specific pool of antimicrobial-resistance genes and organisms, degrade the protective effects of the microbiota against invasion by pathogens, and may impair vaccine efficacy. Through these effects on the microbiome they may affect immune responses. Vaccines that exert protective or therapeutic effects against pathogens may reduce the use of antimicrobials, the development and spread of antimicrobial resistance, and the harmful impacts of these drugs on the microbiome. Other strategies involving manipulation of the microbiome to deplete antibiotic-resistant organisms or to enhance immune responses to vaccines may prove valuable in addressing antimicrobial resistance as well. This article describes the intersections of immunity, microbiome and antimicrobial exposure, and the use of vaccines and other alternative strategies for the control and management of antimicrobial resistance.
Collapse
|
26
|
Contributions and Challenges of High Throughput qPCR for Determining Antimicrobial Resistance in the Environment: A Critical Review. Molecules 2019; 24:molecules24010163. [PMID: 30609875 PMCID: PMC6337382 DOI: 10.3390/molecules24010163] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 12/28/2018] [Accepted: 12/29/2018] [Indexed: 12/12/2022] Open
Abstract
Expansion in whole genome sequencing and subsequent increase in antibiotic resistance targets have paved the way of high throughput qPCR (HT-qPCR) for analyzing hundreds of antimicrobial resistance genes (ARGs) in a single run. A meta-analysis of 51 selected studies is performed to evaluate ARGs abundance trends over the last 7 years. WaferGenTM SmartChip is found to be the most widely used HT-qPCR platform among others for evaluating ARGs. Up till now around 1000 environmental samples (excluding biological replicates) from different parts of the world have been analyzed on HT-qPCR. Calculated detection frequency and normalized ARGs abundance (ARGs/16S rRNA gene) reported in gut microbiome studies have shown a trend of low ARGs as compared to other environmental matrices. Disparities in the HT-qPCR data analysis which are causing difficulties to researchers in precise interpretation of results have been highlighted and a possible way forward for resolving them is also suggested. The potential of other amplification technologies and point of care or field deployable devices for analyzing ARGs have also been discussed in the review. Our review has focused on updated information regarding the role, current status and future perspectives of HT-qPCR in the field of antimicrobial resistance.
Collapse
|
27
|
Gut Bacterial Microbiota and its Resistome Rapidly Recover to Basal State Levels after Short-term Amoxicillin-Clavulanic Acid Treatment in Healthy Adults. Sci Rep 2018; 8:11192. [PMID: 30046129 PMCID: PMC6060159 DOI: 10.1038/s41598-018-29229-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 06/29/2018] [Indexed: 12/23/2022] Open
Abstract
Clinical effects of antimicrobials and probiotics in combination have been reported, however, little is known about their impact on gut microbiota and its resistome. In this study 16S rRNA gene amplicon, shotgun metagenomics sequencing and antibiotic resistance (ABR) microarray were used on fecal samples of 70 healthy participants, taken at four time points in probiotic (Lactobacillus rhamnosus R0011 and Lactobacillus helveticus R0052) and placebo groups to profile the gut bacterial microbiota and its resistome following administration of amoxicillin-clavulanic acid for one week. Significant shifts in microbiota family composition caused by the antimicrobial in both groups that included decreases in the proportion of Lachnospiraceae, Coriobacteriaceae and unidentified Clostridiales; and notable increases for the proportion of Enterobacteriaceae, Bacteroidaceae and Porphyromonadaceae compared to baseline levels. Resistome showed a corresponding enrichment of ABR genes compared to baseline from such classes as aminoglycosides and beta-lactams that were linked, by in silico inference, to the enrichment of the family Enterobacteriaceae. Despite perturbations caused by short-term antibiotic treatment, both gut microbiota and resistome showed prompt recovery to baseline levels one week after cessation of the antimicrobial. This rapid recovery may be explained by the hypothesis of community resilience.
