1
|
Lawrence GW, Garcia-Gutierrez E, O'Mahony AK, Walsh CJ, O'Connor PM, Begley M, Guinane CM, Cotter PD. A gut-derived Streptococcus salivarius produces the novel nisin variant designated nisin G and inhibits Fusobacterium nucleatum in a model of the human distal colon microbiome. mBio 2024:e0157324. [PMID: 39692472 DOI: 10.1128/mbio.01573-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Fusobacterium nucleatum is a human pathogen associated with intestinal conditions including colorectal cancer. Screening for gut-derived strains that exhibit anti-F. nucleatum activity in vitro revealed Streptococcus salivarius DPC6487 as a strain of interest. Whole-genome sequencing of S. salivarius DPC6487 identified a nisin operon with a novel structural variant designated nisin G. The structural nisin G peptide differs from the prototypical nisin A with respect to seven amino acids (Ile4Tyr, Ala15Val, Gly18Ala, Asn20His, Met21Leu, His27Asn, and His31Ile), including differences that have not previously been associated with a natural nisin variant. The nisin G gene cluster consists of nsgGEFABTCPRK with transposases encoded between the nisin G structural gene (nsgA) and nsgF, notably lacking an equivalent to the nisI immunity determinant. S. salivarius DPC6487 exhibited a narrower spectrum of activity in vitro compared to the nisin A-producing Lactococcus lactis NZ9700. Nisin G-producing S. salivarius DPC6487 demonstrated the ability to control F. nucleatum DSM15643 in an ex vivo model colonic environment while exerting minimal impact on the surrounding microbiota. The production of this bacteriocin by a gut-derived S. salivarius, its narrow-spectrum activity, and its anti-F. nucleatum activity in a model colonic environment indicates that this strain merits further attention with a view to harnessing its probiotic potential.IMPORTANCEFusobacterium nucleatum is a human pathogen associated with intestinal conditions, including colorectal cancer, making it a potentially important therapeutic target. Bacteriocin-producing probiotic bacteria demonstrate the potential to target disease-associated taxa in situ in the gut. A gut-derived strain Streptococcus salivarius DPC6487 was found to demonstrate anti-F. nucleatum activity, which was attributable to a gene encoding a novel nisin variant designated nisin G. Nisin G-producing S. salivarius DPC6487 demonstrated the ability to control an infection of F. nucleatum in a simulated model of the human distal colon while exerting minimal impact on the surrounding microbiota. Here, we describe this nisin variant produced by S. salivarius, a species that is frequently a focus for probiotic development. The production of nisin G by a gut-derived S. salivarius, its narrow-spectrum activity against F. nucleatum, and its anti-F. nucleatum activity in a model colonic environment warrants further research to determine its probiotic-related applications.
Collapse
Affiliation(s)
- Garreth W Lawrence
- Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | | | - A Kate O'Mahony
- Teagasc Food Research Centre, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | | | - Paula M O'Connor
- Teagasc Food Research Centre, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
| | - Máire Begley
- Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Caitriona M Guinane
- Department of Biological Sciences, Munster Technological University, Cork, Ireland
| | - Paul D Cotter
- Teagasc Food Research Centre, Cork, Ireland
- APC Microbiome Ireland, Cork, Ireland
- VistaMilk SFI Research Centre, Cork, Ireland
| |
Collapse
|
2
|
Khalil M, Di Ciaula A, Mahdi L, Jaber N, Di Palo DM, Graziani A, Baffy G, Portincasa P. Unraveling the Role of the Human Gut Microbiome in Health and Diseases. Microorganisms 2024; 12:2333. [PMID: 39597722 PMCID: PMC11596745 DOI: 10.3390/microorganisms12112333] [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: 10/28/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
The human gut is a complex ecosystem that supports billions of living species, including bacteria, viruses, archaea, phages, fungi, and unicellular eukaryotes. Bacteria give genes and enzymes for microbial and host-produced compounds, establishing a symbiotic link between the external environment and the host at both the gut and systemic levels. The gut microbiome, which is primarily made up of commensal bacteria, is critical for maintaining the healthy host's immune system, aiding digestion, synthesizing essential nutrients, and protecting against pathogenic bacteria, as well as influencing endocrine, neural, humoral, and immunological functions and metabolic pathways. Qualitative, quantitative, and/or topographic shifts can alter the gut microbiome, resulting in dysbiosis and microbial dysfunction, which can contribute to a variety of noncommunicable illnesses, including hypertension, cardiovascular disease, obesity, diabetes, inflammatory bowel disease, cancer, and irritable bowel syndrome. While most evidence to date is observational and does not establish direct causation, ongoing clinical trials and advanced genomic techniques are steadily enhancing our understanding of these intricate interactions. This review will explore key aspects of the relationship between gut microbiota, eubiosis, and dysbiosis in human health and disease, highlighting emerging strategies for microbiome engineering as potential therapeutic approaches for various conditions.
Collapse
Affiliation(s)
- Mohamad Khalil
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Agostino Di Ciaula
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Laura Mahdi
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Nour Jaber
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| | - Domenica Maria Di Palo
- Division of Hygiene, Department of Interdisciplinary Medicine, University of Bari Aldo Moro, Piazza Giulio Cesare 11, 70124 Bari, Italy;
| | - Annarita Graziani
- Institut AllergoSan Pharmazeutische Produkte Forschungs- und Vertriebs GmbH, 8055 Graz, Austria;
| | - Gyorgy Baffy
- Division of Gastroenterology, Hepatology and Endoscopy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02130, USA;
- Section of Gastroenterology, Department of Medicine, VA Boston Healthcare System, Boston, MA 02130, USA
| | - Piero Portincasa
- Clinica Medica “A. Murri”, Department of Precision and Regenerative Medicine and Ionian Area (DiMePre-J), Medical School, University of Bari Aldo Moro, 70124 Bari, Italy; (M.K.); (A.D.C.); (L.M.); (N.J.)
| |
Collapse
|
3
|
N S SA, Pillai DS, Shanmugam R. The Antifungal Activity of Chitosan Nanoparticle-Incorporated Probiotics Against Oral Candidiasis. Cureus 2024; 16:e70093. [PMID: 39449901 PMCID: PMC11501512 DOI: 10.7759/cureus.70093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 09/24/2024] [Indexed: 10/26/2024] Open
Abstract
BACKGROUND Candida species, especially Candida albicans, cause oral candidiasis, also known as oral thrush. It affects the elderly, newborns, patients under antibiotics, chemotherapeutic agents, and patients with weaker immune systems. Although successful, traditional antifungal medications are available, they may have negative effects and do not prevent recurrence. Conventional antifungal medications have a restricted range of effectiveness; hence, the need for a novel antifungal agent arises. Chitosan, a natural polymer made from chitin found in crustaceans, kills fungal cells by damaging membranes. Excellent biocompatibility and biodegradability make it a promising medical material. Probiotics, live bacteria, compete with pathogenic microbes for adhesion sites and nutrients, produce antimicrobials such as lactic acid, hydrogen peroxide, and bacteriocins, and modulate the host's immunological response. Adding probiotics to chitosan nanoparticles (CSNPs) boosts their antifungal activity against Candida species and increases their stability and transport to the infection site. METHODS The antifungal activity was evaluated through in vitro experiments utilizing Candida strains that are significant to oral candidiasis. Different concentrations of CSNPs and formulations loaded with probiotics were examined to assess their effectiveness. The antifungal properties were assessed using microbiological assays, specifically agar diffusion and minimum inhibitory concentration (MIC). RESULTS The study demonstrated a notable fungicidal impact of CSNPs combined with probiotics against oral candidiasis. Furthermore, the MIC values were lower for the probiotic-loaded CSNPs than for chitosan alone or probiotics alone. CONCLUSION The combination of CSNPs and probiotics shows potential in effectively combating oral candidiasis by inhibiting fungal growth. This novel method offers a promising strategy for creating therapeutic treatments for oral candidiasis by utilizing the combined benefits of chitosan and probiotics to combat fungal infections effectively.
Collapse
Affiliation(s)
- Shree Abiraami N S
- Oral Medicine and Radiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Devika S Pillai
- Oral Medicine and Radiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Rajeshkumar Shanmugam
- Nanobiomedicine, Centre for Global Health Research, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
4
|
Lindon S, Shah S, Gifford DR, Lood C, Gomis Font MA, Kaur D, Oliver A, MacLean RC, Wheatley RM. Antibiotic resistance alters the ability of Pseudomonas aeruginosa to invade bacteria from the respiratory microbiome. Evol Lett 2024; 8:735-747. [PMID: 39328287 PMCID: PMC11424078 DOI: 10.1093/evlett/qrae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 06/06/2024] [Accepted: 06/14/2024] [Indexed: 09/28/2024] Open
Abstract
The emergence and spread of antibiotic resistance in bacterial pathogens is a global health threat. One important unanswered question is how antibiotic resistance influences the ability of a pathogen to invade the host-associated microbiome. Here we investigate how antibiotic resistance impacts the ability of a bacterial pathogen to invade bacteria from the microbiome, using the opportunistic bacterial pathogen Pseudomonas aeruginosa and the respiratory microbiome as our model system. We measure the ability of P. aeruginosa spontaneous antibiotic-resistant mutants to invade pre-established cultures of commensal respiratory microbes in an assay that allows us to link specific resistance mutations with changes in invasion ability. While commensal respiratory microbes tend to provide some degree of resistance to P. aeruginosa invasion, antibiotic resistance is a double-edged sword that can either help or hinder the ability of P. aeruginosa to invade. The directionality of this help or hindrance depends on both P. aeruginosa genotype and respiratory microbe identity. Specific resistance mutations in genes involved in multidrug efflux pump regulation are shown to facilitate the invasion of P. aeruginosa into Staphylococcus lugdunensis, yet impair invasion into Rothia mucilaginosa and Staphylococcus epidermidis. Streptococcus species provide the strongest resistance to P. aeruginosa invasion, and this is maintained regardless of antibiotic resistance genotype. Our study demonstrates how the cost of mutations that provide enhanced antibiotic resistance in P. aeruginosa can crucially depend on community context. We suggest that attempts to manipulate the microbiome should focus on promoting the growth of commensals that can increase the fitness costs associated with antibiotic resistance and provide robust inhibition of both wildtype and antibiotic-resistant pathogen strains.
Collapse
Affiliation(s)
- Selina Lindon
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Sarah Shah
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Danna R Gifford
- Division of Evolution, Infection and Genomics, School of Biological Sciences, The University of Manchester, Manchester, United Kingdom
| | - Cédric Lood
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Maria A Gomis Font
- Servicio de Microbiología, Hospital Universitari Son Espases, Instituto de Investigación Sanitaria Illes Balears (IdISBa), CIBERINFEC, Palma de Mallorca, Spain
| | - Divjot Kaur
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Antonio Oliver
- Servicio de Microbiología, Hospital Universitari Son Espases, Instituto de Investigación Sanitaria Illes Balears (IdISBa), CIBERINFEC, Palma de Mallorca, Spain
| | - R Craig MacLean
- Department of Biology, University of Oxford, Oxford, United Kingdom
| | - Rachel M Wheatley
- Department of Biology, University of Oxford, Oxford, United Kingdom
- School of Biological Sciences, Queen's University Belfast, Belfast, United Kingdom
| |
Collapse
|
5
|
Senthil Kumar S, Johnson MDL, Wilson JE. Insights into the enigma of oral streptococci in carcinogenesis. Microbiol Mol Biol Rev 2024; 88:e0009523. [PMID: 38506551 PMCID: PMC11338076 DOI: 10.1128/mmbr.00095-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/21/2024] Open
Abstract
SUMMARYThe genus Streptococcus consists of a taxonomically diverse group of Gram-positive bacteria that have earned significant scientific interest due to their physiological and pathogenic characteristics. Within the genus Streptococcus, viridans group streptococci (VGS) play a significant role in the oral ecosystem, constituting approximately 80% of the oral biofilm. Their primary role as pioneering colonizers in the oral cavity with multifaceted interactions like adherence, metabolic signaling, and quorum sensing contributes significantly to the complex dynamics of the oral biofilm, thus shaping oral health and disease outcomes. Perturbations in oral streptococci composition drive oral dysbiosis and therefore impact host-pathogen interactions, resulting in oral inflammation and representing VGS as an opportunistic pathogen. The association of oral streptococci in tumors across distant organs, spanning the esophagus, stomach, pancreas, and colon, illuminates a potential association between oral streptococci, inflammation, and tumorigenesis. This finding emphasizes the need for further investigations into the role of oral streptococci in mucosal homeostasis and their involvement in carcinogenesis. Hence, here, we review the significance of oral streptococci in biofilm dynamics and how the perturbation may impact mucosal immunopathogenesis in the context of cancer, with a vision of exploiting oral streptococci for cancer intervention and for the development of non-invasive cancer diagnosis.
Collapse
Affiliation(s)
- Sangeetha Senthil Kumar
- Department of
Immunobiology, The University of
Arizona, Tucson,
Arizona, USA
- The University of
Arizona Cancer Center,
Tucson, Arizona, USA
| | - Michael D. L. Johnson
- Department of
Immunobiology, The University of
Arizona, Tucson,
Arizona, USA
- Valley Fever Center
for Excellence, The University of Arizona College of
Medicine, Tucson,
Arizona, USA
- BIO5 Institute, The
University of Arizona College of
Medicine, Tucson,
Arizona, USA
- Asthma and Airway
Disease Research Center, The University of Arizona College of
Medicine, Tucson,
Arizona, USA
| | - Justin E. Wilson
- Department of
Immunobiology, The University of
Arizona, Tucson,
Arizona, USA
- The University of
Arizona Cancer Center,
Tucson, Arizona, USA
| |
Collapse
|
6
|
Mato EG, Montaño-Barrientos BJ, Rivas-Mundiña B, Aneiros IV, López LS, Posse JL, Lamas LM. Anti-caries Streptococcus spp.: A potential preventive tool for special needs patients. SPECIAL CARE IN DENTISTRY 2024; 44:813-822. [PMID: 37674277 DOI: 10.1111/scd.12920] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 08/16/2023] [Indexed: 09/08/2023]
Abstract
INTRODUCTION Probiotics are living microorganisms that act on the host-microbiome interface to restore the microbiota's physiological homeostasis. Numerous probiotics have been marketed with inhibitory activity against Streptococcus mutans and consequently with a potential anti-caries effect, mainly of the genera Lactobacillus and Bifidobacterium, whose main disadvantage is their limited ability to settle in the oral cavity. METHODS This narrative review describes the main Streptococcus spp. with probiotic anti-Streptococcus mutans activity, whose substantivity is greater than that of Lactobacillus spp. and consequently with anti-caries potentiality. We performed a literature review in the PubMed, Science Direct and Google Scholar databases of articles published in English (without time restriction) related to caries and probiotics. RESULTS The potential identified anti-caries probiotics included Streptococcus spp. A12, Streptococcus oralis (AJ3), Streptococcus oligofermentans, Streptococcus salivarius (K12, M18, JH, LAB813, 24SMB), Streptococcus spp. with arginolytic activity (S. sanguinis, S. gordonii, S. ratti, S. parasanguinis, S. intermedius, S. australis, and S. cristatus), Streptococcus rattus (JH145), Streptococcus dentisani and Streptococcus downii. CONCLUSIONS The possibility of using these Streptococcus spp. as probiotics that inhibit the growth of dental plaque and the development of carious lesions represents a potential tool of particular interest for individuals with physical or intellectual disabilities that impede the routine and effective application of mechanical dental plaque removal techniques.
