1
|
Brown GD, Ballou ER, Bates S, Bignell EM, Borman AM, Brand AC, Brown AJP, Coelho C, Cook PC, Farrer RA, Govender NP, Gow NAR, Hope W, Hoving JC, Dangarembizi R, Harrison TS, Johnson EM, Mukaremera L, Ramsdale M, Thornton CR, Usher J, Warris A, Wilson D. The pathobiology of human fungal infections. Nat Rev Microbiol 2024; 22:687-704. [PMID: 38918447 DOI: 10.1038/s41579-024-01062-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2024] [Indexed: 06/27/2024]
Abstract
Human fungal infections are a historically neglected area of disease research, yet they cause more than 1.5 million deaths every year. Our understanding of the pathophysiology of these infections has increased considerably over the past decade, through major insights into both the host and pathogen factors that contribute to the phenotype and severity of these diseases. Recent studies are revealing multiple mechanisms by which fungi modify and manipulate the host, escape immune surveillance and generate complex comorbidities. Although the emergence of fungal strains that are less susceptible to antifungal drugs or that rapidly evolve drug resistance is posing new threats, greater understanding of immune mechanisms and host susceptibility factors is beginning to offer novel immunotherapeutic options for the future. In this Review, we provide a broad and comprehensive overview of the pathobiology of human fungal infections, focusing specifically on pathogens that can cause invasive life-threatening infections, highlighting recent discoveries from the pathogen, host and clinical perspectives. We conclude by discussing key future challenges including antifungal drug resistance, the emergence of new pathogens and new developments in modern medicine that are promoting susceptibility to infection.
Collapse
Affiliation(s)
- Gordon D Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK.
| | - Elizabeth R Ballou
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Steven Bates
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Elaine M Bignell
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Andrew M Borman
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Alexandra C Brand
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Alistair J P Brown
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Carolina Coelho
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Peter C Cook
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Rhys A Farrer
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Nelesh P Govender
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Neil A R Gow
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - William Hope
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - J Claire Hoving
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Rachael Dangarembizi
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Thomas S Harrison
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Elizabeth M Johnson
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Liliane Mukaremera
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Mark Ramsdale
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | | | - Jane Usher
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Adilia Warris
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| | - Duncan Wilson
- Medical Research Council Centre for Medical Mycology, University of Exeter, Exeter, UK
| |
Collapse
|
2
|
Dricot CEMK, Erreygers I, Cauwenberghs E, De Paz J, Spacova I, Verhoeven V, Ahannach S, Lebeer S. Riboflavin for women's health and emerging microbiome strategies. NPJ Biofilms Microbiomes 2024; 10:107. [PMID: 39420006 PMCID: PMC11486906 DOI: 10.1038/s41522-024-00579-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/06/2024] [Indexed: 10/19/2024] Open
Abstract
Riboflavin (vitamin B2) is an essential water-soluble vitamin that serves as a precursor of flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD). FMN and FAD are coenzymes involved in key enzymatic reactions in energy metabolism, biosynthesis, detoxification and electron scavenging pathways. Riboflavin deficiency is prevalent worldwide and impacts women's health due to riboflavin demands linked to urogenital and reproductive health, hormonal fluctuations during the menstrual cycle, pregnancy, and breastfeeding. Innovative functional foods and nutraceuticals are increasingly developed to meet women's riboflavin needs to supplement dietary sources. An emerging and particularly promising strategy is the administration of riboflavin-producing lactic acid bacteria, combining the health benefits of riboflavin with those of probiotics and in situ riboflavin production. Specific taxa of lactobacilli are of particular interest for women, because of the crucial role of Lactobacillus species in the vagina and the documented health effects of other Lactobacillaceae taxa in the gut and on the skin. In this narrative review, we synthesize the underlying molecular mechanisms and clinical benefits of riboflavin intake for women's health, and evaluate the synergistic potential of riboflavin-producing lactobacilli and other microbiota.
Collapse
Affiliation(s)
- Caroline E M K Dricot
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Isabel Erreygers
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Eline Cauwenberghs
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Jocelyn De Paz
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Irina Spacova
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Veronique Verhoeven
- Department of Family Medicine and Population Health, University of Antwerp, Antwerp, Belgium
- U-MaMi Excellence Centre, University of Antwerp, Antwerp, Belgium
| | - Sarah Ahannach
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Sarah Lebeer
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium.
- U-MaMi Excellence Centre, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
3
|
Vazquez-Munoz R, Thompson A, Sobue T, Dongari-Bagtzoglou A. Lactobacillus johnsonii is a dominant Lactobacillus in the murine oral mucosa and has chitinase activity that compromises fungal cell wall integrity. mBio 2024; 15:e0241624. [PMID: 39287438 PMCID: PMC11481578 DOI: 10.1128/mbio.02416-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
The oral microbiome is a critical determinant of health and disease, as interactions between oral microorganisms can influence their physiology and the development or severity of oral infections. Lactobacilli have a widely recognized antagonistic relationship with Candida albicans and may exhibit probiotic properties that limit oral fungal infection. We previously reported that Lactobacillus johnsonii strain MT4, an oral strain isolated from C57BL/6 mice, can induce global changes in the murine oral microbiome and has anti-Candida activity in vitro. To build on this information, we analyzed its abundance on the mouse oral mucosa, tested its impact on the severity and progression of oropharyngeal candidiasis (OPC) in a mouse model, and further explored the mechanism of antifungal activity in vitro. Our findings reveal that L. johnsonii MT4 is a dominant cultivable Lactobacillus in the oral mucosa of C57BL/6 mice. Strain MT4 has chitinase activity against C. albicans, which damages the cell wall and compromises fungal metabolic activity. Oral inoculation with strain MT4 causes a reduction in the Candida-induced rise in the abundance of oral enterococci and oral mucosal damage. This research underscores the potential of L. johnsonii strain MT4 as a novel probiotic agent in the prevention or management of OPC, and it contributes to a better understanding of the role of oral bacterial microbiota role in the pathogenesis of fungal infections. IMPORTANCE The interactions between the opportunistic pathogen Candida albicans and resident oral bacteria are particularly crucial in maintaining oral health. Emerging antifungal drug-resistant strains, slow-paced drug discovery, and the risk of side effects can compromise the effectiveness of current treatments available for oropharyngeal candidiasis. This study advances the search for alternative microbiome-targeted therapies in oral fungal infections. We report that Lactobacillus johnsonii strain MT4 prevents the Candida-induced bloom of dysbiotic oral enterococci and reduces oral mucosal lesions in an oropharyngeal candidiasis murine model. We also show that this strain directly compromises the cell wall and reduces fungal metabolic activity, partly due to its chitinase activity.
Collapse
Affiliation(s)
- Roberto Vazquez-Munoz
- Department of General Dentistry, the University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Angela Thompson
- Department of General Dentistry, the University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Takanori Sobue
- Department of General Dentistry, the University of Connecticut Health Center, Farmington, Connecticut, USA
| | - Anna Dongari-Bagtzoglou
- Department of General Dentistry, the University of Connecticut Health Center, Farmington, Connecticut, USA
| |
Collapse
|
4
|
Ferreira RLPS, Nova BGV, Carmo MS, Abreu AG. Mechanisms of action of Lactobacillus spp. in the treatment of oral candidiasis. BRAZ J BIOL 2024; 84:e282609. [PMID: 39319927 DOI: 10.1590/1519-6984.282609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 07/16/2024] [Indexed: 09/26/2024] Open
Abstract
Candida albicans is often associated with oral candidiasis, and drug-resistance profiles have contributed to an increase in morbidity and mortality. It is known that Lactobacillus spp. acts by competing for adhesion to the epithelium, absorption of nutrients and modulation of the human microbiota. Therefore, they are important to assist in the host's microbiological balance and reduce the growth of Candida spp. Until now, there have been no reports in the literature of reviews correlating to the use of Lactobacillus spp. in the treatment of oral candidiasis. Thus, this review aims to highlight the mechanisms of action of Lactobacillus spp. and methods that can be used in the treatment of oral candidiasis. This is a study carried out through the databases PubMed Central and Scientific Electronic Library Online, using the following keywords: Oral Candidiasis and Lactobacillus. Original articles about oral candidiasis were included, with both in vitro and in vivo analyses, and published from 2012 to 2022. Lactobacillus rhamnosus was the most common microorganism used in the experiments against Candida, acting mainly in the reduction of biofilm, filamentation, and competing for adhesion sites of Candida spp. Among in vivo studies, most researchers used immunosuppressed mouse modelsof Candida infection. The studies showed that Lactobacillus has a great potential as a probiotic, acting mainly in the prevention and treatment of mucosal diseases. Thus, the use of Lactobacillus may be a good strategy for the treatment of oral candidiasis.
Collapse
Affiliation(s)
- R L P S Ferreira
- Universidade Ceuma - UniCEUMA, Laboratório de Patogenicidade Microbiana, São Luís, MA, Brasil
- Universidade Federal do Maranhão - UFMA, Programa de Pós-graduação em Ciências da Saúde, São Luís, MA, Brasil
| | - B G V Nova
- Universidade Ceuma - UniCEUMA, Laboratório de Patogenicidade Microbiana, São Luís, MA, Brasil
| | - M S Carmo
- Universidade Ceuma - UniCEUMA, Laboratório de Patogenicidade Microbiana, São Luís, MA, Brasil
| | - A G Abreu
- Universidade Ceuma - UniCEUMA, Laboratório de Patogenicidade Microbiana, São Luís, MA, Brasil
- Universidade Federal do Maranhão - UFMA, Programa de Pós-graduação em Ciências da Saúde, São Luís, MA, Brasil
| |
Collapse
|
5
|
Tran DM, Huynh TU, Do TO. Novel domain-structure-containing chitinases A and B of Bacillus velezensis produced by recombinant Escherichia coli cells: Synergism on chitin degradation and their potential in suppressing Candida albicans cell germination. J Genet Eng Biotechnol 2024; 22:100402. [PMID: 39179322 PMCID: PMC11318559 DOI: 10.1016/j.jgeb.2024.100402] [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: 02/25/2024] [Revised: 06/12/2024] [Accepted: 07/10/2024] [Indexed: 08/26/2024]
Abstract
Bacillus velezensis RB.IBE29 harbors two chitinases belonging to the glycoside hydrolase family 18 and exhibiting a novel domain structure. The roles of these chitinases in crop production have been reported; nevertheless, their contribution to controlling human pathogens is unknown. In this initial work, the chitinases A (BvChiA) and B (BvChiB) of strain RB.IBE29 were produced in recombinant Escherichia coli BL21-CodonPlus (DE3)-RIPL cells and subsequently purified using HisTrap FF column. The purified BvChiA and BvChiB exhibited the highest chitinase and binding activities against colloidal chitin. Combining both chitinases for the hydrolysis of powdered chitin increased the reducing sugar content by 88.7 %. Moreover, the purified chitinases remarkably suppressed the germination of Candida albicans VTCC 20568 (=JCM 2070) cells. These results indicated that the novel domain-structure-containing chitinases of strain RB.IBE29 have great potential and can be further developed as a novel therapeutic agent against human pathogenic C. albicans.