Collapse
|
28
|
Häsler R, Kautz C, Rehman A, Podschun R, Gassling V, Brzoska P, Sherlock J, Gräsner JT, Hoppenstedt G, Schubert S, Ferlinz A, Lieb W, Laudes M, Heinsen FA, Scholz J, Harmsen D, Franke A, Eisend S, Kunze T, Fickenscher H, Ott S, Rosenstiel P, Schreiber S. The antibiotic resistome and microbiota landscape of refugees from Syria, Iraq and Afghanistan in Germany. MICROBIOME 2018; 6:37. [PMID: 29458422 PMCID: PMC5819293 DOI: 10.1186/s40168-018-0414-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 01/24/2018] [Indexed: 05/12/2023]
Abstract
BACKGROUND Multidrug-resistant bacteria represent a substantial global burden for human health, potentially fuelled by migration waves: in 2015, 476,649 refugees applied for asylum in Germany mostly as a result of the Syrian crisis. In Arabic countries, multiresistant bacteria cause significant problems for healthcare systems. Currently, no data exist describing antibiotic resistances in healthy refugees. Here, we assess the microbial landscape and presence of antibiotic resistance genes (ARGs) in refugees and German controls. To achieve this, a systematic study was conducted in 500 consecutive refugees, mainly from Syria, Iraq, and Afghanistan and 100 German controls. Stool samples were subjected to PCR-based quantification of 42 most relevant ARGs, 16S ribosomal RNA gene sequencing-based microbiota analysis, and culture-based validation of multidrug-resistant microorganisms. RESULTS The fecal microbiota of refugees is substantially different from that of resident Germans. Three categories of resistance profiles were found: (i) ARGs independent of geographic origin of individuals comprising BIL/LAT/CMA, ErmB, and mefE; (ii) vanB with a high prevalence in Germany; and (iii) ARGs showing substantially increased prevalences in refugees comprising CTX-M group 1, SHV, vanC1, OXA-1, and QnrB. The majority of refugees carried five or more ARGs while the majority of German controls carried three or less ARGs, although the observed ARGs occurred independent of signatures of potential pathogens. CONCLUSIONS Our results, for the first time, assess antibiotic resistance genes in refugees and demonstrate a substantially increased prevalence for most resistances compared to German controls. The antibiotic resistome in refugees may thus require particular attention in the healthcare system of host countries.
Collapse
Affiliation(s)
- Robert Häsler
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Christian Albrecht University of Kiel, Campus Kiel, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany
| | - Christian Kautz
- Pharmaceutical Institute, Department of Clinical Pharmacy, Christian Albrecht University of Kiel, Kiel, Germany
| | - Ateequr Rehman
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Christian Albrecht University of Kiel, Campus Kiel, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany
| | - Rainer Podschun
- Institute of Infection Medicine, Christian Albrecht University of Kiel, Kiel, Germany
| | - Volker Gassling
- Department of Oral and Maxillofacial Surgery, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Pius Brzoska
- Thermo Fisher Scientific, San Francisco, CA, USA
| | - Jon Sherlock
- Thermo Fisher Scientific, San Francisco, CA, USA
| | - Jan-Thorsten Gräsner
- Department of Anaesthesia and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Gesine Hoppenstedt
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Christian Albrecht University of Kiel, Campus Kiel, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany
| | - Sabine Schubert
- Institute of Infection Medicine, Christian Albrecht University of Kiel, Kiel, Germany
| | - Astrid Ferlinz
- Thermo Fisher Scientific, Life Technologies GmbH, Darmstadt, Germany
| | - Wolfgang Lieb
- POPGEN Biobank and Institute of Epidemiology, Christian Albrecht University of Kiel, Kiel, Germany
| | - Matthias Laudes
- POPGEN Biobank and Institute of Epidemiology, Christian Albrecht University of Kiel, Kiel, Germany
| | - Femke-Anouska Heinsen
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Christian Albrecht University of Kiel, Campus Kiel, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany
| | - Jens Scholz
- Department of Anaesthesia and Intensive Care Medicine, University Hospital of Schleswig-Holstein, Kiel, Germany
| | - Dag Harmsen
- Department of Periodontology and Restorative Dentistry, University Hospital Münster, Münster, Germany
| | - Andre Franke
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Christian Albrecht University of Kiel, Campus Kiel, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany
| | - Swantje Eisend
- Department of Internal Medicine, University Hospital of Schleswig-Holstein, Campus Kiel, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany
| | - Thomas Kunze
- Pharmaceutical Institute, Department of Clinical Pharmacy, Christian Albrecht University of Kiel, Kiel, Germany
| | - Helmut Fickenscher
- Institute of Infection Medicine, Christian Albrecht University of Kiel, Kiel, Germany
| | - Stephan Ott
- Department of Internal Medicine, University Hospital of Schleswig-Holstein, Campus Kiel, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Christian Albrecht University of Kiel, Campus Kiel, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany.
| | - Stefan Schreiber
- Institute of Clinical Molecular Biology, University Hospital Schleswig-Holstein, Christian Albrecht University of Kiel, Campus Kiel, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany.
