1
|
Brito JA, Martins SJ, Michaud A, Subbotin SA, Vau S, Pasche JM, Xue R, Stanley JD. Morphological, biochemical, and molecular characterization of the root-knot nematode Meloidogyne spartinae and assessment of bacterial assemblages in infected smooth cordgrass roots. Antonie Van Leeuwenhoek 2025; 118:78. [PMID: 40353944 DOI: 10.1007/s10482-025-02089-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/23/2025] [Indexed: 05/14/2025]
Abstract
Smooth cordgrass (Spartina alterniflora), a keystone species in marine ecosystems is crucial for maintaining the health and stability of coastal habitats. However, the cordgrass root-knot nematode (Meloidogyne spartinae), has been reported infecting this plant and still very little is known about this plant-parasitic nematode species well adapted to coastal saltmarsh. Morphological, biochemical, molecular, and gall microbiome analyses were performed using three M. spartinae populations to better characterized this nematode. M. spartinae females did not produce egg masses; females, eggs and males are found inside of the root galls. No esterase activity was detected using homogenates of one, five or ten females of M. spartinae per well regardless of the population. Nevertheless, one strong band of malate dehydrogenase activity (MDH = N1b phenotype) was detected using macerate of one female. Partial 18S rRNA, the D2-D3 expansion segments of 28S rRNA, partial COI and NAD5 gene sequences were obtained. The phylogenetical analyses showed that M. spartinae was related to M. marylandi and M. graminis. Microbiome analysis of cordgrass root galls revealed Proteobacteria as the most abundant phylum and Vibrio as the most abundant genus, in addition other environmentally important bacterial genera, such as those with nitrogen fixation and sulfate reducing roles. Our findings provide novel information on the bacterial assemblages of cordgrass infected roots and demonstrated via rRNA and mtDNA gene sequences that M. spartinae is clearly different from all other Meloidogyne spp.
Collapse
Affiliation(s)
- Janete A Brito
- Division of Plant Industry, Florida Department of Agriculture and Consumer Services, Gainesville, FL, 32608, USA.
| | - Samuel J Martins
- Department of Plant Pathology, University of Florida, Gainesville, FL, 32611, USA.
| | - Amy Michaud
- Plant Pest Diagnostic Center, California Department of Food and Agriculture, Sacramento, CA, 95832, USA
| | - Sergei A Subbotin
- Plant Pest Diagnostic Center, California Department of Food and Agriculture, Sacramento, CA, 95832, USA
- Center of Parasitology of A.N. Severtsov Institute of Ecology and Evolution of the Russian, Academy of Sciences, Moscow, Russia
| | - Silvia Vau
- Division of Plant Industry, Florida Department of Agriculture and Consumer Services, Gainesville, FL, 32608, USA
| | - Josephine M Pasche
- Department of Plant Pathology, University of Florida, Gainesville, FL, 32611, USA
| | - Ruimin Xue
- Division of Plant Industry, Florida Department of Agriculture and Consumer Services, Gainesville, FL, 32608, USA
| | - Jason D Stanley
- Division of Plant Industry, Florida Department of Agriculture and Consumer Services, Gainesville, FL, 32608, USA
| |
Collapse
|
2
|
El Houari A, Ranchou-Peyruse M, Carlier E, Ranchou-Peyruse A, Hirschler-Réa A, Bennisse R, Bouterfas R, McDonald JE, Guyoneaud R, Qatibi AI. Representatives of the Synergistaceae family, taxonomic description and genome sequence of Caenicola nitritireducens gen nov., sp. nov., a novel fermenting and amino-acid degrading bacterium isolated from a municipal anaerobic digester sludge. Syst Appl Microbiol 2025; 48:126607. [PMID: 40288042 DOI: 10.1016/j.syapm.2025.126607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 04/12/2025] [Accepted: 04/14/2025] [Indexed: 04/29/2025]
Abstract
Members of the phylum Synergistota are important but understudied components of microbial communities during anaerobic digestion. In this study, their diversity was assessed in full-scale anaerobic digester sludge samples from Marrakesh wastewater treatment plant (Morocco), using 16S rRNA gene community profiling, as well as targeted isolation, physiological characterization, and genome sequencing of novel Synergistaceae isolates. The 16S rRNA gene analysis identified 23 operational taxonomic units (OTUs) belonging to the family of Synergistaceae, representing 8.8 % of the total microbial community. 17 of these OTUs belonged to previously uncultured taxa. A dominant OTU19, presumably a new representative of the family of Synergistaceae was isolated in pure culture (strain DS-S4T) and subjected to both culture- and genome-based characterizations. Phylogenetic analysis revealed that strain DZ-S4T was related to Cloacibacillus porcorum CL-84T and Cloacibacillus evryensis 158T but with low sequence similarity of 89.94 % and 88.60 %, respectively. Based on genome relatedness, including Average Nucleotide Identity (ANI) and Amino Acid Identity (AAI), strain DZ-S4T is considered to represent a novel genus for which the name Caenicola gen.nov is proposed. Moreover, several phenotypic and eco-physiological properties differentiated the novel isolate from its related species, indicating that the strain represents a new species for which the name Caenicola nitritireducens sp. nov. is proposed, with strain DZ-S4T (=DSM 104940T = JCM 31897T) being the type strain. Additionally, this study investigates the ecological role of strain DZ-S4T, specifically the protein degradation, the bioconversion of carbohydrates, and the nitrite reduction during anaerobic digestion.
Collapse
Affiliation(s)
- Abdelaziz El Houari
- Anaerobic Microbiology Team (E02B26), Sciences and Techniques Faculty, Cadi Ayyad University, PO Box 549, 40 000 Marrakesh, Morocco; Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM UMR 5254, Pau, France.
| | - Magali Ranchou-Peyruse
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM UMR 5254, Pau, France; Université de Pau et des Pays de l'Adour, E2S UPPA, LaTEP, Pau, France
| | - Elisabeth Carlier
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM UMR 5254, Pau, France
| | | | - Agnès Hirschler-Réa
- Aix Marseille Univ, Université de Toulon, CNRS, IRD MIO UM110, Marseille, France
| | - Rhizlane Bennisse
- Anaerobic Microbiology Team (E02B26), Sciences and Techniques Faculty, Cadi Ayyad University, PO Box 549, 40 000 Marrakesh, Morocco
| | - Radia Bouterfas
- Anaerobic Microbiology Team (E02B26), Sciences and Techniques Faculty, Cadi Ayyad University, PO Box 549, 40 000 Marrakesh, Morocco; Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM UMR 5254, Pau, France
| | - James E McDonald
- Institute of Microbiology and Infection, School of Biosciences, University of Birmingham, Birmingham, UK
| | - Rémy Guyoneaud
- Université de Pau et des Pays de l'Adour, E2S UPPA, CNRS, IPREM UMR 5254, Pau, France
| | - Abdel-Illah Qatibi
- Anaerobic Microbiology Team (E02B26), Sciences and Techniques Faculty, Cadi Ayyad University, PO Box 549, 40 000 Marrakesh, Morocco.
| |
Collapse
|
3
|
Pimenta M, Alexa M, Mabandza DB, Dulaurent S, Huynh BT, Gaschet M, Opatowski L, Breurec S, Ploy MC, Dagot C. Wastewater-based AMR surveillance associated with tourism on a Caribbean island (Guadeloupe). J Glob Antimicrob Resist 2025; 43:27-34. [PMID: 40154781 DOI: 10.1016/j.jgar.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 02/23/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025] Open
Abstract
OBJECTIVES Antimicrobial resistance (AMR) is a major public health concern worldwide. International travel is a risk factor for acquiring antibiotic-resistant bacteria (ARB) and antibiotic-resistance genes (ARGs). Therefore, understanding the transmission of ARB and ARGs is instrumental in tackling AMR. This longitudinal study aimed to assess the benefit of wastewater monitoring in Guadeloupe to evaluate the role of tourism in the spread of AMR. METHODS A wastewater-based surveillance (WBS) study was conducted to monitor AMR in Guadeloupe in 2022 during dry and wet seasons. We characterized the resistome, microbiome and exposome of water samples collected in wastewater treatment facilities of two cities with different levels of tourism activities, in the content of aircraft toilets, and the pumping station receiving effluents from hotels. RESULTS The results show that the WBS approach facilitates the differentiation of various untreated effluents concerning exposome, microbiome, and resistome, offering insights into AMR dissemination. Additionally, the findings reveal that microbiome and exposome are comparable across sites and seasons, while resistome characterisation at specific locations may be pertinent for health surveillance. The microbiome of aircraft was predominantly composed of anaerobic bacteria from human intestinal microbiota, whereas the other locations exhibited a blend of human and environmental bacteria. Notably, individuals arriving by air have not introduced clinically significant resistance genes. Exposome compounds have been shown to influence the resistome's variance. CONCLUSIONS Clear differences were seen between the aircraft and the local sampling sites, indicating that the contribution of tourism to the observed resistance in Guadeloupe is not significant.
Collapse
Affiliation(s)
- Mélanie Pimenta
- INSERM, CHU Limoges, RESINFIT, U1092, University of Limoges, Limoges, France
| | - Maria Alexa
- Institut Pasteur, Université Paris-Cité, Epidemiology and Modelling of Antibiotic Evasion (EMAE), France; Anti-infective Evasion and Pharmacoepidemiology Team, CESP, Université Paris-Saclay, UVSQ, INSERM, Montigny-le-Bretonneux, France
| | - Degrâce Batantou Mabandza
- Transmission, Reservoir and Diversity of Pathogens Unit, Pasteur Institute of Guadeloupe, Pointe-à-Pitre, France
| | - Sylvain Dulaurent
- Department of Pharmacology, Toxicology and Pharmacovigilance, Limoges University Hospital, Limoges, France
| | - Bich-Tram Huynh
- Institut Pasteur, Université Paris-Cité, Epidemiology and Modelling of Antibiotic Evasion (EMAE), France; Anti-infective Evasion and Pharmacoepidemiology Team, CESP, Université Paris-Saclay, UVSQ, INSERM, Montigny-le-Bretonneux, France
| | - Margaux Gaschet
- INSERM, CHU Limoges, RESINFIT, U1092, University of Limoges, Limoges, France
| | - Lulla Opatowski
- Institut Pasteur, Université Paris-Cité, Epidemiology and Modelling of Antibiotic Evasion (EMAE), France; Anti-infective Evasion and Pharmacoepidemiology Team, CESP, Université Paris-Saclay, UVSQ, INSERM, Montigny-le-Bretonneux, France
| | - Sébastien Breurec
- Transmission, Reservoir and Diversity of Pathogens Unit, Pasteur Institute of Guadeloupe, Pointe-à-Pitre, France; Faculty of Medicine Hyacinthe Bastaraud, University of Antilles, Pointe-à-Pitre, France; Centre for Clinical Investigation 1424, INSERM, Pointe-à-Pitre/Les Abymes, France; Laboratory of clinical microbiology, University hospital Center of Guadeloupe, Pointe-à-Pitre/Les Abymes, France
| | - Marie-Cécile Ploy
- INSERM, CHU Limoges, RESINFIT, U1092, University of Limoges, Limoges, France
| | - Christophe Dagot
- INSERM, CHU Limoges, RESINFIT, U1092, University of Limoges, Limoges, France.
| |
Collapse
|
4
|
Mokrani M, Saad N, Nardy L, Sifré E, Despres J, Brochot A, Varon C, Urdaci MC. Biombalance™, an Oligomeric Procyanidins-Enriched Grape Seed Extract, Prevents Inflammation and Microbiota Dysbiosis in a Mice Colitis Model. Antioxidants (Basel) 2025; 14:305. [PMID: 40227242 PMCID: PMC11939601 DOI: 10.3390/antiox14030305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 04/15/2025] Open
Abstract
Inflammatory bowel disease (IBD) results from genetic factors, environmental factors, and intestinal microbiota interactions. This study investigated the effects of Biombalance™ (BB) in dextran sulphate sodium (DSS)-induced colitis in mice. BB extract exhibits high antioxidant activity, as determined by DPPH and ORAC tests. Mice were fed a standard diet, and BB was administered by gavage for ten days, before administration of 2.75% DSS in drinking water. BB significantly protected mice against DSS effects, as assessed by colon length, disease activity index (DAI) scores and colonic pathological damage. In addition, BB inhibited the expression of proinflammatory markers, such as IL-6, IL-17, CXCL1 and TNF-α, and the inflammatory mediators iNOS, TGF-β, FoxP3 and F4/80, while increasing IL-10 expression in the colon. BB modified microbiota composition, attenuating the microbial diversity lost due to DSS, increasing beneficial bacteria like Muribaculum, Lactobacillus, Muscispirillum, Roseburia and Bifidobacterium, and decreasing potentially harmful bacteria such as Proteobacteria and Enterococcus. Interestingly, microbiota-predicted functions using PICRUSt revealed that BB extract increases the antioxidant superpathway of ubiquinol biosynthesis, including ubiquinol-7, 8, 9 and 10 (CoenzymesQ). These findings suggest that Biombalance™ administration may help to reduce gut inflammation and oxidation, at least partly through modifications of the microbiota and its metabolites.
Collapse
Affiliation(s)
- Mohamed Mokrani
- University Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France; (M.M.); (N.S.); (L.N.)
- Bordeaux Sciences Agro, F-33175 Gradignan, France
- Groupe Berkem, 20 Rue Jean Duvert, F-33290 Blanquefort, France; (J.D.); (A.B.)
| | - Naima Saad
- University Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France; (M.M.); (N.S.); (L.N.)
- Univ. Limoges, LABCiS, UR 22722, F-87000 Limoges, France
| | - Ludivine Nardy
- University Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France; (M.M.); (N.S.); (L.N.)
- Bordeaux Sciences Agro, F-33175 Gradignan, France
| | - Elodie Sifré
- INSERM U1312 BRIC Bordeaux Institute of Oncology, Université de Bordeaux, F-33077 Bordeaux, France; (E.S.); (C.V.)
| | - Julie Despres
- Groupe Berkem, 20 Rue Jean Duvert, F-33290 Blanquefort, France; (J.D.); (A.B.)
| | - Amandine Brochot
- Groupe Berkem, 20 Rue Jean Duvert, F-33290 Blanquefort, France; (J.D.); (A.B.)
| | - Christine Varon
- INSERM U1312 BRIC Bordeaux Institute of Oncology, Université de Bordeaux, F-33077 Bordeaux, France; (E.S.); (C.V.)
| | - Maria C. Urdaci
- University Bordeaux, CNRS, Bordeaux INP, CBMN, UMR 5248, F-33600 Pessac, France; (M.M.); (N.S.); (L.N.)
- Bordeaux Sciences Agro, F-33175 Gradignan, France
| |
Collapse
|
5
|
Mittal A, Choudhary N, Kumari A, Yadav K, Maras JS, Sarin SK, Sharma S. Protein supplementation differentially alters gut microbiota and associated liver injury recovery in mouse model of alcohol-related liver disease. Clin Nutr 2025; 46:96-106. [PMID: 39892166 DOI: 10.1016/j.clnu.2025.01.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/10/2025] [Accepted: 01/18/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Patients with Alcohol-related Liver Disease (ALD) are advised increased protein supplementation. These nutrients are also available to gut microbiota. We evaluated the effects of protein supplementation from two sources, soya (veg) or egg, on gut microbiota modulation and ALD remission. METHODS ALD was induced in mice using the Lieber-DeCarli diet and incremental ethanol + thioacetamide (150 mg/kg body-weight,i.p.) twice-a-week. After 8wks, mice were fed standard (std.), egg (ovalbumin) or veg diet (20 % increase protein) for 7days. Biochemical parameters, hepatic proteome and gut microbiota composition were analyzed and correlated to capture liver and intestinal recovery. RESULTS Veg-diet decreased hepatic steatosis and fibrosis compared with std diet (83.3 %, p = 0.001 and 75 %, p = 0.01, respectively) or egg-diet (66.6 %, p = 0.03 and 25 %, p = 0.04, respectively). ALT and AST levels reduced by 40 % (p = 0.04) and 27.3 % (p = 0.04), respectively in veg diet compared to egg diet. Veg-diet increased intestinal claudin-3 (61 %, p = 0.02) and occludin (80 %, p = 0.001) compared to egg-diet. Plasma endotoxin levels in veg were reduced by 64 % and 32 % compared to std. (p = 0.04) or egg (p = 0.06). Veg-diet increased beneficial taxa, Lachnospiraceae UCG-006 (8.06-folds, p = 1.64E-25), Prevotellaceae NK3B31 (9.96-folds, p = 1.58E-36), Kurthia (8.11-folds, p = 3.98E-16) and Akkermansia (5.9-folds, p = 5.01E-75), while decreasing pathogenic Roseburia (-3.28-folds, p = 1.60E-06), Klebsiella (-5.7-folds, p = 1.55E-06), Staphylococcus (-5.3-folds, p = 1.62E-12). Hepatic proteome showed an increase in pyruvate, cysteine, methionine metabolism, bile acid biosynthesis, and glycolysis. CONCLUSION Alteration in protein alone can affect variable outcomes in ALD, with protein from vegetable sources resulting in enhanced improvement in the gut-liver axis. Vegetable protein-supplemented diet enhances fatty acid beta oxidation and energy metabolism accompanied by improvement in gut-dysbiosis and ALD associated hepatic injury.