Collapse
Affiliation(s)
- Eliane García Mato
- Medical-Surgical Dentistry Research Group (OMEQUI), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Bitalio J Montaño-Barrientos
- Clinical Microbiology, Hospital Álvaro Cunqueiro, University Hospital Complex of Vigo, Microbiology and Infectology Group, Galicia Sur Health Research Institute (IISGS), Vigo, Galicia, Spain
| | - Berta Rivas-Mundiña
- Medical-Surgical Dentistry Research Group (OMEQUI), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Iván Varela Aneiros
- Medical-Surgical Dentistry Research Group (OMEQUI), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Lucía Sande López
- Medical-Surgical Dentistry Research Group (OMEQUI), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Jacobo Limeres Posse
- Medical-Surgical Dentistry Research Group (OMEQUI), Health Research Institute of Santiago de Compostela (IDIS), University of Santiago de Compostela (USC), Santiago de Compostela, Spain
| | - Lucía Martínez Lamas
- Clinical Microbiology, Hospital Álvaro Cunqueiro, University Hospital Complex of Vigo, Microbiology and Infectology Group, Galicia Sur Health Research Institute (IISGS), Vigo, Galicia, Spain
| |
Collapse
|
7
|
Laws GA, Harold LK, Tagg JR, Hale JDF. Interferon Gamma Response in Human Saliva Following Exposure to the Oral Probiotic Streptococcus salivarius BLIS K12. Probiotics Antimicrob Proteins 2024; 16:93-98. [PMID: 36477439 DOI: 10.1007/s12602-022-10010-0] [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] [Accepted: 11/15/2022] [Indexed: 12/12/2022]
Abstract
Streptococcus salivarius BLIS K12 is a probiotic strain developed for application to the oral cavity. The strain was originally characterised for its in vitro antibacterial activity against the prominent oral pathogen Streptococcus pyogenes. More recent research has expanded its applications to include reducing halitosis, preventing otitis media and protecting against virus infections of the respiratory tract. A potential mechanism for this anti-viral activity could be the stimulation of salivary interferon gamma (IFN-γ) production in the oral cavity. The aim of this study was to investigate whether the ingestion of and oral cavity colonisation by S. salivarius BLIS K12 is associated with enhancement of IFN-γ levels in saliva. Application of ELISA demonstrated that consumption of S. salivarius BLIS K12 effected an increase in salivary IFN-γ, and this response was more consistent with use of viable cells than following ingestion of heat-killed S. salivarius BLIS K12. Interestingly, those subjects who more successfully colonised with S. salivarius BLIS K12 did not experience a relatively larger increase in their IFN-γ levels, indicating that the observed IFN-γ response occurs independently of colonisation efficacy. In summary, the consumption of S. salivarius BLIS K12 increases salivary levels of IFN-γ, an effect that may contribute to protection of the host against certain virus infections.
Collapse
Affiliation(s)
- Gemma A Laws
- Blis Technologies, 81 Glasgow St, South Dunedin, 9012, New Zealand
| | - Liam K Harold
- Blis Technologies, 81 Glasgow St, South Dunedin, 9012, New Zealand
| | - John R Tagg
- Blis Technologies, 81 Glasgow St, South Dunedin, 9012, New Zealand
| | - John D F Hale
- Blis Technologies, 81 Glasgow St, South Dunedin, 9012, New Zealand.
| |
Collapse
|
8
|
Reichardt E, Shyp V, Alig L, Verna C, Kulik EM, Bornstein MM. Antimicrobial effect of probiotic bacteriocins on Streptococcus mutans biofilm in a dynamic oral flow chamber model - an in vitro study. J Oral Microbiol 2024; 16:2304971. [PMID: 38288293 PMCID: PMC10823884 DOI: 10.1080/20002297.2024.2304971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 01/05/2024] [Indexed: 01/31/2024] Open
Abstract
Aim To determine the antimicrobial activity of the bacteriocin-producing probiotic strains Streptococcus salivarius K12 and Streptococcus salivarius M18 alone or in combination against caries-associated Streptococcus mutans. Methods Antimicrobial activity of S. salivarius K12 and/or S. salivarius M18 against S. mutans ATCC 25175 growth and biofilm formation on hydroxyapatite (HA) discs was determined in a flow chamber model by recording the colony forming units (CFU/ml) after 48 h of co-cultivation. The biofilm was analyzed by scanning electron microscopy (SEM) and by confocal laser scanning microscopy (CLSM). Additionally, the simultaneous antagonism assay was used to assess the inhibitory effect of S. salivarius K12 and/or S. salivarius M18 against S. mutans ATCC 25175 and 21 clinical isolates of S. mutans. Results Co-cultivation of S. mutans and S. salivarius K12 and/or S. salivarius M18 led to the inhibition of S. mutans viability, thereby, preventing its biofilm formation on HA discs. Furthermore, S. salivarius K12 and S. salivarius M18 exhibited antimicrobial activity against most clinical isolates of S. mutans. Conclusion The in vitro flow chamber system used in this study allows the simulation of time-dependent administration of S. salivarius probiotic strains, either alone or in combination, to investigate the prevention of S. mutans biofilm formation in a standardized model.
Collapse
Affiliation(s)
- Elisabeth Reichardt
- Department of Pediatric Oral Health and Orthodontics, University Center for Dental Medicine UZB, University of Basel, Basel, Switzerland
| | - Viktoriya Shyp
- Department Research, University Center for Dental Medicine UZB, University of Basel, Basel, Switzerland
| | - Lea Alig
- Department Research, University Center for Dental Medicine UZB, University of Basel, Basel, Switzerland
| | - Carlalberta Verna
- Department of Pediatric Oral Health and Orthodontics, University Center for Dental Medicine UZB, University of Basel, Basel, Switzerland
| | - Eva M Kulik
- Department Research, University Center for Dental Medicine UZB, University of Basel, Basel, Switzerland
| | - Michael M Bornstein
- Department of Oral Health & Medicine, University Center for Dental Medicine UZB, University of Basel, Basel, Switzerland
| |
Collapse
|
9
|
Tian Z, Liu Y, Zhu D, Cao B, Cui M. Changes in Intestinal Flora and Serum Metabolites Pre- and Post-Antitumor Drug Therapy in Patients with Non-Small Cell Lung Cancer. J Clin Med 2024; 13:529. [PMID: 38256661 PMCID: PMC10816336 DOI: 10.3390/jcm13020529] [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: 12/05/2023] [Revised: 01/04/2024] [Accepted: 01/15/2024] [Indexed: 01/24/2024] Open
Abstract
OBJECTIVE this study aimed to identify the relationships between gut microbiota, metabolism, and non-small cell lung cancer (NSCLC) treatment outcomes, which are presently unclear. METHODS in this single-center prospective cohort study, we investigated changes in the gut microbiota and serum metabolite profile in 60 patients with NSCLC after four cycles of anticancer therapy. RESULTS The microbial landscape of the gut exhibited a surge in Proteobacteria and Verrucomicrobiota populations, alongside a decline in Firmicutes, Actinobacteriota, and Bacteroidota. Furthermore, a significant shift in the prevalence of certain bacterial genera was noted, with an increase in Escherichia/Shigella and Klebsiella, contrasted by a reduction in Bifidobacterium. Metabolomic analysis uncovered significant changes in lipid abundances, with certain metabolic pathways markedly altered post-treatment. Correlation assessments identified strong links between certain gut microbial genera and serum metabolite concentrations. Despite these findings, a subgroup analysis delineating patient responses to therapy revealed no significant shifts in the gut microbiome's composition after four cycles of treatment. CONCLUSIONS This study emphasizes the critical role of gut microbiota changes in NSCLC patients during anticancer treatment. These insights pave the way for managing treatment complications and inform future research to improve patient care by understanding and addressing these microbiota changes.
Collapse
Affiliation(s)
- Zhenyu Tian
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China; (Z.T.); (D.Z.)
| | - Yan’e Liu
- Department of Cancer Chemotherapy and Radiation, Peking University Third Hospital, Beijing 100191, China;
| | - Dan Zhu
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China; (Z.T.); (D.Z.)
| | - Baoshan Cao
- Department of Cancer Chemotherapy and Radiation, Peking University Third Hospital, Beijing 100191, China;
| | - Ming Cui
- Department of Cardiology, Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China; (Z.T.); (D.Z.)
| |
Collapse
|
10
|
Su Y, Xu MY, Cui Y, Chen RZ, Xie LX, Zhang JX, Chen YQ, Ding T. Bacterial quorum sensing orchestrates longitudinal interactions to shape microbiota assembly. MICROBIOME 2023; 11:241. [PMID: 37926838 PMCID: PMC10626739 DOI: 10.1186/s40168-023-01699-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 10/16/2023] [Indexed: 11/07/2023]
Abstract
BACKGROUND The mechanism of microbiota assembly is one of the main problems in microbiome research, which is also the primary theoretical basis for precise manipulation of microbial communities. Bacterial quorum sensing (QS), as the most common means for bacteria to exchange information and interactions, is characterized by universality, specificity, and regulatory power, which therefore may influence the assembly processes of human microbiota. However, the regulating role of QS in microbiota assembly is rarely reported. In this study, we developed an optimized in vitro oral biofilm microbiota assembling (OBMA) model to simulate the time-series assembly of oral biofilm microbiota (OBM), by which to excavate the QS network and its regulating power in the process. RESULTS By using the optimized OBMA model, we were able to restore the assembly process of OBM and generate time-series OBM metagenomes of each day. We discovered a total of 2291 QS protein homologues related to 21 QS pathways. Most of these pathways were newly reported and sequentially enriched during OBM assembling. These QS pathways formed a comprehensive longitudinal QS network that included successively enriched QS hubs, such as Streptococcus, Veillonella-Megasphaera group, and Prevotella-Fusobacteria group, for information delivery. Bidirectional cross-talk among the QS hubs was found to play critical role in the directional turnover of microbiota structure, which in turn, influenced the assembly process. Subsequent QS-interfering experiments accurately predicted and experimentally verified the directional shaping power of the longitudinal QS network in the assembly process. As a result, the QS-interfered OBM exhibited delayed and fragile maturity with prolonged membership of Streptococcus and impeded membership of Prevotella and Fusobacterium. CONCLUSION Our results revealed an unprecedented longitudinal QS network during OBM assembly and experimentally verified its power in predicting and manipulating the assembling process. Our work provides a new perspective to uncover underlying mechanism in natural complex microbiota assembling and a theoretical basis for ultimately precisely manipulating human microbiota through intervention in the QS network. Video Abstract.
Collapse
Affiliation(s)
- Ying Su
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China
| | - Ming-Ying Xu
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Department of Immunology and Pathogenic Biology, Zhaoqing Medical College, Zhaoqing, 526020, China
| | - Ying Cui
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China
| | - Run-Zhi Chen
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China
| | - Li-Xiang Xie
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China
| | - Jing-Xiang Zhang
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China
| | - Yong-Qiu Chen
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China
| | - Tao Ding
- Department of Immunology and Microbiology, Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, China.
- Key Laboratory of Tropical Diseases Control (Sun Yat-Sen University), Ministry of Education, Guangzhou, 510080, China.
| |
Collapse
|
11
|
Begić G, Badovinac IJ, Karleuša L, Kralik K, Cvijanovic Peloza O, Kuiš D, Gobin I. Streptococcus salivarius as an Important Factor in Dental Biofilm Homeostasis: Influence on Streptococcus mutans and Aggregatibacter actinomycetemcomitans in Mixed Biofilm. Int J Mol Sci 2023; 24:ijms24087249. [PMID: 37108414 PMCID: PMC10139097 DOI: 10.3390/ijms24087249] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
A disturbed balance within the dental biofilm can result in the dominance of cariogenic and periodontopathogenic species and disease development. Due to the failure of pharmacological treatment of biofilm infection, a preventive approach to promoting healthy oral microbiota is necessary. This study analyzed the influence of Streptococcus salivarius K12 on the development of a multispecies biofilm composed of Streptococcus mutans, S. oralis and Aggregatibacter actinomycetemcomitans. Four different materials were used: hydroxyapatite, dentin and two dense polytetrafluoroethylene (d-PTFE) membranes. Total bacteria, individual species and their proportions in the mixed biofilm were quantified. A qualitative analysis of the mixed biofilm was performed using scanning electron microscopy (SEM) and confocal laser scanning microscopy (CLSM). The results showed that in the presence of S. salivarius K 12 in the initial stage of biofilm development, the proportion of S. mutans was reduced, which resulted in the inhibition of microcolony development and the complex three-dimensional structure of the biofilm. In the mature biofilm, a significantly lower proportion of the periodontopathogenic species A. actinomycetemcomitans was found in the salivarius biofilm. Our results show that S. salivarius K 12 can inhibit the growth of pathogens in the dental biofilm and help maintain the physiological balance in the oral microbiome.
