201
|
Bailey MJ, Naik NN, Wild LE, Patterson WB, Alderete TL. Exposure to air pollutants and the gut microbiota: a potential link between exposure, obesity, and type 2 diabetes. Gut Microbes 2020; 11:1188-1202. [PMID: 32347153 PMCID: PMC7524284 DOI: 10.1080/19490976.2020.1749754] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Work has shown that increased exposure to air pollutants independently contributes to obesity and type 2 diabetes risk, yet the exact mechanisms underlying these associations have not been fully characterized. The current review summarizes recent findings regarding the impact of inhaled and ingested air pollutants on the gut microbiota. Animal and human studies provide evidence that air pollutants, such as particulate matter, nitrogen oxides, and ozone, have the potential to alter the gut microbiota. Further, studies suggest that such exposure-induced alterations to the gut microbiota may contribute to increased risk for obesity and type 2 diabetes through inflammatory pathways. Future work is needed to fully understand the complex interactions between air pollution, the gut microbiome, and human health. Additionally, advanced sequencing methods for gut microbiome research present unique opportunities to study the underlying pathways that link increased air pollution exposure with obesity and type 2 diabetes risk.
Collapse
Affiliation(s)
- Maximillian J. Bailey
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Noopur N. Naik
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Laura E. Wild
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - William B. Patterson
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA
| | - Tanya L. Alderete
- Department of Integrative Physiology, University of Colorado Boulder, Boulder, CO, USA,CONTACT Tanya L. Alderete University of Colorado Boulder, Department of Integrative Physiology, Ramaley Biology Building, 1800 Colorado Avenue, N379, Boulder, CO80309
| |
Collapse
|
202
|
Yang J, McDowell A, Seo H, Kim S, Min TK, Jee YK, Choi Y, Park HS, Pyun BY, Kim YK. Diagnostic Models for Atopic Dermatitis Based on Serum Microbial Extracellular Vesicle Metagenomic Analysis: A Pilot Study. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2020; 12:792-805. [PMID: 32638560 PMCID: PMC7346989 DOI: 10.4168/aair.2020.12.5.792] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 03/05/2020] [Accepted: 03/07/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE Associations between a wide variety of diseases and the microbiome have been extensively verified. Recently, there has been a rising interest in the role the microbiome plays in atopic dermatitis (AD). Furthermore, metagenomic analysis of microbe-derived extracellular vesicles (EVs) has revealed the importance and relevance of microbial EVs in human health. METHODS We compared the diversity and proportion of microbial EVs in the sera of 24 AD patients and 49 healthy controls, and developed a diagnostic model. After separating microbial EVs from serum, we specifically targeted the V3-V4 hypervariable regions of the 16S rDNA gene for amplification and subsequent sequencing. RESULTS Alpha and beta diversity between controls and AD patients both differed, but only the difference in beta diversity was significant. Proteobacteria, Firmicutes, and Bacteroidetes were the dominant phyla in healthy controls and AD patients, accounting for over 85% of the total serum bacterial EVs. Also, Proteobacteria, Firmicutes, Actinobacteria, Verrucomicrobia, and Cyanobacteria relative abundances were significantly different between the AD and control groups. At the genus level, the proportions of Escherichia-Shigella, Acinetobacter, Pseudomonas, and Enterococcus were drastically altered between the AD and control groups. AD diagnostic models developed using biomarkers selected on the basis of linear discriminant analysis effect size from the class to genus levels all yielded area under the receiver operating characteristic curve, sensitivity, specificity, and accuracy of value 1.00. CONCLUSIONS In summary, microbial EVs demonstrated the potential in their use as novel biomarkers for AD diagnosis. Therefore, future work should investigate larger case and control groups with cross-sectional or longitudinal clinical data to explore the utility and validity of serum microbiota EV-based AD diagnosis.
Collapse
Affiliation(s)
- Jinho Yang
- Institute of MD Healthcare Inc., Seoul, Korea
- Department of Health and Safety Convergence Science, Graduate School of Korea University, Seoul, Korea
| | | | - Hochan Seo
- Institute of MD Healthcare Inc., Seoul, Korea
| | - Sungwon Kim
- Institute of MD Healthcare Inc., Seoul, Korea
| | - Taek Ki Min
- Department of Pediatrics, Soonchunhyang University College of Medicine, Seoul, Korea
| | - Young Koo Jee
- Department of Internal Medicine, Dankook University College of Medicine, Cheonan, Korea
| | - Youngwoo Choi
- Department of Allergy and Clinical Immunology, Ajou University Medical Center, Suwon, Korea
| | - Hae Sim Park
- Department of Allergy and Clinical Immunology, Ajou University Medical Center, Suwon, Korea
| | - Bok Yang Pyun
- Department of Pediatrics, Soonchunhyang University College of Medicine, Seoul, Korea.
| | | |
Collapse
|
203
|
Influence of aerobic fitness on gastrointestinal barrier integrity and microbial translocation following a fixed-intensity military exertional heat stress test. Eur J Appl Physiol 2020; 120:2325-2337. [PMID: 32794058 DOI: 10.1007/s00421-020-04455-w] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Accepted: 07/30/2020] [Indexed: 02/06/2023]
Abstract
PURPOSE Exertional-heat stress adversely disrupts gastrointestinal (GI) barrier integrity, whereby subsequent microbial translocation (MT) can result in potentially serious health consequences. To date, the influence of aerobic fitness on GI barrier integrity and MT following exertional-heat stress is poorly characterised. METHOD Ten untrained (UT; VO2max = 45 ± 3 ml·kg-1·min-1) and ten highly trained (HT; VO2max = 64 ± 4 ml·kg-1·min-1) males completed an ecologically valid (military) 80-min fixed-intensity exertional-heat stress test (EHST). Venous blood was drawn immediately pre- and post-EHST. GI barrier integrity was assessed using the serum dual-sugar absorption test (DSAT) and plasma Intestinal Fatty-Acid Binding Protein (I-FABP). MT was assessed using plasma Bacteroides/total 16S DNA. RESULTS UT experienced greater thermoregulatory, cardiovascular and perceptual strain (p < 0.05) than HT during the EHST. Serum DSAT responses were similar between the two groups (p = 0.59), although Δ I-FABP was greater (p = 0.04) in the UT (1.14 ± 1.36 ng·ml-1) versus HT (0.20 ± 0.29 ng·ml-1) group. Bacteroides/Total 16S DNA ratio was unchanged (Δ; -0.04 ± 0.18) following the EHST in the HT group, but increased (Δ; 0.19 ± 0.25) in the UT group (p = 0.05). Weekly aerobic training hours had a weak, negative correlation with Δ I-FABP and Bacteroides/total 16S DNA responses. CONCLUSION When exercising at the same absolute workload, UT individuals are more susceptible to small intestinal epithelial injury and MT than HT individuals. These responses appear partially attributable to greater thermoregulatory, cardiovascular, and perceptual strain.
Collapse
|
204
|
Balboni A, Franzo G, Bano L, De Arcangeli S, Rizzardi A, Urbani L, Segatore S, Serafini F, Dondi F, Battilani M. Culture-Dependent and Sequencing Methods Revealed the Absence of a Bacterial Community Residing in the Urine of Healthy Cats. Front Vet Sci 2020; 7:438. [PMID: 32851021 PMCID: PMC7419613 DOI: 10.3389/fvets.2020.00438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Accepted: 06/16/2020] [Indexed: 12/12/2022] Open
Abstract
A growing number of studies suggest that the lower urinary tract of humans and dogs can harbor a urinary microbiota. Nevertheless, a certain concern has developed that the microbiota reported could be due to unaccounted contamination, especially in low-biomass samples. The aim of this study was to investigate the bacterial community which populates the urine of healthy cats using two approaches: a culture-dependent approach which consisted of the expanded quantitative urine culture (EQUC) techniques capable of identifying live bacteria not growing in standard urine cultures, and a culture-independent approach which consisted of 16S ribosomal RNA next generation sequencing (16S rRNA NGS) capable of identifying bacterial DNA and exploring microbial diversity with high resolution. To avoid confounding factors of possible bacterial contamination, the urine was sampled using ultrasound-guided cystocentesis, and several sample controls and negative controls were analyzed. The urine sampled from the 10 cats included in the study showed no bacterial growth in the EQUC procedure. Although several reads were successfully originated using 16S rRNA NGS, a comparable pattern was observed between urine samples and the negative control, and no taxa were statistically accepted as non-contaminant. Taken together, the results obtained allowed stating that no viable bacteria were present in the urine of healthy cats without lower urinary tract disease and urinary tract infections, and that the bacterial DNA detected was of contaminant origin.
Collapse
Affiliation(s)
- Andrea Balboni
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Ozzano Emilia, Italy
| | - Giovanni Franzo
- Department of Animal Medicine, Production and Health, University of Padua, Legnaro, Italy
| | - Luca Bano
- Diagnostic and Microbiology Laboratory, Istituto Zooprofilattico Sperimentale delle Venezie, Villorba di Treviso, Italy
| | - Stefano De Arcangeli
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Ozzano Emilia, Italy
| | - Alessia Rizzardi
- Diagnostic and Microbiology Laboratory, Istituto Zooprofilattico Sperimentale delle Venezie, Villorba di Treviso, Italy
| | - Lorenza Urbani
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Ozzano Emilia, Italy
| | - Sofia Segatore
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Ozzano Emilia, Italy
| | - Federica Serafini
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Ozzano Emilia, Italy
| | - Francesco Dondi
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Ozzano Emilia, Italy
| | - Mara Battilani
- Department of Veterinary Medical Sciences, Alma Mater Studiorum-University of Bologna, Ozzano Emilia, Italy
| |
Collapse
|
205
|
Tilg H, Burcelin R, Tremaroli V. Liver tissue microbiome in NAFLD: next step in understanding the gut-liver axis? Gut 2020; 69:1373-1374. [PMID: 32060128 DOI: 10.1136/gutjnl-2019-320490] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2020] [Revised: 02/06/2020] [Accepted: 02/06/2020] [Indexed: 12/20/2022]
Affiliation(s)
- Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, Innsbruck, Austria
| | - Remy Burcelin
- Institut National de la Santé et de la Recherche Médicale (INSERM) Unité 1048, Toulouse, France.,Université Paul Sabatier (UPS), Unité Mixte de Recherche (UMR) 1048, Institut des Maladies Métaboliques et Cardiovasculaires (I2MC), Team 2: 'Intestinal Risk Factors, Diabetes, Dyslipidemia' F-31432, Toulouse Cedex 4, France
| | - Valentina Tremaroli
- Wallenberg Laboratory for Cardiovascular and Metabolic Research, Department of Molecular and Clinical Medicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
206
|
Lichota A, Gwozdzinski K, Szewczyk EM. Microbial Modulation of Coagulation Disorders in Venous Thromboembolism. J Inflamm Res 2020; 13:387-400. [PMID: 32801832 PMCID: PMC7406375 DOI: 10.2147/jir.s258839] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 06/19/2020] [Indexed: 12/24/2022] Open
Abstract
Venous thromboembolism (VTE), including deep vein thrombosis (DVT) and pulmonary embolism (PE), is the third leading cause of cardiovascular death in the world. Important risk factors of thrombosis include bed restraint, surgery, major trauma, long journeys, inflammation, pregnancy, and oral contraceptives, previous venous thromboembolism, cancer, and bacterial infections. Sepsis increases the risk of blood clot formation 2–20 times. In this review, we discussed various mechanisms related to the role of bacteria in venous thrombosis also taking into consideration the role of the human microbiome. Many known bacteria, such as Helicobacter pylori, Chlamydia pneumoniae, Mycoplasma pneumoniae, Haemophilus influenzae, Streptococcus pneumoniae, Staphylococcus aureus, and Escherichia coli, causing infections may increase the risk of thrombotic complications through platelet activation or may lead to an inflammatory reaction involving the fibrinolytic system. Additionally, the bacteria participate in the production of factors causing or increasing the risk of cardiovascular diseases. An example can be trimethylamine N-oxide (TMAO) but also uremic toxins (indoxyl sulfate), short-chain fatty acids (SCFA) phytoestrogens, and bile acids. Finally, we presented the involvement of many bacteria in the development of venous thromboembolism and other cardiovascular diseases.
Collapse
Affiliation(s)
- Anna Lichota
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Faculty of Pharmacy, Medical University of Lodz, Lodz, Poland
| | - Krzysztof Gwozdzinski
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Lodz, Poland
| | - Eligia M Szewczyk
- Department of Pharmaceutical Microbiology and Microbiological Diagnostics, Faculty of Pharmacy, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
207
|
Schierwagen R, Alvarez-Silva C, Servant F, Trebicka J, Lelouvier B, Arumugam M. Trust is good, control is better: technical considerations in blood microbiome analysis. Gut 2020; 69:1362-1363. [PMID: 31203205 PMCID: PMC7306979 DOI: 10.1136/gutjnl-2019-319123] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 05/31/2019] [Accepted: 06/03/2019] [Indexed: 12/12/2022]
Affiliation(s)
- Robert Schierwagen
- Translational Hepatology, Department of Internal Medicine I, Universitätsklinikum/Goethe-Universität, Frankfurt, Germany
| | - Camila Alvarez-Silva
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | | | - Jonel Trebicka
- Translational Hepatology, Department of Internal Medicine I, Universitätsklinikum/Goethe-Universität, Frankfurt, Germany
| | | | - Manimozhiyan Arumugam
- The Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
208
|
Serrano-Villar S, Sanchez-Carrillo S, Talavera-Rodríguez A, Lelouvier B, Gutiérrez C, Vallejo A, Servant F, Bernadino JI, Estrada V, Madrid N, Gosalbes MJ, Bisbal O, de Lagarde M, Martínez-Sanz J, Ron R, Herrera S, Moreno S, Ferrer M. Blood Bacterial Profiles Associated With Human Immunodeficiency Virus Infection and Immune Recovery. J Infect Dis 2020; 223:471-481. [PMID: 32601702 DOI: 10.1093/infdis/jiaa379] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/23/2020] [Indexed: 02/06/2023] Open
Abstract
Human immunodeficiency virus (HIV) infection impairs mucosal immunity and leads to bacterial translocation, fueling chronic inflammation and disease progression. While this is well established, questions remain about the compositional profile of the translocated bacteria, and to what extent it is influenced by antiretroviral therapy (ART). Using 16S ribosomal DNA targeted sequencing and shotgun proteomics, we showed that HIV increases bacterial translocation from the gut to the blood. HIV increased alpha diversity in the blood, which was dominated by aerobic bacteria belonging to Micrococcaceae (Actinobacteria) and Pseudomonadaceae (Proteobacteria) families, and the number of circulating bacterial proteins was also increased. Forty-eight weeks of ART attenuated this phenomenon. We found that enrichment with Lactobacillales order, and depletion of Actinobacteria class and Moraxellaceae and Corynebacteriacae families, were significantly associated with greater immune recovery and correlated with several inflammatory markers. Our findings suggest that the molecular cross talk between the host and the translocated bacterial products could influence ART-mediated immune recovery.