Collapse
Affiliation(s)
- Ashi Mittal
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Nishu Choudhary
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Anupama Kumari
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Kavita Yadav
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Jaswinder Singh Maras
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shiv K Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Shvetank Sharma
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, New Delhi, India.
| |
Collapse
|
6
|
Mathew DE, Soni A, Dhimmar A, Gajjar A, Parab AS, Phakatkar SS, Sahastrabudhe H, Manohar CS, Shinde PB, Mantri VA. Characterization, Bio-Prospection, and Comparative Metagenomics of Bacterial Communities Revealing the Predictive Functionalities in Wild and Cultured Samples of Industrially Important Red Seaweed Gracilaria dura. Curr Microbiol 2025; 82:85. [PMID: 39821458 DOI: 10.1007/s00284-025-04065-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/30/2024] [Indexed: 01/19/2025]
Abstract
The present study explores the microbial community associated with the industrially important red seaweed Gracilaria dura to determine the diversity and biotechnological potential through culture and metagenomics approaches. In the first part of the investigation, we isolated and characterized 75 bacterial morphotypes, with varied colony characteristics and metabolic diversity from the wild seaweed. Phylogenetic analysis identified isolates in Proteobacteria, Firmicutes, and Actinobacteria, with Bacillus sp. being prevalent. B. licheniformis and Streptomyces sp. were notable in producing important enzymes like L-asparaginase, and polysaccharide lyases. Antimicrobial activity was significant in 21% of isolates, effective against seaweed pathogens such as Vibrio and Xanthomonas. Rhodococcus pyridinivorans showed strong pyridine degradation, suggesting bioremediation potential. Several isolates exhibited phosphate solubilization and nitrate indicating the roles of bacteria as algal growth promoters and biocontrol agents. Subsequent metagenome analysis of wild and cultured samples provides insights into bacterial communities associated with G. dura, revealing their distribution and functional roles. Proteobacteria (~ 95%) dominated the communities, further bacterial groups involved in algal growth, carpospore liberation, stress resistance, biogeochemical cycles, and biomedical applications were identified. A notable difference in bacteriomes was observed between the samples, with 25% remaining stable. The samples are cultured in the lab to generate seedlings for farming and serve as germplasm storage during the monsoon season. Microbiome surveys are crucial for understanding the association of pathogens and the overall health of the seedlings, supporting successful seaweed farming. Our findings provide valuable insights into G. dura-associated microbial communities and their role in algal growth, which has aquacultural implications.
Collapse
Affiliation(s)
- Doniya Elze Mathew
- Applied Phycology and Biotechnology Division, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, 364001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Aastha Soni
- Applied Phycology and Biotechnology Division, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, 364001, India
| | - Asmita Dhimmar
- Natural Products and Green Chemistry Division, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, 364001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Apexa Gajjar
- Natural Products and Green Chemistry Division, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, 364001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ashutosh Shankar Parab
- Biological Oceanography Division, CSIR-National Institute of Oceanography, Dona Paula, 403004, India
| | - Sumit Sudhir Phakatkar
- Biological Oceanography Division, CSIR-National Institute of Oceanography, Dona Paula, 403004, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Harshal Sahastrabudhe
- Natural Products and Green Chemistry Division, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, 364001, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Cathrine Sumathi Manohar
- Biological Oceanography Division, CSIR-National Institute of Oceanography, Dona Paula, 403004, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Pramod B Shinde
- Natural Products and Green Chemistry Division, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, 364001, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| | - Vaibhav A Mantri
- Applied Phycology and Biotechnology Division, CSIR-Central Salt and Marine Chemicals Research Institute, Bhavnagar, 364001, India.
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
7
|
Angebault C, Vandenborght L, Bassinet L, Wizla N, Ferroni A, Dessein R, Remus N, Thumerelle C, Fauchet N, Epaud R, Delhaes L, Botterel F. Airway Mycobiota-Microbiota During Pulmonary Exacerbation of Cystic Fibrosis Patients: A Culture and Targeted Sequencing Study. Mycoses 2025; 68:e70024. [PMID: 39816006 PMCID: PMC11736540 DOI: 10.1111/myc.70024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 01/03/2025] [Accepted: 01/06/2025] [Indexed: 01/18/2025]
Abstract
BACKGROUND The airways of patients with cystic fibrosis (pwCF) harbour complex fungal and bacterial microbiota involved in pulmonary exacerbations (PEx) and requiring antimicrobial treatment. Descriptive studies analysing bacterial and fungal microbiota concomitantly are scarce, especially using both culture and high-throughput-sequencing (HTS). OBJECTIVES We analysed bacterial-fungal microbiota and inter-kingdom correlations in two French CF centres according to clinical parameters and antimicrobial choices. METHODS Forty-eight pwCF with PEx from Creteil (n = 24) and Lille (n = 24) CF centres were included over 2 years. Sputa were collected for culture and targeted-HTS (ITS2 and V3-V4 targets). Sequencing and culture data, along with clinical, radiological and treatment data, were analysed. Two-level stratified analysis was performed to study potential confounding factors (age, CF mutation, FEV1 and antibiotics) on the centre factor. Inter-kingdom correlations were analysed. RESULTS Significant differences in the bacterial microbiota profile were found between centres (p-value = 0.03). For mycobiota, the taxonomic distribution and diversity were comparable. HTS provided concordant but more detailed information than culture and increased detection of main CF fungi (> 25% more positive samples for Aspergillus or Scedosporium). FEV1 and systemic antibiotic before PEx influenced bacterial microbiota, but no clinical association was found with the mycobiota. No inter-kingdom correlation between Pseudomonas and fungi was found. CONCLUSIONS Describing concomitant bacterial and fungal communities of pwCF at the beginning of PEx using culture and HTS shows greater diversity in HTS and better detection in case of low microbial load. Interesting inter-kingdom correlations were observed, requiring further research on larger cohorts to understand the potential microbial interactions.
Collapse
Affiliation(s)
- Cécile Angebault
- Unité de Parasitologie‐Mycologie, Département de Prévention, Diagnostic et Traitement Des InfectionsCHU Henri Mondor, Assistance Publique Des Hôpitaux de Paris (APHP)CreteilFrance
- UR Dynamyc 7380, Faculté de Santé, Univ Paris‐Est Creteil (UPEC), Ecole Nationale Vétérinaire d'Alfort (ENVA); USC Anses, Maisons‐Alfort, FranceCreteilFrance
| | | | - Laurence Bassinet
- Service de PneumologieCentre Hospitalier Intercommunal de CreteilCreteilFrance
| | - Nathalie Wizla
- Service de Gastro‐Entérologie, Hépatologie Pédiatrique, CHU LilleLilleFrance
| | - Agnès Ferroni
- Service de Microbiologie CliniqueCHU Necker‐Enfants Malades, Assistance Publique Des Hôpitaux de Paris (APHP)ParisFrance
| | | | - Natacha Remus
- Service de Pédiatrie GénéraleCentre Hospitalier Intercommunal de CreteilCreteilFrance
- Centre Des Maladies Respiratoires RaresCRCM, RespirareCreteilFrance
| | | | - Nathalie Fauchet
- Service de MicrobiologieCentre Hospitalier Intercommunal de CreteilCreteilFrance
| | - Ralph Epaud
- Service de Pédiatrie GénéraleCentre Hospitalier Intercommunal de CreteilCreteilFrance
- Centre Des Maladies Respiratoires RaresCRCM, RespirareCreteilFrance
- INSERM, IMRB, Univ Paris Est CreteilCreteilFrance
| | - Laurence Delhaes
- Service de Parasitologie‐MycologieCHU Bordeaux, Groupe Hospitalier PellegrinBordeauxFrance
- INSERM U1045, Université de BordeauxBordeauxFrance
| | - Françoise Botterel
- Unité de Parasitologie‐Mycologie, Département de Prévention, Diagnostic et Traitement Des InfectionsCHU Henri Mondor, Assistance Publique Des Hôpitaux de Paris (APHP)CreteilFrance
- UR Dynamyc 7380, Faculté de Santé, Univ Paris‐Est Creteil (UPEC), Ecole Nationale Vétérinaire d'Alfort (ENVA); USC Anses, Maisons‐Alfort, FranceCreteilFrance
| |
Collapse
|
8
|
Ketehouli T, Goss EM, Ascunce MS, Martins SJ. Metabolic and physiological effects of antibiotic-induced dysbiosis in citrus. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 287:117325. [PMID: 39541699 DOI: 10.1016/j.ecoenv.2024.117325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 10/21/2024] [Accepted: 11/09/2024] [Indexed: 11/16/2024]
Abstract
Streptomycin (Str) and oxytetracycline (Otc) are widely used antibiotics to manage bacterial diseases in citrus and other crops. However, their impacts on the rhizosphere bacterial assembly and plant physiology are poorly understood. The aim of this study was to examine the effects of Str and Otc on the physiology (assimilation, transpiration rate, intracellular CO2, and stomatal conductance to water vapor), rhizosphere bacterial assemblages (16S rRNA gene high-throughput amplicon sequencing), and rhizosphere metabolite profiles in healthy Citrus reticulata trees. The results indicated a reduction in photosynthesis after Str and Otc treatments, whereas CO2 outflow stayed constant. Both antibiotics decreased the culturable numbers of bacteria. Analysis of the microbiome showed changes in relative abundance of bacterial groups, specifically Pseudomonas, Agrobacterium, and Streptomyces, in response to the antibiotics. Metabolite profiles changed in streptomycin- and oxytetracycline-treated citrus plants suggesting response to microbe targets or induction of stress responses. This study advances knowledge of antibiotic-driven effects on the rhizosphere microbiome, rhizosphere metabolome, and plant physiology, which is essential for managing plant diseases while safeguarding rhizosphere ecology and long-term plant health.
Collapse
Affiliation(s)
- Toi Ketehouli
- Department of Plant Pathology, University of Florida, Gainesville, FL 32611, USA
| | - Erica M Goss
- Department of Plant Pathology, University of Florida, Gainesville, FL 32611, USA; Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610, USA
| | - Marina S Ascunce
- USDA-ARS Center for Medical, Agricultural, and Veterinary Entomology, Gainesville, FL 32608, USA
| | - Samuel J Martins
- Department of Plant Pathology, University of Florida, Gainesville, FL 32611, USA.
| |
Collapse
|
9
|
Reynaud Y, Gelasse A, Multigner L, Quénel P, Talarmin A, Guyomard-Rabenirina S. Looking for Pathogens in Dust from North Africa Arriving in the French West Indies Using Metabarcoding and Cultivable Analysis. Microorganisms 2024; 12:2111. [PMID: 39458420 PMCID: PMC11510511 DOI: 10.3390/microorganisms12102111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 09/25/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Periodically, the French West Indies receive dust originating from North Africa (NA). Microorganisms associated with desert dust can be transported over long distances through the atmosphere and could represent a means for the remote colonization of new habitats by putatively pathogenic microorganisms. The aim of this study was to determine the diversity and frequency of microbial agents (bacteria, eukaryotes) in NA dusts and the potential threat toward human and/or animal health by comparing microbial air composition during dust events and in control samples. In 2017 and 2018, 16 samples were collected during seven NA dust episodes and there were 9 controls. The microbial composition of the samples was characterized using a cultivable approach and by metabarcoding analyses (16S and 18S). A greater bacterial load and greater diversity were observed during the dust events, and some genera were significantly associated with the events. Some, such as Geodermatophilus, can be considered signature species of NA dust. No pathogenic species were found with the cultivable approach, whereas the metabarcoding analyses highlighted the presence of several potentially pathogenic species or known human pathogens such as Naegleria fowleri.
Collapse
Affiliation(s)
- Yann Reynaud
- Unité Transmission Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Guadeloupe, 97139 Les Abymes, France
| | - Andric Gelasse
- Unité Transmission Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Guadeloupe, 97139 Les Abymes, France
| | - Luc Multigner
- Institut de Recherche en Santé, Environnement et Travail, UMR_S 1085, INSERM, EHESP, University Rennes, 35000 Rennes, France (P.Q.)
| | - Philippe Quénel
- Institut de Recherche en Santé, Environnement et Travail, UMR_S 1085, INSERM, EHESP, University Rennes, 35000 Rennes, France (P.Q.)
| | - Antoine Talarmin
- Unité Transmission Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Guadeloupe, 97139 Les Abymes, France
| | - Stéphanie Guyomard-Rabenirina
- Unité Transmission Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Guadeloupe, 97139 Les Abymes, France
| |
Collapse
|
10
|
Vega-Carranza AS, Escamilla-Montes R, Fierro-Coronado JA, Diarte-Plata G, Guo X, García-Gutiérrez C, Luna-González A. Investigating the Effect of Bacilli and Lactic Acid Bacteria on Water Quality, Growth, Survival, Immune Response, and Intestinal Microbiota of Cultured Litopenaeus vannamei. Animals (Basel) 2024; 14:2676. [PMID: 39335265 PMCID: PMC11429436 DOI: 10.3390/ani14182676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 09/06/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
Shrimp is one of the most important aquaculture industries. Therefore, we determined the effect of nitrifying-probiotic bacteria on water quality, growth, survival, immune response, and intestinal microbiota of Litopenaeus vannamei cultured without water exchange. In vitro, only Bacillus licheniformis used total ammonia nitrogen (TAN), nitrites, and nitrates since nitrogen bubbles were produced. TAN decreased significantly in the treatments with B. licheniformis and Pediococcus pentosaceus and Leuconostoc mesenteroides, but no differences were observed in nitrites. Nitrates were significantly higher in the treatments with bacteria. The final weight was higher only with bacilli and bacilli and LAB treatments. The survival of shrimp in the bacterial treatments increased significantly, and superoxide anion increased significantly only in lactic acid bacteria (LAB) treatment. The activity of phenoloxidase decreased significantly in the treatments with bacteria compared to the control. Shrimp treated with bacilli in the water showed lower species richness. The gut bacterial community after treatments was significantly different from that of the control. Linoleic acid metabolism was positively correlated with final weight and superoxide anion, whereas quorum sensing was correlated with survival. Thus, bacilli and LAB in the water of hyperintensive culture systems act as heterotrophic nitrifers, modulate the intestinal microbiota and immune response, and improve the growth and survival of shrimp. This is the first report on P. pentosaceus and L. mesenteroides identified as nitrifying bacteria.
Collapse
Affiliation(s)
- Ana Sofía Vega-Carranza
- Instituto Politécnico Nacional, Centro Interdisciplinario de Investigación para el Desarrollo Integral Regional-Unidad Sinaloa, Departamento de Acuacultura, Boulevard Juan de Dios Bátiz Paredes #250, Col. San Joachín, Guasave 81101, Sinaloa, Mexico; (A.S.V.-C.); (J.A.F.-C.); (G.D.-P.); (C.G.-G.)
| | - Ruth Escamilla-Montes
- Instituto Politécnico Nacional, Centro Interdisciplinario de Investigación para el Desarrollo Integral Regional-Unidad Sinaloa, Departamento de Acuacultura, Boulevard Juan de Dios Bátiz Paredes #250, Col. San Joachín, Guasave 81101, Sinaloa, Mexico; (A.S.V.-C.); (J.A.F.-C.); (G.D.-P.); (C.G.-G.)
| | - Jesús Arturo Fierro-Coronado
- Instituto Politécnico Nacional, Centro Interdisciplinario de Investigación para el Desarrollo Integral Regional-Unidad Sinaloa, Departamento de Acuacultura, Boulevard Juan de Dios Bátiz Paredes #250, Col. San Joachín, Guasave 81101, Sinaloa, Mexico; (A.S.V.-C.); (J.A.F.-C.); (G.D.-P.); (C.G.-G.)
| | - Genaro Diarte-Plata
- Instituto Politécnico Nacional, Centro Interdisciplinario de Investigación para el Desarrollo Integral Regional-Unidad Sinaloa, Departamento de Acuacultura, Boulevard Juan de Dios Bátiz Paredes #250, Col. San Joachín, Guasave 81101, Sinaloa, Mexico; (A.S.V.-C.); (J.A.F.-C.); (G.D.-P.); (C.G.-G.)
| | - Xianwu Guo
- Instituto Politécnico Nacional, Centro de Biotecnología Genómica, Boulevard del Maestro S/N Esquina Elías Piña, Col. Narciso Mendoza, Reynosa 88710, Tamaulipas, Mexico;
| | - Cipriano García-Gutiérrez
- Instituto Politécnico Nacional, Centro Interdisciplinario de Investigación para el Desarrollo Integral Regional-Unidad Sinaloa, Departamento de Acuacultura, Boulevard Juan de Dios Bátiz Paredes #250, Col. San Joachín, Guasave 81101, Sinaloa, Mexico; (A.S.V.-C.); (J.A.F.-C.); (G.D.-P.); (C.G.-G.)
| | - Antonio Luna-González
- Instituto Politécnico Nacional, Centro Interdisciplinario de Investigación para el Desarrollo Integral Regional-Unidad Sinaloa, Departamento de Acuacultura, Boulevard Juan de Dios Bátiz Paredes #250, Col. San Joachín, Guasave 81101, Sinaloa, Mexico; (A.S.V.-C.); (J.A.F.-C.); (G.D.-P.); (C.G.-G.)
| |
Collapse
|
11
|
Ketehouli T, Pasche J, Buttrós VH, Goss EM, Martins SJ. The underground world of plant disease: Rhizosphere dysbiosis reduces above-ground plant resistance to bacterial leaf spot and alters plant transcriptome. Environ Microbiol 2024; 26:e16676. [PMID: 39010309 DOI: 10.1111/1462-2920.16676] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 07/03/2024] [Indexed: 07/17/2024]
Abstract
Just as the human gut microbiome is colonized by a variety of microbes, so too is the rhizosphere of plants. An imbalance in this microbial community, known as dysbiosis, can have a negative impact on plant health. This study sought to explore the effect of rhizosphere dysbiosis on the health of tomato plants (Solanum lycopersicum L.), using them and the foliar bacterial spot pathogen Xanthomonas perforans as model organisms. The rhizospheres of 3-week-old tomato plants were treated with either streptomycin or water as a control, and then spray-inoculated with X. perforans after 24 h. Half of the plants that were treated with both streptomycin and X. perforans received soil microbiome transplants from uninfected plant donors 48 h after the streptomycin was applied. The plants treated with streptomycin showed a 26% increase in disease severity compared to those that did not receive the antibiotic. However, the plants that received the soil microbiome transplant exhibited an intermediate level of disease severity. The antibiotic-treated plants demonstrated a reduced abundance of rhizobacterial taxa such as Cyanobacteria from the genus Cylindrospermum. They also showed a down-regulation of genes related to plant primary and secondary metabolism, and an up-regulation of plant defence genes associated with induced systemic resistance. This study highlights the vital role that beneficial rhizosphere microbes play in disease resistance, even against foliar pathogens.