Collapse
Affiliation(s)
- Gabrijela Begić
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Ivana Jelovica Badovinac
- Faculty of Physics and Centre for Micro- and Nanosciences and Technologies, University of Rijeka, 51000 Rijeka, Croatia
| | - Ljerka Karleuša
- Department of Physiology and Immunology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| | - Kristina Kralik
- Department of Medical Statistics and Medical Informatics, Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
| | | | - Davor Kuiš
- Department of Periodontology, Faculty of Dental Medicine, University of Rijeka, 51000 Rijeka, Croatia
- Department of Dental Medicine, Faculty of Dental Medicine and Health, Josip Juraj Strossmayer University of Osijek, 31000 Osijek, Croatia
- Clinical Hospital Centre, 51000 Rijeka, Croatia
| | - Ivana Gobin
- Department of Microbiology and Parasitology, Faculty of Medicine, University of Rijeka, 51000 Rijeka, Croatia
| |
Collapse
|
12
|
Pauter-Iwicka K, Railean V, Złoch M, Pomastowski P, Szultka-Młyńska M, Błońska D, Kupczyk W, Buszewski B. Characterization of the salivary microbiome before and after antibiotic therapy via separation technique. Appl Microbiol Biotechnol 2023; 107:2515-2531. [PMID: 36843196 PMCID: PMC10033590 DOI: 10.1007/s00253-023-12371-0] [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: 09/26/2022] [Revised: 01/06/2023] [Accepted: 01/09/2023] [Indexed: 02/28/2023]
Abstract
In the present research, the MALDI-TOF MS technique was applied as a tool to rapidly identify the salivary microbiome. In this fact, it has been monitored the changes occurred in molecular profiles under different antibiotic therapy. Significant changes in the composition of the salivary microbiota were noticed not only in relation to the non antibiotic (non-AT) and antibiotic treatment (AT) groups, but also to the used media, the antibiotic therapy and co-existed microbiota. Each antibiotic generates specific changes in molecular profiles. The highest number of bacterial species was isolated in the universal culture medium (72%) followed by the selective medium (48% and 38%). In the case of non-AT patients, the prevalence of Streptococcus salivarius (25%), Streptococcus vestibularis (19%), Streptococcus oralis (13%), and Staphylococcus aureus (6%) was identified while in the case of AT, Streptococcus salivarius (11%), Streptococcus parasanguinis (11%), Staphylococcus epidermidis (12%), Enterococcus faecalis (9%), Staphylococcus hominis (8%), and Candida albicans (6%) were identified. Notable to specified that the Candida albicans was noticed only in AT samples, indicating a negative impact on the antibiotic therapy. The accuracy of the MALDI-TOF MS technique was performed by the 16S rRNA gene sequencing analysis-as a reference method. Conclusively, such an approach highlighted in the present study can help in developing the methods enabling a faster diagnosis of disease changes at the cellular level before clinical changes occur. Once the MALDI tool allows for the distinguishing of the microbiota of non-AT and AT, it may enable to monitor the diseases treatment and develop a treatment regimen for individual patients in relation to each antibiotic. KEY POINTS: The salivary microbiota of antibiotic-treated patients was more bacteria variety MALDI-TOF MS is a promising tool for recording of reproducible molecular profiles Our data can allow to monitor the treatment of bacterial diseases for patients.
Collapse
Affiliation(s)
- Katarzyna Pauter-Iwicka
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University, Gagarina 7, 87-100, Torun, Poland
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Wilenska 4, 87-100, Torun, Poland
| | - Viorica Railean
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Wilenska 4, 87-100, Torun, Poland
- Department of Infectious, Invasive Diseases and Veterinary Administration, Institute of Veterinary Medicine, Nicolaus Copernicus University in Torun, Gagarina 7, 87-100, Toruń, Poland
| | - Michał Złoch
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University, Gagarina 7, 87-100, Torun, Poland
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Wilenska 4, 87-100, Torun, Poland
| | - Paweł Pomastowski
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Wilenska 4, 87-100, Torun, Poland
| | - Małgorzata Szultka-Młyńska
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University, Gagarina 7, 87-100, Torun, Poland
| | - Dominika Błońska
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University, Gagarina 7, 87-100, Torun, Poland
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Wilenska 4, 87-100, Torun, Poland
| | - Wojciech Kupczyk
- Department of General, Gastroenterological&Oncological Surgery Collegium Medicum, Nicolaus Copernicus University, Torun, Poland
| | - Bogusław Buszewski
- Department of Environmental Chemistry and Bioanalytics, Faculty of Chemistry, Nicolaus Copernicus University, Gagarina 7, 87-100, Torun, Poland.
- Centre for Modern Interdisciplinary Technologies, Nicolaus Copernicus University, Wilenska 4, 87-100, Torun, Poland.
| |
Collapse
|
13
|
Tagg JR, Harold LK, Jain R, Hale JDF. Beneficial modulation of human health in the oral cavity and beyond using bacteriocin-like inhibitory substance-producing streptococcal probiotics. Front Microbiol 2023; 14:1161155. [PMID: 37056747 PMCID: PMC10086258 DOI: 10.3389/fmicb.2023.1161155] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Accepted: 03/03/2023] [Indexed: 03/30/2023] Open
Abstract
The human oral cavity contains a diversity of microbial habitats that have been adopted and adapted to as homeland by an amazingly heterogeneous population of microorganisms collectively referred to as the oral microbiota. These microbes generally co-habit in harmonious homeostasis. However, under conditions of imposed stress, as with changes to the host’s physiology or nutritional status, or as a response to foreign microbial or antimicrobial incursions, some components of the oral “microbiome” (viz. the in situ microbiota) may enter a dysbiotic state. This microbiome dysbiosis can manifest in a variety of guises including streptococcal sore throats, dental caries, oral thrush, halitosis and periodontal disease. Most of the strategies currently available for the management or treatment of microbial diseases of the oral cavity focus on the repetitive “broad sweep” and short-term culling of oral microbe populations, hopefully including the perceived principal pathogens. Both physical and chemical techniques are used. However, the application of more focused approaches to the harnessing or elimination of key oral cavity pathogens is now feasible through the use of probiotic strains that are naturally adapted for oral cavity colonization and also are equipped to produce anti-competitor molecules such as the bacteriocins and bacteriocin-like inhibitory substances (viz BLIS). Some of these probiotics are capable of suppressing the proliferation of a variety of recognized microbial pathogens of the human mouth, thereby assisting with the restoration of oral microbiome homeostasis. BLIS K12 and BLIS M18, the progenitors of the BLIS-producing oral probiotics, are members of the human oral cavity commensal species Streptococcus salivarius. More recently however, a number of other streptococcal and some non-streptococcal candidate oral probiotics have also been promoted. What is becoming increasingly apparent is that the future for oral probiotic applications will probably extend well beyond the attempted limitation of the direct pathological consequences of oral microbiome dysbiosis to also encompass a plethora of systemic diseases and disorders of the human host. The background to and the evolving prospects for the beneficial modulation of the oral microbiome via the application of BLIS-producing S. salivarius probiotics comprises the principal focus of the present review.
Collapse
|
14
|
Athanasopoulou K, Adamopoulos PG, Scorilas A. Unveiling the Human Gastrointestinal Tract Microbiome: The Past, Present, and Future of Metagenomics. Biomedicines 2023; 11:biomedicines11030827. [PMID: 36979806 PMCID: PMC10045138 DOI: 10.3390/biomedicines11030827] [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: 01/31/2023] [Revised: 02/26/2023] [Accepted: 03/08/2023] [Indexed: 03/30/2023] Open
Abstract
Over 1014 symbiotic microorganisms are present in a healthy human body and are responsible for the synthesis of vital vitamins and amino acids, mediating cellular pathways and supporting immunity. However, the deregulation of microbial dynamics can provoke diverse human diseases such as diabetes, human cancers, cardiovascular diseases, and neurological disorders. The human gastrointestinal tract constitutes a hospitable environment in which a plethora of microbes, including diverse species of archaea, bacteria, fungi, and microeukaryotes as well as viruses, inhabit. In particular, the gut microbiome is the largest microbiome community in the human body and has drawn for decades the attention of scientists for its significance in medical microbiology. Revolutions in sequencing techniques, including 16S rRNA and ITS amplicon sequencing and whole genome sequencing, facilitate the detection of microbiomes and have opened new vistas in the study of human microbiota. Especially, the flourishing fields of metagenomics and metatranscriptomics aim to detect all genomes and transcriptomes that are retrieved from environmental and human samples. The present review highlights the complexity of the gastrointestinal tract microbiome and deciphers its implication not only in cellular homeostasis but also in human diseases. Finally, a thorough description of the widely used microbiome detection methods is discussed.
Collapse
Affiliation(s)
- Konstantina Athanasopoulou
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece
| | - Panagiotis G Adamopoulos
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece
| | - Andreas Scorilas
- Department of Biochemistry and Molecular Biology, Faculty of Biology, National and Kapodistrian University of Athens, 15701 Athens, Greece
| |
Collapse
|
15
|
Chen Y, Chen X, Chen P, Chen X, Pan L, Han L, Zhu T. Alteration of the Gut Microbiota in Missed Abortion. Indian J Microbiol 2023; 63:106-119. [PMID: 37179577 PMCID: PMC10172435 DOI: 10.1007/s12088-023-01063-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 02/09/2023] [Indexed: 03/03/2023] Open
Abstract
There is a symbiotic relationship between gut microbiota and human beings. Imbalance of the gut microbiota will cause pathological damages to humans. Although many risk factors are associated with missed abortion (MA), the pathological mechanism of it is still unclear. Here, we analyzed gut flora of the patients with MA by S16 high-throughput sequencing. The possible pathogenic mechanisms of the MA were explored. Fecal samples from 14 healthy controls and 16 MA patients were collected to do 16S rRNA gene high-throughput sequencing analysis. The abundance of the Bacteroidetes, Proteobacteria, Actinobacteria, Escherichia, Streptococcus_ Salivarius, and Lactobacillus was significantly reduced in the MA group, while, the abundance of the Klebsiella was significantly increased in the MA patients. The Ruminococcaceae and [Eubacterium]_coprostanoligenes_group were found only in the specimens of the MA patients. The Fabrotax function prediction analysis showed that four photosynthesis function bacteria (cyanobateria, oxygenic_photoautotrophy, photoautotrophy, and phototrophy) only existed in the MA group. In the analysis of the BugBase microbiome function prediction, the Escherichia of the MA group is significantly reduced compared to that of the healthy controls in the items of that Contains_Mobile_Elements, Facultatively_Anaerobic, Forms_Biofilms, Potentially_Pathogenic.png, Gram_Nagative, and Stress_Tolerant_relabundance. These alterations may affect the stability of the host's immune, neural, metabolic and other systems by interfering with the balance of the gut microbiota or by the metabolites of those bacteria, causing the MA. This study explored the possible pathogenic factors of the gut microbiota of the MA. The results provide evidence to figure out the pathogenesis of the MA.
Collapse
Affiliation(s)
- Yi Chen
- Gynaecology Department, The First Hospital of Putian, Putian, 351100 Fujian China
| | - Xianqian Chen
- Gynaecology Department, The First Hospital of Putian, Putian, 351100 Fujian China
| | - Pingyu Chen
- Gynaecology Department, The First Hospital of Putian, Putian, 351100 Fujian China
| | - Xiuxia Chen
- Gynaecology Department, The First Hospital of Putian, Putian, 351100 Fujian China
| | - Lin Pan
- Gynaecology Department, The First Hospital of Putian, Putian, 351100 Fujian China
| | - Lihong Han
- Key Laboratory of Translational Tumor Medicine in Fujian Province, School of Basic Medical Science, Putian University, 450 Dongzhen Road West, Putian, 351100 Fujian China
| | - Tang Zhu
- Key Laboratory of Translational Tumor Medicine in Fujian Province, School of Basic Medical Science, Putian University, 450 Dongzhen Road West, Putian, 351100 Fujian China
- Yujia Biotech., D-201, 3 Juquan Road, Guangzhou, 510700 Guangdong China
| |
Collapse
|
16
|
Aggarwal N, Kitano S, Puah GRY, Kittelmann S, Hwang IY, Chang MW. Microbiome and Human Health: Current Understanding, Engineering, and Enabling Technologies. Chem Rev 2023; 123:31-72. [PMID: 36317983 PMCID: PMC9837825 DOI: 10.1021/acs.chemrev.2c00431] [Citation(s) in RCA: 99] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Indexed: 01/12/2023]
Abstract
The human microbiome is composed of a collection of dynamic microbial communities that inhabit various anatomical locations in the body. Accordingly, the coevolution of the microbiome with the host has resulted in these communities playing a profound role in promoting human health. Consequently, perturbations in the human microbiome can cause or exacerbate several diseases. In this Review, we present our current understanding of the relationship between human health and disease development, focusing on the microbiomes found across the digestive, respiratory, urinary, and reproductive systems as well as the skin. We further discuss various strategies by which the composition and function of the human microbiome can be modulated to exert a therapeutic effect on the host. Finally, we examine technologies such as multiomics approaches and cellular reprogramming of microbes that can enable significant advancements in microbiome research and engineering.