Collapse
Affiliation(s)
- Sergio Serrano-Villar
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, Facultad de Medicina, Universidad de Alcalá, Instituto de Investigación Sanitaria Ramón y Cajal, Madrid, Spain
| | | | - Alba Talavera-Rodríguez
- Bioinformatics Unit, Hospital Universitario Ramón y Cajal, Facultad de Medicina, Universidad de Alcalá, Instituto de Investigación Sanitaria Ramón y Cajal, Madrid, Spain
| | | | - Carolina Gutiérrez
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, Facultad de Medicina, Universidad de Alcalá, Instituto de Investigación Sanitaria Ramón y Cajal, Madrid, Spain
| | - Alejandro Vallejo
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, Facultad de Medicina, Universidad de Alcalá, Instituto de Investigación Sanitaria Ramón y Cajal, Madrid, Spain
| | | | | | | | - Nadia Madrid
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, Facultad de Medicina, Universidad de Alcalá, Instituto de Investigación Sanitaria Ramón y Cajal, Madrid, Spain
| | - María José Gosalbes
- Area of Genomics and Health, FISABIO-Salud Pública, Valencia, Spain.,CIBER Epidemiología y Salud Pública, Madrid, Spain
| | - Otilia Bisbal
- HIV Unit, Hospital Universitario Doce de Octubre, Madrid, Spain
| | | | - Javier Martínez-Sanz
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, Facultad de Medicina, Universidad de Alcalá, Instituto de Investigación Sanitaria Ramón y Cajal, Madrid, Spain
| | - Raquel Ron
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, Facultad de Medicina, Universidad de Alcalá, Instituto de Investigación Sanitaria Ramón y Cajal, Madrid, Spain
| | - Sabina Herrera
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, Facultad de Medicina, Universidad de Alcalá, Instituto de Investigación Sanitaria Ramón y Cajal, Madrid, Spain
| | - Santiago Moreno
- Department of Infectious Diseases, Hospital Universitario Ramón y Cajal, Facultad de Medicina, Universidad de Alcalá, Instituto de Investigación Sanitaria Ramón y Cajal, Madrid, Spain
| | - Manuel Ferrer
- Institute of Catalysis, Consejo Superior de Investigaciones Científicas, Madrid, Spain
| |
Collapse
|
209
|
Panza F, Lozupone M, Solfrizzi V, Watling M, Imbimbo BP. Time to test antibacterial therapy in Alzheimer's disease. Brain 2020; 142:2905-2929. [PMID: 31532495 DOI: 10.1093/brain/awz244] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 05/24/2019] [Accepted: 06/14/2019] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease is associated with cerebral accumulation of amyloid-β peptide and hyperphosphorylated tau. In the past 28 years, huge efforts have been made in attempting to treat the disease by reducing brain accumulation of amyloid-β in patients with Alzheimer's disease, with no success. While anti-amyloid-β therapies continue to be tested in prodromal patients with Alzheimer's disease and in subjects at risk of developing Alzheimer's disease, there is an urgent need to provide therapeutic support to patients with established Alzheimer's disease for whom current symptomatic treatment (acetylcholinesterase inhibitors and N-methyl d-aspartate antagonist) provide limited help. The possibility of an infectious aetiology for Alzheimer's disease has been repeatedly postulated over the past three decades. Infiltration of the brain by pathogens may act as a trigger or co-factor for Alzheimer's disease, with Herpes simplex virus type 1, Chlamydia pneumoniae, and Porphyromonas gingivalis being most frequently implicated. These pathogens may directly cross a weakened blood-brain barrier, reach the CNS and cause neurological damage by eliciting neuroinflammation. Alternatively, pathogens may cross a weakened intestinal barrier, reach vascular circulation and then cross blood-brain barrier or cause low grade chronic inflammation and subsequent neuroinflammation from the periphery. The gut microbiota comprises a complex community of microorganisms. Increased permeability of the gut and blood-brain barrier induced by microbiota dysbiosis may impact Alzheimer's disease pathogenesis. Inflammatory microorganisms in gut microbiota are associated with peripheral inflammation and brain amyloid-β deposition in subjects with cognitive impairment. Oral microbiota may also influence Alzheimer's disease risk through circulatory or neural access to the brain. At least two possibilities can be envisaged to explain the association of suspected pathogens and Alzheimer's disease. One is that patients with Alzheimer's disease are particularly prone to microbial infections. The other is that microbial infection is a contributing cause of Alzheimer's disease. Therapeutic trials with antivirals and/or antibacterials could resolve this dilemma. Indeed, antiviral agents are being tested in patients with Alzheimer's disease in double-blind placebo-controlled studies. Although combined antibiotic therapy was found to be effective in animal models of Alzheimer's disease, antibacterial drugs are not being widely investigated in patients with Alzheimer's disease. This is because it is not clear which bacterial populations in the gut of patients with Alzheimer's disease are overexpressed and if safe, selective antibacterials are available for them. On the other hand, a bacterial protease inhibitor targeting P. gingivalis toxins is now being tested in patients with Alzheimer's disease. Clinical studies are needed to test if countering bacterial infection may be beneficial in patients with established Alzheimer's disease.
Collapse
Affiliation(s)
- Francesco Panza
- Unit of Epidemiological Research on Aging, National Institute of Gastroenterology 'Saverio de Bellis', Research Hospital, Castellana Grotte, Bari, Italy
| | - Madia Lozupone
- Unit of Epidemiological Research on Aging, National Institute of Gastroenterology 'Saverio de Bellis', Research Hospital, Castellana Grotte, Bari, Italy
- Neurodegenerative Disease Unit, Department of Basic Medicine, Neuroscience, and Sense Organs, University of Bari Aldo Moro, Bari, Italy
| | - Vincenzo Solfrizzi
- 'C. Frugoni' Internal and Geriatric Medicine and Memory Unit, University of Bari Aldo Moro, Bari, Italy
| | - Mark Watling
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma, Italy
| |
Collapse
|
210
|
Abstract
This review addresses the interplay between obesity, type 2 diabetes mellitus, and cardiovascular diseases. It is proposed that obesity, generally defined by an excess of body fat causing prejudice to health, can no longer be evaluated solely by the body mass index (expressed in kg/m2) because it represents a heterogeneous entity. For instance, several cardiometabolic imaging studies have shown that some individuals who have a normal weight or who are overweight are at high risk if they have an excess of visceral adipose tissue-a condition often accompanied by accumulation of fat in normally lean tissues (ectopic fat deposition in liver, heart, skeletal muscle, etc). On the other hand, individuals who are overweight or obese can nevertheless be at much lower risk than expected when faced with excess energy intake if they have the ability to expand their subcutaneous adipose tissue mass, particularly in the gluteal-femoral area. Hence, excessive amounts of visceral adipose tissue and of ectopic fat largely define the cardiovascular disease risk of overweight and moderate obesity. There is also a rapidly expanding subgroup of patients characterized by a high accumulation of body fat (severe obesity). Severe obesity is characterized by specific additional cardiovascular health issues that should receive attention. Because of the difficulties of normalizing body fat content in patients with severe obesity, more aggressive treatments have been studied in this subgroup of individuals such as obesity surgery, also referred to as metabolic surgery. On the basis of the above, we propose that we should refer to obesities rather than obesity.
Collapse
Affiliation(s)
- Marie-Eve Piché
- From the Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval (M.-E.P., A.T., J.-P.D.), Université Laval, Québec, QC, Canada.,Department of Medicine, Faculty of Medicine (M.-E.P.), Université Laval, Québec, QC, Canada
| | - André Tchernof
- From the Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval (M.-E.P., A.T., J.-P.D.), Université Laval, Québec, QC, Canada.,School of Nutrition (A.T.), Université Laval, Québec, QC, Canada
| | - Jean-Pierre Després
- From the Centre de recherche de l'Institut universitaire de cardiologie et de pneumologie de Québec - Université Laval (M.-E.P., A.T., J.-P.D.), Université Laval, Québec, QC, Canada.,Vitam - Centre de recherche en santé durable, CIUSSS - Capitale-Nationale (J.-P.D.), Université Laval, Québec, QC, Canada.,Department of Kinesiology, Faculty of Medicine (J.-P.D.), Université Laval, Québec, QC, Canada
| |
Collapse
|
211
|
Abstract
Human health is intimately linked to the ecology and diversity of the human microbiome. Together, the human organism and the human microbiome work as a complex super-organism throughout the human life cycle. Microbiome science provides direct evidence and substantiation of the fundamental principles of homeopathy, including holism, psychosomatics, direction of cure, the Law of Similars, individuality and susceptibility, minimum dose, and homeostasis. Whilst many conventional (allopathic) medical treatments irreversibly damage the ecology of the microbiome and trigger chronic immune dysfunction and inflammation, the future sustainability of the entire field of medicine depends on the ability to recognize these inconvenient biological truths and to embrace a safer approach based on this evidence. Fortunately, one of the oldest forms of clinically verifiable, evidence-based, and ecologically sustainable medicine, that does not harm the microbiome, already exists in the form of homeopathy.
Collapse
Affiliation(s)
- Ronald D Whitmont
- Department of Family and Community Medicine, New York Medical College, Rhinebeck, New York, United States
| |
Collapse
|
212
|
Salehi B, Dimitrijević M, Aleksić A, Neffe-Skocińska K, Zielińska D, Kołożyn-Krajewska D, Sharifi-Rad J, Stojanović-Radić Z, Prabu SM, Rodrigues CF, Martins N. Human microbiome and homeostasis: insights into the key role of prebiotics, probiotics, and symbiotics. Crit Rev Food Sci Nutr 2020; 61:1415-1428. [PMID: 32400169 DOI: 10.1080/10408398.2020.1760202] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The interest in the study of the gut microbiome has grown exponentially. Indeed, its impact on health and disease has been increasingly reported, and the importance of keeping gut microbiome homeostasis clearly highlighted. However, and despite many advances, there are still some gaps, as well as the real discernment on the contribution of some species falls far short of what is needed. Anyway, it is already more than a solid fact of its importance in maintaining health and preventing disease, as well as in the treatment of some pathologies. In this sense, and given the existence of some ambiguous opinions, the present review aims to discuss the importance of gut microbiome in homeostasis maintenance, and even the role of probiotics, prebiotics, and symbiotics in both health promotion and disease prevention.
Collapse
Affiliation(s)
- Bahare Salehi
- Student Research Committee, School of Medicine, Bam University of Medical Sciences, Bam, Iran
| | - Marina Dimitrijević
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Ana Aleksić
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | - Katarzyna Neffe-Skocińska
- Department of Food Gastronomy and Food Hygiene, Warsaw University of Life Sciences (WULS), Warszawa, Poland
| | - Dorota Zielińska
- Department of Food Gastronomy and Food Hygiene, Warsaw University of Life Sciences (WULS), Warszawa, Poland
| | - Danuta Kołożyn-Krajewska
- Department of Food Gastronomy and Food Hygiene, Warsaw University of Life Sciences (WULS), Warszawa, Poland
| | - Javad Sharifi-Rad
- Phytochemistry Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zorica Stojanović-Radić
- Department of Biology and Ecology, Faculty of Science and Mathematics, University of Niš, Niš, Serbia
| | | | - Célia F Rodrigues
- LEPABE - Department of Chemical Engineering, Faculty of Engineering, University of Porto, Porto, Portugal
| | - Natália Martins
- Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, Porto, Portugal.,Institute for Research and Innovation in Health (i3S), University of Porto, Porto, Portugal
| |
Collapse
|
213
|
Microbiome and Schizophrenia: Current Evidence and Future Challenges. Curr Behav Neurosci Rep 2020. [DOI: 10.1007/s40473-020-00206-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
214
|
Theis KR, Romero R, Winters AD, Jobe AH, Gomez-Lopez N. Lack of Evidence for Microbiota in the Placental and Fetal Tissues of Rhesus Macaques. mSphere 2020; 5:e00210-20. [PMID: 32376701 PMCID: PMC7203455 DOI: 10.1128/msphere.00210-20] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/16/2020] [Indexed: 12/31/2022] Open
Abstract
The prevailing paradigm in obstetrics has been the sterile womb hypothesis. However, some are asserting that the placenta, intra-amniotic environment, and fetus harbor microbial communities. The objective of this study was to determine whether the fetal and placental tissues of rhesus macaques harbor bacterial communities. Fetal, placental, and uterine wall samples were obtained from cesarean deliveries without labor (∼130/166 days gestation). The presence of bacteria in the fetal intestine and placenta was investigated through culture. The bacterial burden and profiles of the placenta, umbilical cord, and fetal brain, heart, liver, and colon were determined through quantitative real-time PCR and DNA sequencing. These data were compared with those of the uterine wall as well as to negative and positive technical controls. Bacterial cultures of fetal and placental tissues yielded only a single colony of Cutibacterium acnes This bacterium was detected at a low relative abundance (0.02%) in the 16S rRNA gene profile of the villous tree sample from which it was cultured, yet it was also identified in 12/29 background technical controls. The bacterial burden and profiles of fetal and placental tissues did not exceed or differ from those of background technical controls. By contrast, the bacterial burden and profiles of positive controls exceeded and differed from those of background controls. Among the macaque samples, distinct microbial signals were limited to the uterine wall. Therefore, using multiple modes of microbiologic inquiry, there was not consistent evidence of bacterial communities in the fetal and placental tissues of rhesus macaques.IMPORTANCE Microbial invasion of the amniotic cavity (i.e., intra-amniotic infection) has been causally linked to pregnancy complications, especially preterm birth. Therefore, if the placenta and the fetus are typically populated by low-biomass microbial communities, current understanding of the role of microbes in reproduction and pregnancy outcomes will need to be fundamentally reconsidered. Could these communities be of benefit by competitively excluding potential pathogens or priming the fetal immune system for the microbial bombardment it will experience upon delivery? If so, what properties (e.g., microbial load and community membership) of these microbial communities preclude versus promote intra-amniotic infection? Given the ramifications of the in utero colonization hypothesis, critical evaluation is required. In this study, using multiple modes of microbiologic inquiry (i.e., culture, quantitative real-time PCR [qPCR], and DNA sequencing) and controlling for potential background DNA contamination, we did not find consistent evidence for microbial communities in the placental and fetal tissues of rhesus macaques.