Collapse
Affiliation(s)
- Dinh Minh Tran
- Institute of Biotechnology and Environment, Tay Nguyen University, Buon Ma Thuot, Dak Lak 630000, Viet Nam.
| | - To Uyen Huynh
- Institute of Biotechnology and Environment, Tay Nguyen University, Buon Ma Thuot, Dak Lak 630000, Viet Nam
| | - Tu Oanh Do
- Institute of Biotechnology and Environment, Tay Nguyen University, Buon Ma Thuot, Dak Lak 630000, Viet Nam
| |
Collapse
|
6
|
Elnagar RM. Cross interaction between bacterial and fungal microbiota and their relevance to human health and disease: mechanistic pathways and prospective therapy. BIOSCIENCE OF MICROBIOTA, FOOD AND HEALTH 2024; 43:309-320. [PMID: 39364131 PMCID: PMC11444862 DOI: 10.12938/bmfh.2024-031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Accepted: 06/27/2024] [Indexed: 10/05/2024]
Abstract
Diverse bacterial and fungal microbiota communities inhabit the human body, and their presence is essential for maintaining host homeostasis. The oral cavity, lung, gut, and vagina are just a few of the bodily cavities where these microorganisms communicate with one another, either directly or indirectly. The effects of this interaction can be either useful or detrimental to the host. When the healthy microbial diversity is disturbed, for instance, as a result of prolonged treatment with broad spectrum antibiotics, this allows the growth of specific microbes at the expense of others and alters their pathogenicity, causing a switch of commensal germs into pathogenic germs, which could promote tissue invasion and damage, as occurs in immunocompromised patients. Consequently, antimicrobials that specifically target pathogens may help in minimizing secondary issues that result from the disruption of useful bacterial/fungal interactions (BFIs). The interface between Candida albicans and Aspergillus fumigatus with bacteria at various body sites is emphasized in the majority of the medically important BFIs that have been reported thus far. This interface either supports or inhibits growth, or it enhances or blocks the generation of virulence factors. The aim of this review is to draw attention to the link between the bacterial and fungal microbiota and how they contribute to both normal homeostasis and disease development. Additionally, recent research that has studied microbiota as novel antimicrobials is summarized.
Collapse
Affiliation(s)
- Rasha Mokhtar Elnagar
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
- Medical Microbiology and Immunology Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| |
Collapse
|
7
|
Ismail SA, Fayed B, Abdelhameed RM, Hassan AA. Chitinase-functionalized UiO-66 framework nanoparticles active against multidrug-resistant Candida Auris. BMC Microbiol 2024; 24:269. [PMID: 39030474 PMCID: PMC11264975 DOI: 10.1186/s12866-024-03414-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 07/05/2024] [Indexed: 07/21/2024] Open
Abstract
Candida auris (C. auris) is a yeast that has caused several outbreaks in the last decade. Cell wall chitin plays a primary role in the antifungal resistance of C. auris. Herein, we investigated the potential of chitinase immobilized with UiO-66 to act as a potent antifungal agent against C. auris. Chitinase was produced from Talaromyces varians SSW3 in a yield of 8.97 U/g dry substrate (ds). The yield was statistically enhanced to 120.41 U/g ds by using Plackett-Burman and Box-Behnken design. We synthesized a UiO-66 framework that was characterized by SEM, TEM, XRD, FTIR, a particle size analyzer, and a zeta sizer. The produced framework had a size of 70.42 ± 8.43 nm with a uniform cubic shape and smooth surface. The produced chitinase was immobilized on UiO-66 with an immobilization yield of 65% achieved after a 6 h loading period. The immobilization of UiO-66 increased the enzyme activity and stability, as indicated by the obtained Kd and T1/2 values. Furthermore, the hydrolytic activity of chitinase was enhanced after immobilization on UiO-66, with an increase in the Vmax and a decrease in the Km of 2- and 38-fold, respectively. Interestingly, the antifungal activity of the produced chitinase was boosted against C. auris by loading the enzyme on UiO-66, with an MIC50 of 0.89 ± 0.056 U/mL, compared to 5.582 ± 0.57 U/mL for the free enzyme. This study offers a novel promising alternative approach to combat the new emerging pathogen C. auris.
Collapse
Affiliation(s)
- Shaymaa A Ismail
- Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Institute, National Research Centre, P.O. 12622, 33 El Bohouth Street, Dokki, Giza, Egypt.
| | - Bahgat Fayed
- Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Institute, National Research Centre, P.O. 12622, 33 El Bohouth Street, Dokki, Giza, Egypt.
| | - Reda M Abdelhameed
- Applied Organic Chemistry Department, Chemical Industries Research Institute, National Research Centre, 33 EL Buhouth St, Dokki, Giza, 12622, Egypt
| | - Amira A Hassan
- Department of Chemistry of Natural and Microbial Products, Pharmaceutical and Drug Industries Research Institute, National Research Centre, P.O. 12622, 33 El Bohouth Street, Dokki, Giza, Egypt
| |
Collapse
|
8
|
Chow EW, Pang LM, Wang Y. The impact of the host microbiota on Candida albicans infection. Curr Opin Microbiol 2024; 80:102507. [PMID: 38955050 DOI: 10.1016/j.mib.2024.102507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 07/04/2024]
Abstract
The human microbiota is a complex microbial ecosystem populated by bacteria, fungi, viruses, protists, and archaea. The coexistence of fungi alongside with many billions of bacteria, especially in the gut, involves complex interactions, ranging from antagonistic to beneficial, between the members of these two kingdoms. Bacteria can impact fungi through various means, such as physical interactions, secretion of metabolites, or alteration of the host immune response, thereby affecting fungal growth and virulence. This review summarizes recent progress in this field, delving into the latest understandings of bacterial-fungal-immune interactions and innovative therapeutic approaches addressing the challenges of treating fungal infections associated with microbiota imbalances.
Collapse
Affiliation(s)
- Eve Wl Chow
- A*STAR Infectious Diseases Laboratories (ID Labs), Agency for Science and Technology Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648
| | - Li M Pang
- A*STAR Infectious Diseases Laboratories (ID Labs), Agency for Science and Technology Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648
| | - Yue Wang
- A*STAR Infectious Diseases Laboratories (ID Labs), Agency for Science and Technology Research (A*STAR), 8A Biomedical Grove, #05-13 Immunos, Singapore 138648; Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
9
|
Kaur M, Nagpal M, Dhingra GA, Rathee A. Exploring chitin: novel pathways and structures as promising targets for biopesticides. Z NATURFORSCH C 2024; 79:125-136. [PMID: 38760917 DOI: 10.1515/znc-2024-0027] [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: 02/01/2024] [Accepted: 05/05/2024] [Indexed: 05/20/2024]
Abstract
Chitin, the most prevalent polymer in nature, a significant structural polysaccharide that comes in second only to cellulose. Chitin is a crucial component of fungal cell walls and also present in many other creatures, such as viruses, plants, animals, insect exoskeletons, and crustacean shells. Chitin presents itself as a promising target for the development of biopesticides. It focuses on unraveling the unique structures and biochemical pathways associated with chitin, aiming to identify vulnerabilities that can be strategically leveraged for effective and environmentally sustainable pest control. It involves a comprehensive analysis of chitinase enzymes, chitin biosynthesis, and chitin-related processes across diverse organisms. By elucidating the molecular intricacies involved in chitin metabolism, this review seeks to unveil potential points of intervention that can disrupt essential biological processes in target pests without harming non-target species. This holistic approach to understanding chitin-related pathways aims to inform the design and optimization of biopesticides with enhanced specificity and reduced ecological impact. The outcomes of this study hold great promise for advancing innovative and eco-friendly pest management strategies. By targeting chitin structures and pathways, biopesticides developed based on these findings may offer a sustainable and selective alternative to conventional chemical pesticides, contributing to the ongoing efforts towards more environmentally conscious and effective pest control solutions.
Collapse
Affiliation(s)
- Malkiet Kaur
- 418665 University Institute of Pharma Sciences, Chandigarh University , Mohali, Punjab, India
| | - Manju Nagpal
- Chitkara College of Pharmacy, 154025 Chitkara University , Rajpura, Punjab, India
| | | | - Ankit Rathee
- 418665 University Institute of Pharma Sciences, Chandigarh University , Mohali, Punjab, India
| |
Collapse
|
10
|
Spacova I, Allonsius CN, De Boeck I, Oerlemans E, Tuyaerts I, Van de Vliet N, van den Broek MFL, Jimenez L, Boyer M, Rodriguez B, Ballet N, Lebeer S. Multifactorial inhibition of Candida albicans by combinations of lactobacilli and probiotic Saccharomyces cerevisiae CNCM I-3856. Sci Rep 2024; 14:9365. [PMID: 38654026 DOI: 10.1038/s41598-024-59869-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 04/15/2024] [Indexed: 04/25/2024] Open
Abstract
Strategies against the opportunistic fungal pathogen Candida albicans based on probiotic microorganisms represent a promising alternative to traditional antifungals. Here, we investigated the effects of Lactobacillaceae isolates from fermented foods or the human vagina, alone or in combination with the probiotic yeast Saccharomyces cerevisiae CNCM I-3856, against C. albicans in vitro. Nine out of nineteen tested strains of Lactobacillaceae inhibited growth of C. albicans with inhibition zones of 1-3 mm in spot assays. Five out of nineteen lactobacilli tested as such or in combination with S. cerevisiae CNCM I-3856 also significantly inhibited C. albicans hyphae formation, including Limosilactobacillus fermentum LS4 and L. fermentum LS5 resulting in respectively 62% and 78% hyphae inhibition compared to the control. Thirteen of the tested nineteen lactobacilli aggregated with the yeast form of C. albicans, with Lactiplantibacillus carotarum AMBF275 showing the strongest aggregation. The aggregation was enhanced when lactobacilli were combined with S. cerevisiae CNCM I-3856. No significant antagonistic effects were observed between the tested lactobacilli and S. cerevisiae CNCM I-3856. The multifactorial activity of Lactobacillaceae strains alone or combined with the probiotic S. cerevisiae CNCM I-3856 against C. albicans without antagonistic effects between the beneficial strains, paves the way for developing consortium probiotics for in vivo applications.
Collapse
Affiliation(s)
- Irina Spacova
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Camille Nina Allonsius
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Ilke De Boeck
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Eline Oerlemans
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Ines Tuyaerts
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Nele Van de Vliet
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Marianne F L van den Broek
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium
| | - Luciana Jimenez
- Lesaffre International, Lesaffre Group, Rue Gabriel Péri 137, 59700, Marcq-en-Baroeul, France
| | - Mickaël Boyer
- Lesaffre International, Lesaffre Group, Rue Gabriel Péri 137, 59700, Marcq-en-Baroeul, France
| | - Bertrand Rodriguez
- Gnosis by Lesaffre, Lesaffre Group, Rue Gabriel Péri 137, 59700, Marcq-en-Baroeul, France
| | - Nathalie Ballet
- Lesaffre International, Lesaffre Group, Rue Gabriel Péri 137, 59700, Marcq-en-Baroeul, France
| | - Sarah Lebeer
- Laboratory of Applied Microbiology and Biotechnology, Department of Bioscience Engineering, University of Antwerp, Groenenborgerlaan 171, 2020, Antwerp, Belgium.
| |
Collapse
|
11
|
Leser T, Baker A. Molecular Mechanisms of Lacticaseibacillus rhamnosus, LGG ® Probiotic Function. Microorganisms 2024; 12:794. [PMID: 38674738 PMCID: PMC11051730 DOI: 10.3390/microorganisms12040794] [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: 02/26/2024] [Revised: 04/10/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
To advance probiotic research, a comprehensive understanding of bacterial interactions with human physiology at the molecular and cellular levels is fundamental. Lacticaseibacillus rhamnosus LGG® is a bacterial strain that has long been recognized for its beneficial effects on human health. Probiotic effector molecules derived from LGG®, including secreted proteins, surface-anchored proteins, polysaccharides, and lipoteichoic acids, which interact with host physiological processes have been identified. In vitro and animal studies have revealed that specific LGG® effector molecules stimulate epithelial cell survival, preserve intestinal barrier integrity, reduce oxidative stress, mitigate excessive mucosal inflammation, enhance IgA secretion, and provide long-term protection through epigenetic imprinting. Pili on the cell surface of LGG® promote adhesion to the intestinal mucosa and ensure close contact to host cells. Extracellular vesicles produced by LGG® recapitulate many of these effects through their cargo of effector molecules. Collectively, the effector molecules of LGG® exert a significant influence on both the gut mucosa and immune system, which promotes intestinal homeostasis and immune tolerance.