Collapse
Affiliation(s)
- Nikhil Aggarwal
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Shohei Kitano
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
| | - Ginette Ru Ying Puah
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Wilmar-NUS
(WIL@NUS) Corporate Laboratory, National
University of Singapore, Singapore 117599, Singapore
- Wilmar
International Limited, Singapore 138568, Singapore
| | - Sandra Kittelmann
- Wilmar-NUS
(WIL@NUS) Corporate Laboratory, National
University of Singapore, Singapore 117599, Singapore
- Wilmar
International Limited, Singapore 138568, Singapore
| | - In Young Hwang
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Department
of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
- Singapore
Institute of Technology, Singapore 138683, Singapore
| | - Matthew Wook Chang
- NUS
Synthetic Biology for Clinical and Technological Innovation (SynCTI), National University of Singapore, Singapore 117456, Singapore
- Synthetic
Biology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117456, Singapore
- Wilmar-NUS
(WIL@NUS) Corporate Laboratory, National
University of Singapore, Singapore 117599, Singapore
- Department
of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117596, Singapore
| |
Collapse
|
17
|
Biofilm ecology associated with dental caries: Understanding of microbial interactions in oral communities leads to development of therapeutic strategies targeting cariogenic biofilms. ADVANCES IN APPLIED MICROBIOLOGY 2023; 122:27-75. [PMID: 37085193 DOI: 10.1016/bs.aambs.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/12/2023]
Abstract
A biofilm is a sessile community characterized by cells attached to the surface and organized into a complex structural arrangement. Dental caries is a biofilm-dependent oral disease caused by infection with cariogenic pathogens, such as Streptococcus mutans, and associated with frequent exposure to a sugar-rich diet and poor oral hygiene. The virulence of cariogenic biofilms is often associated with the spatial organization of S. mutans enmeshed with exopolysaccharides on tooth surfaces. However, in the oral cavity, S. mutans does not act alone, and several other microbes contribute to cariogenic biofilm formation. Microbial communities in cariogenic biofilms are spatially organized into complex structural arrangements of various microbes and extracellular matrices. The balance of microbiota diversity with reduced diversity and a high proportion of acidogenic-aciduric microbiota within the biofilm is closely related to the disease state. Understanding the characteristics of polymicrobial biofilms and the association of microbial interactions within the biofilm (e.g., symbiosis, cooperation, and competition) in terms of their potential role in the pathogenesis of oral disease would help develop new strategies for interventions in virulent biofilm formation.
Collapse
|
18
|
Oral microbiota in cancer: could the bad guy turn good with application of polyphenols? Expert Rev Mol Med 2022; 25:e1. [PMID: 36511134 DOI: 10.1017/erm.2022.39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The human oral cavity is comprised of dynamic and polynomial microbes which uniquely reside in the microenvironments of oral cavities. The cumulative functions of the symbiotic microbial communities maintain normal homeostasis; however, a shifted microbiota yields a dysbiosis state, which produces local and systemic diseases including dental caries, periodontitis, cancer, obesity and diabetes. Recent research reports claim that an association occurs between oral dysbiosis and the progression of different types of cancers including oral, gastric and pancreatic ones. Different mechanisms are proposed for the development of cancer, such as induction of inflammatory reactions, production of carcinogenic materials and alteration of the immune system. Medications are available to treat these associated diseases; however, the current strategies may further worsen the disease by unwanted side effects. Natural-derived polyphenol molecules significantly inhibit a wide range of systemic diseases with fewer side effects. In this review, we have displayed the functions of the oral microbes and we have extended the report regarding the role of polyphenols in oral microbiota to maintain healthy conditions and prevention of diseases with emphasis on the treatment of oral microbiota-associated cancer.
Collapse
|
19
|
A Brief Review of Local Bacteriotherapy for Preventing Respiratory Infections. ALLERGIES 2022. [DOI: 10.3390/allergies2040013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Recurrent respiratory infections (RRIs) account for relevant economic and social implications and significantly affect family life. Local Bacteriotherapy (LB) represents an innovative option in preventing RRIs. Local bacteriotherapy consists of administering “good” and safe bacteria (probiotics) by nasal or oral route. In particular, two strains (Streptococcus salivarius 24SMB and Streptococcus oralis 89a) are commonly used. The present article presents and discusses the literature concerning LB. Infections of airways include the upper and lower respiratory tract. A series of clinical trials investigated the preventive role of LB in preventing upper and lower RIs. These studies demonstrated that LB safely reduced the prevalence and severity of RIs, the use of antibiotics, and absences from school. Therefore, Local Bacteriotherapy may be considered an interesting therapeutic option in RRI prevention.
Collapse
|
20
|
Integrative Analysis of the Nasal Microbiota and Serum Metabolites in Bovines with Respiratory Disease by 16S rRNA Sequencing and Gas Chromatography/Mass Selective Detector-Based Metabolomics. Int J Mol Sci 2022; 23:ijms231912028. [PMID: 36233330 PMCID: PMC9569885 DOI: 10.3390/ijms231912028] [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: 09/12/2022] [Revised: 10/01/2022] [Accepted: 10/07/2022] [Indexed: 11/17/2022] Open
Abstract
Bovine respiratory disease (BRD) continues to pose a serious threat to the cattle industry, resulting in substantial economic losses. As a multifactorial disease, pathogen infection and respiratory microbial imbalance are important causative factors in the occurrence and development of BRD. Integrative analyses of 16S rRNA sequencing and metabolomics allow comprehensive identification of the changes in microbiota and metabolism associated with BRD, making it possible to determine which pathogens are responsible for the disease and to develop new therapeutic strategies. In our study, 16S rRNA sequencing and metagenomic analysis were used to describe and compare the composition and diversity of nasal microbes in healthy cattle and cattle with BRD from different farms in Yinchuan, Ningxia, China. We found a significant difference in nasal microbial diversity between diseased and healthy bovines; notably, the relative abundance of Mycoplasma bovis and Pasteurella increased. This indicated that the composition of the microbial community had changed in diseased bovines compared with healthy ones. The data also strongly suggested that the reduced relative abundance of probiotics, including Pasteurellales and Lactobacillales, in diseased samples contributes to the susceptibility to bovine respiratory disease. Furthermore, serum metabolomic analysis showed altered concentrations of metabolites in BRD and that a significant decrease in lactic acid and sarcosine may impair the ability of bovines to generate energy and an immune response to pathogenic bacteria. Based on the correlation analysis between microbial diversity and the metabolome, lactic acid (2TMS) was positively correlated with Gammaproteobacteria and Bacilli and negatively correlated with Mollicutes. In summary, microbial communities and serum metabolites in BRD were characterized by integrative analysis. This study provides a reference for monitoring biomarkers of BRD, which will be critical for the prevention and treatment of BRD in the future.
Collapse
|
21
|
Inchingolo AD, Malcangi G, Semjonova A, Inchingolo AM, Patano A, Coloccia G, Ceci S, Marinelli G, Di Pede C, Ciocia AM, Mancini A, Palmieri G, Barile G, Settanni V, De Leonardis N, Rapone B, Piras F, Viapiano F, Cardarelli F, Nucci L, Bordea IR, Scarano A, Lorusso F, Palermo A, Costa S, Tartaglia GM, Corriero A, Brienza N, Di Venere D, Inchingolo F, Dipalma G. Oralbiotica/Oralbiotics: The Impact of Oral Microbiota on Dental Health and Demineralization: A Systematic Review of the Literature. CHILDREN (BASEL, SWITZERLAND) 2022; 9:1014. [PMID: 35883998 PMCID: PMC9323959 DOI: 10.3390/children9071014] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 12/17/2022]
Abstract
The oral microbiota plays a vital role in the human microbiome and oral health. Imbalances between microbes and their hosts can lead to oral and systemic disorders such as diabetes or cardiovascular disease. The purpose of this review is to investigate the literature evidence of oral microbiota dysbiosis on oral health and discuss current knowledge and emerging mechanisms governing oral polymicrobial synergy and dysbiosis; both have enhanced our understanding of pathogenic mechanisms and aided the design of innovative therapeutic approaches as ORALBIOTICA for oral diseases such as demineralization. PubMed, Web of Science, Google Scholar, Scopus, Cochrane Library, EMBEDDED, Dentistry & Oral Sciences Source via EBSCO, APA PsycINFO, APA PsyArticles, and DRUGS@FDA were searched for publications that matched our topic from January 2017 to 22 April 2022, with an English language constraint using the following Boolean keywords: ("microbio*" and "demineralization*") AND ("oral microbiota" and "demineralization"). Twenty-two studies were included for qualitative analysis. As seen by the studies included in this review, the balance of the microbiota is unstable and influenced by oral hygiene, the presence of orthodontic devices in the oral cavity and poor eating habits that can modify its composition and behavior in both positive and negative ways, increasing the development of demineralization, caries processes, and periodontal disease. Under conditions of dysbiosis, favored by an acidic environment, the reproduction of specific bacterial strains increases, favoring cariogenic ones such as Bifidobacterium dentium, Bifidobacterium longum, and S. mutans, than S. salivarius and A. viscosus, and increasing of Firmicutes strains to the disadvantage of Bacteroidetes. Microbial balance can be restored by using probiotics and prebiotics to manage and treat oral diseases, as evidenced by mouthwashes or dietary modifications that can influence microbiota balance and prevent or slow disease progression.
Collapse
Affiliation(s)
- Alessio Danilo Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Giuseppina Malcangi
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Alexandra Semjonova
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Angelo Michele Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Assunta Patano
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Giovanni Coloccia
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Sabino Ceci
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Grazia Marinelli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Chiara Di Pede
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Anna Maria Ciocia
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Antonio Mancini
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Giulia Palmieri
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Giuseppe Barile
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Vito Settanni
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Nicole De Leonardis
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Biagio Rapone
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Fabio Piras
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Fabio Viapiano
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Filippo Cardarelli
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Ludovica Nucci
- Multidisciplinary Department of Medical-Surgical and Dental Specialties, University of Campania “Luigi Vanvitelli”, Via L. De Crecchio 6, 80138 Naples, Italy;
| | - Ioana Roxana Bordea
- Department of Oral Rehabilitation, Faculty of Dentistry, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania
| | - Antonio Scarano
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy; (A.S.); (F.L.)
| | - Felice Lorusso
- Department of Innovative Technologies in Medicine and Dentistry, University of Chieti-Pescara, 66100 Chieti, Italy; (A.S.); (F.L.)
| | - Andrea Palermo
- Implant Dentistry College of Medicine and Dentistry Birmingham, University of Birmingham, Birmingham B46BN, UK;
| | - Stefania Costa
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, Section of Orthodontics, School of Dentistry, University of Messina, 98125 Messina, Italy;
| | - Gianluca Martino Tartaglia
- UOC Maxillo-Facial Surgery and Dentistry, Department of Biomedical, Surgical and Dental Sciences, School of Dentistry, Fondazione IRCCS Ca’ Granda, Ospedale Maggiore Policlinico, University of Milan, 20100 Milan, Italy;
- Department of Orthodontics, Faculty of Medicine, University of Milan, 20100 Milan, Italy
| | - Alberto Corriero
- Unit of Anesthesia and Resuscitation, Department of Emergencies and Organ Transplantations, Aldo Moro University, 70124 Bari, Italy; (A.C.); (N.B.)
| | - Nicola Brienza
- Unit of Anesthesia and Resuscitation, Department of Emergencies and Organ Transplantations, Aldo Moro University, 70124 Bari, Italy; (A.C.); (N.B.)
| | - Daniela Di Venere
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Francesco Inchingolo
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| | - Gianna Dipalma
- Department of Interdisciplinary Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.D.I.); (G.M.); (A.S.); (A.M.I.); (A.P.); (G.C.); (S.C.); (G.M.); (C.D.P.); (A.M.C.); (A.M.); (G.P.); (G.B.); (V.S.); (N.D.L.); (B.R.); (F.P.); (F.V.); (F.C.); (D.D.V.); (G.D.)
| |
Collapse
|
22
|
Strategies to Combat Caries by Maintaining the Integrity of Biofilm and Homeostasis during the Rapid Phase of Supragingival Plaque Formation. Antibiotics (Basel) 2022; 11:antibiotics11070880. [PMID: 35884135 PMCID: PMC9312143 DOI: 10.3390/antibiotics11070880] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 06/24/2022] [Accepted: 06/28/2022] [Indexed: 02/06/2023] Open
Abstract
Bacteria in the oral cavity, including commensals and opportunistic pathogens, are organized into highly specialized sessile communities, coexisting in homeostasis with the host under healthy conditions. A dysbiotic environment during biofilm evolution, however, allows opportunistic pathogens to become the dominant species at caries-affected sites at the expense of health-associated taxa. Combining tooth brushing with dentifrices or rinses combat the onset of caries by partially removes plaque, but resulting in the biofilm remaining in an immature state with undesirables’ consequences on homeostasis and oral ecosystem. This leads to the need for therapeutic pathways that focus on preserving balance in the oral microbiota and applying strategies to combat caries by maintaining biofilm integrity and homeostasis during the rapid phase of supragingival plaque formation. Adhesion, nutrition, and communication are fundamental in this phase in which the bacteria that have survived these adverse conditions rebuild and reorganize the biofilm, and are considered targets for designing preventive strategies to guide the biofilm towards a composition compatible with health. The present review summarizes the most important advances and future prospects for therapies based on the maintenance of biofilm integrity and homeostasis as a preventive measure of dysbiosis focused on these three key factors during the rapid phase of plaque formation.
Collapse
|
23
|
Goya-Jorge E, Gonza I, Bondue P, Douny C, Taminiau B, Daube G, Scippo ML, Delcenserie V. Human Adult Microbiota in a Static Colon Model: AhR Transcriptional Activity at the Crossroads of Host–Microbe Interaction. Foods 2022; 11:foods11131946. [PMID: 35804761 PMCID: PMC9265634 DOI: 10.3390/foods11131946] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 06/21/2022] [Accepted: 06/25/2022] [Indexed: 02/01/2023] Open
Abstract
Functional symbiotic intestinal microbiota regulates immune defense and the metabolic processing of xenobiotics in the host. The aryl hydrocarbon receptor (AhR) is one of the transcription factors mediating host–microbe interaction. An in vitro static simulation of the human colon was used in this work to analyze the evolution of bacterial populations, the microbial metabolic output, and the potential induction of AhR transcriptional activity in healthy gut ecosystems. Fifteen target taxa were explored by qPCR, and the metabolic content was chromatographically profiled using SPME-GC-MS and UPLC-FLD to quantify short-chain fatty acids (SCFA) and biogenic amines, respectively. Over 72 h of fermentation, the microbiota and most produced metabolites remained stable. Fermentation supernatant induced AhR transcription in two of the three reporter gene cell lines (T47D, HepG2, HT29) evaluated. Mammary and intestinal cells were more sensitive to microbiota metabolic production, which showed greater AhR agonism than the 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) used as a positive control. Some of the SCFA and biogenic amines identified could crucially contribute to the potent AhR induction of the fermentation products. As a fundamental pathway mediating human intestinal homeostasis and as a sensor for several microbial metabolites, AhR activation might be a useful endpoint to include in studies of the gut microbiota.