Collapse
Affiliation(s)
- Kevin R Theis
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
| | - Roberto Romero
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
- Department of Epidemiology and Biostatistics, Michigan State University, East Lansing, Michigan, USA
- Center for Molecular Medicine and Genetics, Wayne State University, Detroit, Michigan, USA
- Detroit Medical Center, Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Florida International University, Miami, Florida, USA
| | - Andrew D Winters
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
| | - Alan H Jobe
- Cincinnati Children's Hospital, University of Cincinnati School of Medicine, Cincinnati, Ohio, USA
| | - Nardhy Gomez-Lopez
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, Michigan, USA
- Perinatology Research Branch, Division of Obstetrics and Maternal-Fetal Medicine, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, U.S. Department of Health and Human Services, Bethesda, Maryland, and Detroit, Michigan, USA
- Department of Obstetrics and Gynecology, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
215
|
Chakaroun RM, Massier L, Kovacs P. Gut Microbiome, Intestinal Permeability, and Tissue Bacteria in Metabolic Disease: Perpetrators or Bystanders? Nutrients 2020; 12:E1082. [PMID: 32295104 PMCID: PMC7230435 DOI: 10.3390/nu12041082] [Citation(s) in RCA: 177] [Impact Index Per Article: 35.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 04/07/2020] [Indexed: 02/06/2023] Open
Abstract
The emerging evidence on the interconnectedness between the gut microbiome and host metabolism has led to a paradigm shift in the study of metabolic diseases such as obesity and type 2 diabetes with implications on both underlying pathophysiology and potential treatment. Mounting preclinical and clinical evidence of gut microbiota shifts, increased intestinal permeability in metabolic disease, and the critical positioning of the intestinal barrier at the interface between environment and internal milieu have led to the rekindling of the "leaky gut" concept. Although increased circulation of surrogate markers and directly measurable intestinal permeability have been linked to increased systemic inflammation in metabolic disease, mechanistic models behind this phenomenon are underdeveloped. Given repeated observations of microorganisms in several tissues with congruent phylogenetic findings, we review current evidence on these unanticipated niches, focusing specifically on the interaction between gut permeability and intestinal as well as extra-intestinal bacteria and their joint contributions to systemic inflammation and metabolism. We further address limitations of current studies and suggest strategies drawing on standard techniques for permeability measurement, recent advancements in microbial culture independent techniques and computational methodologies to robustly develop these concepts, which may be of considerable value for the development of prevention and treatment strategies.
Collapse
Affiliation(s)
- Rima M. Chakaroun
- Medical Department III—Endocrinology, Nephrology, Rheumatology, University of Leipzig Medical Center, 04103 Leipzig, Germany; (L.M.); (P.K.)
| | | | | |
Collapse
|
216
|
Díez López C, Montiel González D, Haas C, Vidaki A, Kayser M. Microbiome-based body site of origin classification of forensically relevant blood traces. Forensic Sci Int Genet 2020; 47:102280. [PMID: 32244163 DOI: 10.1016/j.fsigen.2020.102280] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 03/18/2020] [Accepted: 03/18/2020] [Indexed: 12/12/2022]
Abstract
Human blood traces are amongst the most commonly encountered biological stains collected at crime scenes. Identifying the body site of origin of a forensic blood trace can provide crucial information in many cases, such as in sexual and violent assaults. However, means for reliably and accurately identifying from which body site a forensic blood trace originated are missing, but would be highly valuable in crime scene investigations. With this study, we introduce a taxonomy-independent deep neural network approach based on massively parallel microbiome sequencing, which delivers accurate body site of origin classification of forensically-relevant blood samples, such as menstrual, nasal, fingerprick, and venous blood. A total of 50 deep neural networks were trained using a large 16S rRNA gene sequencing dataset from 773 reference samples, including 220 female urogenital tract, 190 nasal cavity, 213 skin, and 150 venous blood samples. Validation was performed with de-novo generated 16S rRNA gene massively parallel sequencing (MPS) data from 94 blood test samples of four different body sites, and achieved high classification accuracy with AUC values at 0.992 for menstrual blood (N = 23), 0.978 for nasal blood (N = 16), 0.978 for fingerprick blood (N = 30), and 0.990 for venous blood (N = 25). The obtained highly accurate classification of menstrual blood was independent of the day of the menses, as established in additional 86 menstrual blood test samples. Accurate body site of origin classification was also revealed for 45 fresh and aged mock casework blood samples from all four body sites. Our novel microbiome approach works based on the assumption that a sample is from blood, as can be obtained in forensic practise from prior presumptive blood testing, and provides accurate information on the specific body source of blood, with high potentials for future forensic applications.
Collapse
Affiliation(s)
- Celia Díez López
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Diego Montiel González
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands
| | - Cordula Haas
- Zurich Institute of Forensic Medicine, University of Zurich, Zurich, Switzerland
| | - Athina Vidaki
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Manfred Kayser
- Department of Genetic Identification, Erasmus MC University Medical Center Rotterdam, Rotterdam, the Netherlands.
| |
Collapse
|
217
|
Kappel BA, De Angelis L, Heiser M, Ballanti M, Stoehr R, Goettsch C, Mavilio M, Artati A, Paoluzi OA, Adamski J, Mingrone G, Staels B, Burcelin R, Monteleone G, Menghini R, Marx N, Federici M. Cross-omics analysis revealed gut microbiome-related metabolic pathways underlying atherosclerosis development after antibiotics treatment. Mol Metab 2020; 36:100976. [PMID: 32251665 PMCID: PMC7183232 DOI: 10.1016/j.molmet.2020.100976] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2019] [Revised: 02/27/2020] [Accepted: 03/08/2020] [Indexed: 12/24/2022] Open
Abstract
Objective The metabolic influence of gut microbiota plays a pivotal role in the pathogenesis of cardiometabolic diseases. Antibiotics affect intestinal bacterial diversity, and long-term usage has been identified as an independent risk factor for atherosclerosis-driven events. The aim of this study was to explore the interaction between gut dysbiosis by antibiotics and metabolic pathways with the impact on atherosclerosis development. Methods We combined oral antibiotics with different diets in an Apolipoprotein E-knockout mouse model linking gut microbiota to atherosclerotic lesion development via an integrative cross-omics approach including serum metabolomics and cecal 16S rRNA targeted metagenomic sequencing. We further investigated patients with carotid atherosclerosis compared to control subjects with comparable cardiovascular risk. Results Here, we show that increased atherosclerosis by antibiotics was connected to a loss of intestinal diversity and alterations of microbial metabolic functional capacity with a major impact on the host serum metabolome. Pathways that were modulated by antibiotics and connected to atherosclerosis included diminished tryptophan and disturbed lipid metabolism. These pathways were related to the reduction of certain members of Bacteroidetes and Clostridia by antibiotics in the gut. Patients with atherosclerosis presented a similar metabolic signature as those induced by antibiotics in our mouse model. Conclusion Taken together, this work provides insights into the complex interaction between intestinal microbiota and host metabolism. Our data highlight that detrimental effects of antibiotics on the gut flora are connected to a pro-atherogenic metabolic phenotype beyond classical risk factors. Antibiotics exacerbate atherosclerosis independently of diet. Gut microbiota and metabolic alpha diversity are reduced by antibiotics. Pathways connected to atherogenesis are tryptophan and lipid metabolism. Metabolic changes are linked to reduced Clostridia and Bacteroidetes in the gut.
Collapse
Affiliation(s)
- Ben Arpad Kappel
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Department of Internal Medicine 1, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Lorenzo De Angelis
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Michael Heiser
- Metabolomic discoveries GmbH, Potsdam, Germany; Metabolon Inc., Morrisville, NC, USA
| | - Marta Ballanti
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Center for Atherosclerosis, Policlinico Tor Vergata, Rome, Italy
| | - Robert Stoehr
- Department of Internal Medicine 1, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Claudia Goettsch
- Department of Internal Medicine 1, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Maria Mavilio
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Anna Artati
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | | | - Jerzy Adamski
- Research Unit Molecular Endocrinology and Metabolism, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany; Lehrstuhl für Experimentelle Genetik, Technical University of Munich, Freising-Weihenstephan, Germany; German Center for Diabetes Research (DZD e.V.), Neuherberg, Germany; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Geltrude Mingrone
- Department of Internal Medicine, Catholic University, Rome, Italy; Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy; Diabetes and Nutritional Sciences, Hodgkin Building, Guy's Campus, King's College London, London, United Kingdom
| | - Bart Staels
- Université Lille, INSERM, Centre Hospitalier Universitaire de Lille, Institut Pasteur de Lille, U1011-European Genomic Institute for Diabetes, Lille, France
| | - Remy Burcelin
- Institut des Maladies Métaboliques et Cardiovasculaires, INSERM U1048, Toulouse, France; Université Paul Sabatier, Toulouse, France
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Gastroenterology Unit, Policlinico Tor Vergata, Rome, Italy
| | - Rossella Menghini
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nikolaus Marx
- Department of Internal Medicine 1, University Hospital Aachen, RWTH Aachen University, Aachen, Germany
| | - Massimo Federici
- Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy; Center for Atherosclerosis, Policlinico Tor Vergata, Rome, Italy.
| |
Collapse
|
218
|
Ho KM, Kalgudi S, Corbett JM, Litton E. Gut microbiota in surgical and critically ill patients. Anaesth Intensive Care 2020; 48:179-195. [PMID: 32131606 DOI: 10.1177/0310057x20903732] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Microbiota-defined as a collection of microbial organisms colonising different parts of the human body-is now recognised as a pivotal element of human health, and explains a large part of the variance in the phenotypic expression of many diseases. A reduction in microbiota diversity, and replacement of normal microbes with non-commensal, pathogenic or more virulent microbes in the gastrointestinal tract-also known as gut dysbiosis-is now considered to play a causal role in the pathogenesis of many acute and chronic diseases. Results from animal and human studies suggest that dysbiosis is linked to cardiovascular and metabolic disease through changes to microbiota-derived metabolites, including trimethylamine-N-oxide and short-chain fatty acids. Dysbiosis can occur within hours of surgery or the onset of critical illness, even without the administration of antibiotics. These pathological changes in microbiota may contribute to important clinical outcomes, including surgical infection, bowel anastomotic leaks, acute kidney injury, respiratory failure and brain injury. As a strategy to reduce dysbiosis, the use of probiotics (live bacterial cultures that confer health benefits) or synbiotics (probiotic in combination with food that encourages the growth of gut commensal bacteria) in surgical and critically ill patients has been increasingly reported to confer important clinical benefits, including a reduction in ventilator-associated pneumonia, bacteraemia and length of hospital stay, in small randomised controlled trials. However, the best strategy to modulate dysbiosis or counteract its potential harms remains uncertain and requires investigation by a well-designed, adequately powered, randomised controlled trial.
Collapse
Affiliation(s)
- Kwok M Ho
- Department of Intensive Care Medicine, Royal Perth Hospital, Perth, Australia.,School of Veterinary and Life Sciences, Murdoch University, Perth, Australia.,Medical School, University of Western Australia, Perth, Australia
| | - Shankar Kalgudi
- Department of Intensive Care Medicine, Royal Perth Hospital, Perth, Australia
| | - Jade-Marie Corbett
- Department of Intensive Care Medicine, Royal Perth Hospital, Perth, Australia
| | - Edward Litton
- Medical School, University of Western Australia, Perth, Australia.,Department of Intensive Care Medicine, Fiona Stanley Hospital, Murdoch, Australia
| |
Collapse
|
219
|
Rukavina Mikusic NL, Kouyoumdzian NM, Choi MR. Gut microbiota and chronic kidney disease: evidences and mechanisms that mediate a new communication in the gastrointestinal-renal axis. Pflugers Arch 2020; 472:303-320. [PMID: 32064574 DOI: 10.1007/s00424-020-02352-x] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2020] [Revised: 01/06/2020] [Accepted: 01/21/2020] [Indexed: 02/06/2023]
Abstract
Chronic kidney disease (CKD) represents a growing public health problem associated with loss of kidney function and cardiovascular disease (CVD), the main leading cause of morbidity and mortality in CKD. It is well established that CKD is associated with gut dysbiosis. Over the past few years, there has been a growing interest in studying the composition of the gut microbiota in patients with CKD as well as the mechanisms by which gut dysbiosis contributes to CKD progression, in order to identify possible therapeutic targets to improve the morbidity and survival in CKD. The purpose of this review is to explore the clinical evidence and the mechanisms involved in the gut-kidney crosstalk as well as the possible interventions to restore a normal balance of the gut microbiota in CKD. It is well known that the influence of the gut microbiota on the gut-kidney axis acts in a reciprocal way: on the one hand, CKD significantly modifies the composition and functions of the gut microbiota. On the other hand, gut microbiota is able to manipulate the processes leading to CKD onset and progression through inflammatory, endocrine, and neurologic pathways. Understanding the complex interaction between these two organs (gut microbiota and kidney) may provide novel nephroprotective interventions to prevent the progression of CKD by targeting the gut microbiota. The review is divided into three main sections: evidences from clinical studies about the existence of a gut microbiota dysbiosis in CKD; the complex mechanisms that explain the bidirectional relationship between CKD and gut dysbiosis; and reports regarding the effects of prebiotic, probiotic, and synbiotic supplementation to restore gut microbiota balance in CKD.