Collapse
Affiliation(s)
- Thomas Leser
- Future Labs, Human Health Biosolutions, Novonesis, Kogle Alle 6, 2970 Hoersholm, Denmark;
| | | |
Collapse
|
12
|
Wang Y, Liu Z, Chen T. Vaginal microbiota: Potential targets for vulvovaginal candidiasis infection. Heliyon 2024; 10:e27239. [PMID: 38463778 PMCID: PMC10923723 DOI: 10.1016/j.heliyon.2024.e27239] [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: 11/23/2023] [Revised: 01/28/2024] [Accepted: 02/27/2024] [Indexed: 03/12/2024] Open
Abstract
Vulvovaginal candidiasis (VVC) is the second most common cause of vaginal infection globally after bacterial vaginosis (BV) and associated with adverse reproductive and obstetric outcomes, including preterm delivery, sexually transmitted infections and pelvic inflammatory disease. Although effective control of VVC is achievable with the use of traditional treatment strategies (i.e., antifungals), the possibility of drug intolerance, treatment failure and recurrence, as well as the appearance of antifungal-resistant Candida species remain critical challenges. Therefore, alternative therapeutic strategies against VVC are urgently required. In recent years, an improved understanding of the dysbiotic vaginal microbiota (VMB) during VVC has prompted the consideration of administering -biotics to restore the balance of the VMB within the context of VVC prevention and treatment. Here, we aim to summarize the current evidence of the anti-Candida effects of probiotics, postbiotics and synbiotics and their potential use as an alternative/complementary therapy against VVC. Additionally, this review discusses advantages and challenges associated with the application of -biotics in VVC to provide guidance for their later use. We also review new developments in VVC therapy, i.e., vaginal microbiota transplantation (VMT) as an emerging live biotherapeutic therapy against VVC and discuss existing shortcomings associated with this nascent field, expecting to stimulate further investigations for introduction of new therapies against VVC.
Collapse
Affiliation(s)
- Yufei Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University Jiangxi Medical College, No.1299, Xuefu Avenue, Honggutan District, Nanchang City, Jiangxi Province, China
- Queen Mary School, Jiangxi Medical College, Nanchang University, No.1299, Xuefu Avenue, Honggutan District, Nanchang City, Jiangxi Province, China
| | - Zhaoxia Liu
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University Jiangxi Medical College, No.1299, Xuefu Avenue, Honggutan District, Nanchang City, Jiangxi Province, China
| | - Tingtao Chen
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Nanchang University Jiangxi Medical College, No.1299, Xuefu Avenue, Honggutan District, Nanchang City, Jiangxi Province, China
- School of Pharmacy, National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, No.1299, Xuefu Avenue, Honggutan District, Nanchang City, Jiangxi Province, China
| |
Collapse
|
13
|
Netto de Oliveira da Cunha C, Rodeghiero Collares S, Carvalho Rodrigues D, Walcher DL, Quintana de Moura M, Rodrigues Martins LH, Baracy Klafke G, de Oliveira Arias JL, Carapelli R, do Santos Espinelli Junior JB, Scaini CJ, Farias da Costa de Avila L. The larvicidal effect of the supernatant of Lactobacillus acidophilus ATCC 4356 on Toxocara canis. Exp Parasitol 2024; 258:108720. [PMID: 38367945 DOI: 10.1016/j.exppara.2024.108720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2023] [Revised: 02/14/2024] [Accepted: 02/15/2024] [Indexed: 02/19/2024]
Abstract
Human toxocariasis is a parasitic anthropozoonosis that is difficult to treat and control. A previous study carried out with Lactobacillus acidophilus ATCC 4356 revealed that the cell free supernatant (CFS) of this probiotic killed 100% of Toxocara canis larvae in vitro. The present study aimed to investigate the characteristics of the CFS of L. acidophilus ATCC 4356, which may be involved in its larvicidal effects on T. canis. L. acidophilus ATCC 4356 was cultured, and lactic and acetic acids present in the CFS were quantified by high performance liquid chromatography (HPLC). The levels of pH and H2O2 were also analyzed. To assess the larvicidal effect of the CFS, this was tested pure and diluted (1:2 to 1:128) on T. canis larvae. High concentrations of lactic and acetic acids were detected in the CFS. The acidity of the pure CFS was observed at pH 3.8, remaining acidic at dilutions of 1:2 to 1:16. Regarding the in vitro larvicidal effect, 100% death of T. canis larvae was observed using the pure CFS and 1:2 dilution. Based on these results, it can be inferred that the presence of higher concentrations of organic acids and low pH of the medium contributed to the larvicidal activity of the CFS of L. acidophilus ATCC 4356. In addition, the maintenance of the larvicidal effect, even after dilution, suggests a greater chance of the larvicidal effect of this CFS against T. canis in vivo.
Collapse
Affiliation(s)
- Carolina Netto de Oliveira da Cunha
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil.
| | | | - Débora Carvalho Rodrigues
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Débora Liliane Walcher
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Micaele Quintana de Moura
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Lourdes Helena Rodrigues Martins
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Gabriel Baracy Klafke
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Jean Lucas de Oliveira Arias
- Integrated Analysis Center, School of Chemistry and Food, Federal University of Rio Grande, Av. Italia, Km 6 - Campus Carreiros, CEP 96203-900, Rio Grande, Rio Grande do Sul, Brazil
| | - Rodolfo Carapelli
- School of Chemistry and Food, Federal University of Rio Grande, Avenida Itália, Km 08 - Campus Carreiros, CEP 96.203-900, Rio Grande, Rio Grande do Sul, Brazil
| | - João Batista do Santos Espinelli Junior
- School of Chemistry and Food, Federal University of Rio Grande, Avenida Itália, Km 08 - Campus Carreiros, CEP 96.203-900, Rio Grande, Rio Grande do Sul, Brazil
| | - Carlos James Scaini
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| | - Luciana Farias da Costa de Avila
- Faculty of Medicine (FAMED)/Parasitology Laboratory, Federal University of Rio Grande, General Osório, S/N, CEP 96200-190, Rio Grande, RS, Brazil
| |
Collapse
|
14
|
Das S, Konwar BK. Inhibiting pathogenicity of vaginal Candida albicans by lactic acid bacteria and MS analysis of their extracellular compounds. APMIS 2024; 132:161-186. [PMID: 38168754 DOI: 10.1111/apm.13365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Accepted: 11/26/2023] [Indexed: 01/05/2024]
Abstract
Maintaining healthy vaginal microflora post-puberty is critical. In this study we explore the potential of vaginal lactic acid bacteria (LAB) and their extracellular metabolites against the pathogenicity of Candida albicans. The probiotic culture free supernatant (PCFS) from Lactobacillus crispatus, L. gasseri, and L. vaginalis exhibit an inhibitory effect on budding, hyphae, and biofilm formation of C. albicans. LGPCFS manifested the best potential among the LAB PCFS, inhibiting budding for 24 h and restricting hyphae formation post-stimulation. LGPCFS also pre-eminently inhibited biofilm formation. Furthermore, L. gasseri itself grew under RPMI 1640 stimulation suppressing the biofilm formation of C. albicans. The PCFS from the LAB downregulated the hyphal genes of C. albicans, inhibiting the yeast transformation to fungi. Hyphal cell wall proteins HWP1, ALS3, ECE1, and HYR1 and transcription factors BCR1 and CPH1 were downregulated by the metabolites from LAB. Finally, the extracellular metabolome of the LAB was studied by LC-MS/MS analysis. L.gasseri produced the highest antifungal compounds and antibiotics, supporting its best activity against C. albicans. Vaginal LAB and their extracellular metabolites perpetuate C. albicans at an avirulent state. The metabolites produced by these LAB in vitro have been identified, and can be further exploited as a preventive measure against vaginal candidiasis.
Collapse
Affiliation(s)
- Shreaya Das
- Department of MBBT, Tezpur University, Napaam, Assam, India
| | | |
Collapse
|
15
|
Eichelberger KR, Paul S, Peters BM, Cassat JE. Candida-bacterial cross-kingdom interactions. Trends Microbiol 2023; 31:1287-1299. [PMID: 37640601 PMCID: PMC10843858 DOI: 10.1016/j.tim.2023.08.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/14/2023] [Accepted: 08/02/2023] [Indexed: 08/31/2023]
Abstract
While the fungus Candida albicans is a common colonizer of healthy humans, it is also responsible for mucosal infections and severe invasive disease. Understanding the mechanisms that allow C. albicans to exist as both a benign commensal and as an invasive pathogen have been the focus of numerous studies, and recent findings indicate an important role for cross-kingdom interactions on C. albicans biology. This review highlights how C. albicans-bacteria interactions influence healthy polymicrobial community structure, host immune responses, microbial pathogenesis, and how dysbiosis may lead to C. albicans infection. Finally, we discuss how cross-kingdom interactions represent an opportunity to identify new antivirulence compounds that target fungal infections.
Collapse
Affiliation(s)
- Kara R Eichelberger
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Saikat Paul
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Brian M Peters
- Department of Clinical Pharmacy and Translational Science, University of Tennessee Health Science Center, Memphis, TN, USA; Department of Microbiology, Immunology, and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - James E Cassat
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation (VI4), Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
16
|
Temmermans J, Legein M, Zhao Y, Kiekens F, Smagghe G, de Coninck B, Lebeer S. The biocontrol agent Lactiplantibacillus plantarum AMBP214 is dispersible to plants via bumblebees. Appl Environ Microbiol 2023; 89:e0095023. [PMID: 37882529 PMCID: PMC10686056 DOI: 10.1128/aem.00950-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Accepted: 09/11/2023] [Indexed: 10/27/2023] Open
Abstract
IMPORTANCE Plant protection products are essential for ensuring food production, but their use poses a threat to human and environmental health, and their efficacy is decreasing due to the acquisition of resistance by pathogens. Stricter regulations and consumer demand for cleaner produce are driving the search for safer and more sustainable alternatives. Microbial biocontrol agents, such as microorganisms with antifungal activity, have emerged as a promising alternative management strategy, but their commercial use has been limited by poor establishment and spread on crops. This study presents a novel system to overcome these challenges. The biocontrol agent Lactiplantibacillus plantarum AMBP214 was spray-dried and successfully dispersed to strawberry flowers via bumblebees. This is the first report of combining spray-dried, non-spore-forming bacteria with pollinator-dispersal, which scored better than the state-of-the-art in terms of dispersal to the plant (CFU/flower), and resuscitation of the biocontrol agent. Therefore, this new entomovectoring system holds great promise for the use of biocontrol agents for disease management in agriculture.