- Department of Internal Medicine, University Hospital of Schleswig-Holstein, Campus Kiel, Rosalind-Franklin-Straße 12, 24105, Kiel, Germany.
| |
Collapse
|
29
|
Llop P, Latorre A, Moya A. Experimental Epidemiology of Antibiotic Resistance: Looking for an Appropriate Animal Model System. Microbiol Spectr 2018; 6. [PMID: 29637886 DOI: 10.1128/microbiolspec.mtbp-0007-2016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Antibiotic resistance is recognized as one of the major challenges in public health. The global spread of antibiotic resistance is the consequence of a constant flow of information across multi-hierarchical interactions, involving cellular (clones), subcellular (resistance genes located in plasmids, transposons, and integrons), and supracellular (clonal complexes, genetic exchange communities, and microbiotic ensembles) levels. In order to study such multilevel complexity, we propose to establish an experimental epidemiology model for the transmission of antibiotic resistance with the cockroach Blatella germanica. This paper reports the results of five types of preliminary experiments with B. germanica populations that allow us to conclude that this animal is an appropriate model for experimental epidemiology: (i) the composition, transmission, and acquisition of gut microbiota and endosymbionts; (ii) the effect of different diets on gut microbiota; (iii) the effect of antibiotics on host fitness; (iv) the evaluation of the presence of antibiotic resistance genes in natural- and lab-reared populations; and (v) the preparation of plasmids harboring specific antibiotic resistance genes. The basic idea is to have populations with higher and lower antibiotic exposure, simulating the hospital and the community, respectively, and with a certain migration rate of insects between populations. In parallel, we present a computational model based on P-membrane computing that will mimic the experimental system of antibiotic resistance transmission. The proposal serves as a proof of concept for the development of more-complex population dynamics of antibiotic resistance transmission that are of interest in public health, which can help us evaluate procedures and design appropriate interventions in epidemiology.
Collapse
Affiliation(s)
- Pablo Llop
- Foundation for the Promotion of Sanitary and Biomedical Research in the Valencian Region (FISABIO), València, Spain
| | - Amparo Latorre
- Foundation for the Promotion of Sanitary and Biomedical Research in the Valencian Region (FISABIO), València, Spain
- Integrative Systems Biology Institute, Universitat de València, València, Spain
- Network Research Center for Epidemiology and Public Health (CIBERESP), Madrid, Spain
| | - Andrés Moya
- Foundation for the Promotion of Sanitary and Biomedical Research in the Valencian Region (FISABIO), València, Spain
- Integrative Systems Biology Institute, Universitat de València, València, Spain
- Network Research Center for Epidemiology and Public Health (CIBERESP), Madrid, Spain
| |
Collapse
|
30
|
Munir A, Waseem H, Williams MR, Stedtfeld RD, Gulari E, Tiedje JM, Hashsham SA. Modeling Hybridization Kinetics of Gene Probes in a DNA Biochip Using FEMLAB. MICROARRAYS 2017; 6:microarrays6020009. [PMID: 28555058 PMCID: PMC5487956 DOI: 10.3390/microarrays6020009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/15/2017] [Revised: 05/22/2017] [Accepted: 05/26/2017] [Indexed: 11/29/2022]
Abstract
Microfluidic DNA biochips capable of detecting specific DNA sequences are useful in medical diagnostics, drug discovery, food safety monitoring and agriculture. They are used as miniaturized platforms for analysis of nucleic acids-based biomarkers. Binding kinetics between immobilized single stranded DNA on the surface and its complementary strand present in the sample are of interest. To achieve optimal sensitivity with minimum sample size and rapid hybridization, ability to predict the kinetics of hybridization based on the thermodynamic characteristics of the probe is crucial. In this study, a computer aided numerical model for the design and optimization of a flow-through biochip was developed using a finite element technique packaged software tool (FEMLAB; package included in COMSOL Multiphysics) to simulate the transport of DNA through a microfluidic chamber to the reaction surface. The model accounts for fluid flow, convection and diffusion in the channel and on the reaction surface. Concentration, association rate constant, dissociation rate constant, recirculation flow rate, and temperature were key parameters affecting the rate of hybridization. The model predicted the kinetic profile and signal intensities of eighteen 20-mer probes targeting vancomycin resistance genes (VRGs). Predicted signal intensities and hybridization kinetics strongly correlated with experimental data in the biochip (R2 = 0.8131).