Collapse
Affiliation(s)
- Natalia Lucía Rukavina Mikusic
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Buenos Aires, Argentina
- CONICET - Universidad de Buenos Aires, Instituto de Química y Fisicoquímica Biológicas (IQUIFIB), Buenos Aires, Argentina
| | - Nicolás Martín Kouyoumdzian
- CONICET - Universidad de Buenos Aires, Instituto Alberto C. Taquini de Investigaciones en Medicina Translacional (IATIMET), Buenos Aires, Argentina
| | - Marcelo Roberto Choi
- Universidad de Buenos Aires, Facultad de Farmacia y Bioquímica, Departamento de Ciencias Biológicas, Cátedra de Anatomía e Histología, Buenos Aires, Argentina.
- CONICET - Universidad de Buenos Aires, Instituto Alberto C. Taquini de Investigaciones en Medicina Translacional (IATIMET), Buenos Aires, Argentina.
| |
Collapse
|
220
|
Anhê FF, Jensen BAH, Varin TV, Servant F, Van Blerk S, Richard D, Marceau S, Surette M, Biertho L, Lelouvier B, Schertzer JD, Tchernof A, Marette A. Type 2 diabetes influences bacterial tissue compartmentalisation in human obesity. Nat Metab 2020; 2:233-242. [PMID: 32694777 DOI: 10.1038/s42255-020-0178-9] [Citation(s) in RCA: 161] [Impact Index Per Article: 32.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Accepted: 02/05/2020] [Indexed: 12/13/2022]
Abstract
Visceral obesity is a key risk factor for type 2 diabetes (T2D). Whereas gut dysbiosis appears to be instrumental for this relationship, whether gut-associated signatures translocate to extra-intestinal tissues and how this affects host metabolism remain elusive. Here we provide a comparative analysis of the microbial profile found in plasma, liver and in three distinct adipose tissues of individuals with morbid obesity. We explored how these tissue microbial signatures vary between individuals with normoglycaemia and those with T2D that were matched for body mass index. We identified tissue-specific signatures with higher bacterial load in the liver and omental adipose tissue. Gut commensals, but also environmental bacteria, showed tissue- and T2D-specific compartmentalisation. T2D signatures were most evident in mesenteric adipose tissue, in which individuals with diabetes displayed reduced bacterial diversity concomitant with fewer Gram-positive bacteria, such as Faecalibacterium, as opposed to enhanced levels of typically opportunistic Gram-negative Enterobacteriaceae. Plasma samples of individuals with diabetes were similarly enriched in Enterobacteriaceae, including the pathobiont Escherichia-Shigella. Our work provides evidence for the presence of selective plasma and tissue microbial signatures in individuals with severe obesity and identifies new potential microbial targets and biomarkers of T2D.
Collapse
Affiliation(s)
- Fernando F Anhê
- Québec Heart and Lung Research Institute, Laval University, Québec, Québec, Canada
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute and Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Benjamin Anderschou Holbech Jensen
- Québec Heart and Lung Research Institute, Laval University, Québec, Québec, Canada
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Thibault V Varin
- Québec Heart and Lung Research Institute, Laval University, Québec, Québec, Canada
| | | | | | - Denis Richard
- Québec Heart and Lung Research Institute, Laval University, Québec, Québec, Canada
| | - Simon Marceau
- Québec Heart and Lung Research Institute, Laval University, Québec, Québec, Canada
| | - Michael Surette
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute and Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - Laurent Biertho
- Québec Heart and Lung Research Institute, Laval University, Québec, Québec, Canada
| | | | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute and Centre for Metabolism Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada
| | - André Tchernof
- Québec Heart and Lung Research Institute, Laval University, Québec, Québec, Canada
| | - André Marette
- Québec Heart and Lung Research Institute, Laval University, Québec, Québec, Canada.
| |
Collapse
|
221
|
Guglielmetti S, Bernardi S, Del Bo' C, Cherubini A, Porrini M, Gargari G, Hidalgo-Liberona N, Gonzalez-Dominguez R, Peron G, Zamora-Ros R, Winterbone MS, Kirkup B, Kroon PA, Andres-Lacueva C, Riso P. Effect of a polyphenol-rich dietary pattern on intestinal permeability and gut and blood microbiomics in older subjects: study protocol of the MaPLE randomised controlled trial. BMC Geriatr 2020; 20:77. [PMID: 32102662 PMCID: PMC7045478 DOI: 10.1186/s12877-020-1472-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Accepted: 02/12/2020] [Indexed: 12/15/2022] Open
Abstract
Background During aging, alterations of the intestinal microbial ecosystem can occur contributing to immunosenescence, inflamm-aging and impairment of intestinal barrier function (increased intestinal permeability; IP). In the context of a diet-microbiota-IP axis in older subjects, food bioactives such as polyphenols may play a beneficial modulatory role. Methods MaPLE is a project centered on a randomized, controlled cross-over dietary intervention trial [polyphenol-rich diet (PR-diet) versus control diet (C-diet)] targeted to older people (≥ 60 y) living in a well-controlled setting (i.e. nursing home). The 8-week interventions are separated by an 8-week wash-out period. Three small portions per day of selected polyphenol-rich foods are consumed during intervention in substitution of other comparable products within the C-diet. Biological samples are collected before and after each treatment period to evaluate markers related to IP, inflammation, vascular function, oxidative stress, gut and blood microbiomics, metabolomics. A sample size of 50 subjects was defined based on IP as primary outcome. Discussion Evidence that increasing the consumption of polyphenol-rich food products can positively affect intestinal microbial ecosystem resulting in reduced IP and decreased translocation of inflammogenic bacterial factors into the bloodstream will be provided. The integration of data from gut and blood microbiomics, metabolomics and other IP-related markers will improve the understanding of the beneficial effect of the intervention in the context of polyphenols−microbiota−IP interactions. Finally, findings obtained will provide a proof of concept of the reliability of the dietary intervention, also contributing to future implementations of dietary guidelines directed to IP management in the older and other at risk subjects. Trial registration The trial is registered at (ISRCTN10214981); April 28, 2017.
Collapse
Affiliation(s)
- Simone Guglielmetti
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, 20133, Milan, Italy
| | - Stefano Bernardi
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, 20133, Milan, Italy
| | - Cristian Del Bo'
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, 20133, Milan, Italy
| | - Antonio Cherubini
- Geriatria, Accettazione Geriatrica e Centro di Ricerca per l'Invecchiamento, IRCCS INRCA, 60127, Ancona, Italy
| | - Marisa Porrini
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, 20133, Milan, Italy
| | - Giorgio Gargari
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, 20133, Milan, Italy
| | - Nicole Hidalgo-Liberona
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Technology Reference Net (XaRTA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain.,CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 08028, Barcelona, Spain
| | - Raul Gonzalez-Dominguez
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Technology Reference Net (XaRTA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain.,CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 08028, Barcelona, Spain
| | - Gregorio Peron
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Technology Reference Net (XaRTA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain.,CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 08028, Barcelona, Spain
| | - Raul Zamora-Ros
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Technology Reference Net (XaRTA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain.,Unit of Nutrition and Cancer, Cancer Epidemiology Research Programme, Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), Barcelona, Spain
| | - Mark S Winterbone
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Benjamin Kirkup
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Paul A Kroon
- Quadram Institute Bioscience, Norwich Research Park, Norwich, NR4 7UQ, UK
| | - Cristina Andres-Lacueva
- Biomarkers and Nutrimetabolomics Laboratory, Department of Nutrition, Food Sciences and Gastronomy, Food Technology Reference Net (XaRTA), Nutrition and Food Safety Research Institute (INSA), Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028, Barcelona, Spain.,CIBER de Fragilidad y Envejecimiento Saludable (CIBERfes), Instituto de Salud Carlos III, 08028, Barcelona, Spain
| | - Patrizia Riso
- Department of Food, Environmental and Nutritional Sciences (DeFENS), Università degli Studi di Milano, 20133, Milan, Italy.
| |
Collapse
|
222
|
Abstract
The review covers achievements and developments in the field of probiosis and prebiosis originating from sources other than dairy sources, mainly from plant material like cereals. The actual definitions of probiotic microorganisms, prebiotic, and postbiotic compounds and functional food are discussed. The presentation takes into account the relations between selected food components and their effect on probiotic bacteria, as well as effects on some health issues in humans. The review also focuses on the preservation of cereals using probiotic bacteria, adverse effects of probiotics and prebiotics, and novel possibilities for using probiotic bacteria in the food industry.
Collapse
|
223
|
Ogden HB, Child RB, Fallowfield JL, Delves SK, Westwood CS, Layden JD. The Gastrointestinal Exertional Heat Stroke Paradigm: Pathophysiology, Assessment, Severity, Aetiology and Nutritional Countermeasures. Nutrients 2020; 12:E537. [PMID: 32093001 PMCID: PMC7071449 DOI: 10.3390/nu12020537] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 02/14/2020] [Accepted: 02/15/2020] [Indexed: 12/12/2022] Open
Abstract
Exertional heat stroke (EHS) is a life-threatening medical condition involving thermoregulatory failure and is the most severe condition along a continuum of heat-related illnesses. Current EHS policy guidance principally advocates a thermoregulatory management approach, despite growing recognition that gastrointestinal (GI) microbial translocation contributes to disease pathophysiology. Contemporary research has focused to understand the relevance of GI barrier integrity and strategies to maintain it during periods of exertional-heat stress. GI barrier integrity can be assessed non-invasively using a variety of in vivo techniques, including active inert mixed-weight molecular probe recovery tests and passive biomarkers indicative of GI structural integrity loss or microbial translocation. Strenuous exercise is strongly characterised to disrupt GI barrier integrity, and aspects of this response correlate with the corresponding magnitude of thermal strain. The aetiology of GI barrier integrity loss following exertional-heat stress is poorly understood, though may directly relate to localised hyperthermia, splanchnic hypoperfusion-mediated ischemic injury, and neuroendocrine-immune alterations. Nutritional countermeasures to maintain GI barrier integrity following exertional-heat stress provide a promising approach to mitigate EHS. The focus of this review is to evaluate: (1) the GI paradigm of exertional heat stroke; (2) techniques to assess GI barrier integrity; (3) typical GI barrier integrity responses to exertional-heat stress; (4) the aetiology of GI barrier integrity loss following exertional-heat stress; and (5) nutritional countermeasures to maintain GI barrier integrity in response to exertional-heat stress.
Collapse
Affiliation(s)
- Henry B. Ogden
- Faculty of Sport, Health and Wellbeing, Plymouth MARJON University, Derriford Rd, Plymouth PL6 8BH, UK; (C.S.W.); (J.D.L.)
| | - Robert B. Child
- School of Chemical Engineering, University of Birmingham, Birmingham B15 2QU, UK;
| | | | - Simon K. Delves
- Institute of Naval Medicine, Alverstoke PO12 2DW, UK; (J.L.F.); (S.K.D.)
| | - Caroline S. Westwood
- Faculty of Sport, Health and Wellbeing, Plymouth MARJON University, Derriford Rd, Plymouth PL6 8BH, UK; (C.S.W.); (J.D.L.)
| | - Joseph D. Layden
- Faculty of Sport, Health and Wellbeing, Plymouth MARJON University, Derriford Rd, Plymouth PL6 8BH, UK; (C.S.W.); (J.D.L.)
| |
Collapse
|
224
|
Giannoni P, Claeysen S, Noe F, Marchi N. Peripheral Routes to Neurodegeneration: Passing Through the Blood-Brain Barrier. Front Aging Neurosci 2020; 12:3. [PMID: 32116645 PMCID: PMC7010934 DOI: 10.3389/fnagi.2020.00003] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 01/08/2020] [Indexed: 12/21/2022] Open
Abstract
A bidirectional crosstalk between peripheral players of immunity and the central nervous system (CNS) exists. Hence, blood-brain barrier (BBB) breakdown is emerging as a participant mechanism of dysregulated peripheral-CNS interplay, promoting diseases. Here, we examine the implication of BBB damage in neurodegeneration, linking it to peripheral brain-directed autoantibodies and gut-brain axis mechanisms. As BBB breakdown is a factor contributing to, or even anticipating, neuronal dysfunction(s), we here identify contemporary pharmacological strategies that could be exploited to repair the BBB in disease conditions. Developing neurovascular, add on, therapeutic strategies may lead to a more efficacious pre-clinical to clinical transition with the goal of curbing the progression of neurodegeneration.
Collapse
Affiliation(s)
| | - Sylvie Claeysen
- CNRS, INSERM U1191, Institut de Génomique Fonctionnelle, University of Montpellier, Montpellier, France
| | - Francesco Noe
- HiLIFE – Neuroscience Center, University of Helsinki, Helsinki, Finland
| | - Nicola Marchi
- CNRS, INSERM U1191, Institut de Génomique Fonctionnelle, University of Montpellier, Montpellier, France
| |
Collapse
|
225
|
Simões-Silva L, Araujo R, Pestana M, Soares-Silva I, Sampaio-Maia B. Peritoneal Microbiome in End-Stage Renal Disease Patients and the Impact of Peritoneal Dialysis Therapy. Microorganisms 2020; 8:173. [PMID: 31991821 PMCID: PMC7074711 DOI: 10.3390/microorganisms8020173] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 01/09/2020] [Accepted: 01/18/2020] [Indexed: 12/12/2022] Open
Abstract
Factors influencing the occurrence of peritoneal dialysis (PD)-related infections are still far from fully understood. Recent studies described the existence of specific microbiomes in body sites previously considered microbiome-free, unravelling new microbial pathways in the human body. In the present study, we analyzed the peritoneum of end-stage kidney disease (ESKD) patients to determine if they harbored a specific microbiome and if it is altered in patients on PD therapy. We conducted a cross-sectional study where the peritoneal microbiomes from ESKD patients with intact peritoneal cavities (ESKD non-PD, n = 11) and ESKD patients undergoing PD therapy (ESKD PD, n = 9) were analyzed with a 16S rRNA approach. Peritoneal tissue of ESKD patients contained characteristically low-abundance microbiomes dominated by Proteobacteria, Firmicutes, Actinobacteria, and Bacteroidetes. Patients undergoing PD therapy presented lower species richness, with dominance by the Pseudomonadaceae and Prevotelaceae families. This study provides the first characterization of the peritoneal microbiome in ESKD patients, bringing new insight to the human microbiome. Additionally, PD therapy may induce changes in this unique microbiome. The clinical relevance of these observations should be further explored to uncover the role of the peritoneal microbiome as a key element in the onset or aggravation of infection in ESKD patients, especially those undergoing PD.