Collapse
Affiliation(s)
- Elizabeth Goya-Jorge
- Laboratory of Food Quality Management, Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, Av. de Cureghem 10 (B43b), 4000 Liege, Belgium; (E.G.-J.); (I.G.)
| | - Irma Gonza
- Laboratory of Food Quality Management, Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, Av. de Cureghem 10 (B43b), 4000 Liege, Belgium; (E.G.-J.); (I.G.)
| | - Pauline Bondue
- Research & Development, ORTIS S.A., Hinter der Heck 46, 4750 Elsenborn, Belgium;
| | - Caroline Douny
- Laboratory of Food Analysis, Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, Av. de Cureghem 10 (B43b), 4000 Liege, Belgium; (C.D.); (M.-L.S.)
| | - Bernard Taminiau
- Laboratory of Microbiology, Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, Av. de Cureghem 180 (B42), 4000 Liege, Belgium; (B.T.); (G.D.)
| | - Georges Daube
- Laboratory of Microbiology, Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, Av. de Cureghem 180 (B42), 4000 Liege, Belgium; (B.T.); (G.D.)
| | - Marie-Louise Scippo
- Laboratory of Food Analysis, Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, Av. de Cureghem 10 (B43b), 4000 Liege, Belgium; (C.D.); (M.-L.S.)
| | - Véronique Delcenserie
- Laboratory of Food Quality Management, Department of Food Sciences, Faculty of Veterinary Medicine, University of Liege, Av. de Cureghem 10 (B43b), 4000 Liege, Belgium; (E.G.-J.); (I.G.)
- Correspondence: ; Tel.: +32-4-366-51-24
| |
Collapse
|
24
|
Guo H, Xiang X, Lin X, Wang Q, Qin S, Lu X, Xu J, Fang Y, Liu Y, Cui J, Li Z. Oropharyngeal Probiotic ENT-K12 as an Effective Dietary Intervention for Children With Recurrent Respiratory Tract Infections During Cold Season. Front Nutr 2022; 9:900448. [PMID: 35634421 PMCID: PMC9132010 DOI: 10.3389/fnut.2022.900448] [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: 03/20/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Recurrent respiratory tract infections (RRTi) cause a high burden of disease and lead to negative impact on quality of life, frequent school/work absenteeism, and doctor visits, which remain a great challenge to pediatricians because RRTi can increase the risk of various complications including antibiotic overuse and resistance, which is one of the biggest threats to global health, and there is no confirmed effective treatment. In this study, we aimed to assess the clinical efficacy and safety of oropharyngeal probiotic ENT-K12 as a dietary intervention or a complementary treatment along with standard medical treatment during acute respiratory infections among children with RRTi during cold season. The results of this study show that when comparing to practicing of standard medical treatment only, the complementary intake of oropharyngeal probiotic ENT-K12 can effectively reduce episodes of both acute and RRTi in school children, shorten the course of respiratory symptoms onset, reduce the use of antibiotics and antiviral drugs, and reduce the absence days from both children's school and parents' work. Using oropharyngeal probiotics as a complementary dietary intervention to stabilize oropharyngeal microflora, specifically inhibiting respiratory pathogens and enhancing host immunity, could possibly be a promising approach to reduce RRTi burden and combating antibiotic resistance in long term, more clinical studies will be needed to further confirm the clinical practicing guide to ensure its clinical benefit.
Collapse
Affiliation(s)
- Hongyan Guo
- Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Xiaochen Xiang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Xuan Lin
- Department of Endocrinology, CR & WISCO General Hospital Affiliated to Wuhan University of Science and Technology, Wuhan, China
| | - Qiang Wang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Si Qin
- College of Food Science and Technology, Hunan Agricultural University, Changsha, China
| | - Xinyan Lu
- Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Jiawei Xu
- Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Ying Fang
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Yang Liu
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Jing Cui
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| | - Zhi Li
- Institute of Infection, Immunology and Tumor Microenvironment, Hubei Province Key Laboratory of Occupational Hazard Identification and Control, Medical College, Wuhan University of Science and Technology, Wuhan, China
| |
Collapse
|
25
|
Guo S, Zhang H, Chu Y, Jiang Q, Ma Y. A neural network-based framework to understand the type 2 diabetes-related alteration of the human gut microbiome. IMETA 2022; 1:e20. [PMID: 38868565 PMCID: PMC10989819 DOI: 10.1002/imt2.20] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 03/13/2022] [Accepted: 03/15/2022] [Indexed: 06/14/2024]
Abstract
The identification of microbial markers adequate to delineate the disease-related microbiome alterations from the complex human gut microbiota is of great interest. Here, we develop a framework combining neural network (NN) and random forest, resulting in 40 marker species and 90 marker genes identified from the metagenomic data set (185 healthy and 183 type 2 diabetes [T2D] samples), respectively. In terms of these markers, the NN model obtained higher accuracy in classifying the T2D-related samples than other methods; the interaction network analyses identified the key species and functional modules; the regression analysis determined that fasting blood glucose is the most significant factor (p < 0.05) in the T2D-related alteration of the human gut microbiome. We also observed that those marker species varied little across the case and control samples greatly shift in the different stages of the T2D development, suggestive of their important roles in the T2D-related microbiome alteration. Our study provides a new way of identifying the disease-related biomarkers and analyzing the role they may play in the development of the disease.
Collapse
Affiliation(s)
- Shun Guo
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Shenzhen Key Laboratory of Synthetic Genomics; Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Shenzhen Key Lab for High Performance Data Mining, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| | - Haoran Zhang
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Shenzhen Key Laboratory of Synthetic Genomics; Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| | - Yunmeng Chu
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Shenzhen Key Laboratory of Synthetic Genomics; Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| | - Qingshan Jiang
- Shenzhen Key Lab for High Performance Data Mining, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| | - Yingfei Ma
- Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Key Laboratory of Quantitative Engineering Biology, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
- Shenzhen Key Laboratory of Synthetic Genomics; Guangdong Provincial Key Laboratory of Synthetic Genomics, Shenzhen Institutes of Advanced TechnologyChinese Academy of SciencesShenzhenGuangdongChina
| |
Collapse
|
26
|
Diagne AM, Pelletier A, Durmort C, Faure A, Kanonenberg K, Freton C, Page A, Delolme F, Vorac J, Vallet S, Bellard L, Vivès C, Fieschi F, Vernet T, Rousselle P, Guiral S, Grangeasse C, Jault JM, Orelle C. Identification of a two-component regulatory system involved in antimicrobial peptide resistance in Streptococcus pneumoniae. PLoS Pathog 2022; 18:e1010458. [PMID: 35395062 PMCID: PMC9020739 DOI: 10.1371/journal.ppat.1010458] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 04/20/2022] [Accepted: 03/18/2022] [Indexed: 11/24/2022] Open
Abstract
Two-component regulatory systems (TCS) are among the most widespread mechanisms that bacteria use to sense and respond to environmental changes. In the human pathogen Streptococcus pneumoniae, a total of 13 TCS have been identified and many of them have been linked to pathogenicity. Notably, TCS01 strongly contributes to pneumococcal virulence in several infection models. However, it remains one of the least studied TCS in pneumococci and its functional role is still unclear. In this study, we demonstrate that TCS01 cooperates with a BceAB-type ABC transporter to sense and induce resistance to structurally-unrelated antimicrobial peptides of bacterial origin that all target undecaprenyl-pyrophosphate or lipid II, which are essential precursors of cell wall biosynthesis. Even though tcs01 and bceAB genes do not locate in the same gene cluster, disruption of either of them equally sensitized the bacterium to the same set of antimicrobial peptides. We show that the key function of TCS01 is to upregulate the expression of the transporter, while the latter appears the main actor in resistance. Electrophoretic mobility shift assays further demonstrated that the response regulator of TCS01 binds to the promoter region of the bceAB genes, implying a direct control of these genes. The BceAB transporter was overexpressed and purified from E. coli. After reconstitution in liposomes, it displayed substantial ATPase and GTPase activities that were stimulated by antimicrobial peptides to which it confers resistance to, revealing new functional features of a BceAB-type transporter. Altogether, this inducible defense mechanism likely contributes to the survival of the opportunistic microorganism in the human host, in which competition among commensal microorganisms is a key determinant for effective host colonization and invasive path.
Collapse
Affiliation(s)
- Aissatou Maty Diagne
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Anaïs Pelletier
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Claire Durmort
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Agathe Faure
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Kerstin Kanonenberg
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Céline Freton
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Adeline Page
- Protein Science Facility, SFR BioSciences, CNRS, UMS3444, INSERM US8, University of Lyon, Lyon, France
| | - Frédéric Delolme
- Protein Science Facility, SFR BioSciences, CNRS, UMS3444, INSERM US8, University of Lyon, Lyon, France
| | - Jaroslav Vorac
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Sylvain Vallet
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Laure Bellard
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Corinne Vivès
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Franck Fieschi
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Thierry Vernet
- Institute of Structural Biology (IBS), UMR 5075 CNRS/University of Grenoble-Alpes, Grenoble, France
| | - Patricia Rousselle
- Laboratoire de Biologie Tissulaire et Ingénierie Thérapeutique (LBTI), UMR 5305 CNRS/University of Lyon, Lyon, France
| | - Sébastien Guiral
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Christophe Grangeasse
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Jean-Michel Jault
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| | - Cédric Orelle
- Molecular Microbiology and Structural Biochemistry (MMSB), UMR 5086 CNRS/University of Lyon, Lyon, France
| |
Collapse
|
27
|
Li S, Li N, Wang C, Zhao Y, Cao J, Li X, Zhang Z, Li Y, Yang X, Wang X, Che C, Zhao Y, Wang L, Zhao L, Shen J. Gut Microbiota and Immune Modulatory Properties of Human Breast Milk Streptococcus salivarius and S. parasanguinis Strains. Front Nutr 2022; 9:798403. [PMID: 35273986 PMCID: PMC8901577 DOI: 10.3389/fnut.2022.798403] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 01/24/2022] [Indexed: 01/14/2023] Open
Abstract
Human breast milk Streptococcus spp. are transferred to infant guts via breast feeding, but their effects on the gut microbiota and immunity remain unclear. In this study, we characterized gut microbiota and immune modulatory properties of human breast milk S. salivarius F286 and S. parasanguinis F278 that had been shown to be able to colonize gut. The two Streptococcus strains were orally administered to mouse pups individually at 1 × 107 cells/day from postnatal Days 1 to 21. At postnatal week 3 (the weaning period), S. salivarius F286 reduced the colonic microbiota α-diversity, increased 21 amplicon sequence variants (ASVs), including bacteria from Akkermansia, Intestinimonas, and Lachnospiraceae, and decreased 52 ASVs, including bacteria from Eubacterium, Bifidobacterium, Escherichia-Shigella, and Turicibacter; however, S. parasanguinis F278 didn't change the colonic microbiota. Both Streptococcus strains reduced the ileal mRNA expression of cytokine/transcription factor representatives of T helper (Th) cells, including IFN-γ (Th1), Gata3 (Th2), and TGF-β (Treg) in 2-week-old suckling mice, and promoted the ileal expression of Foxp3 and TGF-β, which are representatives of anti-inflammatory Treg cells, in 3-week-old weaning mice. The two Streptococcus strains exhibited anti-inflammatory potential when incubated in vitro with human peripheral blood mononuclear cells and TNF-α-treated gut epithelial HT29 cells. In C. elegans, both strains activated immune response genes, which was associated with their lifespan-prolonging effects. Our results suggest that S. salivarius F286 and S. parasanguinis F278 may exert regulatory (anti-inflammatory) roles in gut immunity and S. salivarius F286 can modulate gut microbiota, and highlight the probiotic potential of milk S. salivarius and S. parasanguinis strains.
Collapse
Affiliation(s)
- Shuo Li
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Na Li
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chenwei Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yi Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jie Cao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Xuejing Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Ziyi Zhang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Yue Li
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xin Yang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Xiaoxin Wang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Chuanyan Che
- Department of Animal Sciences, Anhui Science and Technology University, Chuzhou, China
| | - Yufeng Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| | - Linghua Wang
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Liping Zhao
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China.,State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, Shanghai, China
| | - Jian Shen
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
28
|
Yang N, Xu J, Wang X, Chen N, Su L, Liu Y. The Spatial Landscape of the Bacterial Community and Bile Acids in the Digestive Tract of Patients With Bile Reflux. Front Microbiol 2022; 13:835310. [PMID: 35356519 PMCID: PMC8959417 DOI: 10.3389/fmicb.2022.835310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Accepted: 02/17/2022] [Indexed: 11/13/2022] Open
Abstract
Background Bile reflux can lead to inflammation and increased intestinal metaplasia. Since bile acids can influence the gastrointestinal environment, it is possible that bile reflux may alter the gastric microbiota and potentially the oral or gut microbiota. Bile acids have a very complex interrelationship with microbiota. We aimed to explore the characteristics of the digestive tract microbiota and bile acids profile in bile reflux patients. Methods This study included 20 chronic gastritis patients with bile reflux and 20 chronic gastritis patients without bile reflux. Saliva, gastric fluid, and fecal samples were collected for bile acid testing. Buccal mucosal swabs, gastric mucosal tissues, and feces were collected for bacteria detection. The UPLC-MS/MS examined bile acids profiles. 16S rRNA gene sequencing was used to analyze the bacterial profile. Results Bilirubin in the blood increased in bile reflux patients. No other clinical factors were identified to be significantly associated with bile reflux. 12-DHCA, 6,7-diketo LCA, and βHDCA decreased while TUDCA increased in saliva of bile reflux patients. Streptococcus, Capnocytophaga, Neisseria, and Actinobacillus decreased in oral mucosa of bile reflux patients while Helicobacter, Prevotella, and Veillonella increased. Gastric bile acid levels were generally higher in bile reflux patients. Gastric mucosal microbiota was highly stable. The changes in fecal bile acids were insignificant. Bifidobacterium, Prevotella_2, Ruminococcus, Weissella, Neisseria, and Akkermansia decreased in fecal samples from bile reflux patients; while Alloprevotella, Prevotella_9, Parabacteroides, and Megamonas increased. Conclusion Our results demonstrate that bile reflux significantly alters the oral, gastric, and intestinal bile acids profiles but only influences the oral and gut microbiota composition. These findings indicate that bile reflux can modulate the gastrointestinal microbiota in a site-specific manner.