Collapse
Affiliation(s)
- Jari Temmermans
- Department of Bioscience Engineering, Research Group Environmental Ecology and Applied Microbiology, Antwerp University, Antwerp, Belgium
| | - Marie Legein
- Department of Bioscience Engineering, Research Group Environmental Ecology and Applied Microbiology, Antwerp University, Antwerp, Belgium
- Department of Integrative Biology, University of California, Berkeley, California, USA
| | - Yijie Zhao
- Laboratory of Plant Health and Protection, Department of Biosystems, KU Leuven, Leuven, Belgium
- KU Leuven Plant Institute, Leuven, Belgium
| | - Filip Kiekens
- Laboratory of Pharmaceutical Technology and Biopharmacy, Department of Pharmaceutical Sciences, Antwerp University, Wilrijk, Belgium
| | - Guy Smagghe
- Laboratory of Agrozoology, Department of Plants and Crops, Faculty of Bioscience Engineering, Ghent University, Ghent, Belgium
| | - Barbara de Coninck
- Laboratory of Plant Health and Protection, Department of Biosystems, KU Leuven, Leuven, Belgium
- KU Leuven Plant Institute, Leuven, Belgium
| | - Sarah Lebeer
- Department of Bioscience Engineering, Research Group Environmental Ecology and Applied Microbiology, Antwerp University, Antwerp, Belgium
| |
Collapse
|
17
|
Suissa R, Olender T, Malitsky S, Golani O, Turjeman S, Koren O, Meijler MM, Kolodkin-Gal I. Metabolic inputs in the probiotic bacterium Lacticaseibacillus rhamnosus contribute to cell-wall remodeling and increased fitness. NPJ Biofilms Microbiomes 2023; 9:71. [PMID: 37752249 PMCID: PMC10522624 DOI: 10.1038/s41522-023-00431-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 08/24/2023] [Indexed: 09/28/2023] Open
Abstract
Lacticaseibacillus rhamnosus GG (LGG) is a Gram-positive beneficial bacterium that resides in the human intestinal tract and belongs to the family of lactic acid bacteria (LAB). This bacterium is a widely used probiotic and was suggested to provide numerous benefits for human health. However, as in most LAB strains, the molecular mechanisms that mediate the competitiveness of probiotics under different diets remain unknown. Fermentation is a fundamental process in LAB, allowing the oxidation of simple carbohydrates (e.g., glucose, mannose) for energy production under oxygen limitation, as in the human gut. Our results indicate that fermentation reshapes the metabolome, volatilome, and proteome architecture of LGG. Furthermore, fermentation alters cell envelope remodeling and peptidoglycan biosynthesis, which leads to altered cell wall thickness, aggregation properties, and cell wall composition. In addition, fermentable sugars induced the secretion of known and novel metabolites and proteins targeting the enteric pathogens Enterococcus faecalis and Salmonella enterica Serovar Typhimurium. Overall, our results link simple carbohydrates with cell wall remodeling, aggregation to host tissues, and biofilm formation in probiotic strains and connect them with the production of broad-spectrum antimicrobial effectors.
Collapse
Affiliation(s)
- Ronit Suissa
- Department of Chemistry, Ben-Gurion University of the Negev, Be'er Sheva, Israel
| | - Tsviya Olender
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Sergey Malitsky
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Ofra Golani
- Life Science Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Sondra Turjeman
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel.
| | - Michael M Meijler
- Department of Chemistry, Ben-Gurion University of the Negev, Be'er Sheva, Israel.
| | - Ilana Kolodkin-Gal
- Department of Plant Pathology and Microbiology, Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel.
- The Scojen Institute for Synthetic Biology, Reichman University, Herzliya, Israel.
| |
Collapse
|
18
|
Wang J, Chen Y, Li M, Xia S, Zhao K, Fan H, Ni J, Sun W, Jia X, Lai S. The effects of differential feeding on ileum development, digestive ability and health status of newborn calves. Front Vet Sci 2023; 10:1255122. [PMID: 37745216 PMCID: PMC10514501 DOI: 10.3389/fvets.2023.1255122] [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: 07/08/2023] [Accepted: 08/29/2023] [Indexed: 09/26/2023] Open
Abstract
Pre-weaning is the most important period for the growth and development of calves. Intestinal morphology, microbial community and immunity are initially constructed at this stage, and even have a lifelong impact on calves. Early feeding patterns have a significant impact on gastrointestinal development and microbial communities. This study mainly analyzed the effects of three feeding methods on the gastrointestinal development of calves, and provided a theoretical basis for further improving the feeding mode of calves. it is very important to develop a suitable feeding mode. In this study, we selected nine newborn healthy Holstein bull calves were randomly selected and divided into three groups (n = 3), which were fed with starter + hay + milk (SH group), starter + milk (SF group), total mixed ration + milk (TMR group). After 80 days of feeding Feeding to 80 days of age after, the ileum contents and blood samples were collected, and the differences were compared and analyzed by metagenomic analysis and serum metabolomics analysis. Results show that compared with the other two groups, the intestinal epithelium of the SH group was more complete and the goblet cells developed better. The feeding method of SH group was more conducive to the development of calves, with higher daily gain and no pathological inflammatory reaction. The intestinal microbial community was more conducive to digestion and absorption, and the immunity was stronger. These findings are helpful for us to explore better calf feeding patterns. In the next step, we will set up more biological replicates to study the deep-seated reasons for the differences in the development of pre-weaning calves. At the same time, the new discoveries of neuro microbiology broaden our horizons and are the focus of our future attention.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Songjia Lai
- College of Animal Science and Technology, Sichuan Agricultural University, Chengdu, China
| |
Collapse
|
19
|
Abstract
It has been widely appreciated that numerous bacterial species express chitinases for the purpose of degrading environmental chitin. However, chitinases and chitin-binding proteins are also expressed by pathogenic bacterial species during infection even though mammals do not produce chitin. Alternative molecular targets are therefore likely present within the host. Here, we will describe our current understanding of chitinase/chitin-binding proteins as virulence factors that promote bacterial colonization and infection. The targets of these chitinases in the host have been shown to include immune system components, mucins, and surface glycans. Bacterial chitinases have also been shown to interact with other microorganisms, targeting the peptidoglycan or chitin in the bacterial and fungal cell wall, respectively. This review highlights that even though the name "chitinase" implies activity toward chitin, chitinases can have a wide diversity of targets, including ones relevant to host infection. Chitinases may therefore be useful as a target of future anti-infective therapeutics.
Collapse
Affiliation(s)
- Jason R. Devlin
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, USA
| | - Judith Behnsen
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, Illinois, USA
| |
Collapse
|
20
|
MacAlpine J, Robbins N, Cowen LE. Bacterial-fungal interactions and their impact on microbial pathogenesis. Mol Ecol 2023; 32:2565-2581. [PMID: 35231147 PMCID: PMC11032213 DOI: 10.1111/mec.16411] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 01/14/2022] [Accepted: 02/18/2022] [Indexed: 11/27/2022]
Abstract
Microbial communities of the human microbiota exhibit diverse effects on human health and disease. Microbial homeostasis is important for normal physiological functions and changes to the microbiota are associated with many human diseases including diabetes, cancer, and colitis. In addition, there are many microorganisms that are either commensal or acquired from environmental reservoirs that can cause diverse pathologies. Importantly, the balance between health and disease is intricately connected to how members of the microbiota interact and affect one another's growth and pathogenicity. However, the mechanisms that govern these interactions are only beginning to be understood. In this review, we outline bacterial-fungal interactions in the human body, including examining the mechanisms by which bacteria govern fungal growth and virulence, as well as how fungi regulate bacterial pathogenesis. We summarize advances in the understanding of chemical, physical, and protein-based interactions, and their role in exacerbating or impeding human disease. We focus on the three fungal species responsible for the majority of systemic fungal infections in humans: Candida albicans, Cryptococcus neoformans, and Aspergillus fumigatus. We conclude by summarizing recent studies that have mined microbes for novel antimicrobials and antivirulence factors, highlighting the potential of the human microbiota as a rich resource for small molecule discovery.
Collapse
Affiliation(s)
- Jessie MacAlpine
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Nicole Robbins
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| | - Leah E. Cowen
- Department of Molecular Genetics, University of Toronto, Toronto, Ontario, M5G 1M1, Canada
| |
Collapse
|
21
|
Das S, Bhattacharjee MJ, Mukherjee AK, Khan MR. Recent advances in understanding of multifaceted changes in the vaginal microenvironment: implications in vaginal health and therapeutics. Crit Rev Microbiol 2023; 49:256-282. [PMID: 35312419 DOI: 10.1080/1040841x.2022.2049696] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The vagina endures multifaceted changes from neonatal to menopausal phases due to hormonal flux, metabolite deposition, and microbial colonization. These features have important implications in women's health. Several pre-factors show dynamic characteristics according to the phases that shift the vaginal microbiota from anaerobes to aerobes which is a hallmark of healthy vaginal environment. These factors include oestrogen levels, glycogen deposition, and vaginal microstructure. In the adult phase, Lactobacillus is highly dominant and regulates pH, adherence, aggregation, immune modulation, synthesis of bacteriocins, and biosurfactants (BSs) which are antagonistic to pathogens. Maternal factors are protective by favouring the colonization of lactobacilli in the vagina in the neonatal phase, which diminishes with age. The dominance of lactobacilli and dysbiosis in the adult phase depends on intrinsic and extrinsic factors in women, which vary between ethnicities. Recent developments in probiotics used against vaginal microbiome dysbiosis have shown great promise in restoring the normal microbiota including preventing the loss of beneficial bacteria. However, further in-depth studies are warranted to ensure long-term protection by probiotics. This review highlights various aspects of the vaginal microenvironment in different phases of growth and diverse ethnicities. Furthermore, it discusses future trends for formulating more effective population-specific probiotics and implications of paraprobiotics and postbiotics as effective therapeutics.
Collapse
Affiliation(s)
- Sushmita Das
- Division of Life Science, Institute of Advanced Study in Science and Technology, Guwahati, India
| | | | - Ashis K Mukherjee
- Division of Life Science, Institute of Advanced Study in Science and Technology, Guwahati, India.,Department of Molecular Biology and Biotechnology, Tezpur University, Tezpur, India
| | - Mojibur Rohman Khan
- Division of Life Science, Institute of Advanced Study in Science and Technology, Guwahati, India
| |
Collapse
|
22
|
Guan C, Zhang W, Su J, Li F, Chen D, Chen X, Huang Y, Gu R, Zhang C. Antibacterial and antibiofilm potential of Lacticaseibacillus rhamnosus YT and its cell-surface extract. BMC Microbiol 2023; 23:12. [PMID: 36635630 PMCID: PMC9835366 DOI: 10.1186/s12866-022-02751-3] [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: 10/20/2022] [Accepted: 12/28/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Foodborne pathogens and spoilage bacteria survived in the biofilm pose a serious threat to food safety and human health. It is urgent to find safe and effective methods to control the planktonic bacteria as well as the biofilm formation. Substances with antibacterial and antibiofilm activity found in lactic acid bacteria were mainly metabolites secreted in the cell-free supernatant. Previously, Lacticaseibacillus rhamnosus YT was isolated because its cell pellets displayed distinguished antibacterial activity under neutral conditions. This study aimed to investigate the antibacterial and antibiofilm properties of the L. rhamnosus YT cells and its crude cell-surface extract. RESULTS The antibacterial activity of the L. rhamnosus YT cells constantly increased with cells growth and reached the peak value after the cells grew into stationary phase. After cocultivation with the L. rhamnosus YT cells, the biofilm formation of B. subtilis and S. enterica was reduced. The antibacterial activity of the L. rhamnosus YT cells was varied along with various culture conditions (carbon sources, nitrogen sources, medium pH and cultural temperatures) and the antibacterial intensity (antibacterial activity per cell) was disproportional to the biomass. Furthermore, the cell-surface extract was isolated and displayed broad antimicrobial spectrum with a bacteriostatic mode of action. The antibiofilm activity of the extract was concentration-dependent. In addition, the extract was stable to physicochemical treatments (heat, pH and protease). The extract performed favorable emulsifying property which could reduce the water surface tension from 72.708 mN/m to 51.011 mN/m and the critical micelle concentration (CMC) value was 6.88 mg/mL. Besides, the extract was also able to emulsify hydrocarbon substrates with the emulsification, index (E24) ranged from 38.55% (for n-hexane) to 53.78% (for xylene). The E24 for xylene/extract emulsion was merely decreased by 5.77% after standing for 120 h. The main components of the extract were polysaccharide (684.63 μg/mL) and protein (120.79 μg/mL). CONCLUSION The properties of the extract indicated that it might be a kind of biosurfactant. These data suggested that L. rhamnosus YT and the cell-surface extract could be used as an alternative antimicrobial and antibiofilm agent against foodborne pathogens and spoilage bacteria in food industry.