Collapse
Affiliation(s)
- Liliana Simões-Silva
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-180 Porto, Portugal; (L.S.-S.); (R.A.); (M.P.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-180 Porto, Portugal
- Escola Superior de Saúde Dr. Lopes Dias, Instituto Politécnico de Castelo Branco, 6000-767 Castelo Branco, Portugal
| | - Ricardo Araujo
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-180 Porto, Portugal; (L.S.-S.); (R.A.); (M.P.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-180 Porto, Portugal
- Medical Biotechnology, Flinders University of South Australia, Bedford Park SA 5042, Australia
| | - Manuel Pestana
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-180 Porto, Portugal; (L.S.-S.); (R.A.); (M.P.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-180 Porto, Portugal
- Faculdade de Medicina, Universidade do Porto, 4200-319 Porto, Portugal
- Department of Nephrology, Centro Hospitalar Universitário de São João, EPE, 4200-319 Porto, Portugal
| | - Isabel Soares-Silva
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-180 Porto, Portugal; (L.S.-S.); (R.A.); (M.P.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-180 Porto, Portugal
- Centre of Molecular and Environmental Biology (CBMA), Department of Biology, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Benedita Sampaio-Maia
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-180 Porto, Portugal; (L.S.-S.); (R.A.); (M.P.)
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen, 208, 4200-180 Porto, Portugal
- Faculdade de Medicina Dentária, Universidade do Porto, 4200-393 Porto, Portugal
| |
Collapse
|
226
|
Hammad DBM, Hider SL, Liyanapathirana VC, Tonge DP. Molecular Characterization of Circulating Microbiome Signatures in Rheumatoid Arthritis. Front Cell Infect Microbiol 2020; 9:440. [PMID: 32039040 PMCID: PMC6987042 DOI: 10.3389/fcimb.2019.00440] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/09/2019] [Indexed: 12/12/2022] Open
Abstract
Rheumatoid Arthritis (RA) has been increasingly associated with perturbations to the microbial communities that reside in and on the body (the microbiome), in both human and animal studies. To date, such studies have mainly focused on the microbial communities that inhabit the gut and oral cavity. Mounting evidence suggests that microbial DNA can be detected in the blood circulation using a range of molecular methods. This DNA may represent an untapped pool of biomarkers that have the potential to report on changes to the microbiome of distant sites (e.g., example, the gut and oral cavity). To this end, through amplification and sequencing of the bacterial 16S rRNA variable region four, we evaluated the presence and identity of microbial DNA in blood samples obtained from RA patients (both prior to and 3 months following the instigation of treatment) in comparison to a small number of healthy control subjects and samples obtained from patients with ankylosing spondylitis (AS) and psoriatic arthritis (PA). Bacterial-derived DNA was identified in the majority of our patient samples. Taxonomic classification revealed that the microbiome community in RA was distinct from AS, PA, and the healthy state. Through analysis of paired patient samples obtained prior to and 3 months following treatment (V0 vs. V3), we found the microbiome to be modulated by treatment, and in many cases, this shift reduced the distance between these samples and the healthy control samples, suggesting a partial normalization following treatment in some patients. This effect was especially evident in seronegative arthritis patients. Herein, we provide further evidence for the existence of a blood microbiome in health and identify specific taxa modulated in disease and following treatment. These blood-derived signatures may have significant utility as disease biomarkers and suggest this area warrants further investigation.
Collapse
Affiliation(s)
- Dargham B. M. Hammad
- Faculty of Natural Sciences, School of Life Sciences, Keele University, Keele, United Kingdom
| | - S. L. Hider
- Arthritis Research UK Primary Care Centre, Research Institute for Primary Care and Health Sciences, Keele University, Keele, United Kingdom
- Haywood Academic Rheumatology Group, Midlands Partnership Foundation Trust, Staffordshire, United Kingdom
| | | | - Daniel P. Tonge
- Faculty of Natural Sciences, School of Life Sciences, Keele University, Keele, United Kingdom
| |
Collapse
|
227
|
Rodríguez-Rabassa M, López P, Sánchez R, Hernández C, Rodríguez C, Rodríguez-Santiago RE, Orengo JC, Green V, Yamamura Y, Rivera-Amill V. Inflammatory Biomarkers, Microbiome, Depression, and Executive Dysfunction in Alcohol Users. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2020; 17:ijerph17030689. [PMID: 31973090 PMCID: PMC7037324 DOI: 10.3390/ijerph17030689] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/11/2020] [Accepted: 01/13/2020] [Indexed: 02/07/2023]
Abstract
Alcohol-related disorders (ARD) are highly prevalent among Latin American-Caribbean countries. Mental disorders are common comorbidities in individuals with ARD. However, the etiology of the association between ARD and mental disorders remains unclear. We examined the association of inflammatory cytokines, microbiome, and other biomakers with measures of depression, social anxiety, and executive functions. We observed a significant increase in cytokine and chemokine expression levels in saliva and plasma in the alcohol group (AG) samples. Also, the salivary bacterial composition in the AG revealed an abundance of Prevotella. Depression symptomatology was markedly higher in the AG, but social anxiety levels were negligible. AG also exhibited executive dysfunctions, which negatively correlated with increased plasma levels of pro-inflammatory cytokines and increased salivary concentrations of Prevotella bacteria. Our study suggests that chronic alcohol use correlates with executive dysfunction, immune system dysregulation, and dysbiosis of the salivary microbiota. Additional studies are needed to understand the role of the microbiome and inflammation in alcohol use and mental comorbidities.
Collapse
Affiliation(s)
- Mary Rodríguez-Rabassa
- Center for Research Resources, Ponce Health Sciences University-Ponce Research Institute, Ponce, PR 00716-2348, USA; (M.R.-R.); (P.L.); (R.S.); (R.E.R.-S.); (Y.Y.)
- Clinical Psychology Program, Ponce Health Sciences University-Ponce Research Institute, Ponce, PR 00716-2348, USA; (C.H.); (C.R.)
| | - Pablo López
- Center for Research Resources, Ponce Health Sciences University-Ponce Research Institute, Ponce, PR 00716-2348, USA; (M.R.-R.); (P.L.); (R.S.); (R.E.R.-S.); (Y.Y.)
| | - Raphael Sánchez
- Center for Research Resources, Ponce Health Sciences University-Ponce Research Institute, Ponce, PR 00716-2348, USA; (M.R.-R.); (P.L.); (R.S.); (R.E.R.-S.); (Y.Y.)
| | - Cyanela Hernández
- Clinical Psychology Program, Ponce Health Sciences University-Ponce Research Institute, Ponce, PR 00716-2348, USA; (C.H.); (C.R.)
| | - Cesarly Rodríguez
- Clinical Psychology Program, Ponce Health Sciences University-Ponce Research Institute, Ponce, PR 00716-2348, USA; (C.H.); (C.R.)
| | - Ronald E. Rodríguez-Santiago
- Center for Research Resources, Ponce Health Sciences University-Ponce Research Institute, Ponce, PR 00716-2348, USA; (M.R.-R.); (P.L.); (R.S.); (R.E.R.-S.); (Y.Y.)
| | - Juan C. Orengo
- Public Health Program, Ponce Health Sciences University-Ponce Research Institute, Ponce, PR 00716-2348, USA; (J.C.O.); (V.G.)
| | - Vivian Green
- Public Health Program, Ponce Health Sciences University-Ponce Research Institute, Ponce, PR 00716-2348, USA; (J.C.O.); (V.G.)
| | - Yasuhiro Yamamura
- Center for Research Resources, Ponce Health Sciences University-Ponce Research Institute, Ponce, PR 00716-2348, USA; (M.R.-R.); (P.L.); (R.S.); (R.E.R.-S.); (Y.Y.)
| | - Vanessa Rivera-Amill
- Center for Research Resources, Ponce Health Sciences University-Ponce Research Institute, Ponce, PR 00716-2348, USA; (M.R.-R.); (P.L.); (R.S.); (R.E.R.-S.); (Y.Y.)
- Correspondence: ; Tel.: +1-(787)-841-5150; Fax: +1-(787)-841-5159
| |
Collapse
|
228
|
Wang Q, Zhao L, Han L, Fu G, Tuo X, Ma S, Li Q, Wang Y, Liang D, Tang M, Sun C, Wang Q, Song Q, Li Q. The differential distribution of bacteria between cancerous and noncancerous ovarian tissues in situ. J Ovarian Res 2020; 13:8. [PMID: 31954395 PMCID: PMC6969417 DOI: 10.1186/s13048-019-0603-4] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2019] [Accepted: 12/17/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND With the improvement of bacterial detection, the theory of the sterile female upper reproductive tract has been frequently challenged in recent years. However, thus far, no researchers have used ovaries as study targets. METHODS Six women who were diagnosed with ovarian cancer were included in the cancer group, and ten women who were diagnosed with a noncancerous ovarian condition (including three patients with uterine myoma and seven patients with uterine adenomyosis) were included in the control group. Immunohistochemistry staining using an antibacterial lipopolysaccharide (LPS) antibody was used to confirm the presence of bacteria in the ovarian tissues. In addition, 16S rRNA sequencing was used to compare the differences in the bacteria between ovarian cancer tissues and noncancerous ovarian tissues. BugBase and Phylogenetic Investigation of Communities by Reconstruction of Unobserved States (PICRUSt) were used to predict the functional composition of the bacteria. RESULTS Bacterial LPS was present in ovarian cancer tissue and noncancerous ovarian tissue, which implied the presence of bacteria in ovarian tissue. When compared to the noncancerous ovarian bacteria at the phylum level, the cancerous ovarian bacteria were composed of increased Aquificae and Planctomycetes and decreased Crenarchaeota. When predicting metagenomes, gene functions associated with the potentially pathogenic and the oxidative stress-tolerant phenotype were enriched in the ovaries of the cancer group. Forty-six significantly different KEGG pathways existed in the ovarian bacteria of the cancer group compared to that of the control group. CONCLUSIONS Different bacteria compositions were present in cancerous and noncancerous ovarian tissues. TRIAL REGISTRATION Chines Clinical Trail Registry, CHiCTR1800020018, Registered 11 September 2018, http://www.chictr.org.cn/.
Collapse
Affiliation(s)
- Qi Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- Department of Gynecological Oncology, Shaanxi Provincial Cancer Hospital, Xi'an, Shaanxi, China
| | - Lanbo Zhao
- Guipei 77, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Lu Han
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Guoxing Fu
- Omega Bioservices Inc, Norcross, GA, USA
| | - Xiaoqian Tuo
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Sijia Ma
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qing Li
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Yiran Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Dongxin Liang
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Miaomiao Tang
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Chao Sun
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qing Wang
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China
| | - Qing Song
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
- Cardiovascular Research Institute, Morehouse School of Medicine, Atlanta, Georgia, USA.
| | - Qiling Li
- Department of Obstetrics and Gynecology, First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, Shaanxi, China.
| |
Collapse
|
229
|
Microbiome signatures in neonatal central line associated bloodstream infections. PLoS One 2020; 15:e0227967. [PMID: 31945114 PMCID: PMC6964844 DOI: 10.1371/journal.pone.0227967] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2019] [Accepted: 01/03/2020] [Indexed: 12/14/2022] Open
Abstract
Neonates are at high risk for central line associated bloodstream infections (CLABSI). Biofilm formation is universal on indwelling catheters but why some biofilms seed the bloodstream to cause CLABSI is not clearly understood. With the objective to test the hypothesis that catheter biofilm microbiome in neonates with CLABSI differs than those without infection, we prospectively enrolled neonates (n = 30) with infected and uninfected indwelling central catheters. Catheters were collected at the time of removal, along with blood samples and skin swabs at the catheter insertion sites. Microbiomes of catheter biofilms, skin swabs and blood were evaluated by profiling the V4 region of the bacterial 16S rRNA gene using Illumina MiSeq sequencing platform. The microbial DNA load was higher from catheter biofilms of CLABSI patients without differences in alpha diversity when compared to that of the non-CLABSI neonates. Proteus and unclassified Staphylococcaceae were more abundant in infected catheter biofilms while Bradyrhizobium, Cloacibacterium, and Sphingomonas were more abundant in the uninfected catheters. A blood microbiome was detected in uninfected samples. The blood microbiome in CLABSI neonates clustered separately from the uninfected blood samples in beta diversity plots. We found that the microbiome signature in catheter biofilm and blood of neonates with CLABSI is different than the microbiomes of non-CLABSI neonates.
Collapse
|
230
|
Gao L, Rong X, He M, Zhang L, Li T, Wang W, Candotti D, Allain JP, Fu Y, Li C. Metagenomic analysis of potential pathogens from blood donors in Guangzhou, China. Transfus Med 2019; 30:61-69. [PMID: 31845424 DOI: 10.1111/tme.12657] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2019] [Revised: 10/25/2019] [Accepted: 12/02/2019] [Indexed: 12/16/2022]
Abstract
OBJECTIVES This study aimed to identify the emerging/reemerging pathogens in blood donation samples. BACKGROUND A metagenomic analysis has previously been used to look for pathogens but in this study, the relationship with aminotransferase (ALT) is described. METHODS/MATERIALS Excluding samples reactive to hepatitis B virus, hepatitis C virus, human immunodeficiency syndrome virus or syphilis and plasma samples were stratified into three groups of ALT levels (IU/L): A ≤ 50, B 51 to 69 and C ≥ 70, respectively. Each group was mixed in a pool of 100 samples, from which DNA and cDNA libraries were established for next generation sequencing and analysis. Pathogens of interest were identified by immunoassays, nested-polymerase chain reaction, phylogenetic analysis and pathogen detection in follow-up donors. RESULTS Several new or reemerging transfusion-transmitted pathogens were identified; Streptococcus suis, Babesia species and Toxoplasma gondii were found in the three ALT groups, Epstein-Barr virus (EBV) only in group C. Ten S. suis nucleic acid positive samples were detected, all closely phylogenetically related to reference strains. A donor in group A carried both S. suis genome and specific IgM in follow-up samples. This strain was identified as nontoxic S. suis. Five samples contained a short fragment of Babesia species SpeI-AvaI gene, while T. gondii was identified in 20 samples as a short fragment of 18S rDNA gene. In group C, two samples contained EBV genome. CONCLUSIONS Blood donations that contained S. suis, Babesia species and T. gondii sequences might represent potential transfusion risks. EBV, a potential cause of elevated ALT, was detected. Metagenomic analysis might be a useful technology for monitoring blood safety.