Collapse
Affiliation(s)
- Toi Ketehouli
- Department of Plant Pathology, University of Florida, Gainesville, Florida, USA
| | - Josephine Pasche
- Department of Plant Pathology, University of Florida, Gainesville, Florida, USA
| | - Victor Hugo Buttrós
- Department of Plant Pathology, University of Florida, Gainesville, Florida, USA
| | - Erica M Goss
- Department of Plant Pathology, University of Florida, Gainesville, Florida, USA
- Emerging Pathogens Institute, University of Florida, Gainesville, Florida, USA
| | - Samuel J Martins
- Department of Plant Pathology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
12
|
Achudhan AB, Kannan P, Gupta A, Saleena LM. A Review of Web-Based Metagenomics Platforms for Analysing Next-Generation Sequence Data. Biochem Genet 2024; 62:621-632. [PMID: 37507643 DOI: 10.1007/s10528-023-10467-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 07/18/2023] [Indexed: 07/30/2023]
Abstract
Metagenomics has now evolved as a promising technology for understanding the microbial population in the environment. By metagenomics, a number of extreme and complex environment has been explored for their microbial population. Using this technology, researchers have brought out novel genes and their potential characteristics, which have robust applications in food, pharmaceutical, scientific research, and other biotechnological fields. A sequencing platform can provide a sequence of microbial populations in any given environment. The sequence needs to be analysed computationally to derive meaningful information. It is presumed that only bioinformaticians with extensive computational skills can process the sequencing data till the downstream end. However, numerous open-source software and online servers are available to analyse the metagenomic data developed for a biologist with less computational skills. This review is focused on bioinformatics tools such as Galaxy, CSI-NGS portal, ANASTASIA and SHAMAN, EBI- metagenomics, IDseq, and MG-RAST for analysing metagenomic data.
Collapse
Affiliation(s)
- Arunmozhi Bharathi Achudhan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Priya Kannan
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Annapurna Gupta
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India
| | - Lilly M Saleena
- Department of Biotechnology, School of Bioengineering, College of Engineering and Technology, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, India.
| |
Collapse
|
13
|
d'Humières C, Delavy M, Alla L, Ichou F, Gauliard E, Ghozlane A, Levenez F, Galleron N, Quinquis B, Pons N, Mullaert J, Bridier-Nahmias A, Condamine B, Touchon M, Rainteau D, Lamazière A, Lesnik P, Ponnaiah M, Lhomme M, Sertour N, Devente S, Docquier JD, Bougnoux ME, Tenaillon O, Magnan M, Ruppé E, Grall N, Duval X, Ehrlich D, Mentré F, Denamur E, Rocha EPC, Le Chatelier E, Burdet C. Perturbation and resilience of the gut microbiome up to 3 months after β-lactams exposure in healthy volunteers suggest an important role of microbial β-lactamases. MICROBIOME 2024; 12:50. [PMID: 38468305 DOI: 10.1186/s40168-023-01746-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 12/20/2023] [Indexed: 03/13/2024]
Abstract
BACKGROUND Antibiotics notoriously perturb the gut microbiota. We treated healthy volunteers either with cefotaxime or ceftriaxone for 3 days, and collected in each subject 12 faecal samples up to day 90. Using untargeted and targeted phenotypic and genotypic approaches, we studied the changes in the bacterial, phage and fungal components of the microbiota as well as the metabolome and the β-lactamase activity of the stools. This allowed assessing their degrees of perturbation and resilience. RESULTS While only two subjects had detectable concentrations of antibiotics in their faeces, suggesting important antibiotic degradation in the gut, the intravenous treatment perturbed very significantly the bacterial and phage microbiota, as well as the composition of the metabolome. In contrast, treatment impact was relatively low on the fungal microbiota. At the end of the surveillance period, we found evidence of resilience across the gut system since most components returned to a state like the initial one, even if the structure of the bacterial microbiota changed and the dynamics of the different components over time were rarely correlated. The observed richness of the antibiotic resistance genes repertoire was significantly reduced up to day 30, while a significant increase in the relative abundance of β-lactamase encoding genes was observed up to day 10, consistent with a concomitant increase in the β-lactamase activity of the microbiota. The level of β-lactamase activity at baseline was positively associated with the resilience of the metabolome content of the stools. CONCLUSIONS In healthy adults, antibiotics perturb many components of the microbiota, which return close to the baseline state within 30 days. These data suggest an important role of endogenous β-lactamase-producing anaerobes in protecting the functions of the microbiota by de-activating the antibiotics reaching the colon. Video Abstract.
Collapse
Affiliation(s)
- Camille d'Humières
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Microbial Evolutionary Genomics, Paris, 75015, France
| | - Margot Delavy
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie Et Pathogénicité Fongiques, Paris, F-75015, France
| | - Laurie Alla
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
| | - Farid Ichou
- ICANomics, Foundation of Innovation in Cardiometabolism and Nutrition (IHU ICAN), Paris, F-75013, France
| | - Emilie Gauliard
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, Paris, F-75012, France
| | - Amine Ghozlane
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, F-75015, France
| | - Florence Levenez
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
| | - Nathalie Galleron
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
| | - Benoit Quinquis
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
| | - Nicolas Pons
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
| | - Jimmy Mullaert
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Département d'Epidemiologie, Biostatistique and Recherche Clinique, Hôpital Bichat, Paris, F-75018, France
| | | | | | - Marie Touchon
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Microbial Evolutionary Genomics, Paris, 75015, France
| | - Dominique Rainteau
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, Paris, F-75012, France
| | - Antonin Lamazière
- Sorbonne Université, INSERM U938, Centre de Recherche Saint-Antoine, Paris, F-75012, France
| | - Philippe Lesnik
- INSERM UMR-S 1166, Institute of Cardiometabolism and Nutrition, Sorbonne Université, Hôpital Pitié-Salpêtrière, Paris, F-75013, France
- ICANomics, Foundation of Innovation in Cardiometabolism and Nutrition (IHU ICAN), Paris, F-75013, France
| | - Maharajah Ponnaiah
- ICANomics, Foundation of Innovation in Cardiometabolism and Nutrition (IHU ICAN), Paris, F-75013, France
| | - Marie Lhomme
- ICANomics, Foundation of Innovation in Cardiometabolism and Nutrition (IHU ICAN), Paris, F-75013, France
| | - Natacha Sertour
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie Et Pathogénicité Fongiques, Paris, F-75015, France
| | - Savannah Devente
- Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, I-53100, Italy
| | - Jean-Denis Docquier
- Dipartimento di Biotecnologie Mediche, Università di Siena, Siena, I-53100, Italy
| | - Marie-Elisabeth Bougnoux
- Institut Pasteur, Université Paris Cité, INRAE, USC2019, Unité Biologie Et Pathogénicité Fongiques, Paris, F-75015, France
- AP-HP, Unité de Parasitologie-Mycologie, Service de Microbiologie Clinique, Hôpital Necker-Enfants-Malades, Paris, F-75015, France
| | | | - Mélanie Magnan
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
| | - Etienne Ruppé
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Laboratoire de Bactériologie, Hôpital Bichat, Paris, F-75018, France
| | - Nathalie Grall
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Laboratoire de Bactériologie, Hôpital Bichat, Paris, F-75018, France
| | - Xavier Duval
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Centre d'Investigation Clinique, INSERM CIC 1425, Hôpital Bichat, Paris, F-75018, France
| | - Dusko Ehrlich
- Université Paris-Saclay, INRAE, MetaGenoPolis, Jouy-en-Josas, F-78350, France
- University College London, Institute for Neurology, London, UK
| | - France Mentré
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Département d'Epidemiologie, Biostatistique and Recherche Clinique, Hôpital Bichat, Paris, F-75018, France
| | - Erick Denamur
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France
- AP-HP, Laboratoire de Génétique Moléculaire, Hôpital Bichat, Paris, F-75018, France
| | - Eduardo P C Rocha
- Institut Pasteur, Université Paris Cité, CNRS UMR3525, Microbial Evolutionary Genomics, Paris, 75015, France
| | | | - Charles Burdet
- Université Paris Cité, IAME, INSERM, Paris, F-75018, France.
- AP-HP, Département d'Epidemiologie, Biostatistique and Recherche Clinique, Hôpital Bichat, Paris, F-75018, France.
| |
Collapse
|
14
|
Blondeaux A, Valibouze C, Speca S, Rousseaux C, Dubuquoy C, Blanquart H, Zerbib P, Desreumaux P, Foligné B, Titécat M. Changes in HLA-B27 Transgenic Rat Fecal Microbiota Following Tofacitinib Treatment and Ileocecal Resection Surgery: Implications for Crohn's Disease Management. Int J Mol Sci 2024; 25:2164. [PMID: 38396840 PMCID: PMC10889215 DOI: 10.3390/ijms25042164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 02/02/2024] [Accepted: 02/08/2024] [Indexed: 02/25/2024] Open
Abstract
The therapeutic management of Crohn's disease (CD), a chronic relapsing-remitting inflammatory bowel disease (IBD), is highly challenging. Surgical resection is sometimes a necessary procedure even though it is often associated with postoperative recurrences (PORs). Tofacitinib, an orally active small molecule Janus kinase inhibitor, is an anti-inflammatory drug meant to limit PORs in CD. Whereas bidirectional interactions between the gut microbiota and the relevant IBD drug are crucial, little is known about the impact of tofacitinib on the gut microbiota. The HLA-B27 transgenic rat is a good preclinical model used in IBD research, including for PORs after ileocecal resection (ICR). In the present study, we used shotgun metagenomics to first delineate the baseline composition and determinants of the fecal microbiome of HLA-B27 rats and then to evaluate the distinct impact of either tofacitinib treatment, ileocecal resection or the cumulative effect of both interventions on the gut microbiota in these HLA-B27 rats. The results confirmed that the microbiome of the HLA-B27 rats was fairly different from their wild-type littermates. We demonstrated here that oral treatment with tofacitinib does not affect the gut microbial composition of HLA-B27 rats. Of note, we showed that ICR induced an intense loss of bacterial diversity together with dramatic changes in taxa relative abundances. However, the oral treatment with tofacitinib neither modified the alpha-diversity nor exacerbated significant modifications in bacterial taxa induced by ICR. Collectively, these preclinical data are rather favorable for the use of tofacitinib in combination with ICR to address Crohn's disease management when considering microbiota.
Collapse
Affiliation(s)
- Aurélie Blondeaux
- U1286—INFINITE—Institute for Translational Research in Inflammation, CHU Lille, Inserm, Univ. Lille, F-59000 Lille, France; (A.B.); (C.V.); (S.S.); (P.Z.); (P.D.); (M.T.)
- Department of Hepato-Gastroenterology, Lille University Hospital, 59037 Lille, France
| | - Caroline Valibouze
- U1286—INFINITE—Institute for Translational Research in Inflammation, CHU Lille, Inserm, Univ. Lille, F-59000 Lille, France; (A.B.); (C.V.); (S.S.); (P.Z.); (P.D.); (M.T.)
- Department of Hepato-Gastroenterology, Lille University Hospital, 59037 Lille, France
| | - Silvia Speca
- U1286—INFINITE—Institute for Translational Research in Inflammation, CHU Lille, Inserm, Univ. Lille, F-59000 Lille, France; (A.B.); (C.V.); (S.S.); (P.Z.); (P.D.); (M.T.)
| | - Christel Rousseaux
- Intestinal Biotech Development, 1 Avenue Oscar Lambret, 59045 Lille, France; (C.R.); (C.D.)
| | - Caroline Dubuquoy
- Intestinal Biotech Development, 1 Avenue Oscar Lambret, 59045 Lille, France; (C.R.); (C.D.)
| | | | - Philippe Zerbib
- U1286—INFINITE—Institute for Translational Research in Inflammation, CHU Lille, Inserm, Univ. Lille, F-59000 Lille, France; (A.B.); (C.V.); (S.S.); (P.Z.); (P.D.); (M.T.)
- Department of Hepato-Gastroenterology, Lille University Hospital, 59037 Lille, France
| | - Pierre Desreumaux
- U1286—INFINITE—Institute for Translational Research in Inflammation, CHU Lille, Inserm, Univ. Lille, F-59000 Lille, France; (A.B.); (C.V.); (S.S.); (P.Z.); (P.D.); (M.T.)
- Department of Hepato-Gastroenterology, Lille University Hospital, 59037 Lille, France
| | - Benoît Foligné
- U1286—INFINITE—Institute for Translational Research in Inflammation, CHU Lille, Inserm, Univ. Lille, F-59000 Lille, France; (A.B.); (C.V.); (S.S.); (P.Z.); (P.D.); (M.T.)
| | - Marie Titécat
- U1286—INFINITE—Institute for Translational Research in Inflammation, CHU Lille, Inserm, Univ. Lille, F-59000 Lille, France; (A.B.); (C.V.); (S.S.); (P.Z.); (P.D.); (M.T.)
| |
Collapse
|
15
|
Sadeghi M, Mestivier D, Carbonnelle E, Benamouzig R, Khazaie K, Sobhani I. Loss of symbiotic and increase of virulent bacteria through microbial networks in Lynch syndrome colon carcinogenesis. Front Oncol 2024; 13:1313735. [PMID: 38375206 PMCID: PMC10876293 DOI: 10.3389/fonc.2023.1313735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 12/14/2023] [Indexed: 02/21/2024] Open
Abstract
Purpose Through a pilot study, we performed whole gut metagenomic analysis in 17 Lynch syndrome (LS) families, including colorectal cancer (CRC) patients and their healthy first-degree relatives. In a second asymptomatic LS cohort (n=150) undergoing colonoscopy-screening program, individuals with early precancerous lesions were compared to those with a normal colonoscopy. Since bacteria are organized into different networks within the microbiota, we compared related network structures in patients and controls. Experimental design Fecal prokaryote DNA was extracted prior to colonoscopy for whole metagenome (n=34, pilot study) or 16s rRNA sequencing (validation study). We characterized bacteria taxonomy using Diamond/MEGAN6 and DADA2 pipelines and performed differential abundances using Shaman website. We constructed networks using SparCC inference tools and validated the construction's accuracy by performing qPCR on selected bacteria. Results Significant differences in bacterial communities in LS-CRC patients were identified, with an enrichment of virulent bacteria and a depletion of symbionts compared to their first-degree relatives. Bacteria taxa in LS asymptomatic individuals with colonic precancerous lesions (n=79) were significantly different compared to healthy individuals (n=71). The main bacterial network structures, constructed based on bacteria-bacteria correlations in CRC (pilot study) and in asymptomatic precancerous patients (validation-study), showed a different pattern than in controls. It was characterized by virulent/symbiotic co-exclusion in both studies and illustrated (validation study) by a higher Escherichia/Bifidobacterium ratio, as assessed by qPCR. Conclusion Enhanced fecal virulent/symbiotic bacteria ratios influence bacterial network structures. As an early event in colon carcinogenesis, these ratios can be used to identify asymptomatic LS individual with a higher risk of CRC.
Collapse
Affiliation(s)
- Mohammad Sadeghi
- EA7375 –EC2M3: Early detection of Colonic Cancer by using Microbial & Molecular Markers Paris East Créteil University (UPEC), Créteil, France
| | - Denis Mestivier
- EA7375 –EC2M3: Early detection of Colonic Cancer by using Microbial & Molecular Markers Paris East Créteil University (UPEC), Créteil, France
| | - Etienne Carbonnelle
- Bacteriology, Virology, Hygiene Laboratory, Assistance Publique–Hôpitaux de Paris (APHP), Avicenne Hospital, Bobigny, France
| | - Robert Benamouzig
- Department of Gastroenterology, Assistance Publique–Hôpitaux de Paris (APHP), Avicenne Hospital, Bobigny, France
| | | | - Iradj Sobhani
- EA7375 –EC2M3: Early detection of Colonic Cancer by using Microbial & Molecular Markers Paris East Créteil University (UPEC), Créteil, France
- Department of Gastroenterology, Assistance Publique–Hôpitaux de Paris (APHP), Henri Mondor Hospital, Créteil, France
| |
Collapse
|
16
|
Khan S, Banerjee G, Setua S, Jones DH, Chauhan BV, Dhasmana A, Banerjee P, Yallapu MM, Behrman S, Chauhan SC. Metagenomic analysis unveils the microbial landscape of pancreatic tumors. Front Microbiol 2023; 14:1275374. [PMID: 38179448 PMCID: PMC10764597 DOI: 10.3389/fmicb.2023.1275374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 11/20/2023] [Indexed: 01/06/2024] Open
Abstract
The composition of resident microbes in the human body is linked to various diseases and their treatment outcomes. Although studies have identified pancreatic ductal adenocarcinoma (PDAC)-associated bacterial communities in the oral and gut samples, herein, we hypothesize that the prevalence of microbiota in pancreatic tumor tissues is different as compared with their matched adjacent, histologically normal appearing tissues, and these microbial molecular signatures can be highly useful for PDAC diagnosis/prognosis. In this study, we performed comparative profiling of bacterial populations in pancreatic tumors and their respective adjacent normal tissues using 16S rRNA-based metagenomics analysis. This study revealed a higher abundance of Proteobacteria and Actinomycetota in tumor tissues compared with adjacent normal tissues. Interestingly, the linear discriminant analysis (LDA) scores unambiguously revealed an enrichment of Delftia in tumor tissues, whereas Sphingomonas, Streptococcus, and Citrobacter exhibited a depletion in tumor tissues. Furthermore, we analyzed the microbial composition between different groups of patients with different tumor differentiation stages. The bacterial genera, Delftia and Staphylococcus, were very high at the G1 stages (well differentiated) compared with G2 (well to moderate/moderately differentiated) and G3/G4 (poorly differentiated) stages. However, the abundance of Actinobacter and Cloacibacterium was found to be very high in G2 and G3, respectively. Additionally, we evaluated the correlation of programmed death-ligand (PDL1) expression with the abundance of bacterial genera in tumor lesions. Our results indicated that three genera such as Streptomyces, Cutibacterium, and Delftia have a positive correlation with PD-L1 expression. Collectively, these findings demonstrate that PDAC lesions harbor relatively different microbiota compared with their normal tumor adjacent tissues, and this information may be helpful for the diagnosis and prognosis of PADC patients.