Collapse
Affiliation(s)
- Ahsan Munir
- Department of Civil and Environmental Engineering, Michigan State University, East Lansing, MI 48823,USA.
| | - Hassan Waseem
- Department of Civil and Environmental Engineering, Michigan State University, East Lansing, MI 48823,USA.
| | - Maggie R Williams
- Department of Civil and Environmental Engineering, Michigan State University, East Lansing, MI 48823,USA.
| | - Robert D Stedtfeld
- Department of Civil and Environmental Engineering, Michigan State University, East Lansing, MI 48823,USA.
| | - Erdogan Gulari
- Department of Chemical Engineering, University of Michigan, Ann Arbor, MI 48109, USA.
| | - James M Tiedje
- Center for Microbial Ecology, Michigan State University, East Lansing, MI 48823, USA.
- Department of Plant, Soil, and Microbial Sciences, Michigan State University, East Lansing, MI 48823, USA.
| | - Syed A Hashsham
- Department of Civil and Environmental Engineering, Michigan State University, East Lansing, MI 48823,USA.
- Center for Microbial Ecology, Michigan State University, East Lansing, MI 48823, USA.
- Department of Plant, Soil, and Microbial Sciences, Michigan State University, East Lansing, MI 48823, USA.
| |
Collapse
|
31
|
Stedtfeld RD, Stedtfeld TM, Fader KA, Williams MR, Bhaduri P, Quensen J, Zacharewski TR, Tiedje JM, Hashsham SA. TCDD influences reservoir of antibiotic resistance genes in murine gut microbiome. FEMS Microbiol Ecol 2017; 93:3798199. [PMID: 28475713 PMCID: PMC5458050 DOI: 10.1093/femsec/fix058] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 05/02/2017] [Indexed: 02/06/2023] Open
Abstract
Dysbiosis of the gut microbiome via antibiotics, changes in diet and infection can select for bacterial groups that more frequently harbor antimicrobial resistance genes (ARGs) and mobile genetic elements (MGEs). However, the impact of environmental toxicants on the reservoir of ARGs in the gut microbiome has received less attention. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) is a potent aryl hydrocarbon receptor (AhR) agonist with multiple toxic health effects including immune dysfunction. The selective pressure of TCDD on the abundance of ARG and MGE-harboring gut populations was examined using C57BL/6 mice exposed to 0-30 μg/kg TCDD for 28 and 92 days with the latter having a 30-day recovery period. DNA extracted from temporally collected fecal pellets was characterized using a qPCR array with 384 assays targeting ARGs and MGEs. Fourteen genes, typically observed in Enterobacteriaceae, increased significantly within 8 days of initial dosing, persisted throughout the treatment period, and remained induced 30 days post dosing. A qPCR primer set targeting Enterobacteriaceae also showed 10- to 100-fold higher abundance in TCDD-treated groups, which was further verified using metagenomics. Results show a bloom of ARG-harboring bacterial groups in the gut due to a xenobiotic compound that is not a metal, biocide or antimicrobial.