Collapse
Affiliation(s)
- Lei Gao
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, China
| | - Xia Rong
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,Institute of Blood Transfusion, Guangzhou Blood Center, Guangzhou, China
| | - Miao He
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, China
| | - Ling Zhang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Tingting Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Wenjing Wang
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| | - Daniel Candotti
- Department of Blood Transmitted Agents, National Institute of Blood Transfusion, Paris, France
| | - Jean-Pierre Allain
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,Division of Transfusion Medicine, University of Cambridge, Cambridge, United Kingdom
| | - Yongshui Fu
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China.,Institute of Blood Transfusion, Guangzhou Blood Center, Guangzhou, China
| | - Chengyao Li
- Department of Transfusion Medicine, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, China
| |
Collapse
|
231
|
Abstract
Bacterial translocation is a phenomenon in which live bacteria or their products cross the intestinal barrier to other organs or the circulatory system. Gut translocation of bacteria has been reported in both animal models, and clinical trials often accompany acute pancreatitis and are believed to be linked to patient outcome, especially in severe acute pancreatitis. Therefore, the mechanisms of intestinal bacterial translocation in acute pancreatitis have become a topic of interest in recent years. This review discusses Bacterial translocation in acute pancreatitis, identifies possible mechanisms of action, and provides an overview of the methods used to detect Bacterial translocation in acute pancreatitis. This review also highlights areas that require further research.
Collapse
Affiliation(s)
- Jinbo Liu
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, P.R. China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou,Sichuan, P.R. China
| | - Lin Huang
- Department of Paediatrics, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, P.R. China
| | - Ming Luo
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, P.R. China
| | - Xianming Xia
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, P.R. China.,Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou,Sichuan, P.R. China
| |
Collapse
|
232
|
Hammad DBM, Liyanapathirana V, Tonge DP. Molecular characterisation of the synovial fluid microbiome in rheumatoid arthritis patients and healthy control subjects. PLoS One 2019; 14:e0225110. [PMID: 31751379 PMCID: PMC6871869 DOI: 10.1371/journal.pone.0225110] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022] Open
Abstract
METHODS The presence and identity of bacterial and fungal DNA in the synovial fluid of rheumatoid arthritis (RA) patients and healthy control subjects was investigated through amplification and sequencing of the bacterial 16S rRNA gene and fungal internal transcribed spacer region 2 respectively. Synovial fluid concentrations of the cytokines IL-6, IL-17A, IL22 and IL-23 were determined by ELISA. RESULTS Bacterial 16S rRNA genes were detected in 87.5% RA patients, and all healthy control subjects. At the phylum level, the microbiome was predominated by Proteobacteria (Control = 83.5%, RA = 79.3%) and Firmicutes (Control = 16.1%, RA = 20.3%), and to a much lesser extent, Actinobacteria (Control = 0.2%, RA = 0.3%) and Bacteroidetes (Control = 0.1%, RA = 0.1%). Fungal DNA was identified in 75% RA samples, and 88.8% healthy controls. At the phylum level, synovial fluid was predominated by members of the Basidiomycota (Control = 53.9%, RA = 46.9%) and Ascomycota (Control = 35.1%, RA = 50.8%) phyla. Statistical analysis revealed key taxa that were differentially present or abundant dependent on disease status. CONCLUSIONS This study reports the presence of a synovial fluid microbiome, and determines that this is modulated by disease status (RA) as are other classical microbiome niches.
Collapse
Affiliation(s)
- Dargham Bayan Mohsen Hammad
- School of Life Sciences, Faculty of Natural Sciences, Keele University, Keele, Newcastle, England, United Kingdom
| | | | - Daniel Paul Tonge
- School of Life Sciences, Faculty of Natural Sciences, Keele University, Keele, Newcastle, England, United Kingdom
| |
Collapse
|
233
|
Abstract
The engineering of living cells and microbes is ushering in a new era of cancer therapy. Due to recent microbiome studies indicating the prevalence of bacteria within the human body and specifically in tumor tissue, bacteria have generated significant interest as potential targets for cancer therapy. Notably, a multitude of empirical studies over the past decades have demonstrated that administered bacteria home and grow in tumors due to reduced immune surveillance of tumor necrotic cores. Given their specificity for tumors, bacteria present a unique opportunity to be engineered as intelligent delivery vehicles for cancer therapy with synthetic biology techniques. In this review, we discuss the history, current state, and future challenges associated with using bacteria as a cancer therapy.
Collapse
|
234
|
Watts GS, Thornton JE, Youens-Clark K, Ponsero AJ, Slepian MJ, Menashi E, Hu C, Deng W, Armstrong DG, Reed S, Cranmer LD, Hurwitz BL. Identification and quantitation of clinically relevant microbes in patient samples: Comparison of three k-mer based classifiers for speed, accuracy, and sensitivity. PLoS Comput Biol 2019; 15:e1006863. [PMID: 31756192 PMCID: PMC6897419 DOI: 10.1371/journal.pcbi.1006863] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 12/06/2019] [Accepted: 10/10/2019] [Indexed: 12/15/2022] Open
Abstract
Infections are a serious health concern worldwide, particularly in vulnerable populations such as the immunocompromised, elderly, and young. Advances in metagenomic sequencing availability, speed, and decreased cost offer the opportunity to supplement or even replace culture-based identification of pathogens with DNA sequence-based diagnostics. Adopting metagenomic analysis for clinical use requires that all aspects of the workflow are optimized and tested, including data analysis and computational time and resources. We tested the accuracy, sensitivity, and resource requirements of three top metagenomic taxonomic classifiers that use fast k-mer based algorithms: Centrifuge, CLARK, and KrakenUniq. Binary mixtures of bacteria showed all three reliably identified organisms down to 1% relative abundance, while only the relative abundance estimates of Centrifuge and CLARK were accurate. All three classifiers identified the organisms present in their default databases from a mock bacterial community of 20 organisms, but only Centrifuge had no false positives. In addition, Centrifuge required far less computational resources and time for analysis. Centrifuge analysis of metagenomes obtained from samples of VAP, infected DFUs, and FN showed Centrifuge identified pathogenic bacteria and one virus that were corroborated by culture or a clinical PCR assay. Importantly, in both diabetic foot ulcer patients, metagenomic sequencing identified pathogens 4-6 weeks before culture. Finally, we show that Centrifuge results were minimally affected by elimination of time-consuming read quality control and host screening steps.
Collapse
Affiliation(s)
- George S. Watts
- University of Arizona Cancer Center and Department of Pharmacology, University of Arizona, Tucson, Arizona, United States of America
| | - James E. Thornton
- Department of Biosystems Engineering, University of Arizona, Tucson, Arizona, United States of America
| | - Ken Youens-Clark
- Department of Biosystems Engineering, University of Arizona, Tucson, Arizona, United States of America
| | - Alise J. Ponsero
- Department of Biosystems Engineering, University of Arizona, Tucson, Arizona, United States of America
| | - Marvin J. Slepian
- Department of Medicine, University of Arizona, Tucson, Arizona, United States of America
- Department of Biomedical Engineering, University of Arizona, Tucson, Arizona, United States of America
- Arizona Center for Accelerated Biomedical Innovation, University of Arizona, Tucson, Arizona, United States of America
| | - Emmanuel Menashi
- Honor Health Hospital, Scottsdale, Arizona, United States of America
| | - Charles Hu
- Dignity Health Chandler Regional Medical Center, Chandler, Arizona, United States of America
| | - Wuquan Deng
- Department of Endocrinology, Multidisciplinary Diabetic Foot Medical Center, Affiliated Central Hospital of Chongqing University, Chongqing, China
| | - David G. Armstrong
- Southwestern Academic Limb Salvage Alliance (SALSA), Department of Surgery, Keck School of Medicine of University of Southern California, Los Angeles, California, United States of America
| | - Spenser Reed
- University of Arizona Department of Family and Community Medicine, Tucson, Arizona, United States of America
| | - Lee D. Cranmer
- Department of Medicine, University of Washington and Fred Hutchinson Cancer Research Center, and Seattle Cancer Care Alliance, Seattle, Washington, United States of America
| | - Bonnie L. Hurwitz
- Department of Biosystems Engineering, University of Arizona, Tucson, Arizona, United States of America
- BIO5 Institute, University of Arizona, Tucson, Arizona, United States of America
| |
Collapse
|
235
|
Amar J, Lelouvier B, Servant F, Lluch J, Burcelin R, Bongard V, Elbaz M. Blood Microbiota Modification After Myocardial Infarction Depends Upon Low-Density Lipoprotein Cholesterol Levels. J Am Heart Assoc 2019; 8:e011797. [PMID: 31566105 PMCID: PMC6806051 DOI: 10.1161/jaha.118.011797] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background The role of bacteria on the onset of cardiovascular disease has been suggested. Reciprocally, increased intestinal bacterial translocation and bloodstream infection are common comorbidities associated with heart failure and myocardial infarction (MI). In this context, the aim of this study was to analyze the blood microbiome in patients shortly after acute myocardial infarction. Methods and Results We carried out a case control study comparing 103 patients at high cardiovascular risk but free of coronary disease and 99 patients who had an MI. The blood microbiome was analyzed both quantitatively by 16S quantitative polymerase chain reaction and qualitatively by 16S targeted metagenomic sequencing specifically optimized for blood samples. A significant increase in blood bacterial 16S rDNA concentration was observed in patients admitted for MI. This increase in blood bacterial DNA concentration was independent of post‐MI left ventricular function and was more marked in patients with low‐density lipoprotein cholesterol ≥1 g/L. In addition, differences in the proportion of numerous bacterial taxa in blood were significantly modified with the onset of MI, thus defining a blood microbiota signature of MI. Among the bacterial taxa whose proportions are decreased in patients with MI, at least 6 are known to include species able to metabolize cholesterol. Conclusions These results could provide the basis for the identification of blood microbiome‐based biomarkers for the stratification of MI patients. Furthermore, these findings should provide insight into the mechanism underlying the negative correlation reported between low‐density lipoprotein cholesterol concentration and the prognosis at the acute onset of MI and mortality. Clinical Trial Registration URL: http://www.clinicaltrials.gov. Unique identifier: NCT02405468.
Collapse
Affiliation(s)
- Jacques Amar
- Vaiomer Labège France.,Department of Therapeutics Rangueil Hospital CHU Toulouse Toulouse France.,INSERM U1048 I2MC Toulouse France
| | | | | | | | - Rémy Burcelin
- Vaiomer Labège France.,INSERM U1048 I2MC Toulouse France
| | - Vanina Bongard
- Epidemiology Department Rangueil Hospital CHU Toulouse Toulouse France.,Toulouse 3 Paul Sabatier University Toulouse France.,INSERM UMR 1027 Toulouse France
| | - Meyer Elbaz
- Metabolic and Cardiovascular Diseases Department Rangueil Hospital CHU Toulouse Toulouse France.,INSERM U1048 I2MC Toulouse France
| |
Collapse
|
236
|
Berthelot JM, Sellam J, Maugars Y, Berenbaum F. Cartilage-gut-microbiome axis: a new paradigm for novel therapeutic opportunities in osteoarthritis. RMD Open 2019; 5:e001037. [PMID: 31673418 PMCID: PMC6803002 DOI: 10.1136/rmdopen-2019-001037] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2019] [Revised: 07/31/2019] [Accepted: 09/06/2019] [Indexed: 12/12/2022] Open
Abstract
DNA of gut microbiota can be found in synovium of osteoarthritis and rheumatoid arthritis. This finding could result from the translocation of still alive bacteria from gut to joints through blood, since the diversified dormant microbiota of healthy human blood can be transiently resuscitated in vitro. The recent finding of gut microbiome in human cartilage, which differed between osteoarthritis and controls, suggests that a similar trafficking of dead or alive bacteria from gut microbiota physiologically occurs between gut and epiphysial bone marrow. Subchondral microbiota could enhance cartilage healing and transform components of deep cartilage matrix in metabolites with immunosuppressive properties. The differences of microbiome observed between hip and knee cartilage, either in osteoarthritis or controls, might be the counterpart of subtle differences in chondrocyte metabolism, themselves in line with differences in DNA methylation according to joints. Although bacteria theoretically cannot reach chondrocytes from the surface of intact cartilage, some bacteria enter the vascular channels of the epiphysial growth cartilage in young animals, whereas others can infect chondrocytes in vitro. In osteoarthritis, the early osteochondral plate angiogenesis may further enhance the ability of microbiota to locate close to the deeper layers of cartilage, and this might lead to focal dysbiosis, low-grade inflammation, cartilage degradation, epigenetic changes in chondrocytes and worsening of osteoarthritis. More studies on cartilage across different ethnic groups, weights, and according to age, are needed, to confirm the silent presence of gut microbiota close to human cartilage and better understand its physiologic and pathogenic significance.