Collapse
Affiliation(s)
- Sheema Khan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, the University of Texas Rio Grande Valley, McAllen, TX, United States
| | - Goutam Banerjee
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 105 Agricultural Bioprocess Laboratory, Urbana, IL, United States
| | - Saini Setua
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, United States
- Center for Blood Oxygen Transport and Hemostasis (CBOTH), Department of Pediatrics, University of Maryland, Baltimore, MD, United States
| | - Daleniece Higgins Jones
- Division of Epidemiology, Biostatistics, and Environmental Health, University of Memphis, Memphis, TN, United States
- Department of Public Health, University of Tennessee, Knoxville, TN, United States
| | - Bhavin V. Chauhan
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 105 Agricultural Bioprocess Laboratory, Urbana, IL, United States
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, United States
| | - Pratik Banerjee
- Department of Food Science and Human Nutrition, University of Illinois at Urbana-Champaign, 105 Agricultural Bioprocess Laboratory, Urbana, IL, United States
- Division of Epidemiology, Biostatistics, and Environmental Health, University of Memphis, Memphis, TN, United States
| | - Murali Mohan Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, the University of Texas Rio Grande Valley, McAllen, TX, United States
| | - Stephen Behrman
- Department of Surgery, Baptist Memorial Hospital and Medical Education, Memphis, TN, United States
| | - Subhash C. Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, Edinburg, TX, United States
- South Texas Center of Excellence in Cancer Research, School of Medicine, the University of Texas Rio Grande Valley, McAllen, TX, United States
| |
Collapse
|
17
|
Medeot DB, Nilson A, Miazzo RD, Grosso V, Ferrari W, Jofré E, Soltermann A, Peralta MF. Stevia as a natural additive on gut health and cecal microbiota in broilers. Vet Anim Sci 2023; 22:100322. [PMID: 38045012 PMCID: PMC10692954 DOI: 10.1016/j.vas.2023.100322] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2023] Open
Abstract
Stevia mash (SM), leaves of Stevia rebaudiana Bertoni plant, is an additive used in poultry that enhances growth and health. Objective: to determine the effect of 1 % SM on productive parameters, gut health, and the cecal microbiome in broilers between the first 15 and 21 days old. One hundred sixty male, 1-day-old broilers (48.5 ± 2.5 g) were divided into Control (C) without SM and Treated (T) with 1 % SM on diet, during 15/21 days. Each subgroup had eight broilers/five repetitions/treatment. At day 15 or 21, all broilers were dissected, Fabricius Bursa and Gut removed and processed for histomorphometry, followed by Villi Height/Crypt Deep (VH/CD) ratio. Conversion Index (CI) was determined. The V3-V4 region of 16S rRNA gene was amplified from DNA obtained from pooled cecal contents and sequenced on Illumina Miseq PE 2 × 250 platform. Sequence processing and taxonomic assignments were performed using the SHAMAN pipeline. Both T groups have better VH/CD Ratios than C groups (p ≤ 0.05). In guts, increased plasmatic and goblet cells number and thicker mucus layer were found in T15 and T21. All groups received SM showed early immunological maturity in Fabricius Bursa. IC was similar between all treatments. Faecalibacterium, Ruminococcus torques group, and Bacteroides were the major genera modulated by SM addition. At 15 and 21 days old, SM exerts a impact on diversity and evenness of the cecal microbiome. Conclusion: SM (1 %) produced early immunologic maturity on Fabricius Bursa, increased intestinal functionality, and modified the microbiota, increasing beneficial microbial genera and microbial diversity.
Collapse
Affiliation(s)
- Daniela B. Medeot
- Laboratorio de Biología Molecular de las Interacciones Planta-Bacteria-Instituto de Biotecnología Ambiental y de Salud (INBIAS)- Universidad Nacional de Rio Cuarto (UNRC)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) UNRC: ruta 36 Km 601-5800-Rio Cuarto, Córdoba, Argentina
| | - Armando Nilson
- Unidad de Investigación Aviar, Producción Avícola, Facultad de Agronomía y Veterinaria-UNRC, Argentina
| | - Raul D. Miazzo
- Unidad de Investigación Aviar, Producción Avícola, Facultad de Agronomía y Veterinaria-UNRC, Argentina
| | - Viviana Grosso
- Laboratorio de Vinculación Tecnológica, Facultad de Ciencias Exactas, Físico-Químicas y Naturales-UNRC, Argentina
| | - Walter Ferrari
- Laboratorio de Biología Molecular de las Interacciones Planta-Bacteria-Instituto de Biotecnología Ambiental y de Salud (INBIAS)- Universidad Nacional de Rio Cuarto (UNRC)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) UNRC: ruta 36 Km 601-5800-Rio Cuarto, Córdoba, Argentina
| | - Edgardo Jofré
- Laboratorio de Biología Molecular de las Interacciones Planta-Bacteria-Instituto de Biotecnología Ambiental y de Salud (INBIAS)- Universidad Nacional de Rio Cuarto (UNRC)-Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET) UNRC: ruta 36 Km 601-5800-Rio Cuarto, Córdoba, Argentina
| | - Arnaldo Soltermann
- Laboratorio de Vinculación Tecnológica, Facultad de Ciencias Exactas, Físico-Químicas y Naturales-UNRC, Argentina
| | - María Fernanda Peralta
- Unidad de Investigación Aviar, Producción Avícola, Facultad de Agronomía y Veterinaria-UNRC, Argentina
| |
Collapse
|
18
|
Delavy M, Sertour N, Patin E, Le Chatelier E, Cole N, Dubois F, Xie Z, Saint-André V, Manichanh C, Walker AW, Quintana-Murci L, Duffy D, d’Enfert C, Bougnoux ME, Consortium MI. Unveiling Candida albicans intestinal carriage in healthy volunteers: the role of micro- and mycobiota, diet, host genetics and immune response. Gut Microbes 2023; 15:2287618. [PMID: 38017705 PMCID: PMC10732203 DOI: 10.1080/19490976.2023.2287618] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 11/21/2023] [Indexed: 11/30/2023] Open
Abstract
Candida albicans is a commensal yeast present in the gut of most healthy individuals but with highly variable concentrations. However, little is known about the host factors that influence colonization densities. We investigated how microbiota, host lifestyle factors, and genetics could shape C. albicans intestinal carriage in 695 healthy individuals from the Milieu Intérieur cohort. C. albicans intestinal carriage was detected in 82.9% of the subjects using quantitative PCR. Using linear mixed models and multiway-ANOVA, we explored C. albicans intestinal levels with regard to gut microbiota composition and lifestyle factors including diet. By analyzing shotgun metagenomics data and C. albicans qPCR data, we showed that Intestinimonas butyriciproducens was the only gut microbiota species whose relative abundance was negatively correlated with C. albicans concentration. Diet is also linked to C. albicans growth, with eating between meals and a low-sodium diet being associated with higher C. albicans levels. Furthermore, by Genome-Wide Association Study, we identified 26 single nucleotide polymorphisms suggestively associated with C. albicans colonization. In addition, we found that the intestinal levels of C. albicans might influence the host immune response, specifically in response to fungal challenge. We analyzed the transcriptional levels of 546 immune genes and the concentration of 13 cytokines after whole blood stimulation with C. albicans cells and showed positive associations between the extent of C. albicans intestinal levels and NLRP3 expression, as well as secreted IL-2 and CXCL5 concentrations. Taken together, these findings open the way for potential new interventional strategies to curb C. albicans intestinal overgrowth.
Collapse
Affiliation(s)
- Margot Delavy
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, Université Paris Cité INRAE, Paris, France
| | - Natacha Sertour
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, Université Paris Cité INRAE, Paris, France
| | - Etienne Patin
- Human Evolutionary Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR2000, Paris, France
| | | | - Nathaniel Cole
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Florian Dubois
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Institut Pasteur, Université Paris Cité, CBUTechS, Paris, France
| | - Zixuan Xie
- Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Gut Microbiome Group, Barcelona, Spain
| | - Violaine Saint-André
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Bioinformatics and Biostatistics HUB, Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
| | - Chaysavanh Manichanh
- Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Gut Microbiome Group, Barcelona, Spain
| | - Alan W. Walker
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Lluis Quintana-Murci
- Human Evolutionary Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR2000, Paris, France
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Institut Pasteur, Université Paris Cité, CBUTechS, Paris, France
| | - Christophe d’Enfert
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, Université Paris Cité INRAE, Paris, France
| | - Marie-Elisabeth Bougnoux
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, Université Paris Cité INRAE, Paris, France
- APHP, Hôpital Necker-Enfants-Malades, Service de Microbiologie Clinique, Unité de Parasitologie-Mycologie, Paris, France
| | - Milieu Intérieur Consortium
- Unité Biologie et Pathogénicité Fongiques, Institut Pasteur, Université Paris Cité INRAE, Paris, France
- Human Evolutionary Genetics Unit, Institut Pasteur, Université Paris Cité, CNRS UMR2000, Paris, France
- MGP MetaGénoPolis, INRA, Université Paris-Saclay, Jouy-en-Josas, France
- The Rowett Institute, University of Aberdeen, Aberdeen, UK
- Translational Immunology Unit, Institut Pasteur, Université Paris Cité, Paris, France
- Institut Pasteur, Université Paris Cité, CBUTechS, Paris, France
- Vall d’Hebron Institut de Recerca (VHIR), Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Gut Microbiome Group, Barcelona, Spain
- Bioinformatics and Biostatistics HUB, Department of Computational Biology, Institut Pasteur, Université Paris Cité, Paris, France
- APHP, Hôpital Necker-Enfants-Malades, Service de Microbiologie Clinique, Unité de Parasitologie-Mycologie, Paris, France
| |
Collapse
|
19
|
Maigné É, Noirot C, Henry J, Adu Kesewaah Y, Badin L, Déjean S, Guilmineau C, Krebs A, Mathevet F, Segalini A, Thomassin L, Colongo D, Gaspin C, Liaubet L, Vialaneix N. Asterics: a simple tool for the ExploRation and Integration of omiCS data. BMC Bioinformatics 2023; 24:391. [PMID: 37853347 PMCID: PMC10583411 DOI: 10.1186/s12859-023-05504-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023] Open
Abstract
BACKGROUND The rapid development of omics acquisition techniques has induced the production of a large volume of heterogeneous and multi-level omics datasets, which require specific and sometimes complex analyses to obtain relevant biological information. Here, we present ASTERICS (version 2.5), a publicly available web interface for the analyses of omics datasets. RESULTS ASTERICS is designed to make both standard and complex exploratory and integration analysis workflows easily available to biologists and to provide high quality interactive plots. Special care has been taken to provide a comprehensive documentation of the implemented analyses and to guide users toward sound analysis choices regarding some specific omics data. Data and analyses are organized in a comprehensive graphical workflow within ASTERICS workspace to facilitate the understanding of successive data editions and analyses leading to a given result. CONCLUSION ASTERICS provides an easy to use platform for omics data exploration and integration. The modular organization of its open source code makes it easy to incorporate new workflows and analyses by external contributors. ASTERICS is available at https://asterics.miat.inrae.fr and can also be deployed using provided docker images.
Collapse
Affiliation(s)
- Élise Maigné
- Université de Toulouse, INRAE, UR MIAT, 31326, Castanet-Tolosan, France
| | - Céline Noirot
- Université de Toulouse, INRAE, UR MIAT, 31326, Castanet-Tolosan, France
- Université Fédérale de Toulouse, INRAE, Bioinfomics, Genotoul Bioinformatics Facility, 31326, Castanet-Tolosan, France
| | - Julien Henry
- Université de Toulouse, INRAE, UR MIAT, 31326, Castanet-Tolosan, France
- Plateforme Biostatistique, Genotoul, Toulouse, France
| | - Yaa Adu Kesewaah
- Université de Toulouse, INRAE, UR MIAT, 31326, Castanet-Tolosan, France
- Plateforme Biostatistique, Genotoul, Toulouse, France
| | | | - Sébastien Déjean
- Plateforme Biostatistique, Genotoul, Toulouse, France
- IMT, UMR 5219, Université de Toulouse, CNRS, UPS, 31062, Toulouse, France
| | - Camille Guilmineau
- Université de Toulouse, INRAE, UR MIAT, 31326, Castanet-Tolosan, France
- Plateforme Biostatistique, Genotoul, Toulouse, France
| | - Arielle Krebs
- Université de Toulouse, INRAE, UR MIAT, 31326, Castanet-Tolosan, France
- Université Fédérale de Toulouse, INRAE, Bioinfomics, Genotoul Bioinformatics Facility, 31326, Castanet-Tolosan, France
| | - Fanny Mathevet
- Université de Toulouse, INRAE, UR MIAT, 31326, Castanet-Tolosan, France
- Plateforme Biostatistique, Genotoul, Toulouse, France
| | | | | | | | - Christine Gaspin
- Université de Toulouse, INRAE, UR MIAT, 31326, Castanet-Tolosan, France
- Université Fédérale de Toulouse, INRAE, Bioinfomics, Genotoul Bioinformatics Facility, 31326, Castanet-Tolosan, France
| | - Laurence Liaubet
- GenPhySE, Université de Toulouse, INRAE, ENVT, 31326, Castanet-Tolosan, France
| | - Nathalie Vialaneix
- Université de Toulouse, INRAE, UR MIAT, 31326, Castanet-Tolosan, France.
- Plateforme Biostatistique, Genotoul, Toulouse, France.
| |
Collapse
|
20
|
Delannoy S, Hoffer C, Tran ML, Madec JY, Brisabois A, Fach P, Haenni M. High throughput qPCR analyses suggest that Enterobacterales of French sheep and cow cheese rarely carry genes conferring resistances to critically important antibiotics for human medicine. Int J Food Microbiol 2023; 403:110303. [PMID: 37384974 DOI: 10.1016/j.ijfoodmicro.2023.110303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/19/2023] [Accepted: 06/22/2023] [Indexed: 07/01/2023]
Abstract
Bacteria present in raw milk can carry acquired or intrinsic antimicrobial resistance genes (ARGs) and mobile genetic elements (MGEs). However, only a few studies have evaluated raw milk cheese as a potential reservoir of ARGs. This study thus aimed at providing new data regarding resistance markers present in raw milk cheese. Sheep (n = 360) and cow (n = 360) cheese samples produced in France were incubated in buffered peptone water supplemented with acriflavin or novobiocin; as corroborated by 16S metabarcoding, samples were enriched in Gram-negative bacteria since Escherichia coli and Hafnia alvei respectively accounted for 40 % and 20 % of the samples' microbiota. Screening of the samples for the presence of 30 ARGs and 16 MGEs by high throughput qPCR array showed that nine ARGs conferring resistances to 1st-generation beta-lactams, aminoglycosides, trimethoprim/sulfonamides and tetracyclines occurred in >75 % of both sheep and cow samples. This is neither surprising nor alarming since these resistance genes are widely spread across the One Health human, animal and environmental sectors. Conversely, genes conferring resistances to last-generations cephalosporins were rarely identified, while those conferring resistances to carbapenems or amikacin, which are restricted to human use, were never detected. Multiple MGEs were detected, the most frequent ones being IncF plasmids, confirming the potential transmission of ARGs. Our results are in line with the few studies of the resistome of milk or milk cheese showing that genes conferring resistances to 1st-generation beta-lactams, aminoglycosides and tetracyclines families are widespread, while those conferring resistances to critically important antibiotics are rare or absent.
Collapse
Affiliation(s)
- Sabine Delannoy
- COLiPATH Unit & Genomics Platform IdentyPath, Laboratory for Food Safety, ANSES, 94700 Maisons-Alfort, France.
| | - Corine Hoffer
- COLiPATH Unit & Genomics Platform IdentyPath, Laboratory for Food Safety, ANSES, 94700 Maisons-Alfort, France
| | - Maï-Lan Tran
- COLiPATH Unit & Genomics Platform IdentyPath, Laboratory for Food Safety, ANSES, 94700 Maisons-Alfort, France
| | - Jean-Yves Madec
- ANSES - Université de Lyon, Unité Antibiorésistance et Virulence Bactériennes, 69007 Lyon, France
| | - Anne Brisabois
- Strategy and Programs Department, ANSES, 94700 Maisons-Alfort, France
| | - Patrick Fach
- COLiPATH Unit & Genomics Platform IdentyPath, Laboratory for Food Safety, ANSES, 94700 Maisons-Alfort, France
| | - Marisa Haenni
- ANSES - Université de Lyon, Unité Antibiorésistance et Virulence Bactériennes, 69007 Lyon, France
| |
Collapse
|
21
|
Bennouna A, Tantely ML, Raharinosy V, Andriamandimby SF, Bigot T, Chrétien D, Jacquemet E, Volant S, Temmam S, Dussart P, Lacoste V, Girod R, Eloit M. Comprehensive Characterization of Viral Diversity of Female Mosquitoes in Madagascar. Viruses 2023; 15:1852. [PMID: 37766259 PMCID: PMC10537517 DOI: 10.3390/v15091852] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/29/2023] Open
Abstract
The diversity and circulation of arboviruses are not much studied in Madagascar. The fact is that arboviral emergences are rarely detected. The existing surveillance system primarily relies on serological detection and records only a few human infections annually. The city of Mahajanga, however, experienced a confirmed dengue fever epidemic in 2020 and 2021. This study aimed to characterize and analyze the virome of mosquitoes collected in Mahajanga, near patients with dengue-like syndromes to detect known and unknown viruses as well as investigate the factors contributing to the relative low circulation of arboviruses in the area. A total of 4280 mosquitoes representing at least 12 species from the Aedes, Anopheles, and Culex genera were collected during the dry and the rainy seasons from three sites, following an urbanization gradient. The virome analysis of 2192 female mosquitoes identified a diverse range of viral families and genera and revealed different patterns that are signatures of the influence of the mosquito genus or the season of collection on the composition and abundance of the virome. Despite the absence of known human or veterinary arboviruses, the identification and characterization of viral families, genera, and species in the mosquito virome contribute to our understanding of viral ecology and diversity within mosquito populations in Madagascar. This study serves as a foundation for ongoing surveillance efforts and provides a basis for the development of preventive strategies against various mosquito-borne viral diseases, including known arboviruses.