Collapse
Affiliation(s)
- Chengran Guan
- grid.268415.cKey Lab of Dairy Biotechnology and Safety Control, College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu China
| | - Wenjuan Zhang
- grid.268415.cKey Lab of Dairy Biotechnology and Safety Control, College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu China
| | - Jianbo Su
- grid.268415.cKey Lab of Dairy Biotechnology and Safety Control, College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu China
| | - Feng Li
- grid.268415.cKey Lab of Dairy Biotechnology and Safety Control, College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu China
| | - Dawei Chen
- grid.268415.cKey Lab of Dairy Biotechnology and Safety Control, College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu China
| | - Xia Chen
- grid.268415.cKey Lab of Dairy Biotechnology and Safety Control, College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu China
| | - Yujun Huang
- grid.268415.cKey Lab of Dairy Biotechnology and Safety Control, College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu China
| | - Ruixia Gu
- grid.268415.cKey Lab of Dairy Biotechnology and Safety Control, College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu China
| | - Chenchen Zhang
- grid.268415.cKey Lab of Dairy Biotechnology and Safety Control, College of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu China
| |
Collapse
|
23
|
van Thiel I, de Jonge W, van den Wijngaard R. Fungal feelings in the irritable bowel syndrome: the intestinal mycobiome and abdominal pain. Gut Microbes 2023; 15:2168992. [PMID: 36723172 PMCID: PMC9897793 DOI: 10.1080/19490976.2023.2168992] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Although the gut microbiota consists of bacteria, viruses, and fungi, most publications addressing the microbiota-gut-brain axis in irritable bowel syndrome (IBS) have a sole focus on bacteria. This may relate to the relatively low presence of fungi and viruses as compared to bacteria. Yet, in the field of inflammatory bowel disease research, the publication of several papers addressing the role of the intestinal mycobiome now suggested that these low numbers do not necessarily translate to irrelevance. In this review, we discuss the available clinical and preclinical IBS mycobiome data, and speculate how these recent findings may relate to earlier observations in IBS. By surveying literature from the broader mycobiome research field, we identified questions open to future IBS-oriented investigations.
Collapse
Affiliation(s)
- Iam van Thiel
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,Amsterdam UMC, University of Amsterdam, Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands
| | - Wj de Jonge
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,Amsterdam UMC, University of Amsterdam, Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands,Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands,Department of General, Visceral-, Thoracic and Vascular Surgery, University Hospital Bonn, Bonn, Germany
| | - Rm van den Wijngaard
- Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Amsterdam, The Netherlands,Amsterdam UMC, University of Amsterdam, Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, The Netherlands,Department of Gastroenterology and Hepatology, Amsterdam University Medical Centers, Amsterdam, The Netherlands,CONTACT RM van den Wijngaard Tytgat Institute for Liver and Intestinal Research, Amsterdam University Medical Centers, Meibergdreef 69-71, Amsterdam1105 BK, The Netherlands
| |
Collapse
|
24
|
Bacillus Metabolites: Compounds, Identification and Anti-Candida albicans Mechanisms. MICROBIOLOGY RESEARCH 2022. [DOI: 10.3390/microbiolres13040070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022] Open
Abstract
Candida albicans seriously threatens human health, especially for immunosuppressed groups. The antifungal agents mainly include azoles, polyenes and echinocandins. However, the few types of existing antifungal drugs and their resistance make it necessary to develop new antifungal drugs. Bacillus and its metabolites has antifungal activity against pathogenic fungi. This review introduces the application of Bacillus metabolites in the control of C. albicans in recent years. Firstly, several compounds produced by Bacillus spp. are listed. Then the isolation and identification techniques of Bacillus metabolites in recent years are described, including high-precision separation technology and omics technology for the separation of similar components of Bacillus metabolites. The mechanisms of Bacillus metabolites against C. albicans are distinguished from the inhibition of pathogenic fungi and inhibition of the fungal virulence factors. The purpose of this review is to systematically summarize the recent studies on the inhibition of pathogenic fungi by Bacillus metabolites. The review is expected to become the reference for the control of pathogenic fungi such as C. albicans and the application of Bacillus metabolites in the future.
Collapse
|
25
|
Menichini D, Chiossi G, Monari F, De Seta F, Facchinetti F. Supplementation of Probiotics in Pregnant Women Targeting Group B Streptococcus Colonization: A Systematic Review and Meta-Analysis. Nutrients 2022; 14:nu14214520. [PMID: 36364782 PMCID: PMC9657808 DOI: 10.3390/nu14214520] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/14/2022] [Accepted: 10/24/2022] [Indexed: 11/26/2022] Open
Abstract
This systematic review and meta-analysis aimed to determine if probiotic supplementation in pregnancy reduced maternal Group B streptococcus (GBS) recto-vaginal colonization in pregnant women at 35–37 weeks of gestation. Electronic databases (i.e., PubMed, MEDLINE, ClinicalTrials.gov, ScienceDirect, and the Cochrane Library) were searched from inception up to February 2022. We included RCTs assessing the effects of probiotic supplementation in pregnancy on GBS recto-vaginal colonization. The primary outcome was GBS-positive recto-vaginal cultures performed at 35–37 weeks of gestation. Secondarily, we evaluated obstetric and short-term neonatal outcomes. A total of 132 publications were identified; 9 full-length articles were reviewed to finally include 5 studies. Probiotic supplementation reduced vaginal GBS colonization: the GBS positive culture rate was estimated at 31.9% (96/301) in the intervention group compared to 38.6% (109/282) in the control group (OR = 0.62, 95% CI 0.40–0.94, I2 4.8%, p = 0.38). The treatment started after 30 weeks of gestation and was more effective in reducing GBS colonization (OR 0.41, 95% CI 0.21–0.78, I2 0%, p = 0.55). Probiotic administration during pregnancy, namely in the third trimester, was associated with a reduced GBS recto-vaginal colonization at 35–37 weeks and a safe perinatal profile. Whether this new strategy could reduce the exposition of pregnant women to significant doses of antibiotics in labor needs to be evaluated in other trials.
Collapse
Affiliation(s)
- Daniela Menichini
- Department of Biomedical, Metabolic and Neural Sciences, International Doctorate School in Clinical and Experimental Medicine, University of Modena and Reggio Emilia, 41124 Modena, Italy
- Unit of Obstetrics and Gynecology, Mother-Infant Department, University of Modena and Reggio Emilia, 41124 Modena, Italy
- Correspondence: ; Tel.: +39-0594225826
| | - Giuseppe Chiossi
- Unit of Obstetrics and Gynecology, Mother-Infant Department, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Francesca Monari
- Unit of Obstetrics and Gynecology, Mother-Infant Department, University of Modena and Reggio Emilia, 41124 Modena, Italy
| | - Francesco De Seta
- Institute for Maternal and Child Health “IRCCS Burlo Garofolo”, 34137 Trieste, Italy
| | - Fabio Facchinetti
- Unit of Obstetrics and Gynecology, Mother-Infant Department, University of Modena and Reggio Emilia, 41124 Modena, Italy
| |
Collapse
|
26
|
Lactobacilli, a Weapon to Counteract Pathogens through the Inhibition of Their Virulence Factors. J Bacteriol 2022; 204:e0027222. [PMID: 36286515 PMCID: PMC9664955 DOI: 10.1128/jb.00272-22] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To date, several studies have reported an alarming increase in pathogen resistance to current antibiotic therapies and treatments. Therefore, the search for effective alternatives to counter their spread and the onset of infections is becoming increasingly important.
Collapse
|
27
|
Gut Non-Bacterial Microbiota: Emerging Link to Irritable Bowel Syndrome. Toxins (Basel) 2022; 14:toxins14090596. [PMID: 36136534 PMCID: PMC9503233 DOI: 10.3390/toxins14090596] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Revised: 08/18/2022] [Accepted: 08/25/2022] [Indexed: 11/20/2022] Open
Abstract
As a common functional gastrointestinal disorder, irritable bowel syndrome (IBS) significantly affects personal health and imposes a substantial economic burden on society, but the current understanding of its occurrence and treatment is still inadequate. Emerging evidence suggests that IBS is associated with gut microbial dysbiosis, but most studies focus on the bacteria and neglect other communities of the microbiota, including fungi, viruses, archaea, and other parasitic microorganisms. This review summarizes the latest findings that link the nonbacterial microbiota with IBS. IBS patients show less fungal and viral diversity but some alterations in mycobiome, virome, and archaeome, such as an increased abundance of Candida albicans. Moreover, fungi and methanogens can aid in diagnosis. Fungi are related to distinct IBS symptoms and induce immune responses, intestinal barrier disruption, and visceral hypersensitivity via specific receptors, cells, and metabolites. Novel therapeutic methods for IBS include fungicides, inhibitors targeting fungal pathogenic pathways, probiotic fungi, prebiotics, and fecal microbiota transplantation. Additionally, viruses, methanogens, and parasitic microorganisms are also involved in the pathophysiology and treatment. Therefore, the gut nonbacterial microbiota is involved in the pathogenesis of IBS, which provides a novel perspective on the noninvasive diagnosis and precise treatment of this disease.
Collapse
|
28
|
Anticandidal and Antibiofilm Effect of Synbiotics including Probiotics and Inulin-Type Fructans. Antibiotics (Basel) 2022; 11:antibiotics11081135. [PMID: 36010004 PMCID: PMC9405293 DOI: 10.3390/antibiotics11081135] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/09/2022] [Accepted: 08/17/2022] [Indexed: 11/17/2022] Open
Abstract
Background: There is great interest in the search for new alternatives to antimicrobial drugs, and the use of synbiotics is a promising approach to this problem. This study evaluated the growth inhibition and antibiofilm activity of the short-chain fatty acids produced by Lacticaseibacillus rhamnosus and Pediococcus acidilactici in combination with inulin-type fructans against Candida albicans. Methods: The growth inhibition of Candida was evaluated using microdilution analysis in 96-well microtiter plates; different concentrations of cell-free supernatants of Lacticaseibacillus rhamnosus and Pediococcus acidilactici were exposed to Candida albicans. The antibiofilm assessment was carried out using the crystal violet staining assay. The short-chain fatty acids were analyzed by gas chromatography. Results: The clinically isolated Candida albicans interacted with supernatants from Lacticaseibacillus rhamnosus and Pediococcus acidilactici and showed significant growth inhibition and antibiofilm formation versus the controls. Lactate and acetic acid were elevated in the supernatants. The results suggest that the supernatants obtained from the synbiotic combinations of Lacticaseibacillus rhamnosus and Pediococcus acidilactici with inulin-type fructans can inhibit the growth and biofilm formation against a clinically isolated Candida albicans strain. Conclusions: These results suggest that synbiotic formulations could be a promising alternative to antifungal drugs in candidiasis therapy.