Collapse
Affiliation(s)
- Jean-Marie Berthelot
- Rheumatology Unit, Nantes University Hospital, CHU Nantes, 44093 Nantes Cedex 01, France
| | - Jérémie Sellam
- Sorbonne University, Paris, France.,INSERM UMRS_938, CRSA, Paris, France.,Department of Rheumatology, Assistance Publique - Hôpitaux de Paris (AP-HP), Saint- Antoine Hospital, DMU 3iD, Paris, France
| | - Yves Maugars
- Rheumatology Unit, Nantes University Hospital, CHU Nantes, 44093 Nantes Cedex 01, France
| | - Francis Berenbaum
- Sorbonne University, Paris, France.,INSERM UMRS_938, CRSA, Paris, France.,Department of Rheumatology, Assistance Publique - Hôpitaux de Paris (AP-HP), Saint- Antoine Hospital, DMU 3iD, Paris, France
| |
Collapse
|
237
|
Thenappan T, Khoruts A, Chen Y, Weir EK. Can intestinal microbiota and circulating microbial products contribute to pulmonary arterial hypertension? Am J Physiol Heart Circ Physiol 2019; 317:H1093-H1101. [PMID: 31490732 DOI: 10.1152/ajpheart.00416.2019] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pulmonary arterial hypertension (PAH) is a fatal disease with a median survival of only 5-7 yr. PAH is characterized by remodeling of the pulmonary vasculature causing reduced pulmonary arterial compliance (PAC) and increased pulmonary vascular resistance (PVR), ultimately resulting in right ventricular failure and death. Better therapies for PAH will require a paradigm shift in our understanding of the early pathophysiology. PAC decreases before there is an increase in the PVR. Unfortunately, present treatment has little effect on PAC. The loss of compliance correlates with extracellular matrix remodeling and fibrosis in the pulmonary vessels, which have been linked to chronic perivascular inflammation and immune dysregulation. However, what initiates the perivascular inflammation and immune dysregulation in PAH is unclear. Alteration of the gut microbiota composition and function underlies the level of immunopathogenic involvement in several diseases, including atherosclerosis, obesity, diabetes mellitus, and depression, among others. In this review, we discuss evidence that raises the possibility of an etiologic role for changes in the gut and circulating microbiome in the initiation of perivascular inflammation in the early pathogenesis of PAH.
Collapse
Affiliation(s)
- Thenappan Thenappan
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - Alexander Khoruts
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Minnesota, Minneapolis, Minnesota.,Center for Immunology, University of Minnesota, Minneapolis, Minnesota.,BioTechnology Institute, University of Minnesota, Minneapolis, Minnesota
| | - Yingjie Chen
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| | - E Kenneth Weir
- Cardiovascular Division, Department of Medicine, University of Minnesota, Minneapolis, Minnesota
| |
Collapse
|
238
|
Costantini A, Takam Kamga P, Dumenil C, Chinet T, Emile JF, Giroux Leprieur E. Plasma Biomarkers and Immune Checkpoint Inhibitors in Non-Small Cell Lung Cancer: New Tools for Better Patient Selection? Cancers (Basel) 2019; 11:cancers11091269. [PMID: 31470546 PMCID: PMC6769436 DOI: 10.3390/cancers11091269] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 08/21/2019] [Accepted: 08/22/2019] [Indexed: 02/06/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) have transformed the treatment landscape for patients with non-small cell lung cancer (NSCLC). Although some patients can experience important response rates and improved survival, many others do not benefit from ICIs developing hyper-progressive disease or immune-related adverse events. This underlines the need to select biomarkers for ICIs use in order to better select patients. There is currently no universally validated robust biomarker for daily use of ICIs. Programmed death-ligand 1 (PD-L1) or tumor mutational burden (TMB) are sometimes used but still have several limitations. Plasma biomarkers are a promising approach in ICI treatment. This review will describe the development of novel plasma biomarkers such as soluble proteins, circulating tumor DNA (ctDNA), blood TMB, and blood microbiome in NSCLC patients treated with ICIs and their potential use in predicting response and toxicity.
Collapse
Affiliation(s)
- Adrien Costantini
- Department of Respiratory Diseases and Thoracic Oncology, APHP-Hôpital Ambroise Paré, 92100 Boulogne-Billancourt, France
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France
| | - Paul Takam Kamga
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France
| | - Coraline Dumenil
- Department of Respiratory Diseases and Thoracic Oncology, APHP-Hôpital Ambroise Paré, 92100 Boulogne-Billancourt, France
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France
| | - Thierry Chinet
- Department of Respiratory Diseases and Thoracic Oncology, APHP-Hôpital Ambroise Paré, 92100 Boulogne-Billancourt, France
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France
| | - Jean-François Emile
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France
- Department of Pathology, APHP-Hôpital Ambroise Pare, 92100 Boulogne-Billancourt, France
| | - Etienne Giroux Leprieur
- Department of Respiratory Diseases and Thoracic Oncology, APHP-Hôpital Ambroise Paré, 92100 Boulogne-Billancourt, France.
- EA 4340 BECCOH, UVSQ, Université Paris Saclay, 92100 Boulogne-Billancourt, France.
| |
Collapse
|
239
|
Abstract
PURPOSE OF REVIEW The host-microbiota relationship is integral in human health and can be rapidly disrupted in ways that may contribute to poor recovery from surgery or acute illness. We review key studies by organ system to understand the effect of perioperative and critical illness stress on the microbiota. Throughout the review, our focus is on potential interventions that may be mediated by the microbiome. RECENT FINDINGS Although any perioperative intervention can have a profound impact on the gut microbiota, it is less clear how such changes translate into altered health outcomes. Preoperative stress (anxiety, lack of sleep, fasting), intraoperative stress (surgery itself, volatile anesthetics, perioperative antibiotics, blood transfusions), and postoperative stress (sepsis, surgical site infections, acute respiratory distress syndrome, catecholamines, antibiotics, opioids, proton pump inhibitors) have all been associated with alterations of the commensal microflora. These factors (e.g. administration of antibiotics or opioids) can create a favorable environment for emergence of pathogen virulence and development of serious infections and multiorgan failure. Data to recommend therapies aimed at restoring a disrupted microbiota, such as probiotics/prebiotics and fecal microbiota transplants is currently scarce. SUMMARY The microbiome is likely to play an important role in the perioperative and ICU setting but existing data is largely descriptive. There is an expanding number of mechanistic studies that attempt to disentangle the complicated bi-directional relationship between the host and the resident microbiota. When these results are combined with ongoing clinical studies, we should be able to offer better therapies aimed at restoring the microbiota in the future.
Collapse
|
240
|
Merlini E, Cerrone M, van Wilgenburg B, Swadling L, Cannizzo ES, d'Arminio Monforte A, Klenerman P, Marchetti G. Association Between Impaired Vα7.2+CD161++CD8+ (MAIT) and Vα7.2+CD161-CD8+ T-Cell Populations and Gut Dysbiosis in Chronically HIV- and/or HCV-Infected Patients. Front Microbiol 2019; 10:1972. [PMID: 31555223 PMCID: PMC6722213 DOI: 10.3389/fmicb.2019.01972] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Accepted: 08/12/2019] [Indexed: 01/06/2023] Open
Abstract
Both HIV and HCV infections feature increased microbial translocation (MT) and gut dysbiosis that affect immune homeostasis and disease outcome. Given their commitment to antimicrobial mucosal immunity, we investigated mucosal-associated invariant T (MAIT) cells and Vα7.2+CD161- T-cell frequency/function and their possible associations with MT and gut dysbiosis, in chronic HIV and/or HCV infections. We enrolled 56 virally infected (VI) patients (pts): 13 HIV+ on suppressive cART (HIV-RNA < 40cp/ml), 13 HCV+ naive to DAA (direct-acting antiviral) anti-HCV agents; 30 HCV+/HIV+ on suppressive cART and naive to anti-HCV. 13 age-matched healthy controls (HC) were enrolled. For Vα7.2+CD161++ and Vα7.2+CD161-CD8+ T cells we assessed: activation (CD69), exhaustion (PD1/CD39), and cytolytic activity (granzymeB/perforin). Following PMA/ionomycin and Escherichia coli stimulation we measured intracellular IL17/TNFα/IFNγ. Markers of microbial translocation (Plasma LPS, 16S rDNA, EndoCAb and I-FABP) were quantified. In 5 patients per group we assessed stool microbiota composition by 16S targeted metagenomics sequencing (alpha/beta diversity, relative abundance). Compared to controls, virally infected pts displayed significantly lower circulating Vα7.2+CD161++CD8+ MAIT cells (p = 0.001), yet expressed higher perforin (p = 0.004) and granzyme B (p = 0.002) on CD8+ MAIT cells. Upon E. coli stimulation, the residual MAIT cells are less functional particularly those from HIV+/HCV+ patients. Conversely, in virally infected pts, Vα7.2+CD161-CD8+ cells were comparable in frequency, highly activated/exhausted (CD69+: p = 0.002; PD-1+: p = 0.030) and with cytolytic potential (perforin+: p < 0.0001), yet were poorly responsive to ex vivo stimulation. A profound gut dysbiosis characterized virally infected pts, especially HCV+/HIV+ co-infected patients, delineating a Firmicutes-poor/Bacteroidetes-rich microbiota, with significant associations with MAIT cell frequency/function. Irrespective of mono/dual infection, HIV+ and HCV+ patients display depleted, yet activated/cytolytic MAIT cells with reduced ex vivo function, suggesting an impoverished pool, possibly due to continuous bacterial challenge. The MAIT cell ability to respond to bacterial stimulation correlates with the presence of Firmicutes and Bacteroidetes, possibly suggesting an association between gut dysbiosis and MAIT cell function and posing viral-mediated dysbiosis as a potential key player in the hampered anti-bacterial MAIT ability.
Collapse
Affiliation(s)
- Esther Merlini
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Maddalena Cerrone
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy.,Chelsea and Westminster Hospital NHS Foundation Trust, London, United Kingdom
| | - Bonnie van Wilgenburg
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Leo Swadling
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - E Stefania Cannizzo
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Antonella d'Arminio Monforte
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Paul Klenerman
- Peter Medawar Building for Pathogen Research, University of Oxford, Oxford, United Kingdom
| | - Giulia Marchetti
- Department of Health Sciences, Clinic of Infectious Diseases, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| |
Collapse
|
241
|
Huggins LG, Koehler AV, Ng-Nguyen D, Wilcox S, Schunack B, Inpankaew T, Traub RJ. Assessment of a metabarcoding approach for the characterisation of vector-borne bacteria in canines from Bangkok, Thailand. Parasit Vectors 2019; 12:394. [PMID: 31395073 PMCID: PMC6686542 DOI: 10.1186/s13071-019-3651-0] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/01/2019] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Globally, bacterial vector-borne disease (VBD) exerts a large toll on dogs in terms of morbidity and mortality but nowhere is this more pronounced than in the tropics. Tropical environments permit a burgeoning diversity and abundance of ectoparasites some of which can transmit an extensive range of infectious agents, including bacteria, amongst others. Although some of these vector-borne bacteria are responsible for both animal and human diseases in the tropics, there is a scarcity of epidemiological investigation into these pathogens' prevalence. The situation is further exacerbated by frequent canine co-infection, complicating symptomatology that regular diagnostic techniques may miss or be unable to fully characterise. Such limitations draw attention to the need to develop screening tools capable of detecting a wide range of pathogens from a host simultaneously. RESULTS Here, we detail the employment of a next-generation sequencing (NGS) metabarcoding methodology to screen for the spectrum of bacterial VBD that are infecting semi-domesticated dogs across temple communities in Bangkok, Thailand. Our NGS detection protocol was able to find high levels of Ehrlichia canis, Mycoplasma haemocanis and Anaplasma platys infection rates as well as less common pathogens, such as "Candidatus Mycoplasma haematoparvum", Mycoplasma turicensis and Bartonella spp. We also compared our high-throughput approach to conventional endpoint PCR methods, demonstrating an improved detection ability for some bacterial infections, such as A. platys but a reduced ability to detect Rickettsia. CONCLUSIONS Our methodology demonstrated great strength at detecting coinfections of vector-borne bacteria and rare pathogens that are seldom screened for in canines in the tropics, highlighting its advantages over traditional diagnostics to better characterise bacterial pathogens in environments where there is a dearth of research.
Collapse
Affiliation(s)
- Lucas G. Huggins
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 3052 Australia
| | - Anson V. Koehler
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 3052 Australia
| | - Dinh Ng-Nguyen
- Faculty of Animal Sciences and Veterinary Medicine, Tay Nguyen University, Buon Ma Thuot, Dak Lak 630000 Vietnam
| | - Stephen Wilcox
- Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052 Australia
| | | | - Tawin Inpankaew
- Faculty of Veterinary Medicine, Kasetsart University, Bangkok, 10900 Thailand
| | - Rebecca J. Traub
- Faculty of Veterinary and Agricultural Sciences, University of Melbourne, Parkville, VIC 3052 Australia
| |
Collapse
|
242
|
Wang C, Li Q, Ren J. Microbiota-Immune Interaction in the Pathogenesis of Gut-Derived Infection. Front Immunol 2019; 10:1873. [PMID: 31456801 PMCID: PMC6698791 DOI: 10.3389/fimmu.2019.01873] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Accepted: 07/24/2019] [Indexed: 12/12/2022] Open
Abstract
Gut-derived infection is among the most common complications in patients who underwent severe trauma, serious burn, major surgery, hemorrhagic shock or severe acute pancreatitis (SAP). It could cause sepsis and multiple organ dysfunction syndrome (MODS), which are regarded as a leading cause of mortality in these cases. Gut-derived infection is commonly caused by pathological translocation of intestinal bacteria or endotoxins, resulting from the dysfunction of the gut barrier. In the last decades, the studies regarding to the pathogenesis of gut-derived infection mainly focused on the breakdown of intestinal epithelial tight junction and increased permeability. Limited information is available on the roles of intestinal microbial barrier in the development of gut-derived infection. Recently, advances of next-generation DNA sequencing techniques and its utilization has revolutionized the gut microecology, leading to novel views into the composition of the intestinal microbiota and its connections with multiple diseases. Here, we reviewed the recent progress in the research field of intestinal barrier disruption and gut-derived infection, mainly through the perspectives of the dysbiosis of intestinal microbiota and its interaction with intestinal mucosal immune cells. This review presents novel insights into how the gut microbiota collaborates with mucosal immune cells to involve the development of pathological bacterial translocation. The data might have important implication to better understand the mechanism underlying pathological bacterial translocation, contributing us to develop new strategies for prevention and treatment of gut-derived sepsis.