Collapse
Affiliation(s)
- Ni Yang
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing, China
| | - Jun Xu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing, China
| | - Xuemei Wang
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing, China
| | - Ning Chen
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing, China
| | - Lin Su
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing, China
| | - Yulan Liu
- Department of Gastroenterology, Peking University People’s Hospital, Beijing, China
- Clinical Center of Immune-Mediated Digestive Diseases, Peking University People’s Hospital, Beijing, China
- *Correspondence: Yulan Liu,
| |
Collapse
|
29
|
Droz L, Jannel R, Rupprecht CDD. Living through multispecies societies: Approaching the microbiome with Imanishi Kinji. ENDEAVOUR 2022; 46:100814. [PMID: 35697549 DOI: 10.1016/j.endeavour.2022.100814] [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: 05/06/2021] [Revised: 09/03/2021] [Accepted: 05/04/2022] [Indexed: 06/15/2023]
Abstract
Recent research about the microbiome points to a picture in which we, humans, are 'living through' nature, and nature itself is living in us. Our bodies are hosting-and depend on-the multiple species that constitute human microbiota. This article will discuss current research on the microbiome through the ideas of Japanese ecologist Imanishi Kinji (1902-1992). First, some of Imanishi's key ideas regarding the world of living beings and multispecies societies are presented. Second, seven types of relationships concerning the human microbiome, human beings, and the environment are explored. Third, inspired by Imanishi's work, this paper develops the idea of dynamic, porous, and complex multispecies societies in which different living beings or species are codependent on others, including microbiota and human beings.
Collapse
Affiliation(s)
- Laÿna Droz
- Basque Center for Climate Change, Spain.
| | - Romaric Jannel
- Kyoto University, Institute for Research in Humanities, Japan.
| | - Christoph D D Rupprecht
- Ehime Daigaku, Faculty of Collaborative Regional Innovation, Department of Environmental Design, Matsuyama, Ehime, Japan.
| |
Collapse
|
30
|
Zhang WX, Xiao CL, Li SY, Bai XC, Qi H, Tian H, Wang N, Yang B, Li XM, Sun Y. Streptococcus strain C17 T as a potential probiotic candidate to modulate oral health. Lett Appl Microbiol 2022; 74:901-908. [PMID: 35218013 DOI: 10.1111/lam.13680] [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: 02/03/2022] [Revised: 02/19/2022] [Accepted: 02/21/2022] [Indexed: 11/28/2022]
Abstract
In the microbiome, probiotics modulate oral diseases. In this study, Streptococcus strain C17T was isolated from the oropharynx of a five-year-old healthy child, and its potential probiotic properties were analysed using human bronchial epithelial cells (16-HBE) used as an in vitro oropharyngeal mucosal model. The results demonstrated that the C17T strain showed tolerance to moderate pH ranges of 4-5 and 0.5-1% bile. However, it was more tolerant to 0.5% bile than 1% bile. It also demonstrated an ability to accommodate maladaptive oropharyngeal conditions (i.e., tolerating lysozyme at 200 μg mL-1) . It was also resistant to hydrogen peroxide at 0.8 mM . In addition, we found out that the strain possesses inhibitory activities against various common pathogenic bacteria. Furthermore, C17T was not cytotoxic to 16-HBE cells at different multiplicities of infection. Scanning electron microscopy disclosed that C17T adhesion to 16-HBE cells. Competition, exclusion, and displacement assays showed that it had good anti-adhesive effect against S. aureus. The present study revealed that Streptococcus strain C17T is a potentially efficacious oropharyngeal probiotic.
Collapse
Affiliation(s)
- Wen Xiao Zhang
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Chun Ling Xiao
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Shu Yin Li
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Xiao Cui Bai
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - He Qi
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Han Tian
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Nan Wang
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Biao Yang
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Xin Ming Li
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| | - Ye Sun
- Key Lab of Environmental Pollution and Microecology of Liaoning Province, Shenyang Medical College, No. 146, Huang he North Street, Shenyang, Liao Ning, People's Republic of China
| |
Collapse
|
31
|
Characterization and Viability Prediction of Commercial Probiotic Supplements under Temperature and Concentration Conditioning Factors by NIR Spectroscopy. FERMENTATION-BASEL 2022. [DOI: 10.3390/fermentation8020066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
The quality of probiotics has been associated with bacteria and yeast strains’ contents and their stability against conditioning factors. Near-infrared spectroscopy (NIRS), as a non-destructive, fast, real-time, and cost-effective analytical technique, can provide some advantages over more traditional food quality control methods in quality evaluation. The aim of our study was to evaluate the applicability of NIRS to the characterization and viability prediction of three commercial probiotic food supplement powders containing lactic acid bacteria (LAB) subjected to concentration and temperature conditioning factors. For each probiotic, 3 different concentrations were considered, and besides normal preparation (25 °C, control), samples were subjected to heat treatment at 60 or 90 °C and left to cool down until reaching room temperature prior to further analysis. Overall, after applying chemometrics to the NIR spectra, the obtained principal component analysis-based linear discriminant analysis (PCA-LDA) classification models showed a high accuracy in both recognition and prediction. The temperature has an important impact on the discrimination of samples. According to the concentration, the best models were identified for the 90 °C temperature treatment, reaching 100% average correct classification for recognition and over 90% for prediction. However, the prediction accuracy decreased substantially at lower temperatures. For the 25°C temperature treatment, the prediction accuracy decreased to nearly 60% for 2 of the 3 probiotics. Moreover, according to the temperature level, both the recognition and prediction accuracies were close to 100%. Additionally, the partial least square regression (PLSR) model achieved respectable values for the prediction of the colony-forming units (log CFU/g) of the probiotic samples, with a determination coefficient for prediction (R2Pr) of 0.82 and root mean square error for prediction (RMSEP) of 0.64. The results of our study show that NIRS is a fast, reliable, and promising alternative to the conventional microbiology technique for the characterization and prediction of the viability of probiotic supplement drink preparations.
Collapse
|
32
|
Hale JDF, Jain R, Wescombe PA, Burton JP, Simon RR, Tagg JR. Safety assessment of Streptococcus salivarius M18 a probiotic for oral health. Benef Microbes 2022; 13:47-60. [PMID: 35098909 DOI: 10.3920/bm2021.0107] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The development of probiotics targeting non-intestinal body sites continues to generate interest amongst researchers, biotech companies and consumers alike. A key consideration for any bacterial strain to be developed into a probiotic is a robust assessment of its safety profile. Streptococcus salivarius strain M18 was originally isolated from a healthy adult and evaluated for its probiotic capabilities targeted to dental and oral health applications. This publication presents the safety characterisation of strain M18. Application of a diverse range of techniques showed that strain M18 can be specifically distinguished from other S. salivarius using a variety of molecular and phenotypic methodologies and that it lacks any relevant antibiotic resistance or virulence determinants. Direct comparison of the strain M18 safety profile with that of the prototype S. salivarius probiotic, S. salivarius strain K12, supports the proposition that strain M18 is indeed safe for probiotic application in humans.
Collapse
Affiliation(s)
- J D F Hale
- Blis Technologies Ltd, 81 Glasgow Street, South Dunedin, Dunedin 9012, New Zealand
| | - R Jain
- Blis Technologies Ltd, 81 Glasgow Street, South Dunedin, Dunedin 9012, New Zealand
| | - P A Wescombe
- Yili Innovation Center Oceania, Lincoln University, Christchurch, New Zealand
| | - J P Burton
- Lawson Health Research Institute, 268 Grosvenor St, London, ON, Canada
| | - R R Simon
- Intertek Health Sciences Inc, Mississauga, ON, Canada
| | - J R Tagg
- Blis Technologies Ltd, 81 Glasgow Street, South Dunedin, Dunedin 9012, New Zealand
| |
Collapse
|
33
|
Dong J, Li W, Wang Q, Chen J, Zu Y, Zhou X, Guo Q. Relationships Between Oral Microecosystem and Respiratory Diseases. Front Mol Biosci 2022; 8:718222. [PMID: 35071321 PMCID: PMC8767498 DOI: 10.3389/fmolb.2021.718222] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 12/09/2021] [Indexed: 02/05/2023] Open
Abstract
Oral microecosystem is a very complicated ecosystem that is located in the mouth and comprises oral microbiome, diverse anatomic structures of oral cavity, saliva and interactions between oral microbiota and between oral microbiota and the host. More and more evidence from studies of epidemiology, microbiology and molecular biology is establishing a significant link between oral microecosystem and respiratory diseases. Microbiota settling down in oral microecosystem is known as the main source of lung microbiome and has been associated with the occurrence and development of respiratory diseases like pneumonia, chronic obstructive pulmonary disease, lung cancer, cystic fibrosis lung disease and asthma. In fact, it is not only indigenous oral microbes promote or directly cause respiratory infection and inflammation when inhaled into the lower respiratory tract, but also internal environment of oral microecosystem serves as a reservoir for opportunistic respiratory pathogens. Moreover, poor oral health and oral diseases caused by oral microecological dysbiosis (especially periodontal disease) are related with risk of multiple respiratory diseases. Here, we review the research status on the respiratory diseases related with oral microecosystem. Potential mechanisms on how respiratory pathogens colonize oral microecosystem and the role of indigenous oral microbes in pathogenesis of respiratory diseases are also summarized and analyzed. Given the importance of oral plaque control and oral health interventions in controlling or preventing respiratory infection and diseases, we also summarize the oral health management measures and attentions, not only for populations susceptible to respiratory infection like the elderly and hospitalized patients, but also for dentist or oral hygienists who undertake oral health care. In conclusion, the relationship between respiratory diseases and oral microecosystem has been established and supported by growing body of literature. However, etiological evidence on the role of oral microecosystem in the development of respiratory diseases is still insufficient. Further detailed studies focusing on specific mechanisms on how oral microecosystem participate in the pathogenesis of respiratory diseases could be helpful to prevent and treat respiratory diseases.
Collapse
Affiliation(s)
- Jiajia Dong
- Department of Pulmonary and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Wei Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qi Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Jiahao Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yue Zu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xuedong Zhou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qiang Guo
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
34
|
Passali D, Passali FM, Damiani V. Recurrent Respiratory Infections in Childhood: The Importance of Local Microbiota Modulation. PEDIATRIC ENT INFECTIONS 2022:55-62. [DOI: 10.1007/978-3-030-80691-0_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
35
|
Qi C, Zhou J, Tu H, Tu R, Chang H, Chen J, Li D, Sun J, Yu R. Lactation-dependent vertical transmission of natural probiotics from the mother to the infant gut through breast milk. Food Funct 2021; 13:304-315. [PMID: 34889924 DOI: 10.1039/d1fo03131g] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The transmission of certain bacteria from the mother's gut to the infant's gut via breast milk (BM) is critical for the offspring's immune system development. Dysbiosis of the BM microbiota can be caused by a variety of reasons, which can be influenced by probiotics delivered via the enteromammary route. The goal of this study was to investigate the bacteria that can be transmitted from the mother to the infant's intestine during various lactation periods in 19 mother-child dyads. Bacterial transmission is most common during the colostrum phase when bacteria with certain amplicon sequence variants (ASVs) enter the newborn intestine and inhabit it permanently. We have established that anaerobic gut-associated bacteria, such as Faecalibacterium, Blautia and Lachnoclostridium, transfer from the mother to the infant's gut with lactation dependence using the idea of weighted transfer ratios. Streptococcus salivarius, Bifidobacterium longum, and Lactobacillus gasseri are transferred from the maternal gut to the BM, as well as from the BM to the newborn gut, depending on different ASVs. These findings suggest that isolation of key microorganisms from breast milk could be utilized to modify the microbiota of BM or newborns by giving the mother a probiotic or adding it to artificial milk to promote neonatal health.
Collapse
Affiliation(s)
- Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Jingbo Zhou
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Huayu Tu
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Rundan Tu
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Hong Chang
- Department of Pediatric Cardiology, Nephrology and Rheumatism, The Affiliated Hospital of Qingdao University Medical College, Qingdao 266003, PR China
| | - Jie Chen
- Department of Obstetrics, Affiliated Hospital of Qingdao University, Qingdao 266000, PR China
| | - Duo Li
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao 266071, PR China.
| | - Renqiang Yu
- Department of Neonatology, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, Wuxi 214002, PR China. E-mail:.
| |
Collapse
|
36
|
Biogeography of Bacterial Communities and Specialized Metabolism in Human Aerodigestive Tract Microbiomes. Microbiol Spectr 2021; 9:e0166921. [PMID: 34704787 PMCID: PMC8549736 DOI: 10.1128/spectrum.01669-21] [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: 11/30/2022] Open
Abstract
The aerodigestive tract (ADT) is the primary portal through which pathogens and other invading microbes enter the body. As the direct interface with the environment, we hypothesize that the ADT microbiota possess biosynthetic gene clusters (BGCs) for antibiotics and other specialized metabolites to compete with both endogenous and exogenous microbes. From 1,214 bacterial genomes, representing 136 genera and 387 species that colonize the ADT, we identified 3,895 BGCs. To determine the distribution of BGCs and bacteria in different ADT sites, we aligned 1,424 metagenomes, from nine different ADT sites, onto the predicted BGCs. We show that alpha diversity varies across the ADT and that each site is associated with distinct bacterial communities and BGCs. We identify specific BGC families enriched in the buccal mucosa, external naris, gingiva, and tongue dorsum despite these sites harboring closely related bacteria. We reveal BGC enrichment patterns indicative of the ecology at each site. For instance, aryl polyene and resorcinol BGCs are enriched in the gingiva and tongue, which are colonized by many anaerobes. In addition, we find that streptococci colonizing the tongue and cheek possess different ribosomally synthesized and posttranslationally modified peptide BGCs. Finally, we highlight bacterial genera with BGCs but are underexplored for specialized metabolism and demonstrate the bioactivity of Actinomyces against other bacteria, including human pathogens. Together, our results demonstrate that specialized metabolism in the ADT is extensive and that by exploring these microbiomes further, we will better understand the ecology and biogeography of this system and identify new bioactive natural products. IMPORTANCE Bacteria produce specialized metabolites to compete with other microbes. Though the biological activities of many specialized metabolites have been determined, our understanding of their ecology is limited, particularly within the human microbiome. As the aerodigestive tract (ADT) faces the external environment, bacteria colonizing this tract must compete both among themselves and with invading microbes, including human pathogens. We analyzed the genomes of ADT bacteria to identify biosynthetic gene clusters (BGCs) for specialized metabolites. We found that the majority of ADT BGCs are uncharacterized and the metabolites they encode are unknown. We mapped the distribution of BGCs across the ADT and determined that each site is associated with its own distinct bacterial community and BGCs. By further characterizing these BGCs, we will inform our understanding of ecology and biogeography across the ADT, and we may uncover new specialized metabolites, including antibiotics.