Collapse
|
29
|
Rodríguez-Arias RJ, Guachi-Álvarez BO, Montalvo-Vivero DE, Machado A. Lactobacilli displacement and Candida albicans inhibition on initial adhesion assays: a probiotic analysis. BMC Res Notes 2022; 15:239. [PMID: 35799214 PMCID: PMC9264498 DOI: 10.1186/s13104-022-06114-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 06/15/2022] [Indexed: 11/10/2022] Open
Abstract
Objective This study evaluates the probiotic activity of three vaginal Lactobacillus gasseri (H59.2, IMAUFB014, and JCM1131) and one non-vaginal L. plantarum ATCC14917 against three Candida albicans (ATCC10231, candidiasis, and healthy vaginal microbiota). Displacement of lactobacilli and adhesion inhibition of C. albicans were evaluated on an abiotic surface through adhesion assays with different experimental settings (ES) through low (1.0E + 03 CFU/ml) and high (1.00E + 09 CFU/ml) levels of colonization. ES simulated dysbiosis (ES1 and ES4), candidiasis (ES2), and healthy vaginal microbiota (ES3). Results At ES2 and ES3, L. gasseri H59.2 showed discrepant inhibition values among C. albicans isolates (ES2: P = 0.008, ES3: P = 0.030; two‐way ANOVA). L. plantarum was only displaced by 23%, 31%, 54%, and 94% against low and high levels of C. albicans ATCC10231. L. plantarum was less displaced, when compared to L. gasseri strains (ES1: 61–84%, ES2: 82–96%, ES3: 83–95%, and ES4: 73–97%), showing multiple statistical differences (ES1: P = < 0.001, ES2: P = 0.003, and ES3: P = < 0.001; two‐way ANOVA). L. plantarum also showed a superior inhibition of C. albicans ATCC10231 in ES1 (81%) and ES2 (58%) when compared to L. gasseri strains (ES1: 27–73%, P < 0.001; and ES2:1–49%, P < 0.001; two‐way ANOVA). Supplementary Information The online version contains supplementary material available at 10.1186/s13104-022-06114-z.
Collapse
Affiliation(s)
- Robert Josue Rodríguez-Arias
- Colegio de Ciencias Biológicas y Ambientales COCIBA, Instituto de Microbiología, Laboratorio de Bacteriología, Calle Diego de Robles y Pampite, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Bryan Omar Guachi-Álvarez
- Colegio de Ciencias Biológicas y Ambientales COCIBA, Instituto de Microbiología, Laboratorio de Bacteriología, Calle Diego de Robles y Pampite, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - Dominique Esther Montalvo-Vivero
- Colegio de Ciencias Biológicas y Ambientales COCIBA, Instituto de Microbiología, Laboratorio de Bacteriología, Calle Diego de Robles y Pampite, Universidad San Francisco de Quito USFQ, Quito, Ecuador
| | - António Machado
- Colegio de Ciencias Biológicas y Ambientales COCIBA, Instituto de Microbiología, Laboratorio de Bacteriología, Calle Diego de Robles y Pampite, Universidad San Francisco de Quito USFQ, Quito, Ecuador.
| |
Collapse
|
30
|
Lactobacillus rhamnosus colonisation antagonizes Candida albicans by forcing metabolic adaptations that compromise pathogenicity. Nat Commun 2022; 13:3192. [PMID: 35680868 PMCID: PMC9184479 DOI: 10.1038/s41467-022-30661-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 05/12/2022] [Indexed: 01/09/2023] Open
Abstract
Intestinal microbiota dysbiosis can initiate overgrowth of commensal Candida species - a major predisposing factor for disseminated candidiasis. Commensal bacteria such as Lactobacillus rhamnosus can antagonize Candida albicans pathogenicity. Here, we investigate the interplay between C. albicans, L. rhamnosus, and intestinal epithelial cells by integrating transcriptional and metabolic profiling, and reverse genetics. Untargeted metabolomics and in silico modelling indicate that intestinal epithelial cells foster bacterial growth metabolically, leading to bacterial production of antivirulence compounds. In addition, bacterial growth modifies the metabolic environment, including removal of C. albicans' favoured nutrient sources. This is accompanied by transcriptional and metabolic changes in C. albicans, including altered expression of virulence-related genes. Our results indicate that intestinal colonization with bacteria can antagonize C. albicans by reshaping the metabolic environment, forcing metabolic adaptations that reduce fungal pathogenicity.
Collapse
|
31
|
Kinetic, Thermodynamic and Bio-applicable Studies on Aspergillus niger Mk981235 Chitinase. Catal Letters 2022. [DOI: 10.1007/s10562-022-04045-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
AbstractChitinases have many applications in food, agricultural, medical, and pharmaceutical fields. This study succeeded in investigating Aspergillus niger MK981235 chitinase in the spot of its physiochemical, kinetic, thermodynamic, and application. The optimum temperature, pH and p-nitrophenyl-β-d-N-acetyl glucosaminide (PNP-β-GlcNAc) concentration to obtain the highest chitinase activity of 2334.79 U ml−1 were at 60 °C, 5 and 0.25%, respectively. The kinetic parameters, including Km and Vmax were determined to be 0.78 mg ml−1 and 2222.22 µmol ml−1 min−1, respectively. Furthermore, the thermodynamic parameters T1/2, D-values, ΔH, ΔG and ΔS at 40, 50 and 60 °C were determined to be (864.10, 349.45, 222.34 min), (2870.99, 1161.07, 738.74 min), (126.40, 126.36, 126.32 kJ mol−1), (101.59, 100.62, 100.86 kJ mol−1), (74.50, 76.17, 47.24 J mol−1 K−1), respectively. A. niger chitinase showed, insecticidal activity on Galleria mellonella by feeding and spraying treatments (72 and 52%, respectively), anti-lytic activity against Candida albicans, and effectiveness in improving the dye removal in the presence of crab shell powder as bio-absorbant. A. niger chitinase can be used in the pharmaceutical field for the bio-control of diseases caused by C. albicans and for the pretreatment of wastewater from the textile industry.
Graphical Abstract
Collapse
|
32
|
Balakrishnan SN, Yamang H, Lorenz MC, Chew SY, Than LTL. Role of Vaginal Mucosa, Host Immunity and Microbiota in Vulvovaginal Candidiasis. Pathogens 2022; 11:pathogens11060618. [PMID: 35745472 PMCID: PMC9230866 DOI: 10.3390/pathogens11060618] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022] Open
Abstract
Vulvovaginal candidiasis (VVC) is a prevalent gynaecological disease characterised by vaginal wall inflammation that is caused by Candida species. VVC impacts almost three-quarters of all women throughout their reproductive years. As the vaginal mucosa is the first point of contact with microbes, vaginal epithelial cells are the first line of defence against opportunistic Candida infection by providing a physical barrier and mounting immunological responses. The mechanisms of defence against this infection are displayed through the rapid shedding of epithelial cells, the presence of pattern recognition receptors, and the release of inflammatory cytokines. The bacterial microbiota within the mucosal layer presents another form of defence mechanism within the vagina through acidic pH regulation, the release of antifungal peptides and physiological control against dysbiosis. The significant role of the microbiota in maintaining vaginal health promotes its application as one of the potential treatment modalities against VVC with the hope of alleviating the burden of VVC, especially the recurrent disease. This review discusses and summarises current progress in understanding the role of vaginal mucosa and host immunity upon infection, together with the function of vaginal microbiota in VVC.
Collapse
Affiliation(s)
- Subatrra Nair Balakrishnan
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang 43300, Selangor, Malaysia; (S.N.B.); (H.Y.)
| | - Haizat Yamang
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang 43300, Selangor, Malaysia; (S.N.B.); (H.Y.)
| | - Michael C. Lorenz
- Department of Microbiology and Molecular Genetics, University of Texas McGovern Medical School, Houston, TX 77030, USA;
| | - Shu Yih Chew
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang 43300, Selangor, Malaysia; (S.N.B.); (H.Y.)
- Correspondence: (S.Y.C.); (L.T.L.T.)
| | - Leslie Thian Lung Than
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang 43300, Selangor, Malaysia; (S.N.B.); (H.Y.)
- Correspondence: (S.Y.C.); (L.T.L.T.)
| |
Collapse
|
33
|
Archambault LS, Dongari-Bagtzoglou A. Probiotics for Oral Candidiasis: Critical Appraisal of the Evidence and a Path Forward. FRONTIERS IN ORAL HEALTH 2022; 3:880746. [PMID: 35495563 PMCID: PMC9046664 DOI: 10.3389/froh.2022.880746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 03/23/2022] [Indexed: 11/29/2022] Open
Abstract
Oropharyngeal Candidiasis (OPC) is a mucosal fungal infection that is prevalent among patients with compromised immunity. The success of probiotics in treating chronic diseases with a microbial etiology component at other mucosal sites (i.e., gastro-intestinal, genitourinary and alveolar mucosae) has inspired research into the use of probiotics in the treatment of OPC. A growing body of research in vitro and in animal models indicates that some probiotic species and strains have inhibitory activities against Candida albicans growth, morphological switching, and biofilm formation. However, recent review and meta-analysis studies reveal a dearth of human randomized, controlled clinical trials on the efficacy of probiotics to treat or prevent OPC, while the majority of these have not based their selection of probiotic strains or the type of administration on sound pre-clinical evidence. In this mini-review, we assess the state of the field, outline some of the difficulties in translating lab results to clinical efficacy, and make recommendations for future research needed in order to move the field forward.
Collapse
Affiliation(s)
- Linda S. Archambault
- Department of Craniofacial Sciences, University of Connecticut Health Center, Farmington, CT, United States
- Center for Quantitative Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Anna Dongari-Bagtzoglou
- Department of Craniofacial Sciences, University of Connecticut Health Center, Farmington, CT, United States
- *Correspondence: Anna Dongari-Bagtzoglou
| |
Collapse
|
34
|
Vazquez-Munoz R, Thompson A, Russell JT, Sobue T, Zhou Y, Dongari-Bagtzoglou A. Insights From the Lactobacillus johnsonii Genome Suggest the Production of Metabolites With Antibiofilm Activity Against the Pathobiont Candida albicans. Front Microbiol 2022; 13:853762. [PMID: 35330775 PMCID: PMC8940163 DOI: 10.3389/fmicb.2022.853762] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 02/14/2022] [Indexed: 01/08/2023] Open
Abstract
Lactobacillus johnsonii is a probiotic bacterial species with broad antimicrobial properties; however, its antimicrobial activities against the pathobiont Candida albicans are underexplored. The aim of this study was to study the interactions of L. johnsonii with C. albicans and explore mechanisms of bacterial anti-fungal activities based on bacterial genomic characterization coupled with experimental data. We isolated an L. johnsonii strain (MT4) from the oral cavity of mice and characterized its effect on C. albicans growth in the planktonic and biofilm states. We also identified key genetic and phenotypic traits that may be associated with a growth inhibitory activity exhibited against C. albicans. We found that L. johnsonii MT4 displays pH-dependent and pH-independent antagonistic interactions against C. albicans, resulting in inhibition of C. albicans planktonic growth and biofilm formation. This antagonism is influenced by nutrient availability and the production of soluble metabolites with anticandidal activity.