Collapse
Affiliation(s)
- Amal Bennouna
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, 75015 Paris, France; (A.B.); (S.T.)
- The WOAH (OIE) Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Michael Luciano Tantely
- Medical Entomology Unit, Institut Pasteur de Madagascar, Antananarivo 101 1274, Madagascar; (M.L.T.)
| | | | | | - Thomas Bigot
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, 75015 Paris, France; (A.B.); (S.T.)
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Delphine Chrétien
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, 75015 Paris, France; (A.B.); (S.T.)
- The WOAH (OIE) Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Elise Jacquemet
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Stevenn Volant
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Sarah Temmam
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, 75015 Paris, France; (A.B.); (S.T.)
- The WOAH (OIE) Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Institut Pasteur, Université Paris Cité, 75015 Paris, France
| | - Philippe Dussart
- Virology Unit, Institut Pasteur de Madagascar, Antananarivo 101 1274, Madagascar
| | - Vincent Lacoste
- Virology Unit, Institut Pasteur de Madagascar, Antananarivo 101 1274, Madagascar
| | - Romain Girod
- Medical Entomology Unit, Institut Pasteur de Madagascar, Antananarivo 101 1274, Madagascar; (M.L.T.)
| | - Marc Eloit
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, 75015 Paris, France; (A.B.); (S.T.)
- The WOAH (OIE) Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Institut Pasteur, Université Paris Cité, 75015 Paris, France
- Ecole Nationale Vétérinaire d’Alfort, University of Paris-Est, 94700 Maisons-Alfort, France
| |
Collapse
|
22
|
Mohamed Ali S, Rakotonirina A, Heng K, Jacquemet E, Volant S, Temmam S, Boyer S, Eloit M. Longitudinal Study of Viral Diversity Associated with Mosquito Species Circulating in Cambodia. Viruses 2023; 15:1831. [PMID: 37766237 PMCID: PMC10535147 DOI: 10.3390/v15091831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/24/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Arthropod-borne viruses (arboviruses) pose a significant global health threat and are primarily transmitted by mosquitoes. In Cambodia, there are currently 290 recorded mosquito species, with at least 17 of them considered potential vectors of arboviruses to humans. Effective surveillance of virome profiles in mosquitoes from Cambodia is vital, as it could help prevent and control arbovirus diseases in a country where epidemics occur frequently. The objective of this study was to identify and characterize the viral diversity in mosquitoes collected during a one-year longitudinal study conducted in various habitats across Cambodia. For this purpose, we used a metatranscriptomics approach and detected the presence of chikungunya virus in the collected mosquitoes. Additionally, we identified viruses categorized into 26 taxa, including those known to harbor arboviruses such as Flaviviridae and Orthomyxoviridae, along with a group of viruses not yet taxonomically identified and provisionally named "unclassified viruses". Interestingly, the taxa detected varied in abundance and composition depending on the mosquito genus, with no significant influence of the collection season. Furthermore, most of the identified viruses were either closely related to viruses found exclusively in insects or represented new viruses belonging to the Rhabdoviridae and Birnaviridae families. The transmission capabilities of these novel viruses to vertebrates remain unknown.
Collapse
Affiliation(s)
- Souand Mohamed Ali
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, 75015 Paris, France; (S.M.A.); (S.T.)
| | - Antsa Rakotonirina
- Medical and Veterinary Entomology Unit, Institut Pasteur du Cambodge, Phnom Penh 12201, Cambodia; (A.R.); (S.B.)
| | - Kimly Heng
- Medical and Veterinary Entomology Unit, Institut Pasteur du Cambodge, Phnom Penh 12201, Cambodia; (A.R.); (S.B.)
| | - Elise Jacquemet
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France (S.V.)
| | - Stevenn Volant
- Bioinformatics and Biostatistics Hub, Institut Pasteur, Université Paris Cité, 75015 Paris, France (S.V.)
| | - Sarah Temmam
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, 75015 Paris, France; (S.M.A.); (S.T.)
| | - Sebastien Boyer
- Medical and Veterinary Entomology Unit, Institut Pasteur du Cambodge, Phnom Penh 12201, Cambodia; (A.R.); (S.B.)
- Ecology and Emergence of Arthropod-Borne Diseases, Institut Pasteur, 75015 Paris, France
| | - Marc Eloit
- Pathogen Discovery Laboratory, Institut Pasteur, Université de Paris, 75015 Paris, France; (S.M.A.); (S.T.)
- Ecole Nationale Vétérinaire d’Alfort, University of Paris-Est, 94704 Maisons-Alfort, France
| |
Collapse
|
23
|
Potes D, Benavides ID, Rivera-Franco N, Portilla CA, Ramirez O, Castillo A, López-Medina E. Effect of Antibiotics and Gut Microbiota on the Development of Sepsis in Children with Hematopoietic Stem Cell Transplants. J PEDIAT INF DIS-GER 2023; 18:186-192. [DOI: 10.1055/s-0043-57249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
Abstract
Objective To describe the association between antibiotic use, gut microbiota composition, and the development of sepsis in pediatric patients undergoing hematopoietic stem cell transplantation (HSCT) to treat acute lymphoblastic leukemia.
Methods A cohort of pediatric patients was followed up between days −30 (pre-HSCT) and +30 (post-HSCT), and sequential stool samples were collected for analysis of the taxonomic composition of bacterial communities by comparing the sequences of the 16s ribosomal RNA gene. Clinically, patients were divided into those with or without sepsis according to their clinical and laboratory data. Gut microbiota was categorized as potentially pathogenic or commensal and was described according to antibiotic use in patients with and without sepsis.
Results A cohort of eight patients provided 34 stool samples at different time points during their pre- and post-HSCT periods. There was a greater diversity in the microbial composition in patients who did not develop sepsis. In contrast, patients who developed sepsis had low microbiota diversity, a slight dominance of the genus Bacteroides and order Enterobacterales, and a low abundance of the genus Akkermansia. The use of antibiotics was associated with a low relative abundance of commensal bacteria, a high relative abundance of potentially pathogenic microbiota, and a risk of sepsis.
Conclusion Our results suggest that gut microbiota sequencing in pediatric HSCT recipients could predict the clinical course and guide direct interventions to improve patient outcomes. Accordingly, short-spectrum, tailored antibiotic therapy could be provided to patients with fever pre- and post-HSCT to prevent dysbiosis and reduce the risk of sepsis.
Collapse
Affiliation(s)
- Daniela Potes
- TAO-Lab/CiBioFi Laboratory, Department of Biology, Faculty of Natural and Exact Sciences, Universidad del Valle, Cali, Colombia
| | - Iván Darío Benavides
- Department of Pediatrics, Universidad del Valle, Cali, Colombia
- Bone Marrow Transplant Unit, Clínica Imbanaco Grupo Quironsalud, Cali, Colombia
| | - Nelson Rivera-Franco
- TAO-Lab/CiBioFi Laboratory, Department of Biology, Faculty of Natural and Exact Sciences, Universidad del Valle, Cali, Colombia
| | - Carlos A. Portilla
- Department of Pediatrics, Universidad del Valle, Cali, Colombia
- Bone Marrow Transplant Unit, Clínica Imbanaco Grupo Quironsalud, Cali, Colombia
- Fundación POHEMA, Cali, Colombia
| | - Oscar Ramirez
- Bone Marrow Transplant Unit, Clínica Imbanaco Grupo Quironsalud, Cali, Colombia
- Fundación POHEMA, Cali, Colombia
| | - Andrés Castillo
- TAO-Lab/CiBioFi Laboratory, Department of Biology, Faculty of Natural and Exact Sciences, Universidad del Valle, Cali, Colombia
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Eduardo López-Medina
- Department of Pediatrics, Universidad del Valle, Cali, Colombia
- Bone Marrow Transplant Unit, Clínica Imbanaco Grupo Quironsalud, Cali, Colombia
- Centro de Estudios en Infectología Pediátrica, Cali, Colombia
| |
Collapse
|
24
|
Graf M, Greenfield LM, Reay MK, Bargiela R, Williams GB, Onyije C, Lloyd CEM, Bull ID, Evershed RP, Golyshin PN, Chadwick DR, Jones DL. Increasing concentration of pure micro- and macro-LDPE and PP plastic negatively affect crop biomass, nutrient cycling, and microbial biomass. JOURNAL OF HAZARDOUS MATERIALS 2023; 458:131932. [PMID: 37390687 DOI: 10.1016/j.jhazmat.2023.131932] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 05/29/2023] [Accepted: 06/23/2023] [Indexed: 07/02/2023]
Abstract
Over the last 50 years, the intense use of agricultural plastic in the form of mulch films has led to an accumulation of plastic in soil, creating a legacy of plastic in agricultural fields. Plastic often contains additives, however it is still largely unknown how these compounds affect soil properties, potentially influencing or masking effects of the plastic itself. Therefore, the aim of this study was to investigate the effects of pure plastics of varying sizes and concentrations, to improve our understanding of plastic-only interactions within soil-plant mesocosms. Maize (Zea mays L.) was grown over eight weeks following the addition of micro and macro low-density polyethylene and polypropylene at increasing concentrations (equivalent to 1, 10, 25, and 50 years mulch film use) and the effects of plastic on key soil and plant properties were measured. We found the effect of both macro and microplastic on soil and plant health is negligible in the short-term (1 to <10 years). However, ≥ 10 years of plastic application for both plastic types and sizes resulted in a clear negative effect on plant growth and microbial biomass. This study provides vital insight into the effect of both macro and microplastics on soil and plant properties.
Collapse
Affiliation(s)
- Martine Graf
- School of Natural Sciences, Bangor University, Bangor, Gwynedd LL57 2UW, UK.
| | - Lucy M Greenfield
- School of Natural Sciences, Bangor University, Bangor, Gwynedd LL57 2UW, UK
| | - Michaela K Reay
- Organic Geochemistry Unit, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Rafael Bargiela
- Centre of Environmental Biotechnology, Bangor University, Bangor, Gwynedd LL57 2UW, UK
| | - Gwion B Williams
- Centre of Environmental Biotechnology, Bangor University, Bangor, Gwynedd LL57 2UW, UK
| | - Charles Onyije
- Organic Geochemistry Unit, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Charlotte E M Lloyd
- Organic Geochemistry Unit, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Ian D Bull
- Organic Geochemistry Unit, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Richard P Evershed
- Organic Geochemistry Unit, School of Chemistry, University of Bristol, Bristol BS8 1TS, UK
| | - Peter N Golyshin
- Centre of Environmental Biotechnology, Bangor University, Bangor, Gwynedd LL57 2UW, UK
| | - David R Chadwick
- School of Natural Sciences, Bangor University, Bangor, Gwynedd LL57 2UW, UK
| | - Davey L Jones
- School of Natural Sciences, Bangor University, Bangor, Gwynedd LL57 2UW, UK; Centre of Environmental Biotechnology, Bangor University, Bangor, Gwynedd LL57 2UW, UK; SoilsWest, Centre for Sustainable Farming Systems, Food Futures Institute, Murdoch University, Murdoch, WA 6150, Australia
| |
Collapse
|
25
|
Khalfi P, Suspène R, Raymond KA, Caval V, Caignard G, Berry N, Thiers V, Combredet C, Rufie C, Rigaud S, Ghozlane A, Volant S, Komarova AV, Tangy F, Vartanian JP. Antagonism of ALAS1 by the Measles Virus V protein contributes to degradation of the mitochondrial network and promotes interferon response. PLoS Pathog 2023; 19:e1011170. [PMID: 36802406 PMCID: PMC9983871 DOI: 10.1371/journal.ppat.1011170] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 03/03/2023] [Accepted: 02/01/2023] [Indexed: 02/23/2023] Open
Abstract
Viruses have evolved countless mechanisms to subvert and impair the host innate immune response. Measles virus (MeV), an enveloped, non-segmented, negative-strand RNA virus, alters the interferon response through different mechanisms, yet no viral protein has been described as directly targeting mitochondria. Among the crucial mitochondrial enzymes, 5'-aminolevulinate synthase (ALAS) is an enzyme that catalyzes the first step in heme biosynthesis, generating 5'-aminolevulinate from glycine and succinyl-CoA. In this work, we demonstrate that MeV impairs the mitochondrial network through the V protein, which antagonizes the mitochondrial enzyme ALAS1 and sequesters it to the cytosol. This re-localization of ALAS1 leads to a decrease in mitochondrial volume and impairment of its metabolic potential, a phenomenon not observed in MeV deficient for the V gene. This perturbation of the mitochondrial dynamics demonstrated both in culture and in infected IFNAR-/- hCD46 transgenic mice, causes the release of mitochondrial double-stranded DNA (mtDNA) in the cytosol. By performing subcellular fractionation post infection, we demonstrate that the most significant source of DNA in the cytosol is of mitochondrial origin. Released mtDNA is then recognized and transcribed by the DNA-dependent RNA polymerase III. The resulting double-stranded RNA intermediates will be captured by RIG-I, ultimately initiating type I interferon production. Deep sequencing analysis of cytosolic mtDNA editing divulged an APOBEC3A signature, primarily analyzed in the 5'TpCpG context. Finally, in a negative feedback loop, APOBEC3A an interferon inducible enzyme will orchestrate the catabolism of mitochondrial DNA, decrease cellular inflammation, and dampen the innate immune response.
Collapse
Affiliation(s)
- Pierre Khalfi
- Virus and Cellular Stress Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, Paris, France
- Sorbonne Université, Complexité du Vivant, ED515, Paris, France
| | - Rodolphe Suspène
- Virus and Cellular Stress Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Kyle A. Raymond
- Virus and Cellular Stress Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, Paris, France
- Sorbonne Université, Complexité du Vivant, ED515, Paris, France
| | - Vincent Caval
- Virus and Cellular Stress Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, Paris, France
| | | | - Noémie Berry
- Virus and Cellular Stress Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, Paris, France
- Sorbonne Université, Complexité du Vivant, ED515, Paris, France
- UMR1161 Virologie, ANSES-INRAE-ENVA, Maisons-Alfort, France
| | - Valérie Thiers
- Virus and Cellular Stress Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Chantal Combredet
- Vaccines Innovation Laboratory, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Claude Rufie
- Vaccines Innovation Laboratory, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Stéphane Rigaud
- Image Analysis Hub, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Amine Ghozlane
- Bioinformatics and Biostatistics HUB, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Stevenn Volant
- Bioinformatics and Biostatistics HUB, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Anastassia V. Komarova
- Interactomics, RNA and Immunity Laboratory, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Frédéric Tangy
- Vaccines Innovation Laboratory, Institut Pasteur, Université de Paris Cité, Paris, France
| | - Jean-Pierre Vartanian
- Virus and Cellular Stress Unit, Department of Virology, Institut Pasteur, Université de Paris Cité, Paris, France
- * E-mail:
| |
Collapse
|
26
|
A Clinical Study Provides the First Direct Evidence That Interindividual Variations in Fecal β-Lactamase Activity Affect the Gut Mycobiota Dynamics in Response to β-Lactam Antibiotics. mBio 2022; 13:e0288022. [PMID: 36448778 PMCID: PMC9765473 DOI: 10.1128/mbio.02880-22] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/05/2022] Open
Abstract
Antibiotics disturb the intestinal bacterial microbiota, leading to gut dysbiosis and an increased risk for the overgrowth of opportunistic pathogens. It is not fully understood to what extent antibiotics affect the fungal fraction of the intestinal microbiota, the mycobiota. There is no report of the direct role of antibiotics in the overgrowth in healthy humans of the opportunistic pathogenic yeast Candida albicans. Here, we have explored the gut mycobiota of 22 healthy subjects before, during, and up to 6 months after a 3-day regimen of third-generation cephalosporins (3GCs). Using ITS1-targeted metagenomics, we highlighted the strong intra- and interindividual diversity of the healthy gut mycobiota. With a specific quantitative approach, we showed that C. albicans prevalence was much higher than previously reported, with all subjects but one being carriers of C. albicans, although with highly variable burdens. 3GCs significantly altered the mycobiota composition and the fungal load was increased both at short and long term. Both C. albicans relative and absolute abundances were increased but 3GCs did not reduce intersubject variability. Variations in C. albicans burden in response to 3GC treatment could be partly explained by changes in the levels of endogenous fecal β-lactamase activity, with subjects characterized by a high increase of β-lactamase activity displaying a lower increase of C. albicans levels. A same antibiotic treatment might thus affect differentially the gut mycobiota and C. albicans carriage, depending on the treated subject, suggesting a need to adjust the current risk factors for C. albicans overgrowth after a β-lactam treatment. IMPORTANCE Fungal infections are redoubtable healthcare-associated complications in immunocompromised patients. Particularly, the commensal intestinal yeast Candida albicans causes invasive infections in intensive care patients and is, therefore, associated with high mortality. These infections are preceded by an intestinal expansion of C. albicans before its translocation into the bloodstream. Antibiotics are a well-known risk factor for C. albicans overgrowth but the impact of antibiotic-induced dysbiosis on the human gut mycobiota-the fungal microbiota-and the understanding of the mechanisms involved in C. albicans overgrowth in humans are very limited. Our study shows that antibiotics increase the fungal proportion in the gut and disturb the fungal composition, especially C. albicans, in a subject-dependent manner. Indeed, variations across subjects in C. albicans burden in response to β-lactam treatment could be partly explained by changes in the levels of endogenous fecal β-lactamase activity. This highlighted a potential new key factor for C. albicans overgrowth. Thus, the significance of our research is in providing a better understanding of the factors behind C. albicans intestinal overgrowth, which might lead to new means to prevent life-threatening secondary infections.