Collapse
Affiliation(s)
- Robert D. Stedtfeld
- Department of Civil and Environmental Engineering, East Lansing, MI 48824, USA
| | | | - Kelly A. Fader
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - Maggie R. Williams
- Department of Civil and Environmental Engineering, East Lansing, MI 48824, USA
| | - Prianca Bhaduri
- Department of Civil and Environmental Engineering, East Lansing, MI 48824, USA
| | - John Quensen
- Center for Microbial Ecology, Michigan State University, East Lansing, MI 48824, USA
| | - Timothy R. Zacharewski
- Institute for Integrative Toxicology, Michigan State University, East Lansing, MI 48824, USA
- Department of Biochemistry and Molecular Biology, Michigan State University, East Lansing, MI 48824, USA
| | - James M. Tiedje
- Center for Microbial Ecology, Michigan State University, East Lansing, MI 48824, USA
| | - Syed A. Hashsham
- Department of Civil and Environmental Engineering, East Lansing, MI 48824, USA
- Center for Microbial Ecology, Michigan State University, East Lansing, MI 48824, USA
| |
Collapse
|
32
|
Sohail M, Rashid A, Aslam B, Waseem M, Shahid M, Akram M, Khurshid M, Rasool MH. Antimicrobial susceptibility of Acinetobacter clinical isolates and emerging antibiogram trends for nosocomial infection management. Rev Soc Bras Med Trop 2017; 49:300-4. [PMID: 27384826 DOI: 10.1590/0037-8682-0111-2016] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Accepted: 05/26/2016] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION The drug resistant Acinetobacter strains are important causes of nosocomial infections that are difficult to control and treat. This study aimed to determine the antimicrobial susceptibility patterns of Acinetobacter strains isolated from different clinical specimens obtained from patients belonging to different age groups. METHODS In total, 716 non-duplicate Acinetobacter isolates were collected from the infected patients admitted to tertiary-care hospitals at Lahore, Pakistan, over a period of 28 months. The Acinetobacter isolates were identified using API 20E, and antimicrobial susceptibility testing was performed and interpreted according to Clinical and Laboratory Standards Institute (CLSI) guidelines. RESULTS The isolation rate of Acinetobacter was high from the respiratory specimens, followed by wound samples. Antibiotic susceptibility analyses of the isolates revealed that the resistance to cefotaxime and ceftazidime was the most common, in 710 (99.2%) specimens each, followed by the resistance to gentamicin in 670 (93.6%) isolates, and to imipenem in 651 (90.9%) isolates. However, almost all isolates were susceptible to tigecycline, colistin, and polymyxin B. CONCLUSIONS The present study showed the alarming trends of resistance of Acinetobacter strains isolated from clinical specimens to the various classes of antimicrobials. The improvement of microbiological techniques for earlier and more accurate identification of bacteria is necessary for the selection of appropriate treatments.
Collapse
Affiliation(s)
- Muhammad Sohail
- Department of Microbiology and Molecular Genetics, Quaid-e-Azam Campus, University of the Punjab, Lahore, Pakistan.,Chughtais Lahore Lab, Lahore, Pakistan
| | - Abid Rashid
- College of Allied Health Professionals, Directorate of Medical Sciences, Government College University, Faisalabad, Pakistan
| | - Bilal Aslam
- Department of Microbiology, Government College University, Faisalabad, Pakistan
| | - Muhammad Waseem
- Department of Microbiology, Government College University, Faisalabad, Pakistan
| | - Muhammad Shahid
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Muhammad Akram
- Department of Biosciences, COMSATS Institute of Information Technology, Islamabad, Pakistan
| | - Mohsin Khurshid
- College of Allied Health Professionals, Directorate of Medical Sciences, Government College University, Faisalabad, Pakistan.,Department of Microbiology, Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
33
|
Imperial ICVJ, Ibana JA. Addressing the Antibiotic Resistance Problem with Probiotics: Reducing the Risk of Its Double-Edged Sword Effect. Front Microbiol 2016; 7:1983. [PMID: 28018315 PMCID: PMC5156686 DOI: 10.3389/fmicb.2016.01983] [Citation(s) in RCA: 131] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Accepted: 11/28/2016] [Indexed: 12/27/2022] Open
Abstract
Antibiotic resistance is a global public health problem that requires our attention. Indiscriminate antibiotic use is a major contributor in the introduction of selective pressures in our natural environments that have significantly contributed in the rapid emergence of antibiotic-resistant microbial strains. The use of probiotics in lieu of antibiotic therapy to address certain health conditions in both animals and humans may alleviate these antibiotic-mediated selective pressures. Probiotic use is defined as the actual application of live beneficial microbes to obtain a desired outcome by preventing diseased state or improving general health. Multiple studies have confirmed the beneficial effects of probiotic use in the health of both livestock and humans. As such, probiotics consumption is gaining popularity worldwide. However, concerns have been raised in the use of some probiotics strains that carry antibiotic resistance genes themselves, as they have the potential to pass the antibiotic resistance genes to pathogenic bacteria through horizontal gene transfer. Therefore, with the current public health concern on antibiotic resistance globally, in this review, we underscore the need to screen probiotic strains that are used in both livestock and human applications to assure their safety and mitigate their potential in significantly contributing to the spread of antibiotic resistance genes in our natural environments.