Collapse
Affiliation(s)
- Roberto Vazquez-Munoz
- Department of Periodontology, University of Connecticut Health Center, Farmington, CT, United States
| | - Angela Thompson
- Department of Periodontology, University of Connecticut Health Center, Farmington, CT, United States
| | - Jordan T Russell
- Department of Psychiatry/Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Takanori Sobue
- Department of Periodontology, University of Connecticut Health Center, Farmington, CT, United States
| | - Yanjiao Zhou
- Department of Psychiatry/Medicine, University of Connecticut Health Center, Farmington, CT, United States
| | - Anna Dongari-Bagtzoglou
- Department of Periodontology, University of Connecticut Health Center, Farmington, CT, United States
| |
Collapse
|
35
|
Ventin-Holmberg R, Saqib S, Korpela K, Nikkonen A, Peltola V, Salonen A, de Vos WM, Kolho KL. The Effect of Antibiotics on the Infant Gut Fungal Microbiota. J Fungi (Basel) 2022; 8:328. [PMID: 35448562 PMCID: PMC9032081 DOI: 10.3390/jof8040328] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 03/15/2022] [Accepted: 03/18/2022] [Indexed: 02/05/2023] Open
Abstract
Antibiotics are commonly used drugs in infants, causing disruptions in the developing gut microbiota with possible detrimental long-term effects such as chronic inflammatory diseases. The focus has been on bacteria, but research shows that fungi might have an important role as well. There are only a few studies on the infant gut fungal microbiota, the mycobiota, in relation to antibiotic treatment. Here, the aim was to investigate the impact of antibiotics on the infant gut mycobiota, and the interkingdom associations between bacteria and fungi. We had 37 antibiotic-naïve patients suffering from respiratory syncytial virus, of which 21 received one to four courses of antibiotics due to complications, and 16 remained antibiotic-naïve throughout the study. Fecal samples were collected before, during and after antibiotic treatment with a follow-up period of up to 9.5 months. The gut mycobiota was studied by Illumina MiSeq sequencing of the ITS1 region. We found that antibiotic use affected the gut mycobiota, most prominently seen as a higher relative abundance of Candida (p < 0.001), and a higher fungal diversity (p = 0.005−0.04) and richness (p = 0.03) in the antibiotic-treated infants compared to the antibiotic-naïve ones at multiple timepoints. This indicates that the gut mycobiota could contribute to the long-term consequences of antibiotic treatments.
Collapse
Affiliation(s)
- Rebecka Ventin-Holmberg
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (R.V.-H.); (S.S.); (K.K.); (A.S.); (W.M.d.V.)
- Folkhälsan Research Center, 00250 Helsinki, Finland
| | - Schahzad Saqib
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (R.V.-H.); (S.S.); (K.K.); (A.S.); (W.M.d.V.)
| | - Katri Korpela
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (R.V.-H.); (S.S.); (K.K.); (A.S.); (W.M.d.V.)
| | - Anne Nikkonen
- Children’s Hospital, Helsinki University, 00029 Helsinki, Finland;
| | - Ville Peltola
- Department of Paediatrics and Adolescent Medicine, Turku University Hospital, University of Turku, 20014 Turku, Finland;
| | - Anne Salonen
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (R.V.-H.); (S.S.); (K.K.); (A.S.); (W.M.d.V.)
| | - Willem M. de Vos
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (R.V.-H.); (S.S.); (K.K.); (A.S.); (W.M.d.V.)
- Laboratory of Microbiology, Wageningen University, 6708 WE Wageningen, The Netherlands
| | - Kaija-Leena Kolho
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, 00014 Helsinki, Finland; (R.V.-H.); (S.S.); (K.K.); (A.S.); (W.M.d.V.)
- Children’s Hospital, Helsinki University, 00029 Helsinki, Finland;
- Department of Pediatrics, Tampere University, 33520 Tampere, Finland
| |
Collapse
|
36
|
Suissa R, Oved R, Jankelowitz G, Turjeman S, Koren O, Kolodkin-Gal I. Molecular genetics for probiotic engineering: dissecting lactic acid bacteria. Trends Microbiol 2022; 30:293-306. [PMID: 34446338 DOI: 10.1016/j.tim.2021.07.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 07/28/2021] [Accepted: 07/29/2021] [Indexed: 02/08/2023]
Abstract
The composition of the gut microbiome is greatly influenced by nutrition and dietary alterations which can also induce large temporary microbial shifts. However, the molecular mechanisms that promote these changes remain to be determined. Species of the family Lactobacillaceae and Bacillus species are genetically manipulatable bacteria that are naturally found in the human gastrointestinal (GI) tract and are often considered models of beneficial microbiota. Here, we identify specific conserved molecular pathways that play a key role in host colonization by beneficial members of the microbiota. In particular, we highlight three pathways important to the success of lactic acid bacteria (LAB) in the GI tract: glycolysis and fermentation, microbial communication via membrane vesicles, and condition-dependent antibiotic production. We elaborate on how the understanding of these circuits can lead to the development of novel therapeutic approaches to combat GI tract infections.
Collapse
Affiliation(s)
- Ronit Suissa
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | - Rela Oved
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | | - Sondra Turjeman
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefad, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar-Ilan University, Zefad, Israel.
| | - Ilana Kolodkin-Gal
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
37
|
Lebeer S, Oerlemans EFM, Claes I, Henkens T, Delanghe L, Wuyts S, Spacova I, van den Broek MFL, Tuyaerts I, Wittouck S, De Boeck I, Allonsius CN, Kiekens F, Lambert J. Selective targeting of skin pathobionts and inflammation with topically applied lactobacilli. Cell Rep Med 2022; 3:100521. [PMID: 35243421 PMCID: PMC8861818 DOI: 10.1016/j.xcrm.2022.100521] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 10/29/2021] [Accepted: 01/16/2022] [Indexed: 01/04/2023]
Abstract
Tailored skin microbiome modulation approaches with probiotics are highly challenging. Here, we show that lactobacilli are underestimated members of the skin microbiota. We select specific strains of nomadic lactobacilli for their functional applicability on the skin and capacity to inhibit growth and inflammation by skin pathobionts. The strains are formulated as microcapsules for topical formulations and tested in patients with mild-to-moderate acne. The selected lactobacilli are able to reduce inflammatory lesions in a pilot and placebo-controlled study. Daily application for 8 weeks is associated with an in vivo temporary modulation of the microbiome, including a reduction in relative abundance of staphylococci and Cutibacterium acnes, and an increase in lactobacilli. The reduction in inflammatory lesions is still apparent 4 weeks after the topical application of the lactobacilli ended, indicating a possible additional immunomodulatory effect. This study shows that carefully selected and formulated lactobacilli are a viable therapeutic option for common acne lesions.
Collapse
Affiliation(s)
- Sarah Lebeer
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Eline F M Oerlemans
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Ingmar Claes
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Tim Henkens
- University of Antwerp, Department of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Pharmaceutical Technology and Biopharmacy, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Lize Delanghe
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Sander Wuyts
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Irina Spacova
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Marianne F L van den Broek
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Ines Tuyaerts
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Stijn Wittouck
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Ilke De Boeck
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Camille N Allonsius
- University of Antwerp, Department of Bioscience Engineering, Groenenborgerlaan 171, B-2020 Antwerp, Belgium
| | - Filip Kiekens
- University of Antwerp, Department of Pharmaceutical, Biomedical and Veterinary Sciences, Laboratory of Pharmaceutical Technology and Biopharmacy, Universiteitsplein 1, B-2610 Wilrijk, Belgium
| | - Julien Lambert
- University Hospital Antwerp/University of Antwerp, Department of Dermatology and Venereology, Wilrijkstraat 10, 2650 Edegem, Belgium
| |
Collapse
|
38
|
The Interactions among Isolates of Lactiplantibacillus plantarum and Dairy Yeast Contaminants: Towards Biocontrol Applications. FERMENTATION-BASEL 2021. [DOI: 10.3390/fermentation8010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Yeast diversity in the cheese manufacturing process and in the cheeses themselves includes indispensable species for the production of specific cheeses and undesired species that cause cheese defects and spoilage. The control of yeast contaminants is problematic due to limitations in sanitation methods and chemicals used in the food industry. The utilisation of lactic acid bacteria and their antifungal products is intensively studied. Lactiplantibacillus plantarum is one of the most frequently studied species producing a wide spectrum of bioactive by-products. In the present study, twenty strains of L. plantarum from four sources were tested against 25 species of yeast isolated from cheeses, brines, and dairy environments. The functional traits of L. plantarum strains, such as the presence of class 2a bacteriocin and chitinase genes and in vitro production of organic acids, were evaluated. The extracellular production of bioactive peptides and proteins was tested using proteomic methods. Antifungal activity against yeast was screened using in vitro tests. Testing of antifungal activity on artificial media and reconstituted milk showed significant variability within the strains of L. plantarum and its group of origin. Strains from sourdoughs (CCDM 3018, K19-3) and raw cheese (L12, L24, L32) strongly inhibited the highest number of yeast strains on medium with reconstituted milk. These strains showed a consistent spectrum of genes belonging to class 2a bacteriocins, the gene of chitinase and its extracellular product 9 LACO Chitin-binding protein. Strain CCDM 3018 with the spectrum of class 2a bacteriocin gene, chitinase and significant production of lactic acid in all media performed significant antifungal effects in artificial and reconstituted milk-based media.
Collapse
|
39
|
Interplay between Candida albicans and Lactic Acid Bacteria in the Gastrointestinal Tract: Impact on Colonization Resistance, Microbial Carriage, Opportunistic Infection, and Host Immunity. Clin Microbiol Rev 2021; 34:e0032320. [PMID: 34259567 PMCID: PMC8404691 DOI: 10.1128/cmr.00323-20] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Emerging studies have highlighted the disproportionate role of Candida albicans in influencing both early community assembly of the bacterial microbiome and dysbiosis during allergic diseases and intestinal inflammation. Nonpathogenic colonization of the human gastrointestinal (GI) tract by C. albicans is common, and the role of this single fungal species in modulating bacterial community reassembly after broad-spectrum antibiotics can be readily recapitulated in mouse studies. One of the most notable features of C. albicans-associated dysbiotic states is a marked change in the levels of lactic acid bacteria (LAB). C. albicans and LAB share metabolic niches throughout the GI tract, and in vitro studies have identified various interactions between these microbes. The two predominant LAB affected are Lactobacillus species and Enterococcus species. Lactobacilli can antagonize enterococci and C. albicans, while Enterococcus faecalis and C. albicans have been reported to exhibit a mutualistic relationship. E. faecalis and C. albicans are also causative agents of a variety of life-threatening infections, are frequently isolated together from mixed-species infections, and share certain similarities in clinical presentation-most notably their emergence as opportunistic pathogens following disruption of the microbiota. In this review, we discuss and model the mechanisms used by Lactobacillus species, E. faecalis, and C. albicans to modulate each other's growth and virulence in the GI tract. With multidrug-resistant E. faecalis and C. albicans strains becoming increasingly common in hospital settings, examining the interplay between these three microbes may provide novel insights for enhancing the efficacy of existing antimicrobial therapies.
Collapse
|
40
|
Poria V, Rana A, Kumari A, Grewal J, Pranaw K, Singh S. Current Perspectives on Chitinolytic Enzymes and Their Agro-Industrial Applications. BIOLOGY 2021; 10:1319. [PMID: 34943233 PMCID: PMC8698876 DOI: 10.3390/biology10121319] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Revised: 12/03/2021] [Accepted: 12/09/2021] [Indexed: 12/16/2022]
Abstract
Chitinases are a large and diversified category of enzymes that break down chitin, the world's second most prevalent polymer after cellulose. GH18 is the most studied family of chitinases, even though chitinolytic enzymes come from a variety of glycosyl hydrolase (GH) families. Most of the distinct GH families, as well as the unique structural and catalytic features of various chitinolytic enzymes, have been thoroughly explored to demonstrate their use in the development of tailor-made chitinases by protein engineering. Although chitin-degrading enzymes may be found in plants and other organisms, such as arthropods, mollusks, protozoans, and nematodes, microbial chitinases are a promising and sustainable option for industrial production. Despite this, the inducible nature, low titer, high production expenses, and susceptibility to severe environments are barriers to upscaling microbial chitinase production. The goal of this study is to address all of the elements that influence microbial fermentation for chitinase production, as well as the purifying procedures for attaining high-quality yield and purity.