Collapse
|
37
|
Karimova M, Moyes D, Ide M, Setterfield JF. The human microbiome in immunobullous disorders and lichen planus. Clin Exp Dermatol 2021; 47:522-528. [PMID: 34669983 DOI: 10.1111/ced.14987] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 10/05/2021] [Accepted: 10/11/2021] [Indexed: 12/18/2022]
Abstract
For several decades, there has been a significant growth in the incidence of autoimmune diseases. Studies indicate that genetic factors may not be the only trigger for disease development and that dysbiosis of the microbiome may be another mechanism involved in the pathogenesis of autoimmune diseases. The role of the microbiome in the development of common skin disorders such as psoriasis, atopic dermatitis, acne and rosacea is increasingly well understood. However, few studies have focused on lichen planus and the rare acquired immunobullous diseases, both mucocutaneous groups of disorders linked to skin, oral and gut microbiomes. This review provides an insight into the current understanding of how the microbiome may contribute to the development of autoimmunity and to the maintenance and exacerbation of acquired immunobullous and lichenoid diseases. These mechanisms may have implications for future preventive and therapeutic approaches.
Collapse
Affiliation(s)
- M Karimova
- Centre for Host Microbiome Interaction (CHMI), Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - D Moyes
- Centre for Host Microbiome Interaction (CHMI), Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - M Ide
- Centre for Host Microbiome Interaction (CHMI), Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK.,Department of Periodontology, Centre for Host-Microbiome Interactions (CHMI), Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, Guy's Hospital, London, UK
| | - J F Setterfield
- Centre for Host Microbiome Interaction (CHMI), Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK.,St John's Institute of Dermatology, Guy's and St Thomas' NHS Foundation Trust, London, UK.,Department of Oral Medicine, Guy's and St Thomas' NHS Foundation Trust, London, UK
| |
Collapse
|
38
|
Chen JW, Wu JH, Chiang WF, Chen YL, Wu WS, Wu LW. Taxonomic and Functional Dysregulation in Salivary Microbiomes During Oral Carcinogenesis. Front Cell Infect Microbiol 2021; 11:663068. [PMID: 34604102 PMCID: PMC8482814 DOI: 10.3389/fcimb.2021.663068] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Accepted: 08/23/2021] [Indexed: 01/04/2023] Open
Abstract
Exploring microbial community compositions in humans with healthy versus diseased states is crucial to understand the microbe-host interplay associated with the disease progression. Although the relationship between oral cancer and microbiome was previously established, it remained controversial, and yet the ecological characteristics and their responses to oral carcinogenesis have not been well studied. Here, using the bacterial 16S rRNA gene amplicon sequencing along with the in silico function analysis by PICRUSt2 (Phylogenetic Investigation of Communities by Reconstruction of Unobserved States 2), we systematically characterized the compositions and the ecological drivers of saliva microbiome in the cohorts of orally healthy, non-recurrent oral verrucous hyperplasia (a pre-cancer lesion), and oral verrucous hyperplasia–associated oral cancer at taxonomic and function levels, and compared them with the re-analysis of publicly available datasets. Diversity analyses showed that microbiome dysbiosis in saliva was significantly linked to oral health status. As oral health deteriorated, the number of core species declined, and metabolic pathways predicted by PICRUSt2 were dysregulated. Partitioned beta-diversity revealed an extremely high species turnover but low function turnover. Functional beta-diversity in saliva microbiome shifted from turnover to nestedness during oral carcinogenesis, which was not observed at taxonomic levels. Correspondingly, the quantitative analysis of stochasticity ratios showed that drivers of microbial composition and functional gene content of saliva microbiomes were primarily governed by the stochastic processes, yet the driver of functional gene content shifted toward deterministic processes as oral cancer developed. Re-analysis of publicly accessible datasets supported not only the distinctive family taxa of Veillonellaceae and Actinomycetaceae present in normal cohorts but also that Flavobacteriaceae and Peptostreptococcaceae as well as the dysregulated metabolic pathways of nucleotides, amino acids, fatty acids, and cell structure were related to oral cancer. Using predicted functional profiles to elucidate the correlations to the oral health status shows superior performance than using taxonomic data among different studies. These findings advance our understanding of the oral ecosystem in relation to oral carcinogenesis and provide a new direction to the development of microbiome-based tools to study the interplay of the oral microbiome, metabolites, and host health.
Collapse
Affiliation(s)
- Jiung-Wen Chen
- Department of Environmental Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Jer-Horng Wu
- Department of Environmental Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Fan Chiang
- Department of Oral & Maxillofacial Surgery, Chi-Mei Medical Center, Liouying, Taiwan.,School of Dentistry, National Yang-Ming University, Taipei, Taiwan
| | - Yuh-Ling Chen
- Institute of Oral Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Wei-Sheng Wu
- Department of Electrical Engineering, National Cheng Kung University, Tainan, Taiwan
| | - Li-Wha Wu
- Institute of Molecular Medicine, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| |
Collapse
|
39
|
Probiotics maintain the gut microbiome homeostasis during Indian Antarctic expedition by ship. Sci Rep 2021; 11:18793. [PMID: 34552104 PMCID: PMC8458292 DOI: 10.1038/s41598-021-97890-4] [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: 06/02/2021] [Accepted: 08/31/2021] [Indexed: 02/08/2023] Open
Abstract
Ship voyage to Antarctica is a stressful journey for expedition members. The response of human gut microbiota to ship voyage and a feasible approach to maintain gut health, is still unexplored. The present findings describe a 24-day long longitudinal study involving 19 members from 38th Indian Antarctic Expedition, to investigate the impact of ship voyage and effect of probiotic intervention on gut microbiota. Fecal samples collected on day 0 as baseline and at the end of ship voyage (day 24), were analyzed using whole genome shotgun sequencing. Probiotic intervention reduced the sea sickness by 10% compared to 44% in placebo group. The gut microbiome in placebo group members on day 0 and day 24, indicated significant alteration compared to a marginal change in the microbial composition in probiotic group. Functional analysis revealed significant alterations in carbohydrate and amino acid metabolism. Carbohydrate-active enzymes analysis represented functional genes involved in glycoside hydrolases, glycosyltransferases and carbohydrate binding modules, for maintaining gut microbiome homeostasis. Suggesting thereby the possible mechanism of probiotic in stabilizing and restoring gut microflora during stressful ship journey. The present study is first of its kind, providing a feasible approach for protecting gut health during Antarctic expedition involving ship voyage.
Collapse
|
40
|
Athayde LA, de Aguiar SLF, Miranda MCG, Brito RVJ, de Faria AMC, Nobre SAM, Andrade MC. Lactococcus lactis Administration Modulates IgE and IL-4 Production and Promotes Enterobacteria Growth in the Gut from Ethanol-Intake Mice. Protein Pept Lett 2021; 28:1164-1179. [PMID: 34315363 DOI: 10.2174/0929866528666210727102019] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND It is well known that alcohol can trigger inflammatory effects in the gastrointestinal tract (GIT) interfering with mucosal homeostasis. OBJECTIVE This study evaluated the effectiveness of Lactococcus lactis treatment in controlling the increase in molecular biomarkers related to allergic inflammation, and the effect on the diversity and abundance of the Enterobacteriaceae family in the GIT after high-dose acute administration of ethanol. METHODS Mice received ethanol or saline solution by gavage for four consecutive days, and 24 h after the last administration the animals were given L. lactis or M17 broth orally ad libitum for two consecutive days. The animals were subsequently sacrificed and dissected. RESULTS L. lactis treatment was able to restore basal levels of secretory immunoglobulin A in the gastric mucosa, serum total immunoglobulin E, interleukin (IL)-4 production in gastric and intestinal tissues, and IL-10 levels in gastric tissue. L. lactis treatment encouraged the diversification of the Enterobacteriaceae population, particularly the commensal species, in the GIT. CONCLUSION This research opens a field of studies regarding the modulatory effect of L. lactis on immunological and microbial changes induced after alcohol intake.
Collapse
|
41
|
Barbour A, Elebyary O, Fine N, Oveisi M, Glogauer M. Metabolites of the Oral Microbiome: Important Mediators of Multi-Kingdom Interactions. FEMS Microbiol Rev 2021; 46:6316110. [PMID: 34227664 DOI: 10.1093/femsre/fuab039] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 07/02/2021] [Indexed: 12/12/2022] Open
Abstract
The oral cavity hosts over 700 different microbial species that produce a rich reservoir of bioactive metabolites critical to oral health maintenance. Over the last two decades, new insights into the oral microbiome and its importance in health and disease have emerged mainly due to the discovery of new oral microbial species using next-generation sequencing (NGS). This advancement has revolutionized the documentation of unique microbial profiles associated with different niches and health/disease states within the oral cavity and the relation of the oral bacteria to systemic diseases. However, less work has been done to identify and characterize the unique oral microbial metabolites that play critical roles in maintaining equilibrium between the various oral microbial species and their human hosts. This article discusses the most significant microbial metabolites produced by these diverse communities of oral bacteria that can either foster health or contribute to disease. Finally, we shed light on how advances in genomics and genome mining can provide a high throughput platform for discovering novel bioactive metabolites derived from the human oral microbiome to tackle emerging human infections and systemic diseases.
Collapse
Affiliation(s)
- Abdelahhad Barbour
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada, M5G 1G6, Canada
| | - Omnia Elebyary
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada, M5G 1G6, Canada
| | - Noah Fine
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada, M5G 1G6, Canada
| | - Morvarid Oveisi
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada, M5G 1G6, Canada
| | - Michael Glogauer
- Faculty of Dentistry, University of Toronto, Toronto, ON, Canada, M5G 1G6, Canada.,Department of Dental Oncology, Maxillofacial and Ocular Prosthetics, Princess Margaret Cancer Centre, Toronto, ON, Canada, M5G 2M9, Canada
| |
Collapse
|
42
|
Shanmugaraj B, Bulaon CJI, Malla A, Phoolcharoen W. Biotechnological Insights on the Expression and Production of Antimicrobial Peptides in Plants. Molecules 2021; 26:4032. [PMID: 34279372 PMCID: PMC8272150 DOI: 10.3390/molecules26134032] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 12/31/2022] Open
Abstract
The emergence of drug-resistant pathogens poses a serious critical threat to global public health and requires immediate action. Antimicrobial peptides (AMPs) are a class of short peptides ubiquitously found in all living forms, including plants, insects, mammals, microorganisms and play a significant role in host innate immune system. These peptides are considered as promising candidates to treat microbial infections due to its distinct advantages over conventional antibiotics. Given their potent broad spectrum of antimicrobial action, several AMPs are currently being evaluated in preclinical/clinical trials. However, large quantities of highly purified AMPs are vital for basic research and clinical settings which is still a major bottleneck hindering its application. This can be overcome by genetic engineering approaches to produce sufficient amount of diverse peptides in heterologous host systems. Recently plants are considered as potential alternatives to conventional protein production systems such as microbial and mammalian platforms due to their unique advantages such as rapidity, scalability and safety. In addition, AMPs can also be utilized for development of novel approaches for plant protection thereby increasing the crop yield. Hence, in order to provide a spotlight for the expression of AMP in plants for both clinical or agricultural use, the present review presents the importance of AMPs and efforts aimed at producing recombinant AMPs in plants for molecular farming and plant protection so far.
Collapse
Affiliation(s)
| | - Christine Joy I Bulaon
- Research Unit for Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | | | - Waranyoo Phoolcharoen
- Research Unit for Plant-Produced Pharmaceuticals, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
43
|
Renye JA, Steinberg DH. Thermophilin 110 inhibits growth and biofilm formation of Streptococcus mutans. ACTA ACUST UNITED AC 2021; 31:e00647. [PMID: 34307072 PMCID: PMC8258636 DOI: 10.1016/j.btre.2021.e00647] [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: 06/11/2020] [Revised: 06/10/2021] [Accepted: 06/12/2021] [Indexed: 11/06/2022]
Abstract
S. thermophilus B59671 naturally produces thermophilin 110, a bacteriocin that inhibits the growth of the oral pathogen Streptococcus mutans Thermophilin 110 was shown to prevent biofilm formation by S. mutans UA159 Co-culturing S. thermophilus B59671 with S. mutans UA159 prevented biofilm formation.
Dental caries continues to occur in both children and adults worldwide resulting in significant economic burden, and consumers have expressed interest in natural products that can prevent these recurrent infections. In this study, S. thermophilus B59671, which produces thermophilin 110, was shown to inhibit the growth of S. mutans UA159. A thermophilin concentration ≥ 80 AU ml−1 prevented the growth of S. mutans UA159 in batch culture, while ≥ 160 AU ml−1 was required to prevent biofilm growth. Co-culturing S. thermophilus B59671 and S. mutans UA159 also resulted in impaired biofilm growth. Thermophillin 110 was also shown inhibit additional S. mutans strains and commensal oral streptococci at higher concentrations (640-1280 AU ml−1). These results suggest that thermophilin 110 could be used as a natural antimicrobial in oral care products and support the need for additional studies to assess the probiotic potential of S. thermophilus B59671.