Collapse
Affiliation(s)
- Ivan C V J Imperial
- Immunopharmacology Research Laboratory, Institute of Biology, College of Science, University of the Philippines Diliman Quezon City, Philippines
| | - Joyce A Ibana
- Immunopharmacology Research Laboratory, Institute of Biology, College of Science, University of the Philippines Diliman Quezon City, Philippines
| |
Collapse
|
34
|
Becattini S, Taur Y, Pamer EG. Antibiotic-Induced Changes in the Intestinal Microbiota and Disease. Trends Mol Med 2016; 22:458-478. [PMID: 27178527 DOI: 10.1016/j.molmed.2016.04.003] [Citation(s) in RCA: 528] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 04/13/2016] [Accepted: 04/13/2016] [Indexed: 12/12/2022]
Abstract
The gut microbiota is a key player in many physiological and pathological processes occurring in humans. Recent investigations suggest that the efficacy of some clinical approaches depends on the action of commensal bacteria. Antibiotics are invaluable weapons to fight infectious diseases. However, by altering the composition and functions of the microbiota, they can also produce long-lasting deleterious effects for the host. The emergence of multidrug-resistant pathogens raises concerns about the common, and at times inappropriate, use of antimicrobial agents. Here we review the most recently discovered connections between host pathophysiology, microbiota, and antibiotics highlighting technological platforms, mechanistic insights, and clinical strategies to enhance resistance to diseases by preserving the beneficial functions of the microbiota.
Collapse
Affiliation(s)
- Simone Becattini
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Ying Taur
- Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Lucille Castori Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Eric G Pamer
- Immunology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Infectious Diseases Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Lucille Castori Center for Microbes, Inflammation and Cancer, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA.
| |
Collapse
|
35
|
Millan B, Park H, Hotte N, Mathieu O, Burguiere P, Tompkins TA, Kao D, Madsen KL. Fecal Microbial Transplants Reduce Antibiotic-resistant Genes in Patients With Recurrent Clostridium difficile Infection. Clin Infect Dis 2016; 62:1479-1486. [PMID: 27025836 PMCID: PMC4885654 DOI: 10.1093/cid/ciw185] [Citation(s) in RCA: 150] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Accepted: 02/15/2016] [Indexed: 01/13/2023] Open
Abstract
Patients with recurrent C. difficile infection harbor large numbers of microbes with antibiotic resistance genes. Fecal microbial transplantation eradicates pathogenic organisms and eliminates antibiotic-resistance genes suggesting this may be a viable treatment option to eradicate multidrug resistant bacteria from patients. Background. Recurrent Clostridium difficile infection (RCDI) is associated with repeated antibiotic treatment and the enhanced growth of antibiotic-resistant microbes. This study tested the hypothesis that patients with RCDI would harbor large numbers of antibiotic-resistant microbes and that fecal microbiota transplantation (FMT) would reduce the number of antibiotic-resistant genes. Methods. In a single center study, patients with RCDI (n = 20) received FMT from universal donors via colonoscopy. Stool samples were collected from donors (n = 3) and patients prior to and following FMT. DNA was extracted and shotgun metagenomics performed. Results as well as assembled libraries from a healthy cohort (n = 87) obtained from the Human Microbiome Project were aligned against the NCBI bacterial taxonomy database and the Comprehensive Antibiotic Resistance Database. Results were corroborated through a DNA microarray containing 354 antibiotic resistance (ABR) genes. Results. RCDI patients had a greater number and diversity of ABR genes compared with donors and healthy controls. Beta-lactam, multidrug efflux pumps, fluoroquinolone, and antibiotic inactivation ABR genes were increased in RCDI patients, although donors primarily had tetracycline resistance. RCDI patients were dominated by Proteobacteria with Escherichia coli and Klebsiella most prevalent. FMT resulted in a resolution of symptoms that correlated directly with a decreased number and diversity of ABR genes and increased Bacteroidetes and Firmicutes with reduced Proteobacteria. ABR gene profiles were maintained in recipients for up to a year following FMT. Conclusions. RCDI patients have increased numbers of antibiotic-resistant organisms. FMT is effective in the eradication of pathogenic antibiotic-resistant organisms and elimination of ABR genes.