Collapse
|
27
|
Bhar A, Gierse LC, Meene A, Wang H, Karte C, Schwaiger T, Schröder C, Mettenleiter TC, Urich T, Riedel K, Kaderali L. Application of a maximal-clique based community detection algorithm to gut microbiome data reveals driver microbes during influenza A virus infection. Front Microbiol 2022; 13:979320. [PMID: 36338082 PMCID: PMC9630851 DOI: 10.3389/fmicb.2022.979320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/21/2022] [Indexed: 11/20/2022] Open
Abstract
Influenza A Virus (IAV) infection followed by bacterial pneumonia often leads to hospitalization and death in individuals from high risk groups. Following infection, IAV triggers the process of viral RNA replication which in turn disrupts healthy gut microbial community, while the gut microbiota plays an instrumental role in protecting the host by evolving colonization resistance. Although the underlying mechanisms of IAV infection have been unraveled, the underlying complex mechanisms evolved by gut microbiota in order to induce host immune response following IAV infection remain evasive. In this work, we developed a novel Maximal-Clique based Community Detection algorithm for Weighted undirected Networks (MCCD-WN) and compared its performance with other existing algorithms using three sets of benchmark networks. Moreover, we applied our algorithm to gut microbiome data derived from fecal samples of both healthy and IAV-infected pigs over a sequence of time-points. The results we obtained from the real-life IAV dataset unveil the role of the microbial families Ruminococcaceae, Lachnospiraceae, Spirochaetaceae and Prevotellaceae in the gut microbiome of the IAV-infected cohort. Furthermore, the additional integration of metaproteomic data enabled not only the identification of microbial biomarkers, but also the elucidation of their functional roles in protecting the host following IAV infection. Our network analysis reveals a fast recovery of the infected cohort after the second IAV infection and provides insights into crucial roles of Desulfovibrionaceae and Lactobacillaceae families in combating Influenza A Virus infection. Source code of the community detection algorithm can be downloaded from https://github.com/AniBhar84/MCCD-WN.
Collapse
Affiliation(s)
- Anirban Bhar
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| | | | - Alexander Meene
- Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Haitao Wang
- Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Claudia Karte
- Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Greifswald, Germany
| | - Theresa Schwaiger
- Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Greifswald, Germany
| | - Charlotte Schröder
- Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Greifswald, Germany
| | | | - Tim Urich
- Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Katharina Riedel
- Institute of Microbiology, University of Greifswald, Greifswald, Germany
| | - Lars Kaderali
- Institute of Bioinformatics, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
28
|
Patrier J, Villageois-Tran K, Szychowiak P, Ruckly S, Gschwind R, Wicky PH, Gueye S, Armand-Lefevre L, Marzouk M, Sonneville R, Bouadma L, Petitjean M, Lamara F, de Montmollin E, Timsit JF, Ruppé E, The French COVID Cohort Study Group AbelLaurentAbrousAmalAndrejakClaireAngoulvantFrançoisBacheletDelphineBartoliMarieBehilillSylvieBeluzeMarineBhavsarKrishnaBouadmaLilaCervantes-GonzalezMinervaChairAnissaCharpentierCharlotteChenardLéoChirouzeCatherineCouffin-CadierguesSandrineCouffignalCamilleDebrayMarie-PierreDeplanqueDominiqueDescampsDianeDialloAlphada SilvaFernanda DiasDorivalCélineDuvalXavierEloyPhilippineEnoufVincentEsperouHélèneEsposito-FareseMarinaEtienneManuelFlorenceAline-MarieGaymardAlexandreGhosnJadeGiganteTristanGilgMorganeGoehringerFrançoisGuedjJérémieHouasIkramHoffmannIsabelleHulotJean-SébastienJaafouraSalmaKafifOuifiyaKhalilAntoineLafhejNadhemLaouénanCédricLaribiSamiraLeMinhLe HingratQuentinLe MestreSoizicLetrouSophieLevyYvesLinaBrunoLingasGuillaumeMalvyDenisMentréFranceMouquetHugoNeantNadègePaulChristellePapadopoulosAuréliePaulChristellePetrov-SanchezVentzislavaPeytavinGillesPiquardValentinePiconeOlivierRosa-CalatravaManuelRossignolBénédicteRossignolPatrickRoyCarineSchneiderMarionSuRichaTardivonCoralieTimsitJean-FrançoisTubianaSarahVan Der WerfSylvieVisseauxBenoitWiedemannAurélie. Oropharyngeal and intestinal concentrations of opportunistic pathogens are independently associated with death of SARS-CoV-2 critically ill adults. Crit Care 2022; 26:300. [PMID: 36192756 PMCID: PMC9527714 DOI: 10.1186/s13054-022-04164-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 08/21/2022] [Indexed: 11/30/2022] Open
Abstract
BACKGROUND The composition of the digestive microbiota may be associated with outcome and infections in patients admitted to the intensive care unit (ICU). The dominance by opportunistic pathogens (such as Enterococcus) has been associated with death. However, whether this association remains all throughout the hospitalization are lacking. METHODS We performed a single-center observational prospective cohort study in critically ill patients admitted with severe SARS-CoV-2 infection. Oropharyngeal and rectal swabs were collected at admission and then twice weekly until discharge or death. Quantitative cultures for opportunistic pathogens were performed on oropharyngeal and rectal swabs. The composition of the intestinal microbiota was assessed by 16S rDNA sequencing. Oropharyngeal and intestinal concentrations of opportunistic pathogens, intestinal richness and diversity were entered into a multivariable Cox model as time-dependent covariates. The primary outcome was death at day 90. RESULTS From March to September 2020, 95 patients (765 samples) were included. The Simplified Acute Physiology Score 2 (SAPS 2) at admission was 33 [24; 50] and a Sequential Organ Failure Assessment score (SOFA score) at 6 [4; 8]. Day 90 all-cause mortality was 44.2% (42/95). We observed that the oropharyngeal and rectal concentrations of Enterococcus spp., Staphylococcus aureus and Candida spp. were associated with a higher risk of death. This association remained significant after adjustment for prognostic covariates (age, chronic disease, daily antimicrobial agent use and daily SOFA score). A one-log increase in Enterococcus spp., S. aureus and Candida spp. in oropharyngeal or rectal swabs was associated with a 17% or greater increase in the risk of death. CONCLUSION We found that elevated oropharyngeal/intestinal Enterococcus spp. S. aureus and Candida spp. concentrations, assessed by culture, are associated with mortality, independent of age, organ failure, and antibiotic therapy, opening prospects for simple and inexpensive microbiota-based markers for the prognosis of critically ill SARS-CoV-2 patients.
Collapse
Affiliation(s)
- Juliette Patrier
- grid.411119.d0000 0000 8588 831XAP-HP, Bichat Hospital, Medical and Infectious Diseases ICU (MI2), 75018 Paris, France
| | - Khanh Villageois-Tran
- grid.411599.10000 0000 8595 4540AP-HP, Service de Microbiologie, Hôpital Beaujon, 75018 Paris, France ,grid.508487.60000 0004 7885 7602IAME, INSERM, Université de Paris, 75018 Paris, France
| | - Piotr Szychowiak
- grid.411119.d0000 0000 8588 831XAP-HP, Bichat Hospital, Medical and Infectious Diseases ICU (MI2), 75018 Paris, France ,grid.508487.60000 0004 7885 7602IAME, INSERM, Université de Paris, 75018 Paris, France
| | - Stéphane Ruckly
- grid.508487.60000 0004 7885 7602IAME, INSERM, Université de Paris, 75018 Paris, France ,grid.508487.60000 0004 7885 7602OUTCOME REA Research Network, IAME, INSERM, Université de Paris, 75018 Paris, France
| | - Rémi Gschwind
- grid.508487.60000 0004 7885 7602IAME, INSERM, Université de Paris, 75018 Paris, France
| | - Paul-Henri Wicky
- grid.411119.d0000 0000 8588 831XAP-HP, Bichat Hospital, Medical and Infectious Diseases ICU (MI2), 75018 Paris, France
| | - Signara Gueye
- grid.411119.d0000 0000 8588 831XAP-HP, Service de Bactériologie, Hôpital Bichat-Claude Bernard, 75018 Paris, France
| | - Laurence Armand-Lefevre
- grid.508487.60000 0004 7885 7602IAME, INSERM, Université de Paris, 75018 Paris, France ,grid.411119.d0000 0000 8588 831XAP-HP, Service de Bactériologie, Hôpital Bichat-Claude Bernard, 75018 Paris, France
| | - Mehdi Marzouk
- grid.411119.d0000 0000 8588 831XAP-HP, Bichat Hospital, Medical and Infectious Diseases ICU (MI2), 75018 Paris, France
| | - Romain Sonneville
- grid.411119.d0000 0000 8588 831XAP-HP, Bichat Hospital, Medical and Infectious Diseases ICU (MI2), 75018 Paris, France ,grid.508487.60000 0004 7885 7602IAME, INSERM, Université de Paris, 75018 Paris, France ,grid.508487.60000 0004 7885 7602OUTCOME REA Research Network, IAME, INSERM, Université de Paris, 75018 Paris, France
| | - Lila Bouadma
- grid.411119.d0000 0000 8588 831XAP-HP, Bichat Hospital, Medical and Infectious Diseases ICU (MI2), 75018 Paris, France ,grid.508487.60000 0004 7885 7602IAME, INSERM, Université de Paris, 75018 Paris, France ,grid.508487.60000 0004 7885 7602OUTCOME REA Research Network, IAME, INSERM, Université de Paris, 75018 Paris, France
| | - Marie Petitjean
- grid.508487.60000 0004 7885 7602IAME, INSERM, Université de Paris, 75018 Paris, France
| | - Fariza Lamara
- grid.411119.d0000 0000 8588 831XAP-HP, Bichat Hospital, Medical and Infectious Diseases ICU (MI2), 75018 Paris, France ,grid.508487.60000 0004 7885 7602OUTCOME REA Research Network, IAME, INSERM, Université de Paris, 75018 Paris, France
| | - Etienne de Montmollin
- grid.411119.d0000 0000 8588 831XAP-HP, Bichat Hospital, Medical and Infectious Diseases ICU (MI2), 75018 Paris, France ,grid.508487.60000 0004 7885 7602IAME, INSERM, Université de Paris, 75018 Paris, France ,grid.508487.60000 0004 7885 7602OUTCOME REA Research Network, IAME, INSERM, Université de Paris, 75018 Paris, France
| | - Jean-Francois Timsit
- grid.411119.d0000 0000 8588 831XAP-HP, Bichat Hospital, Medical and Infectious Diseases ICU (MI2), 75018 Paris, France ,grid.508487.60000 0004 7885 7602IAME, INSERM, Université de Paris, 75018 Paris, France ,grid.508487.60000 0004 7885 7602OUTCOME REA Research Network, IAME, INSERM, Université de Paris, 75018 Paris, France
| | - Etienne Ruppé
- grid.508487.60000 0004 7885 7602IAME, INSERM, Université de Paris, 75018 Paris, France ,grid.411119.d0000 0000 8588 831XAP-HP, Service de Bactériologie, Hôpital Bichat-Claude Bernard, 75018 Paris, France
| | | |
Collapse
|
29
|
Mokrani M, Charradi K, Limam F, Aouani E, Urdaci MC. Grape seed and skin extract, a potential prebiotic with anti-obesity effect through gut microbiota modulation. Gut Pathog 2022; 14:30. [PMID: 35794638 PMCID: PMC9258160 DOI: 10.1186/s13099-022-00505-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 06/11/2022] [Indexed: 12/16/2022] Open
Abstract
Background Obesity is a worldwide health problem and a significant risk factor for diabetes and cardiovascular diseases. Gut microbiota (GM) plays an essential role in obesity, and prebiotics such as polyphenols could be one way to improve microbial dysbiosis-induced obesity. Objective This study was designed to assess the effectiveness of grape seed and skin extract (GSSE), and/or orlistat on obese rats fed with high fat diet by targeting GM modulations. The impact of treatments was also studied in non-obese rats. Material and methods Rats were rendered obese or kept with a standard diet for three months. Then they were treated either with GSSE or orlistat or with the combined treatment (GSOR) during three months and then sacrificed. Adipose tissues, blood and faeces were collected and analyzed. Results In obese rats and to a lesser extent in non-obese rats, treatments decreased the weight of various adipose tissues and the serum levels of cholesterol, LDL, triglycerides, lipase, and CRP and increased HDL and adiponectin. GSOR treatment was even more efficient that orlistat. Obese rats had less GM diversity than non-obese rats and orlistat reduced it even more. However, diversity was restored with GSSE and GSOR treatments. Potential pathogenic Streptococcus alactolyticus/gallolyticus species were greatly increased in obese rats and drastically reduced with the treatments, as wells as other potential pathobionts. Conclusions GSSE exerts beneficial effects in obese rats and restores, at least partially, the observed dysbiosis. GSOR induced the highest beneficial effect. Moreover, the various treatments could also enhance physiological and GM modifications in non obese rats. Supplementary Information The online version contains supplementary material available at 10.1186/s13099-022-00505-0.
Collapse
|
30
|
An extensive description of the microbiological effects of silver diamine fluoride on dental biofilms using an oral in situ model. Sci Rep 2022; 12:7435. [PMID: 35523839 PMCID: PMC9076617 DOI: 10.1038/s41598-022-11477-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 04/15/2022] [Indexed: 12/23/2022] Open
Abstract
Silver diamine fluoride (SDF) has been long studied in laboratories, and its clinical effectiveness in the treatment and prevention of root caries has been reported. In the present study, we assessed the microbiological effects of SDF on dental biofilms grown on demineralized dentin in situ. Specifically, demineralized bovine root dentin slabs used as biofilm substrates were treated with 38% SDF, and the biofilms formed after this treatment were analyzed via real-time PCR, DEAD/LIVE cell staining, and SEM. Next, the viable cell count was determined, and microbial profiles were compared using 16S rRNA gene sequencing. Untreated slabs were used as controls. We observed significant decreases in viable cell counts (p < 0.05), number of biofilm-forming cells (p < 0.01), biofilm thickness (p < 0.01), and high proportion of dead cells with SDF treatment (p < 0.01). The microcolonies in the SDF-treated biofilms showed less complexity, and only a limited number of genera were differentially abundant between the groups. Microbial diversity index comparisons showed no significant differences between the groups with respect to treatments days (p = 0.362). Thus, SDF negatively influenced dental biofilm growth on demineralized root dentin in situ; however, its antimicrobial action did not target a specific oral taxon.
Collapse
|
31
|
Couvin D, Dereeper A, Meyer DF, Noroy C, Gaete S, Bhakkan B, Poullet N, Gaspard S, Bezault E, Marcelino I, Pruneau L, Segretier W, Stattner E, Cazenave D, Garnier M, Pot M, Tressières B, Deloumeaux J, Breurec S, Ferdinand S, Gonzalez-Rizzo S, Reynaud Y. KaruBioNet: a network and discussion group for a better collaboration and structuring of bioinformatics in Guadeloupe (French West Indies). BIOINFORMATICS ADVANCES 2022; 2:vbac010. [PMID: 36699379 PMCID: PMC9710593 DOI: 10.1093/bioadv/vbac010] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/24/2022] [Accepted: 02/09/2022] [Indexed: 01/28/2023]
Abstract
Summary Sequencing and other biological data are now more frequently available and at a lower price. Mutual tools and strategies are needed to analyze the huge amount of heterogeneous data generated by several research teams and devices. Bioinformatics represents a growing field in the scientific community globally. This multidisciplinary field provides a great amount of tools and methods that can be used to conduct scientific studies in a more strategic way. Coordinated actions and collaborations are needed to find more innovative and accurate methods for a better understanding of real-life data. A wide variety of organizations are contributing to KaruBioNet in Guadeloupe (French West Indies), a Caribbean archipelago. The purpose of this group is to foster collaboration and mutual aid among people from different disciplines using a 'one health' approach, for a better comprehension and surveillance of humans, plants or animals' health and diseases. The KaruBioNet network particularly aims to help researchers in their studies related to 'omics' data, but also more general aspects concerning biological data analysis. This transdisciplinary network is a platform for discussion, sharing, training and support between scientists interested in bioinformatics and related fields. Starting from a little archipelago in the Caribbean, we envision to facilitate exchange between other Caribbean partners in the future, knowing that the Caribbean is a region with non-negligible biodiversity which should be preserved and protected. Joining forces with other Caribbean countries or territories would strengthen scientific collaborative impact in the region. Information related to this network can be found at: http://www.pasteur-guadeloupe.fr/karubionet.html. Furthermore, a dedicated 'Galaxy KaruBioNet' platform is available at: http://calamar.univ-ag.fr/c3i/galaxy_karubionet.html. Availability and implementation Information about KaruBioNet is availabe at: http://www.pasteur-guadeloupe.fr/karubionet.html. Contact dcouvin@pasteur-guadeloupe.fr. Supplementary information Supplementary data are available at Bioinformatics Advances online.