Collapse
Affiliation(s)
| | - Qiurong Li
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| | - Jianan Ren
- Research Institute of General Surgery, Jinling Hospital, Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
243
|
Tilg H, Zmora N, Adolph TE, Elinav E. The intestinal microbiota fuelling metabolic inflammation. Nat Rev Immunol 2019; 20:40-54. [DOI: 10.1038/s41577-019-0198-4] [Citation(s) in RCA: 377] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2019] [Indexed: 02/06/2023]
|
244
|
Ouaknine Krief J, Helly de Tauriers P, Dumenil C, Neveux N, Dumoulin J, Giraud V, Labrune S, Tisserand J, Julie C, Emile JF, Chinet T, Giroux Leprieur E. Role of antibiotic use, plasma citrulline and blood microbiome in advanced non-small cell lung cancer patients treated with nivolumab. J Immunother Cancer 2019; 7:176. [PMID: 31292005 PMCID: PMC6621972 DOI: 10.1186/s40425-019-0658-1] [Citation(s) in RCA: 68] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 06/27/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Recent data suggested a role of gut microbiota and antibiotic use on immune checkpoint inhibitors efficacy. We aimed to evaluate the impact of early use of antibiotic (EUA), blood microbiome and plasmatic citrulline (marker of the intestinal barrier) on nivolumab efficacy in non-small cell lung cancer (NSCLC). METHODS We included all consecutive patients with advanced NSCLC treated with nivolumab in our Department between 2014 and 2017. Blood microbiome was analyzed at month (M) M0 and M2. Citrulline rates were evaluated at M0, M2, M4 and M6. RESULTS Seventy-two patients were included (EUA in 42%). Overall survival (OS) was longer without EUA (median 13.4 months) than with EUA (5.1 months, p = 0.03). Thirty-five patients (49%) had plasma samples available. High citrulline rate (≥20 μM) at M0 was associated with tumor response (p = 0.084) and clinical benefit (nivolumab > 6 months) (p = 0.002). Median progression-free survival (PFS) was 7.9 months (high citrulline) vs 1.6 months (low citrulline) (p < 0.0001), and median OS were respectively non reached vs 2.2 months (p < 0.0001). Patients with EUA had lower median citrulline rates at M0: 21 μM (IQR 15.0-30.8) vs 32 μM (IQR 24.0-42.0) without EUA (p = 0.044). The presence of specific bacterial DNA in blood at M0 was associated with response and clinical benefit (Peptostreptococcae, Paludibaculum, Lewinella) or with tumor progression (Gemmatimonadaceae). Multivariate analyses on PFS and OS confirmed the prognostic role of citrulline and blood microbiome. CONCLUSIONS EUA is associated with shorter OS with nivolumab and lower citrulline rates. Plasma citrulline and blood microbiome appear to be promising predictive factors of nivolumab efficacy.
Collapse
Affiliation(s)
- Julia Ouaknine Krief
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Hopital Ambroise Pare, Boulogne-Billancourt, France
| | - Pierre Helly de Tauriers
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Hopital Ambroise Pare, Boulogne-Billancourt, France
| | - Coraline Dumenil
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Hopital Ambroise Pare, Boulogne-Billancourt, France
- EA 4340, UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Nathalie Neveux
- Department of Biochemistry, APHP – Hopital Cochin, Paris, France
| | - Jennifer Dumoulin
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Hopital Ambroise Pare, Boulogne-Billancourt, France
| | - Violaine Giraud
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Hopital Ambroise Pare, Boulogne-Billancourt, France
| | - Sylvie Labrune
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Hopital Ambroise Pare, Boulogne-Billancourt, France
| | - Julie Tisserand
- EA 4340, UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Catherine Julie
- EA 4340, UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
- Department of Pathology, APHP – Hopital Ambroise Pare, Boulogne-Billancourt, France
| | - Jean-François Emile
- EA 4340, UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
- Department of Pathology, APHP – Hopital Ambroise Pare, Boulogne-Billancourt, France
| | - Thierry Chinet
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Hopital Ambroise Pare, Boulogne-Billancourt, France
- EA 4340, UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| | - Etienne Giroux Leprieur
- Department of Respiratory Diseases and Thoracic Oncology, APHP – Hopital Ambroise Pare, Boulogne-Billancourt, France
- EA 4340, UVSQ, Université Paris-Saclay, Boulogne-Billancourt, France
| |
Collapse
|
245
|
You YA, Yoo JY, Kwon EJ, Kim YJ. Blood Microbial Communities During Pregnancy Are Associated With Preterm Birth. Front Microbiol 2019; 10:1122. [PMID: 31214131 PMCID: PMC6558066 DOI: 10.3389/fmicb.2019.01122] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 05/03/2019] [Indexed: 12/15/2022] Open
Abstract
Microbial infection of the placenta, amniotic fluid, vaginal canal, and oral cavity is known to significantly contribute to preterm birth (PTB). Although microbes can be translocated into the blood, little is known regarding the blood microbiota during pregnancy. To assess changes in the microbiome during pregnancy, blood samples were obtained 2 or 3 times during pregnancy from a cohort of 45 pregnant women enrolled between 2008 and 2010. To analyze the association with PTB, we conducted a case-control study involving 41 pregnant women upon admission for preterm labor and rupture of membrane (20 with term delivery; 21 with PTB). Bacterial diversity was assessed in number and composition between the first, second, and third trimesters in term delivered women according to 16S rRNA gene amplicon sequencing, and data were analyzed using Quantitative Insight Into Microbial Ecology (QIIME). Taxonomy was assigned using the GreenGenes 8.15.13 database. Dominant microorganisms at the phylum level in all pregnant women were identified as Firmicutes, Proteobacteria, Bacteroidetes, and Actinobacteria. However, the number and composition of bacteria in women with PTB differed from that in women with term delivery. Firmicutes and Bacteroidetes were more abundant in women with PTB than in women with term delivery, while Proteobacteria was less prevalent in women with PTB. At the genus level, Bacteroides, Lactobacillus, Sphingomonas, Fastidiosipila, Weissella, and Butyricicoccus were enriched in PTB samples. These observational results suggest that several taxa in the maternal blood microbiome are associated with PTB. Further studies are needed to confirm the composition of the blood microbiota in women with PTB. Additionally, the mechanism by which pathogenic microbes in maternal blood cause infection and PTB requires further analysis.
Collapse
Affiliation(s)
- Young-Ah You
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, Ewha Womans University School of Medicine, Seoul, South Korea
| | - Jae Young Yoo
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, Ewha Womans University School of Medicine, Seoul, South Korea
| | - Eun Jin Kwon
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, Ewha Womans University School of Medicine, Seoul, South Korea
| | - Young Ju Kim
- Department of Obstetrics and Gynecology, Ewha Medical Research Institute, Ewha Womans University School of Medicine, Seoul, South Korea
| |
Collapse
|
246
|
Circulating Microbiota-Based Metagenomic Signature for Detection of Hepatocellular Carcinoma. Sci Rep 2019; 9:7536. [PMID: 31101866 PMCID: PMC6525191 DOI: 10.1038/s41598-019-44012-w] [Citation(s) in RCA: 56] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Accepted: 05/07/2019] [Indexed: 02/07/2023] Open
Abstract
Circulating microbial dysbiosis is associated with chronic liver disease including nonalcoholic steatohepatitis and alcoholic liver disease. In this study, we evaluated whether disease-specific alterations of circulating microbiome are present in patients with cirrhosis and hepatocellular carcinoma (HCC), and their potential as diagnostic biomarkers for HCC. We performed cross-sectional metagenomic analyses of serum samples from 79 patients with HCC, 83 with cirrhosis, and 201 matching healthy controls, and validated the results in the same number of subjects. Serum bacterial DNA was analyzed using high-throughput pyrosequencing after amplification of the V3-V4 hypervariable regions of 16S rDNA. Blood microbial diversity was significantly reduced in HCC, compared with cirrhosis and control. There were significant differences in the relative abundances of several bacterial taxa that correlate with the presence of HCC, thus defining a specific blood microbiome-derived metagenomic signature of HCC. We identified 5 microbial gene markers-based model which distinguished HCC from controls with an area under the receiver-operating curve (AUC) of 0.879 and a balanced accuracy of 81.6%. In the validation, this model accurately distinguished HCC with an AUC of 0.875 and an accuracy of 79.8%. In conclusion, circulating microbiome-based signatures may be potential biomarkers for the detection HCC.
Collapse
|
247
|
Castillo DJ, Rifkin RF, Cowan DA, Potgieter M. The Healthy Human Blood Microbiome: Fact or Fiction? Front Cell Infect Microbiol 2019; 9:148. [PMID: 31139578 PMCID: PMC6519389 DOI: 10.3389/fcimb.2019.00148] [Citation(s) in RCA: 205] [Impact Index Per Article: 34.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 04/23/2019] [Indexed: 12/21/2022] Open
Abstract
The blood that flows perpetually through our veins and arteries performs numerous functions essential to our survival. Besides distributing oxygen, this vast circulatory system facilitates nutrient transport, deters infection and dispenses heat throughout our bodies. Since human blood has traditionally been considered to be an entirely sterile environment, comprising only blood-cells, platelets and plasma, the detection of microbes in blood was consistently interpreted as an indication of infection. However, although a contentious concept, evidence for the existence of a healthy human blood-microbiome is steadily accumulating. While the origins, identities and functions of these unanticipated micro-organisms remain to be elucidated, information on blood-borne microbial phylogeny is gradually increasing. Given recent advances in microbial-hematology, we review current literature concerning the composition and origin of the human blood-microbiome, focusing on bacteria and their role in the configuration of both the diseased and healthy human blood-microbiomes. Specifically, we explore the ways in which dysbiosis in the supposedly innocuous blood-borne bacterial microbiome may stimulate pathogenesis. In addition to exploring the relationship between blood-borne bacteria and the development of complex disorders, we also address the matter of contamination, citing the influence of contaminants on the interpretation of blood-derived microbial datasets and urging the routine analysis of laboratory controls to ascertain the taxonomic and metabolic characteristics of environmentally-derived contaminant-taxa.
Collapse
Affiliation(s)
- Diego J Castillo
- Department of Biochemistry, Genetics and Microbiology, Centre for Microbial Ecology and Genomics, University of Pretoria, Pretoria, South Africa
| | - Riaan F Rifkin
- Department of Biochemistry, Genetics and Microbiology, Centre for Microbial Ecology and Genomics, University of Pretoria, Pretoria, South Africa.,Human Origins and Palaeo Environmental Research Group, Department of Anthropology and Geography, Oxford Brookes University, Oxford, United Kingdom
| | - Don A Cowan
- Department of Biochemistry, Genetics and Microbiology, Centre for Microbial Ecology and Genomics, University of Pretoria, Pretoria, South Africa
| | - Marnie Potgieter
- Department of Biochemistry, Genetics and Microbiology, Centre for Microbial Ecology and Genomics, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
248
|
Affiliation(s)
- Robert D. Mair
- Department of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, California; and
- Department of Medicine, Stanford University, Palo Alto, California
| | - Tammy L. Sirich
- Department of Medicine, Veterans Affairs Palo Alto Health Care System, Palo Alto, California; and
- Department of Medicine, Stanford University, Palo Alto, California
| |
Collapse
|
249
|
The mucosal-luminal interface: an ideal sample to study the mucosa-associated microbiota and the intestinal microbial biogeography. Pediatr Res 2019; 85:895-903. [PMID: 30758325 DOI: 10.1038/s41390-019-0326-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Revised: 12/22/2018] [Accepted: 01/28/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Alterations in gastrointestinal microbial communities have been linked to human disease. Most studies use fecal samples as a proxy for the intestinal microbiota; however, the fecal microbiome is not fully representative of the mucosa-associated microbiota at the site of disease. While mucosal biopsies can be used instead, they often contain a high proportion of host DNA that can confound 16S ribosomal RNA (rRNA) gene sequencing studies. METHODS To overcome these limitations, we sampled the mucosal-luminal interface (MLI) to study the mucosa-associated microbiota. We also employed a simple bioinformatics workflow to remove contaminants from 16S rRNA gene profiling results. RESULTS Our results indicate that the microbial differences between individuals are greater than those between different microenvironments within the same individual. Moreover, biopsy samples frequently contained contaminants that could significantly impact biopsy profiling results. CONCLUSIONS Our findings highlight the utility of collecting MLI aspirates to complement biopsies and stools for characterizing human microbial communities.
Collapse
|
250
|
Li H, Li T, Qu J. Stochastic processes govern bacterial communities from the blood of pikas and from their arthropod vectors. FEMS Microbiol Ecol 2019; 94:4990947. [PMID: 29722798 DOI: 10.1093/femsec/fiy082] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 05/01/2018] [Indexed: 02/06/2023] Open
Abstract
Vector-borne microbes influence pathogen transmission and blood microbiomes, thereby affecting the emergence of infectious diseases. Thus, understanding the relationship between host and vector microbiomes is of importance. In this study, we investigated the bacterial community composition, diversity and assembly of the flea (Rhadinopsylla dahurica vicina), torsalo (Hypoderma curzonial), and the blood and gut of their shared pika host, Ochotona curzoniae. Bartonella, Sphingomonas and Bradyrhizobium were enriched in blood, while Wolbachia and Fusobacterium were more abundant in fleas and torsaloes. Most of potential pathogenic microbes (belonging to Fusobacterium, Rickettsia, Kingella, Porphyromonas, Bartonella and Mycoplasma) were present in the blood of pikas and their vectors. Blood communities were more similar to those from fleas than other sample types and were independent of host factors or geographical sites. Notably, blood microbes originate mainly from fleas rather than gut or torsaloes. Interestingly, the community assembly of blood, fleas or torsaloes was primarily governed by stochastic processes, while the gut microbiome was determined by deterministic processes. Ecological drift plays a dominant role in the assembly of blood and flea microbiomes. These results reflect the difficulty for predicting and regulating the microbial ecology of fleas for the prevention of potential microbiome-associated diseases.
Collapse
Affiliation(s)
- Huan Li
- Institute of Occupational Health and Environmental Health, School of Public Health, Lanzhou University, Lanzhou 730000, China
| | - Tongtong Li
- Department of Applied Biology, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou 310014, China
| | - Jiapeng Qu
- Key Laboratory of Adaptation and Evolution of Plateau Biota, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, Qinghai 810008, China.,Qinghai Provincial Key Laboratory of Restoration Ecology in Cold Region, Qinghai 810008, China
| |
Collapse
|