Collapse
Affiliation(s)
- John A Renye
- Dairy and Functional Foods Research Unit, Agricultural Research Service, USDA, 600 E. Mermaid Lane, Wyndmoor, PA 19038
| | - Dennis H Steinberg
- Dairy and Functional Foods Research Unit, Agricultural Research Service, USDA, 600 E. Mermaid Lane, Wyndmoor, PA 19038
| |
Collapse
|
44
|
MacDonald KW, Chanyi RM, Macklaim JM, Cadieux PA, Reid G, Burton JP. Streptococcus salivarius inhibits immune activation by periodontal disease pathogens. BMC Oral Health 2021; 21:245. [PMID: 33962608 PMCID: PMC8103598 DOI: 10.1186/s12903-021-01606-z] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Accepted: 04/29/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Periodontal disease represents a major health concern. The administration of beneficial microbes has been increasing in popularity over efforts to manipulate the microbes using antimicrobial agents. This study determined the ability of Streptococcus salivarius to inhibit IL-6 and IL-8 production by gingival fibroblasts when activated by periodontal pathogens and their effect on the salivary microbiome. METHODS Primary human gingival fibroblasts were challenged with Porphyromonas gingivalis, Aggregatibacter actinomycetemcomitans and Fusobacterium nucleatum and a combination of all three. IL-6 and IL-8 cytokine release were measured. Using this same model, S. salivarius K12, M18 and different supernatant and whole-cell lysate fractions of S. salivarius K12 were administered to pathogen-induced fibroblasts. A patient study of healthy participants was also conducted to determine the effect S. salivarius K12 had on the native microbiome using 16S next generation sequence analysis. RESULTS All pathogens tested induced a significant IL-6 and IL-8 response. S. salivarius K12 or M18, did not exhibit an increase in inflammatory cytokines. When either of the probiotic strains were co-administered with a pathogen, there were significant reductions in both IL-6 and IL-8 release. This effect was also observed when gingival fibroblasts were pre-treated with either S. salivarius K12 or M18 and then stimulated with the oral pathogens. Chewing gum containing S. salivarius K12 did not alter the salivary microbiome and did not increase inflammatory markers in the oral cavity. CONCLUSION S. salivarius K12 and M18 prevented immune activation induced by periodontal disease pathogens. S. salivarius K12 did not alter the salivary microbiome or induce immune activation when administered as a chewing gum. These results warrant further study to determine if it may be an effective treatment in a model of periodontal disease.
Collapse
Affiliation(s)
- Kyle W MacDonald
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Canadian Centre for Human Microbiome and Probiotic Research, Lawson Health Research Institute, London, ON, Canada
| | - Ryan M Chanyi
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Canadian Centre for Human Microbiome and Probiotic Research, Lawson Health Research Institute, London, ON, Canada
| | - Jean M Macklaim
- Canadian Centre for Human Microbiome and Probiotic Research, Lawson Health Research Institute, London, ON, Canada.,Department of Biochemistry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada
| | - Peter A Cadieux
- Canadian Centre for Human Microbiome and Probiotic Research, Lawson Health Research Institute, London, ON, Canada.,School of Health Sciences, Fanshawe College, London, ON, Canada
| | - Gregor Reid
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.,Canadian Centre for Human Microbiome and Probiotic Research, Lawson Health Research Institute, London, ON, Canada
| | - Jeremy P Burton
- Department of Microbiology and Immunology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada. .,Canadian Centre for Human Microbiome and Probiotic Research, Lawson Health Research Institute, London, ON, Canada. .,Department of Surgery, Division of Urology, Schulich School of Medicine and Dentistry, University of Western Ontario, London, ON, Canada.
| |
Collapse
|
45
|
Chang AM, Kantrong N, Darveau RP. Maintaining homeostatic control of periodontal epithelial tissue. Periodontol 2000 2021; 86:188-200. [PMID: 33690934 DOI: 10.1111/prd.12369] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Years of coevolution with resident microbes has made them an essential component of health. Yet, little is known about oral commensal bacteria's contribution to and role in the maintenance of oral health and homeostasis. Commensal bacteria are speculated to play a host protective role in the maintenance of health. In this review, we describe and provide examples of the coordinate regulation that occurs between oral commensal bacteria and the host innate immune response to modulate and maintain oral homeostasis.
Collapse
Affiliation(s)
- Ana M Chang
- Department of Periodontics, University of Washington, Seattle, Washington, USA
| | - Nutthapong Kantrong
- Department of Periodontics, University of Washington, Seattle, Washington, USA.,Oral Biology Research Unit, Faculty of Dentistry, Khon Kaen University, Khon Kaen, Thailand
| | - Richard P Darveau
- Department of Periodontics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
46
|
García-Curiel L, Del Rocío López-Cuellar M, Rodríguez-Hernández AI, Chavarría-Hernández N. Toward understanding the signals of bacteriocin production by Streptococcus spp. and their importance in current applications. World J Microbiol Biotechnol 2021; 37:15. [PMID: 33394178 DOI: 10.1007/s11274-020-02973-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 12/01/2020] [Indexed: 12/25/2022]
Abstract
Microorganisms have developed quorum sensing (QS) systems to detect small signaling molecules that help to control access to additional nutrients and space in highly competitive polymicrobial niches. Many bacterial processes are QS-regulated; two examples are the highly related traits of the natural genetic competence state and the production of antimicrobial peptides such as bacteriocins. The Streptococcus genus is widely studied for its competence and for its ability to produce bacteriocins, as these antimicrobial peptides have significant potential in the treatment of infections caused by multiple-resistant pathogens, a severe public health issue. The transduction of a two-component system controls competence in streptococci: (1) ComD/E, which controls the competence in the Mitis and Anginosus groups, and (2) ComR/S, which performs the same function in the Bovis, Mutans, Salivarius, and Pyogenic groups. The cell-to-cell communication required for bacteriocin production in the Streptococcus groups is controlled mainly by a paralog of the ComD/E system. The relationships between pheromone signals and induction pathways are related to the bacteriocin production systems. In this review, we discuss the recent advances in the understanding of signaling and the induction of bacteriocin biosynthesis by QS regulation in streptococci. This information could aid in the design of better methods for the development and production of these antimicrobial peptides. It could also contribute to the analysis and emerging applications of bacteriocins in terms of their safety, quality, and human health benefits.
Collapse
Affiliation(s)
- Laura García-Curiel
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México
| | - Ma Del Rocío López-Cuellar
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México.
| | - Adriana Inés Rodríguez-Hernández
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México
| | - Norberto Chavarría-Hernández
- Cuerpo Académico de Biotecnología Agroalimentaria, Instituto de Ciencias Agropecuarias-Universidad Autónoma del Estado de Hidalgo, Tulancingo de Bravo, México
| |
Collapse
|
47
|
Martorano-Fernandes L, Rodrigues NC, de Souza Borges MH, Cavalcanti YW, de Almeida LDFD. Interkingdom interaction between C. albicans and S. salivarius on titanium surfaces. BMC Oral Health 2020; 20:349. [PMID: 33261593 PMCID: PMC7706213 DOI: 10.1186/s12903-020-01334-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 11/17/2020] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND In oral candidiasis models, Candida albicans and Streptococcus salivarius sp. biofilms have an antagonistic relationship. Due to this, S. salivarius have been used experimentally as probiotic. However, the interaction between these microorganisms in the peri-implantitis-like microenvironment remains unknown. This study aimed to evaluate the interaction between C. albicans and S. salivarius biofilms developed on titanium surfaces, under reduced oxygen levels. METHODS Titanium specimens were pre-conditioned with artificial saliva (1 h, 37 °C). Single-species biofilms of C. albicans (ATCC 90028) and co-culture biofilms of C. albicans and S. salivarius (ATCC 7073) was developed for 24 and 72 h on titanium specimens. Subsequently, the effect of these intervals of biofilm formation and the interactions among the cells were evaluated. Biofilms from cultures were collected and analyzed for cell viability (CFU/mL), biofilm biomass, and total protein dosage. Data were analyzed using Mann-Whitney test (α = 5%). In addition, co-culture biofilms were analyzed using fluorescence microscopy. RESULTS C. albicans growth did not change due to the presence of S. salivarius. Besides, co-culture biofilms showed a significant difference in the number of viable cells between 24 and 72 h of biofilm development (p < 0.05). The highest biofilm biomass and protein dosage were observed in co-cultures at 72 h of biofilm development. Fluorescence microscopy showed that co-cultures biofilms at 24 h have limited number of pseudo-hyphal and hyphae cells of C. albicans. At 72 h, these types of cells have increased. S. salivarius in both stages of development was present in some clusters surrounded by C. albicans. CONCLUSIONS Co-cultivation of C. albicans with S. salivarius in biofilms developed on titanium surfaces, under lower oxygen levels, did not affect fungus growth. In addition, S. salivarius did not hind C. albicans virulence. These findings suggest that the use of S. salivarius as a probiotic would be ineffective in peri-implant disease treatment.
Collapse
Affiliation(s)
- Loyse Martorano-Fernandes
- Postgraduate Program in Dentistry, Federal University of Paraíba, Cidade Universitária, João Pessoa, Paraiba, Brazil
| | - Nadiny Cezar Rodrigues
- School of Dentistry, Federal University of Paraíba, Cidade Universitária, João Pessoa, Paraiba, Brazil
| | | | - Yuri Wanderley Cavalcanti
- Department of Clinic and Social Dentistry, Federal University of Paraíba, Cidade Universitária, João Pessoa, Paraiba, Brazil
| | | |
Collapse
|
48
|
Ciprandi G, La Mantia I, Damiani V, Passali D. Local Bacteriotherapy - a promising preventive tool in recurrent respiratory infections. Expert Rev Clin Immunol 2020; 16:1047-1052. [PMID: 33022191 DOI: 10.1080/1744666x.2021.1833720] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
INTRODUCTION Children with recurrent respiratory infections (RRI) represent a social issue for the economic burden and the negative family impact. Local Bacteriotherapy is an attractive therapeutic strategy that could be potentially effective in preventing infections. The current article remarks on the existing evidence of preventing RRI by Local Bacteriotherapy. AREAS COVERED The literature search methodology was based on the articles cited by PubMed from 1980 to 2020. Respiratory infections include rhino-pharyngitis, otitis media, rhinosinusitis, pharyngo-tracheitis, bronchitis, and pneumonia. Several studies were performed to investigate the effects of Local Bacteriotherapy in children with RRI. Both intranasal and oral Local Bacteriotherapy were evaluated. The findings showed that Local Bacteriotherapy significantly reduced the number of RI episodes, their severity, the use of antibiotics, and school absences. EXPERT OPINION Local Bacteriotherapy is a promising approach to RRI prevention and could be a profitable strategy to contrast infections in the future.
Collapse
Affiliation(s)
| | | | - Valerio Damiani
- Medical Department, Drugs Minerals and Generics , Pomezia, Italy
| | | |
Collapse
|
49
|
Wei YS, Chang YR, Tsai YT, Yang YT, Weng SH, Tseng LF, Chou HC, Hu AT, Liao EC, Chen HY, Lin GY, Cheng WC, Chan HL. The distribution of cultivable oral anaerobic microbiota identified by MALDI-TOF MS in healthy subjects and in patients with periodontal disease. J Pharm Biomed Anal 2020; 192:113647. [PMID: 33010501 DOI: 10.1016/j.jpba.2020.113647] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 09/12/2020] [Accepted: 09/15/2020] [Indexed: 12/22/2022]
Abstract
In this study, we aimed to identify the cultivatable oral anaerobic bacterial distribution in oral cavity by MALDI-TOF Biotyper. The bacterial distribution of three groups, including subjects with/without periodontal disease, two clusters of age (60 years as the cutoff), and before/after treatment, were investigated in this study. There were 38 participants recruited in this study, involving 18 subjects with moderate to severe periodontal-infected patients and 20 healthy controls. Total number of 126 bacterial species were identified by MALDI-TOF MS. The relative abundance of Streptococcus gordonii and Streptococcus intermedius in periodontal patients is higher than healthy controls indicating potential biomarkers for periodontal disease. Participants with periodontal disease were subdivided in to two clusters of age (60 years as the cutoff), 11 and 7 participants were age <60 years and>60 years, respectively. Meanwhile, the incidence of Streptococcus pneumoniae and Streptococcus oralis infection were higher in the subjects above 60 years old than below. Moreover, the bacterial distribution between pre-treatment and post-treatment was similar indicating that basic treatment without the ability to redistribute the microbiota. In summary, the cultivable oral anaerobic bacteria were identified by MALDI-TOF MS and the bacterial distribution shifting was shown to be associated with the progress of periodontal disease to aging and basic treatment. This study provided information for diagnosis and treatment guidelines for oral healthcare.
Collapse
Affiliation(s)
- Yu-Shan Wei
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ru Chang
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan; General Biologicals Corporation, Hsinchu, Taiwan
| | - Yi-Ting Tsai
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Yi-Ting Yang
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Shang-Hui Weng
- Department of Life Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Lin-Fang Tseng
- Life Science in Department of Applied Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsiu-Chuan Chou
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Taiwan
| | - Alice Tinyu Hu
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - En-Chi Liao
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Hsin-Yi Chen
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan
| | - Guan-Yu Lin
- Institute of Molecular and Cellular Biology, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Chi Cheng
- SDGs Teaching and Research Headquarters, Tzu Chi University, Hualien, Taiwan.
| | - Hong-Lin Chan
- Institute of Bioinformatics and Structural Biology and Department of Medical Science, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
50
|
Changes in the Bacterial Diversity of Human Milk during Late Lactation Period (Weeks 21 to 48). Foods 2020; 9:foods9091184. [PMID: 32867028 PMCID: PMC7554819 DOI: 10.3390/foods9091184] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/28/2022] Open
Abstract
Breast milk from a single mother was collected during a 28-week lactation period. Bacterial diversity was studied by amplicon sequencing analysis of the V3-V4 variable region of the 16S rRNA gene. Firmicutes and Proteobacteria were the main phyla detected in the milk samples, followed by Actinobacteria and Bacteroidetes. The proportion of Firmicutes to Proteobacteria changed considerably depending on the sampling week. A total of 411 genera or higher taxons were detected in the set of samples. Genus Streptococcus was detected during the 28-week sampling period, at relative abundances between 2.0% and 68.8%, and it was the most abundant group in 14 of the samples. Carnobacterium and Lactobacillus had low relative abundances. At the genus level, bacterial diversity changed considerably at certain weeks within the studied period. The weeks or periods with lowest relative abundance of Streptococcus had more diverse bacterial compositions including genera belonging to Proteobacteria that were poorly represented in the rest of the samples.
Collapse
|