Collapse
Affiliation(s)
- Braden Millan
- Department of Medicine, University of Alberta, Edmonton
| | - Heekuk Park
- Department of Medicine, University of Alberta, Edmonton
| | - Naomi Hotte
- Department of Medicine, University of Alberta, Edmonton
| | | | | | | | - Dina Kao
- Department of Medicine, University of Alberta, Edmonton
| | | |
Collapse
|
36
|
Vaccines for Antibiotic-Resistant Bacteria: Possibility or Pipe Dream? Trends Pharmacol Sci 2016; 37:143-152. [DOI: 10.1016/j.tips.2015.10.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2015] [Revised: 10/13/2015] [Accepted: 10/16/2015] [Indexed: 11/19/2022]
|
37
|
Abstract
In recent decades, the emergence and spread of antibiotic resistance among bacterial pathogens has become a major threat to public health. Bacteria can acquire antibiotic resistance genes by the mobilization and transfer of resistance genes from a donor strain. The human gut contains a densely populated microbial ecosystem, termed the gut microbiota, which offers ample opportunities for the horizontal transfer of genetic material, including antibiotic resistance genes. Recent technological advances allow microbiota-wide studies into the diversity and dynamics of the antibiotic resistance genes that are harboured by the gut microbiota (‘the gut resistome’). Genes conferring resistance to antibiotics are ubiquitously present among the gut microbiota of humans and most resistance genes are harboured by strictly anaerobic gut commensals. The horizontal transfer of genetic material, including antibiotic resistance genes, through conjugation and transduction is a frequent event in the gut microbiota, but mostly involves non-pathogenic gut commensals as these dominate the microbiota of healthy individuals. Resistance gene transfer from commensals to gut-dwelling opportunistic pathogens appears to be a relatively rare event but may contribute to the emergence of multi-drug resistant strains, as is illustrated by the vancomycin resistance determinants that are shared by anaerobic gut commensals and the nosocomial pathogen Enterococcus faecium.
Collapse
Affiliation(s)
- Willem van Schaik
- Department of Medical Microbiology, University Medical Center Utrecht, 3584 CX Utrecht, The Netherlands
| |
Collapse
|
38
|
Francino MP. Antibiotics and the Human Gut Microbiome: Dysbioses and Accumulation of Resistances. Front Microbiol 2016; 6:1543. [PMID: 26793178 PMCID: PMC4709861 DOI: 10.3389/fmicb.2015.01543] [Citation(s) in RCA: 459] [Impact Index Per Article: 57.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Accepted: 12/21/2015] [Indexed: 12/12/2022] Open
Abstract
The human microbiome is overly exposed to antibiotics, due, not only to their medical use, but also to their utilization in farm animals and crops. Microbiome composition can be rapidly altered by exposure to antibiotics, with potential immediate effects on health, for instance through the selection of resistant opportunistic pathogens that can cause acute disease. Microbiome alterations induced by antibiotics can also indirectly affect health in the long-term. The mutualistic microbes in the human body interact with many physiological processes, and participate in the regulation of immune and metabolic homeostasis. Therefore, antibiotic exposure can alter many basic physiological equilibria, promoting long-term disease. In addition, excessive antibiotic use fosters bacterial resistance, and the overly exposed human microbiome has become a significant reservoir of resistance genes, contributing to the increasing difficulty in controlling bacterial infections. Here, the complex relationships between antibiotics and the human microbiome are reviewed, with focus on the intestinal microbiota, addressing (1) the effects of antibiotic use on the composition and function of the gut microbiota, (2) the impact of antibiotic-induced microbiota alterations on immunity, metabolism, and health, and (3) the role of the gut microbiota as a reservoir of antibiotic resistances.
Collapse
Affiliation(s)
- M P Francino
- Unitat Mixta d'Investigació en Genòmica i Salut, Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO)-Salud Pública/Institut Cavanilles de Biodiversitat i Biologia Evolutiva, Universitat de ValènciaValència, Spain; Consorcio de Investigación Biomédica en Red de Epidemiología y Salud PúblicaMadrid, Spain
| |
Collapse
|
39
|
Santiago-Rodriguez TM, Ly M, Daigneault MC, Brown IHL, McDonald JAK, Bonilla N, Vercoe EA, Pride DT. Chemostat culture systems support diverse bacteriophage communities from human feces. MICROBIOME 2015; 3:58. [PMID: 26549756 PMCID: PMC4638026 DOI: 10.1186/s40168-015-0124-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2015] [Accepted: 10/27/2015] [Indexed: 06/05/2023]
Abstract
BACKGROUND Most human microbiota studies focus on bacteria inhabiting body surfaces, but these surfaces also are home to large populations of viruses. Many are bacteriophages, and their role in driving bacterial diversity is difficult to decipher without the use of in vitro ecosystems that can reproduce human microbial communities. RESULTS We used chemostat culture systems known to harbor diverse fecal bacteria to decipher whether these cultures also are home to phage communities. We found that there are vast viral communities inhabiting these ecosystems, with estimated concentrations similar to those found in human feces. The viral communities are composed entirely of bacteriophages and likely contain both temperate and lytic phages based on their similarities to other known phages. We examined the cultured phage communities at five separate time points over 24 days and found that they were highly individual-specific, suggesting that much of the subject-specificity found in human viromes also is captured by this culture-based system. A high proportion of the community membership is conserved over time, but the cultured communities maintain more similarity with other intra-subject cultures than they do to human feces. In four of the five subjects, estimated viral diversity between fecal and cultured communities was highly similar. CONCLUSIONS Because the diversity of phages in these cultured fecal communities have similarities to those found in humans, we believe these communities can serve as valuable ecosystems to help uncover the role of phages in human microbial communities.