Collapse
Affiliation(s)
- David Couvin
- Unité Transmission, Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Les Abymes, Guadeloupe 97139, France,To whom correspondence should be addressed
| | - Alexis Dereeper
- Unité Transmission, Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Les Abymes, Guadeloupe 97139, France
| | - Damien F Meyer
- CIRAD, UMR ASTRE, Petit-Bourg, Guadeloupe 97170, France,ASTRE, Univ Montpellier, CIRAD, INRAE, Montpellier 34000, France
| | - Christophe Noroy
- Développement, Analyse, Transfert et Application (DATA), Lamentin, Guadeloupe 97129, France
| | - Stanie Gaete
- Karubiotec Centre de Ressources Biologiques-UF 0216, CHU de la Guadeloupe, Pointe-à-Pitre 97110, France
| | - Bernard Bhakkan
- Registre des cancers de Guadeloupe, CHU de la Guadeloupe, Pointe-à-Pitre 97110, France
| | - Nausicaa Poullet
- URZ Recherches Zootechniques, INRAE, Petit-Bourg, Guadeloupe 97170, France
| | - Sarra Gaspard
- Laboratoire COVACHIMM2E EA3592, Université des Antilles, Pointe-à-Pitre, Guadeloupe 97110, France
| | - Etienne Bezault
- UMR BOREA (MNHN, CNRS-7208, IRD-207, Sorbonne Université, UCN, UA), Université des Antilles, Pointe-à-Pitre, Guadeloupe 97110, France
| | - Isabel Marcelino
- Unité Transmission, Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Les Abymes, Guadeloupe 97139, France
| | - Ludovic Pruneau
- Équipe « Biologie de la mangrove » UMR7205 « ISYEB » MNHN-CNRS-Sorbonne Université-EPHE-UA, UFR SEN Département de Biologie, Université des Antilles, Pointe-à-Pitre, Guadeloupe 97110, France
| | - Wilfried Segretier
- Laboratoire de Mathématiques Informatique et Applications (LAMIA), Université des Antilles, Pointe-à-Pitre, Guadeloupe 97110, France
| | - Erick Stattner
- Laboratoire de Mathématiques Informatique et Applications (LAMIA), Université des Antilles, Pointe-à-Pitre, Guadeloupe 97110, France
| | - Damien Cazenave
- Unité Transmission, Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Les Abymes, Guadeloupe 97139, France
| | - Maëlle Garnier
- Unité Transmission, Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Les Abymes, Guadeloupe 97139, France
| | - Matthieu Pot
- Unité Transmission, Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Les Abymes, Guadeloupe 97139, France
| | - Benoît Tressières
- Centre d’Investigation Clinique Antilles Guyane, Inserm CIC 1424, Les Abymes, Pointe-à-Pitre, Guadeloupe 97110, France
| | - Jacqueline Deloumeaux
- Karubiotec Centre de Ressources Biologiques-UF 0216, CHU de la Guadeloupe, Pointe-à-Pitre 97110, France,Registre des cancers de Guadeloupe, CHU de la Guadeloupe, Pointe-à-Pitre 97110, France
| | - Sébastien Breurec
- Unité Transmission, Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Les Abymes, Guadeloupe 97139, France,Centre d’Investigation Clinique Antilles Guyane, Inserm CIC 1424, Les Abymes, Pointe-à-Pitre, Guadeloupe 97110, France,Faculté de Médecine Hyacinthe Bastaraud, Université des Antilles, Pointe-à-Pitre, Guadeloupe 97110, France
| | - Séverine Ferdinand
- Unité Transmission, Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Les Abymes, Guadeloupe 97139, France
| | - Silvina Gonzalez-Rizzo
- Équipe « Biologie de la mangrove » UMR7205 « ISYEB » MNHN-CNRS-Sorbonne Université-EPHE-UA, UFR SEN Département de Biologie, Université des Antilles, Pointe-à-Pitre, Guadeloupe 97110, France
| | - Yann Reynaud
- Unité Transmission, Réservoir et Diversité des Pathogènes, Institut Pasteur de Guadeloupe, Les Abymes, Guadeloupe 97139, France
| |
Collapse
|
32
|
Lauka L, Sobhani I, Brunetti F, Mestivier D, de’Angelis N. Human Colonic Microbiota and Short-Term Postoperative Outcomes in Colorectal Cancer Patients: A Pilot Study. Microorganisms 2021; 10:microorganisms10010041. [PMID: 35056490 PMCID: PMC8779772 DOI: 10.3390/microorganisms10010041] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/22/2021] [Accepted: 12/22/2021] [Indexed: 12/23/2022] Open
Abstract
Despite the advances in surgical techniques and perioperative care, the complication rates after colorectal cancer surgery have remained stable. Recently, it has been suggested that colon microbiota may be implicated in several pathways that can lead to impaired colonic homeostasis and, thereby, to the development of complications after colorectal surgery. The aim of this study was to evaluate the potential impact of colonic dysbiosis on postoperative course. This prospective human clinical study recruited patients operated on for left colon, sigmoid colon or rectal cancer. Colon mucosa and fecal samples were collected to study mucosa associated microbiota (MAM) and luminal microbiota (LM), accordingly. Preliminary analysis for the first 25 consecutive patients with V3–V4 16S rRNA metagenomic analysis was performed. Bacterial composition and abundance in patients who developed postoperative complications over a 90-day follow-up period were compared to those without postoperative complications. Abundance and distribution of genera in MAM differed significantly when compared to LM with a significant impact on neoadjuvant therapy on bacterial composition. Preliminary analysis revealed no statistically significant differences in LM nor in MAM composition when individuals with and without postoperative surgical complications were compared. In cases of postoperative complications, LM and MAM showed significantly decreased diversity. Composition of the colonic microbiota is altered by neoadjuvant therapy. Results on the impact of colonic dysbiosis on postoperative complications are pending the end of the present study, with 50 patients enrolled.
Collapse
Affiliation(s)
- Lelde Lauka
- Department of Digestive and Hepatobiliary Surgery, APHP-Henri Mondor University Hospital, 94000 Creteil, France; (F.B.); (N.d.)
- EC2M-EA7375 Research Team, Henri Modor Campus, Paris East University, 94000 Creteil, France;
- Correspondence: (L.L.); (I.S.); Tel.: +33-6129-63749 (L.L.); +33-6163-42062 (I.S.)
| | - Iradj Sobhani
- EC2M-EA7375 Research Team, Henri Modor Campus, Paris East University, 94000 Creteil, France;
- Department of Gastroenterology, APHP-Henri Mondor University Hospital, 94000 Creteil, France
- Correspondence: (L.L.); (I.S.); Tel.: +33-6129-63749 (L.L.); +33-6163-42062 (I.S.)
| | - Francesco Brunetti
- Department of Digestive and Hepatobiliary Surgery, APHP-Henri Mondor University Hospital, 94000 Creteil, France; (F.B.); (N.d.)
| | - Denis Mestivier
- EC2M-EA7375 Research Team, Henri Modor Campus, Paris East University, 94000 Creteil, France;
| | - Nicola de’Angelis
- Department of Digestive and Hepatobiliary Surgery, APHP-Henri Mondor University Hospital, 94000 Creteil, France; (F.B.); (N.d.)
- EC2M-EA7375 Research Team, Henri Modor Campus, Paris East University, 94000 Creteil, France;
| |
Collapse
|
33
|
Shi LD, Lv PL, McIlroy SJ, Wang Z, Dong XL, Kouris A, Lai CY, Tyson GW, Strous M, Zhao HP. Methane-dependent selenate reduction by a bacterial consortium. THE ISME JOURNAL 2021; 15:3683-3692. [PMID: 34183781 PMCID: PMC8630058 DOI: 10.1038/s41396-021-01044-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/08/2021] [Accepted: 06/15/2021] [Indexed: 02/06/2023]
Abstract
Methanotrophic microorganisms play a critical role in controlling the flux of methane from natural sediments into the atmosphere. Methanotrophs have been shown to couple the oxidation of methane to the reduction of diverse electron acceptors (e.g., oxygen, sulfate, nitrate, and metal oxides), either independently or in consortia with other microbial partners. Although several studies have reported the phenomenon of methane oxidation linked to selenate reduction, neither the microorganisms involved nor the underlying trophic interaction has been clearly identified. Here, we provide the first detailed evidence for interspecies electron transfer between bacterial populations in a bioreactor community where the reduction of selenate is linked to methane oxidation. Metagenomic and metaproteomic analyses of the community revealed a novel species of Methylocystis as the most abundant methanotroph, which actively expressed proteins for oxygen-dependent methane oxidation and fermentation pathways, but lacked the genetic potential for selenate reduction. Pseudoxanthomonas, Piscinibacter, and Rhodocyclaceae populations appeared to be responsible for the observed selenate reduction using proteins initially annotated as periplasmic nitrate reductases, with fermentation by-products released by the methanotrophs as electron donors. The ability for the annotated nitrate reductases to reduce selenate was confirmed by gene knockout studies in an isolate of Pseudoxanthomonas. Overall, this study provides novel insights into the metabolic flexibility of the aerobic methanotrophs that likely allows them to thrive across natural oxygen gradients, and highlights the potential role for similar microbial consortia in linking methane and other biogeochemical cycles in environments where oxygen is limited.
Collapse
Affiliation(s)
- Ling-Dong Shi
- grid.13402.340000 0004 1759 700XMOE Key Lab of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Science, Zhejiang University, Hangzhou, China
| | - Pan-Long Lv
- grid.13402.340000 0004 1759 700XMOE Key Lab of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Science, Zhejiang University, Hangzhou, China
| | - Simon J. McIlroy
- grid.489335.00000000406180938Centre for Microbiome Research, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, Woolloongabba, QLD Australia ,grid.1003.20000 0000 9320 7537Australian Centre for Ecogenomics, University of Queensland, Brisbane, QLD Australia
| | - Zhen Wang
- grid.13402.340000 0004 1759 700XMOE Key Lab of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Science, Zhejiang University, Hangzhou, China
| | - Xiao-Li Dong
- grid.22072.350000 0004 1936 7697Department of Geoscience, University of Calgary, Calgary, AB Canada
| | - Angela Kouris
- grid.22072.350000 0004 1936 7697Department of Geoscience, University of Calgary, Calgary, AB Canada
| | - Chun-Yu Lai
- grid.13402.340000 0004 1759 700XMOE Key Lab of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Science, Zhejiang University, Hangzhou, China ,grid.1003.20000 0000 9320 7537Australian Centre for Ecogenomics, University of Queensland, Brisbane, QLD Australia
| | - Gene W. Tyson
- grid.489335.00000000406180938Centre for Microbiome Research, School of Biomedical Sciences, Queensland University of Technology (QUT), Translational Research Institute, Woolloongabba, QLD Australia
| | - Marc Strous
- grid.22072.350000 0004 1936 7697Department of Geoscience, University of Calgary, Calgary, AB Canada
| | - He-Ping Zhao
- grid.13402.340000 0004 1759 700XMOE Key Lab of Environmental Remediation and Ecosystem Health, College of Environmental and Resource Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
34
|
Prophylactic Administration of a Bacteriophage Cocktail Is Safe and Effective in Reducing Salmonella enterica Serovar Typhimurium Burden in Vivo. Microbiol Spectr 2021; 9:e0049721. [PMID: 34431719 PMCID: PMC8552648 DOI: 10.1128/spectrum.00497-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Nontyphoidal Salmonella bacteria are the causative agent of salmonellosis, which accounts for the majority of foodborne illness of bacterial etiology in humans. Here, we demonstrate the safety and efficacy of the prophylactic administration of a bacteriophage preparation termed FOP (foodborne outbreak pill), which contains lytic phages targeting Salmonella (SalmoFresh phage cocktail), Shiga toxin-producing Escherichia coli (STEC), and Listeria monocytogenes, for lowering Salmonella burdens in OMM12 gnotobiotic mice. Prophylactic administration of FOP significantly reduced the levels of Salmonella in feces and in intestinal sections compared to the levels in controls. Moreover, the overall symptoms of the disease were also considerably lessened. Dose-dependent administration of FOP showed that phage amplification reached similarly high levels in less than 48 h independent of dose. In addition, 16S rRNA gene analysis showed that FOP did not alter the intestinal microbiota of healthy OMM12 mice and reduced microbiota perturbations induced by Salmonella. FOP maintained its full potency against Salmonella in comparison to that of SalmoFresh, its Salmonella-targeting component phages alone. Altogether, the data support that preventive administration of FOP may offer a safe and effective approach for reducing the risk of foodborne infections caused by Salmonella and, potentially, other foodborne bacteria (namely, STEC and L. monocytogenes) targeted by the FOP preparation. IMPORTANCE Foodborne bacterial infections cause worldwide economic loss. During an epidemic, the use of antibiotics to slow down the spread of the disease is not recommended because of their side effects on the resident microbiota and the selection of antibiotic-resistant bacteria. Here, we investigated the potential for the prophylactic administration of bacteriophages (viruses infecting bacteria) to reduce the burden of Salmonella in vivo using mice colonized by a synthetic microbiota. We found that the repeated administration of bacteriophages was safe and efficient in lowering the Salmonella burden. Perturbations of the microbiota by the Salmonella infection were also reduced when mice received bacteriophages. Altogether, these data support the use of bacteriophages as a prophylactic intervention to lower the spread of foodborne epidemics.
Collapse
|
35
|
Dellière S, Dannaoui E, Fieux M, Bonfils P, Gricourt G, Demontant V, Podglajen I, Woerther PL, Angebault C, Botterel F. Analysis of Microbiota and Mycobiota in Fungal Ball Rhinosinusitis: Specific Interaction between Aspergillus fumigatus and Haemophilus influenza? J Fungi (Basel) 2021; 7:550. [PMID: 34356929 PMCID: PMC8305266 DOI: 10.3390/jof7070550] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 12/22/2022] Open
Abstract
Fungal ball (FB) rhinosinusitis (RS) is the main type of non-invasive fungal RS. Despite positive direct examination (DE) of biopsies, culture remains negative in more than 60% of cases. The aim of the study was to evaluate the performance/efficacy of targeted metagenomics (TM) to analyze microbiota and mycobiota in FB and find microbial associations. Forty-five sinus biopsies from patients who underwent surgery for chronic RS were included. After DE and culture, DNA was extracted, then fungal ITS1-ITS2 and bacterial V3-V4 16S rDNA loci were sequenced (MiSeqTM Illumina). Operational taxonomic units (OTUs) were defined via QIIME and assigned to SILVA (16S) and UNITE (ITS) databases. Statistical analyses were performed using SHAMAN. Thirty-eight patients had FB and seven had non-fungal rhinosinusitis (NFRS). DE and culture of FB were positive for fungi in 97.3 and 31.6% of patients, respectively. TM analysis of the 38 FB yielded more than one fungal genus in 100% of cases, with Aspergillus in 89.5% (34/38). Haemophilus was over-represented in FB with >1000 reads/sample in 47.3% (18/38) compared to NFRS (p < 0.001). TM allowed fungal identification in biopsies with negative culture. Haemophilus was associated with FB. Pathogenesis could result from fungi-bacteria interactions in a mixed biofilm-like structure.
Collapse
Affiliation(s)
- Sarah Dellière
- Unité de Parasitologie-Mycologie, Département de Prévention, Diagnostic et Traitement des Infections, APHP, GHU Hôpitaux Universitaires Henri-Mondor, 94010 Créteil, France; (S.D.); (C.A.)
- Unité de Parasitologie-Mycologie, Hôpital Saint-Louis, Assistance Publique des Hôpitaux de Paris, Université de Paris, 75010 Paris, France
| | - Eric Dannaoui
- UR DYNAMiC 7380, Faculté de Santé, Université Paris-Est Créteil, 94010 Créteil, France; (E.D.); (P.-L.W.)
- UR DYNAMiC 7380, Ecole Nationale Vétérinaire d’Alfort, USC Anses, 94700 Maison-Alfort, France
- Unité de Parasitologie-Mycologie, Département de Microbiologie, Hôpital Européen George Pompidou, APHP, Université de Paris, 75015 Paris, France
| | - Maxime Fieux
- Département d’Otorhinolaryngologie, Hôpital Européen George Pompidou, APHP, Université de Paris, 75015 Paris, France; (M.F.); (P.B.)
- Service d’Otorhinolaryngologie, d’Otoneurochirurgie et de Chirurgie Cervico-Faciale, Centre Hospitalier Lyon Sud, Hospices Civils de Lyon, 69310 Pierre Bénite, France
| | - Pierre Bonfils
- Département d’Otorhinolaryngologie, Hôpital Européen George Pompidou, APHP, Université de Paris, 75015 Paris, France; (M.F.); (P.B.)
| | - Guillaume Gricourt
- Plate-Forme Genomiques, APHP-IMRB, GHU Hôpitaux Universitaires Henri-Mondor, UPEC, 94010 Créteil, France; (G.G.); (V.D.)
| | - Vanessa Demontant
- Plate-Forme Genomiques, APHP-IMRB, GHU Hôpitaux Universitaires Henri-Mondor, UPEC, 94010 Créteil, France; (G.G.); (V.D.)
| | - Isabelle Podglajen
- Unité de Bactériologie, Département de Microbiologie, Hôpital Européen George Pompidou, APHP, Université de Paris, 75015 Paris, France;
| | - Paul-Louis Woerther
- UR DYNAMiC 7380, Faculté de Santé, Université Paris-Est Créteil, 94010 Créteil, France; (E.D.); (P.-L.W.)
- Unité de Bactériologie, Département de Prévention, Diagnostic et Traitement des Infections, APHP, GHU Hôpitaux Universitaires Henri-Mondor, 94010 Créteil, France
| | - Cécile Angebault
- Unité de Parasitologie-Mycologie, Département de Prévention, Diagnostic et Traitement des Infections, APHP, GHU Hôpitaux Universitaires Henri-Mondor, 94010 Créteil, France; (S.D.); (C.A.)
- UR DYNAMiC 7380, Faculté de Santé, Université Paris-Est Créteil, 94010 Créteil, France; (E.D.); (P.-L.W.)
- UR DYNAMiC 7380, Ecole Nationale Vétérinaire d’Alfort, USC Anses, 94700 Maison-Alfort, France
| | - Françoise Botterel
- Unité de Parasitologie-Mycologie, Département de Prévention, Diagnostic et Traitement des Infections, APHP, GHU Hôpitaux Universitaires Henri-Mondor, 94010 Créteil, France; (S.D.); (C.A.)
- UR DYNAMiC 7380, Faculté de Santé, Université Paris-Est Créteil, 94010 Créteil, France; (E.D.); (P.-L.W.)