Collapse
Affiliation(s)
- Tasha M Santiago-Rodriguez
- Department of Pathology, University of California, 9500 Gilman Drive, MC 0612, La Jolla, CA, 92093-0612, USA
| | - Melissa Ly
- Department of Pathology, University of California, 9500 Gilman Drive, MC 0612, La Jolla, CA, 92093-0612, USA
| | - Michelle C Daigneault
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Ian H L Brown
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Julie A K McDonald
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - Natasha Bonilla
- Department of Biology, San Diego State University, San Diego, CA, USA
| | - Emma Allen Vercoe
- Department of Molecular and Cellular Biology, University of Guelph, Guelph, Ontario, Canada
| | - David T Pride
- Department of Pathology, University of California, 9500 Gilman Drive, MC 0612, La Jolla, CA, 92093-0612, USA.
- Department of Medicine, University of California, San Diego, CA, USA.
| |
Collapse
|
40
|
High frequencies of antibiotic resistance genes in infants' meconium and early fecal samples. J Dev Orig Health Dis 2015; 7:35-44. [PMID: 26353938 DOI: 10.1017/s2040174415001506] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The gastrointestinal tract (GIT) microbiota has been identified as an important reservoir of antibiotic resistance genes (ARGs) that can be horizontally transferred to pathogenic species. Maternal GIT microbes can be transmitted to the offspring, and recent work indicates that such transfer starts before birth. We have used culture-independent genetic screenings to explore whether ARGs are already present in the meconium accumulated in the GIT during fetal life and in feces of 1-week-old infants. We have analyzed resistance to β-lactam antibiotics (BLr) and tetracycline (Tcr), screening for a variety of genes conferring each. To evaluate whether ARGs could have been inherited by maternal transmission, we have screened perinatal fecal samples of the 1-week-old babies' mothers, as well as a mother-infant series including meconium, fecal samples collected through the infant's 1st year, maternal fecal samples and colostrum. Our results reveal a high prevalence of BLr and Tcr in both meconium and early fecal samples, implying that the GIT resistance reservoir starts to accumulate even before birth. We show that ARGs present in the mother may reach the meconium and colostrum and establish in the infant GIT, but also that some ARGs were likely acquired from other sources. Alarmingly, we identified in both meconium and 1-week-olds' samples a particularly elevated prevalence of mecA (>45%), six-fold higher than that detected in the mothers. The mecA gene confers BLr to methicillin-resistant Staphylococcus aureus, and although its detection does not imply the presence of this pathogen, it does implicate the young infant's GIT as a noteworthy reservoir of this gene.
Collapse
|
41
|
Abstract
![]()
Bacteria
possess a remarkable ability to rapidly adapt and evolve
in response to antibiotics. Acquired antibiotic resistance can arise
by multiple mechanisms but commonly involves altering the target site
of the drug, enzymatically inactivating the drug, or preventing the
drug from accessing its target. These mechanisms involve new genetic
changes in the pathogen leading to heritable resistance. This recognition
underscores the importance of understanding how such
genetic changes can arise. Here, we review recent advances in our
understanding of the processes that contribute to the evolution of
antibiotic resistance, with a particular focus on hypermutation mediated
by the SOS pathway and horizontal gene transfer. We explore the molecular
mechanisms involved in acquired resistance and discuss their viability
as potential targets. We propose that additional studies into these
adaptive mechanisms not only can provide insights into evolution but
also can offer a strategy for potentiating our current antibiotic
arsenal.
Collapse
|