- UR DYNAMiC 7380, Ecole Nationale Vétérinaire d’Alfort, USC Anses, 94700 Maison-Alfort, France
| |
Collapse
|
36
|
Characterization of biliary microbiota dysbiosis in extrahepatic cholangiocarcinoma. PLoS One 2021; 16:e0247798. [PMID: 33690612 PMCID: PMC7943025 DOI: 10.1371/journal.pone.0247798] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
Extrahepatic cholangiocarcinoma (CCA) accounts for 3% of digestive cancers. The role of biliary microbiota as an environment-related modulator has been scarcely investigated in CCA, and the putative impact of associated diseases has not been yet assessed. We characterized the biliary microbiota in CCA patients in order to identify a specific CCA-related dysbiosis. The biliary effluents were collected through an endoscopic retrograde pancreatic cholangiography (ERCP) examination involving 28 CCA and 47 patients with gallstones, herein considered as controls. The biliary effluents were submitted to bacterial DNA extraction and 16S rRNA sequencing, using Illumina technology. Overall, 32% of CCA and 22% of controls displayed another associated disease, such as diabetes, pancreatitis, inflammatory bowel disease, or primary sclerosing cholangitis. Such associated diseases were considered in the comparisons that were made. Principal coordinate analysis (PCoA) detected a significant disparity of biliary microbiota composition between CCA patients and controls without an associated disease. Amongst the most abundant phyla, Proteobacteria did not significantly differ between CCA patients and controls, whereas Firmicutes levels were lower and Bacteroidetes higher in CCAs’ biliary microbiota than in the controls’ microbiota. The most abundant genera were Enterococcus, Streptococcus, Bacteroides, Klebsiella, and Pyramidobacter in CCA’s biliary microbiota. Additionally, levels of Bacteroides, Geobacillus, Meiothermus, and Anoxybacillus genera were significantly higher in CCA patients’ biliary microbiota, without an associated disease, in comparison with controls. A specific CCA-related dysbiosis was identified as compared to controls independently from associated diseases. This suggests that a microorganism community may be involved in CCA pathogenesis.
Collapse
|
37
|
Smith N, Goncalves P, Charbit B, Grzelak L, Beretta M, Planchais C, Bruel T, Rouilly V, Bondet V, Hadjadj J, Yatim N, Pere H, Merkling SH, Ghozlane A, Kernéis S, Rieux-Laucat F, Terrier B, Schwartz O, Mouquet H, Duffy D, Di Santo JP. Distinct systemic and mucosal immune responses during acute SARS-CoV-2 infection. Nat Immunol 2021; 22:1428-1439. [PMID: 34471264 PMCID: PMC8553615 DOI: 10.1038/s41590-021-01028-7] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 08/12/2021] [Indexed: 01/20/2023]
Abstract
Coordinated local mucosal and systemic immune responses following severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection either protect against coronavirus disease 2019 (COVID-19) pathologies or fail, leading to severe clinical outcomes. To understand this process, we performed an integrated analysis of SARS-CoV-2 spike-specific antibodies, cytokines, viral load and bacterial communities in paired nasopharyngeal swabs and plasma samples from a cohort of clinically distinct patients with COVID-19 during acute infection. Plasma viral load was associated with systemic inflammatory cytokines that were elevated in severe COVID-19, and also with spike-specific neutralizing antibodies. By contrast, nasopharyngeal viral load correlated with SARS-CoV-2 humoral responses but inversely with interferon responses, the latter associating with protective microbial communities. Potential pathogenic microorganisms, often implicated in secondary respiratory infections, were associated with mucosal inflammation and elevated in severe COVID-19. Our results demonstrate distinct tissue compartmentalization of SARS-CoV-2 immune responses and highlight a role for the nasopharyngeal microbiome in regulating local and systemic immunity that determines COVID-19 clinical outcomes.
Collapse
Affiliation(s)
- Nikaïa Smith
- grid.428999.70000 0001 2353 6535Translational Immunology Lab, Institut Pasteur, Paris, France
| | - Pedro Goncalves
- grid.428999.70000 0001 2353 6535Innate Immunity Unit, Institut Pasteur, INSERM U1223, Paris, France
| | - Bruno Charbit
- grid.428999.70000 0001 2353 6535Cytometry and Biomarkers UTechS, Institut Pasteur, Paris, France
| | - Ludivine Grzelak
- grid.428999.70000 0001 2353 6535Virus and Immunity Unit, Institut Pasteur, Paris, France ,grid.508487.60000 0004 7885 7602Sorbonne Paris Cité, Université de Paris, Paris, France
| | - Maxime Beretta
- grid.428999.70000 0001 2353 6535Humoral Immunology Laboratory, Institut Pasteur, INSERM U1222, Paris, France
| | - Cyril Planchais
- grid.428999.70000 0001 2353 6535Humoral Immunology Laboratory, Institut Pasteur, INSERM U1222, Paris, France
| | - Timothée Bruel
- grid.428999.70000 0001 2353 6535Virus and Immunity Unit, Institut Pasteur, Paris, France
| | | | - Vincent Bondet
- grid.428999.70000 0001 2353 6535Translational Immunology Lab, Institut Pasteur, Paris, France
| | - Jérôme Hadjadj
- grid.508487.60000 0004 7885 7602Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), Université de Paris, Paris, France ,grid.508487.60000 0004 7885 7602Imagine Institute, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Université de Paris, Paris, France
| | - Nader Yatim
- grid.428999.70000 0001 2353 6535Translational Immunology Lab, Institut Pasteur, Paris, France ,grid.508487.60000 0004 7885 7602Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), Université de Paris, Paris, France
| | - Helene Pere
- grid.417925.cUnité de Génomique Fonctionnelle des Tumeurs Solides, Centre de Recherche des Cordeliers, INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Sarah H. Merkling
- grid.428999.70000 0001 2353 6535Insect-Virus Interactions Unit, Institut Pasteur, CNRS UMR2000, Paris, France
| | - Amine Ghozlane
- grid.428999.70000 0001 2353 6535Hub de Bioinformatique et Biostatistique, Institut Pasteur, Paris, France
| | - Solen Kernéis
- grid.411784.f0000 0001 0274 3893Equipe Mobile d’Infectiologie, Hôpital Cochin, AP-HP, APHP-CUP, Paris, France ,Université de Paris, INSERM, IAME, Paris, France ,grid.428999.70000 0001 2353 6535Epidemiology and Antimicrobial Resistance Modeling Laboratory, Institut Pasteur, Paris, France
| | - Frederic Rieux-Laucat
- grid.508487.60000 0004 7885 7602Imagine Institute, Laboratory of Immunogenetics of Pediatric Autoimmune Diseases, INSERM UMR 1163, Université de Paris, Paris, France
| | - Benjamin Terrier
- grid.508487.60000 0004 7885 7602Department of Internal Medicine, National Referral Center for Rare Systemic Autoimmune Diseases, Assistance Publique Hôpitaux de Paris-Centre (APHP-CUP), Université de Paris, Paris, France
| | - Olivier Schwartz
- grid.428999.70000 0001 2353 6535Virus and Immunity Unit, Institut Pasteur, Paris, France
| | - Hugo Mouquet
- grid.428999.70000 0001 2353 6535Humoral Immunology Laboratory, Institut Pasteur, INSERM U1222, Paris, France
| | - Darragh Duffy
- grid.428999.70000 0001 2353 6535Translational Immunology Lab, Institut Pasteur, Paris, France
| | - James P. Di Santo
- grid.428999.70000 0001 2353 6535Innate Immunity Unit, Institut Pasteur, INSERM U1223, Paris, France
| |
Collapse
|
38
|
Bergsten E, Mestivier D, Sobhani I. The Limits and Avoidance of Biases in Metagenomic Analyses of Human Fecal Microbiota. Microorganisms 2020; 8:microorganisms8121954. [PMID: 33317070 PMCID: PMC7764459 DOI: 10.3390/microorganisms8121954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/04/2020] [Accepted: 12/07/2020] [Indexed: 01/02/2023] Open
Abstract
An increasing body of evidence highlights the role of fecal microbiota in various human diseases. However, more than two-thirds of fecal bacteria cannot be cultivated by routine laboratory techniques. Thus, physicians and scientists use DNA sequencing and statistical tools to identify associations between bacterial subgroup abundances and disease. However, discrepancies between studies weaken these results. In the present study, we focus on biases that might account for these discrepancies. First, three different DNA extraction methods (G’NOME, QIAGEN, and PROMEGA) were compared with regard to their efficiency, i.e., the quality and quantity of DNA recovered from feces of 10 healthy volunteers. Then, the impact of the DNA extraction method on the bacteria identification and quantification was evaluated using our published cohort of sample subjected to both 16S rRNA sequencing and whole metagenome sequencing (WMS). WMS taxonomical assignation employed the universal marker genes profiler mOTU-v2, which is considered the gold standard. The three standard pipelines for 16S RNA analysis (MALT and MEGAN6, QIIME1, and DADA2) were applied for comparison. Taken together, our results indicate that the G’NOME-based method was optimal in terms of quantity and quality of DNA extracts. 16S rRNA sequence-based identification of abundant bacteria genera showed acceptable congruence with WMS sequencing, with the DADA2 pipeline yielding the highest congruent levels. However, for low abundance genera (<0.5% of the total abundance) two pipelines and/or validation by quantitative polymerase chain reaction (qPCR) or WMS are required. Hence, 16S rRNA sequencing for bacteria identification and quantification in clinical and translational studies should be limited to diagnostic purposes in well-characterized and abundant genera. Additional techniques are warranted for low abundant genera, such as WMS, qPCR, or the use of two bio-informatics pipelines.
Collapse
Affiliation(s)
- Emma Bergsten
- EA7375 (EC2M3 Research Team), Université Paris Est, 94010 Créteil, France; (E.B.); (D.M.)
| | - Denis Mestivier
- EA7375 (EC2M3 Research Team), Université Paris Est, 94010 Créteil, France; (E.B.); (D.M.)
- Bioinformatics Core Facility, Institut Mondor de Recherche Biomédicale, UMR 955—Institut National de la Santé et de la Recherche Médicale—UPEC, 94010 Créteil, France
| | - Iradj Sobhani
- EA7375 (EC2M3 Research Team), Université Paris Est, 94010 Créteil, France; (E.B.); (D.M.)
- Service de Gastroenterologie, Hôpital Henri Mondor, Assistance Publique-Hôpitaux de Paris, 94010 Créteil, France
- Correspondence: ; Tel.: +33-149814358
| |
Collapse
|
39
|
Stressmann FA, Bernal-Bayard J, Perez-Pascual D, Audrain B, Rendueles O, Briolat V, Bruchmann S, Volant S, Ghozlane A, Häussler S, Duchaud E, Levraud JP, Ghigo JM. Mining zebrafish microbiota reveals key community-level resistance against fish pathogen infection. ISME JOURNAL 2020; 15:702-719. [PMID: 33077888 DOI: 10.1038/s41396-020-00807-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 09/30/2020] [Accepted: 10/05/2020] [Indexed: 02/08/2023]
Abstract
The long-known resistance to pathogens provided by host-associated microbiota fostered the notion that adding protective bacteria could prevent or attenuate infection. However, the identification of endogenous or exogenous bacteria conferring such protection is often hindered by the complexity of host microbial communities. Here, we used zebrafish and the fish pathogen Flavobacterium columnare as a model system to study the determinants of microbiota-associated colonization resistance. We compared infection susceptibility in germ-free, conventional and reconventionalized larvae and showed that a consortium of 10 culturable bacterial species are sufficient to protect zebrafish. Whereas survival to F. columnare infection does not rely on host innate immunity, we used antibiotic dysbiosis to alter zebrafish microbiota composition, leading to the identification of two different protection strategies. We first identified that the bacterium Chryseobacterium massiliae individually protects both larvae and adult zebrafish. We also showed that an assembly of 9 endogenous zebrafish species that do not otherwise protect individually confer a community-level resistance to infection. Our study therefore provides a rational approach to identify key endogenous protecting bacteria and promising candidates to engineer resilient microbial communities. It also shows how direct experimental analysis of colonization resistance in low-complexity in vivo models can reveal unsuspected ecological strategies at play in microbiota-based protection against pathogens.
Collapse
Affiliation(s)
- Franziska A Stressmann
- Genetics of Biofilms Laboratory, Institut Pasteur, UMR CNRS2001, 75015, Paris, France.,Department of Chemical Analytics and Biogeochemistry, Leibniz-Institute of Freshwater Ecology and Inland Fisheries, Müggelseedamm 310, 12587, Berlin, Germany
| | - Joaquín Bernal-Bayard
- Genetics of Biofilms Laboratory, Institut Pasteur, UMR CNRS2001, 75015, Paris, France.,Departamento de Genética, Facultad de Biología, Universidad de Sevilla, Apartado 1095, 41080, Sevilla, Spain
| | - David Perez-Pascual
- Genetics of Biofilms Laboratory, Institut Pasteur, UMR CNRS2001, 75015, Paris, France
| | - Bianca Audrain
- Genetics of Biofilms Laboratory, Institut Pasteur, UMR CNRS2001, 75015, Paris, France
| | - Olaya Rendueles
- Genetics of Biofilms Laboratory, Institut Pasteur, UMR CNRS2001, 75015, Paris, France.,Microbial Evolutionary Genomics Laboratory, Institut Pasteur, UMR3525, 75015, Paris, France
| | - Valérie Briolat
- Macrophages and Development of Immunity Laboratory, Institut Pasteur, UMR3738 CNRS, 75015, Paris, France
| | - Sebastian Bruchmann
- Department of Molecular Bacteriology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department of Veterinary Medicine, University of Cambridge, Madingley Road, Cambridge, CB3 0ES, UK
| | - Stevenn Volant
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, Paris, France
| | - Amine Ghozlane
- Hub de Bioinformatique et Biostatistique - Département Biologie Computationnelle, Institut Pasteur, USR 3756 CNRS, Paris, France
| | - Susanne Häussler
- Department of Molecular Bacteriology, Helmholtz Centre for Infection Research, Braunschweig, Germany.,Department of Clinical Microbiology, Rigshospitalet, 2100, Copenhagen, Denmark
| | - Eric Duchaud
- Unité VIM, INRAE, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Jean-Pierre Levraud
- Macrophages and Development of Immunity Laboratory, Institut Pasteur, UMR3738 CNRS, 75015, Paris, France
| | - Jean-Marc Ghigo
- Genetics of Biofilms Laboratory, Institut Pasteur, UMR CNRS2001, 75015, Paris, France.
| |
Collapse
|
40
|
Guenin-Macé L, Morel JD, Doisne JM, Schiavo A, Boulet L, Mayau V, Goncalves P, Duchatelet S, Hovnanian A, Bondet V, Duffy D, Ungeheuer MN, Delage M, Nassif A, Di Santo JP, Demangel C. Dysregulation of tryptophan catabolism at the host-skin microbiota interface in hidradenitis suppurativa. JCI Insight 2020; 5:140598. [PMID: 32970636 PMCID: PMC7605522 DOI: 10.1172/jci.insight.140598] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 09/10/2020] [Indexed: 12/30/2022] Open
Abstract
Hidradenitis suppurativa (HS) is a chronic skin disorder of unknown etiology that manifests as recurrent, painful lesions. Cutaneous dysbiosis and unresolved inflammation are hallmarks of active HS, but their origin and interplay remain unclear. Our metabolomic profiling of HS skin revealed an abnormal induction of the kynurenine pathway of tryptophan catabolism in dermal fibroblasts, correlating with the release of kynurenine pathway–inducing cytokines by inflammatory cell infiltrates. Notably, overactivation of the kynurenine pathway in lesional skin was associated with local and systemic depletion in tryptophan. Yet the skin microbiota normally degrades host tryptophan into indoles regulating tissue inflammation via engagement of the aryl hydrocarbon receptor (AHR). In HS skin lesions, we detected contextual defects in AHR activation coinciding with impaired production of bacteria-derived AHR agonists and decreased incidence of AHR ligand-producing bacteria in the resident flora. Dysregulation of tryptophan catabolism at the skin-microbiota interface thus provides a mechanism linking the immunological and microbiological features of HS lesions. In addition to revealing metabolic alterations in patients with HS, our study suggests that correcting AHR signaling would help restore immune homeostasis in HS skin. Loss of homeostasis of tryptophan metabolism at the host-microbiota interface may contribute to Hidradenitis Suppurativa.
Collapse
Affiliation(s)
- Laure Guenin-Macé
- Immunobiology of Infection Unit, Institut Pasteur, INSERM U1221, Paris, France
| | - Jean-David Morel
- Immunobiology of Infection Unit, Institut Pasteur, INSERM U1221, Paris, France.,Université Paris 7, Sorbonne Paris Cité, Paris, France.,ENS de Lyon, Lyon, France
| | - Jean-Marc Doisne
- Innate Immunity Unit, Institut Pasteur, INSERM U1223, Paris, France
| | - Angèle Schiavo
- Innate Immunity Unit, Institut Pasteur, INSERM U1223, Paris, France
| | - Lysiane Boulet
- Laboratoire de Biochimie Hormonale et Nutritionnelle, CHU Grenoble-Alpes, La Tronche, France
| | - Véronique Mayau
- Immunobiology of Infection Unit, Institut Pasteur, INSERM U1221, Paris, France
| | - Pedro Goncalves
- Innate Immunity Unit, Institut Pasteur, INSERM U1223, Paris, France
| | - Sabine Duchatelet
- Université de Paris, Imagine Institute, Laboratory of Genetic Skin Diseases, INSERM UMR 1163, Paris, France
| | - Alain Hovnanian
- Université de Paris, Imagine Institute, Laboratory of Genetic Skin Diseases, INSERM UMR 1163, Paris, France.,Department of Genetics, Assistance Publique-Hôpitaux de Paris, Hôpital Necker-Enfants Malades, Paris, France
| | - Vincent Bondet
- Immunobiology of Dendritic Cells, Institut Pasteur, INSERM U1223, Paris, France
| | - Darragh Duffy
- Immunobiology of Dendritic Cells, Institut Pasteur, INSERM U1223, Paris, France
| | | | - Maïa Delage
- Centre Médical, Institut Pasteur, Paris, France
| | - Aude Nassif
- Centre Médical, Institut Pasteur, Paris, France
| | - James P Di Santo
- Innate Immunity Unit, Institut Pasteur, INSERM U1223, Paris, France
| | - Caroline Demangel
- Immunobiology of Infection Unit, Institut Pasteur, INSERM U1221, Paris, France
| |
Collapse
|