Collapse
Affiliation(s)
- Vikram Poria
- Department of Microbiology, Central University of Haryana, Mahendargarh 123031, India; (V.P.); (A.K.)
| | - Anuj Rana
- Department of Microbiology (COBS & H), CCS Haryana Agricultural University, Hisar 125004, India;
| | - Arti Kumari
- Department of Microbiology, Central University of Haryana, Mahendargarh 123031, India; (V.P.); (A.K.)
| | - Jasneet Grewal
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Miecznikowa, 102-096 Warsaw, Poland; (J.G.); (K.P.)
| | - Kumar Pranaw
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Miecznikowa, 102-096 Warsaw, Poland; (J.G.); (K.P.)
| | - Surender Singh
- Department of Microbiology, Central University of Haryana, Mahendargarh 123031, India; (V.P.); (A.K.)
| |
Collapse
|
41
|
Baldewijns S, Sillen M, Palmans I, Vandecruys P, Van Dijck P, Demuyser L. The Role of Fatty Acid Metabolites in Vaginal Health and Disease: Application to Candidiasis. Front Microbiol 2021; 12:705779. [PMID: 34276639 PMCID: PMC8282898 DOI: 10.3389/fmicb.2021.705779] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2021] [Accepted: 05/31/2021] [Indexed: 12/28/2022] Open
Abstract
Although the vast majority of women encounters at least one vaginal infection during their life, the amount of microbiome-related research performed in this area lags behind compared to alternative niches such as the intestinal tract. As a result, effective means of diagnosis and treatment, especially of recurrent infections, are limited. The role of the metabolome in vaginal health is largely elusive. It has been shown that lactate produced by the numerous lactobacilli present promotes health by limiting the chance of infection. Short chain fatty acids (SCFA) have been mainly linked to dysbiosis, although the causality of this relationship is still under debate. In this review, we aim to bring together information on the role of the vaginal metabolome and microbiome in infections caused by Candida. Vulvovaginal candidiasis affects near to 70% of all women at least once in their life with a significant proportion of women suffering from the recurrent variant. We assess the role of fatty acid metabolites, mainly SCFA and lactate, in onset of infection and virulence of the fungal pathogen. In addition, we pinpoint where lack of research limits our understanding of the molecular processes involved and restricts the possibility of developing novel treatment strategies.
Collapse
Affiliation(s)
- Silke Baldewijns
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| | - Mart Sillen
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| | - Ilse Palmans
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| | - Paul Vandecruys
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| | - Patrick Van Dijck
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| | - Liesbeth Demuyser
- Laboratory of Molecular Cell Biology, Institute of Botany and Microbiology, KU Leuven, Leuven-Heverlee, Belgium
- VIB-KU Leuven Center for Microbiology, Leuven, Belgium
| |
Collapse
|
42
|
Gomaa EZ. Microbial chitinases: properties, enhancement and potential applications. PROTOPLASMA 2021; 258:695-710. [PMID: 33483852 DOI: 10.1007/s00709-021-01612-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 01/08/2021] [Indexed: 06/12/2023]
Abstract
Chitinases are a category of hydrolytic enzymes that catalyze chitin and are formed by a wide variety of microorganisms. In nature, microbial chitinases are primarily responsible for chitin decomposition and play a vital role in the balance of carbon and nitrogen ratio in the ecosystem. The physicochemical attributes and the source of chitinase are the main bases that determine their functional characteristics and hydrolyzed products. Several chitinases have been reported and characterized, and they obtain a wider consideration for their utilization in a large number of uses such as in agriculture, food, environment, medicine and pharmaceutical companies. The antifungal and insecticidal impacts of several chitinases have been extensively studied, aiming to protect crops from phytopathogenic fungi and insects. Chitooligosaccharides synthesized by chitin degradation have been shown to improve human health through their antimicrobial, antioxidant, anti-inflammatory and antitumor properties. This review aims at investigating chitinase production, properties and their potential applications in various biotechnological fields.
Collapse
Affiliation(s)
- Eman Zakaria Gomaa
- Department of Biological and Geological Sciences, Faculty of Education, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
43
|
d'Enfert C, Kaune AK, Alaban LR, Chakraborty S, Cole N, Delavy M, Kosmala D, Marsaux B, Fróis-Martins R, Morelli M, Rosati D, Valentine M, Xie Z, Emritloll Y, Warn PA, Bequet F, Bougnoux ME, Bornes S, Gresnigt MS, Hube B, Jacobsen ID, Legrand M, Leibundgut-Landmann S, Manichanh C, Munro CA, Netea MG, Queiroz K, Roget K, Thomas V, Thoral C, Van den Abbeele P, Walker AW, Brown AJP. The impact of the Fungus-Host-Microbiota interplay upon Candida albicans infections: current knowledge and new perspectives. FEMS Microbiol Rev 2021; 45:fuaa060. [PMID: 33232448 PMCID: PMC8100220 DOI: 10.1093/femsre/fuaa060] [Citation(s) in RCA: 157] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 11/18/2020] [Indexed: 12/11/2022] Open
Abstract
Candida albicans is a major fungal pathogen of humans. It exists as a commensal in the oral cavity, gut or genital tract of most individuals, constrained by the local microbiota, epithelial barriers and immune defences. Their perturbation can lead to fungal outgrowth and the development of mucosal infections such as oropharyngeal or vulvovaginal candidiasis, and patients with compromised immunity are susceptible to life-threatening systemic infections. The importance of the interplay between fungus, host and microbiota in driving the transition from C. albicans commensalism to pathogenicity is widely appreciated. However, the complexity of these interactions, and the significant impact of fungal, host and microbiota variability upon disease severity and outcome, are less well understood. Therefore, we summarise the features of the fungus that promote infection, and how genetic variation between clinical isolates influences pathogenicity. We discuss antifungal immunity, how this differs between mucosae, and how individual variation influences a person's susceptibility to infection. Also, we describe factors that influence the composition of gut, oral and vaginal microbiotas, and how these affect fungal colonisation and antifungal immunity. We argue that a detailed understanding of these variables, which underlie fungal-host-microbiota interactions, will present opportunities for directed antifungal therapies that benefit vulnerable patients.
Collapse
Affiliation(s)
- Christophe d'Enfert
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Ann-Kristin Kaune
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Leovigildo-Rey Alaban
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Sayoni Chakraborty
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
- Institute of Microbiology, Friedrich Schiller University, Neugasse 25, 07743 Jena, Germany
| | - Nathaniel Cole
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Margot Delavy
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Daria Kosmala
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
- Université de Paris, Sorbonne Paris Cité, 25, rue du Docteur Roux, 75015 Paris, France
| | - Benoît Marsaux
- ProDigest BV, Technologiepark 94, B-9052 Gent, Belgium
- Center for Microbial Ecology and Technology (CMET), Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links, 9000 Ghent, Belgium
| | - Ricardo Fróis-Martins
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Moran Morelli
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Diletta Rosati
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Marisa Valentine
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Zixuan Xie
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Yoan Emritloll
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Peter A Warn
- Magic Bullet Consulting, Biddlecombe House, Ugbrook, Chudleigh Devon, TQ130AD, UK
| | - Frédéric Bequet
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Marie-Elisabeth Bougnoux
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Stephanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF0545, 20 Côte de Reyne, 15000 Aurillac, France
| | - Mark S Gresnigt
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Bernhard Hube
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Ilse D Jacobsen
- Microbial Immunology Research Group, Emmy Noether Junior Research Group Adaptive Pathogenicity Strategies, and the Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knöll Institute, Beutenbergstraße 11a, 07745 Jena, Germany
| | - Mélanie Legrand
- Unité Biologie et Pathogénicité Fongiques, Département de Mycologie, Institut Pasteur, USC 2019 INRA, 25, rue du Docteur Roux, 75015 Paris, France
| | - Salomé Leibundgut-Landmann
- Immunology Section, Vetsuisse Faculty, University of Zurich, Winterthurerstrasse 266a, Zurich 8057, Switzerland
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, Zürich 8057, Switzerland
| | - Chaysavanh Manichanh
- Gut Microbiome Group, Vall d'Hebron Institut de Recerca (VHIR), Vall d'Hebron Hospital Universitari, Vall d'Hebron Barcelona Hospital Campus, Passeig Vall d'Hebron 119–129, 08035 Barcelona, Spain
| | - Carol A Munro
- Aberdeen Fungal Group, Institute of Medical Sciences, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Geert Grooteplein 28, 6525 GA Nijmegen, The Netherlands
| | - Karla Queiroz
- Mimetas, Biopartner Building 2, J.H. Oortweg 19, 2333 CH Leiden, The Netherlands
| | - Karine Roget
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | - Vincent Thomas
- BIOASTER Microbiology Technology Institute, 40 avenue Tony Garnier, 69007 Lyon, France
| | - Claudia Thoral
- NEXBIOME Therapeutics, 22 allée Alan Turing, 63000 Clermont-Ferrand, France
| | | | - Alan W Walker
- Gut Microbiology Group, Rowett Institute, University of Aberdeen, Ashgrove Road West, Foresterhill, Aberdeen AB25 2ZD, UK
| | - Alistair J P Brown
- MRC Centre for Medical Mycology, Department of Biosciences, University of Exeter, Geoffrey Pope Building, Stocker Road, Exeter EX4 4QD, UK
| |
Collapse
|
44
|
Scillato M, Spitale A, Mongelli G, Privitera GF, Mangano K, Cianci A, Stefani S, Santagati M. Antimicrobial properties of Lactobacillus cell-free supernatants against multidrug-resistant urogenital pathogens. Microbiologyopen 2021; 10:e1173. [PMID: 33970542 PMCID: PMC8483400 DOI: 10.1002/mbo3.1173] [Citation(s) in RCA: 45] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Revised: 01/22/2021] [Accepted: 01/29/2021] [Indexed: 01/23/2023] Open
Abstract
The healthy vaginal microbiota is dominated by Lactobacillus spp., which provide an important critical line of defense against pathogens, as well as giving beneficial effects to the host. We characterized L. gasseri 1A‐TV, L. fermentum 18A‐TV, and L. crispatus 35A‐TV, from the vaginal microbiota of healthy premenopausal women, for their potential probiotic activities. The antimicrobial effects of the 3 strains and their combination against clinical urogenital bacteria were evaluated together with the activities of their metabolites produced by cell‐free supernatants (CFSs). Their beneficial properties in terms of ability to interfere with vaginal pathogens (co‐aggregation, adhesion to HeLa cells, biofilm formation) and antimicrobial activity mediated by CFSs were assessed against multidrug urogenital pathogens (S. agalactiae, E. coli, KPC‐producing K. pneumoniae, S. aureus, E. faecium VRE, E. faecalis, P. aeruginosa, P. mirabilis, P. vulgaris, C. albicans, C. glabrata). The Lactobacilli tested exhibited an extraordinary ability to interfere and co‐aggregate with urogenital pathogens, except for Candida spp., as well as to adhere to HeLa cells and to produce biofilm in the Lactobacillus combination. Lactobacillus CFSs and their combination revealed a strong bactericidal effect on the multidrug resistant indicator strains tested, except for E. faecium and E. faecalis. The antimicrobial activity was maintained after heat treatment but decreased after enzymatic treatment. All Lactobacilli showed lactic dehydrogenase activity and production of D‐ and L‐lactic acid isomers on Lactobacillus CFSs, while only 1A‐TV and 35A‐TV released hydrogen peroxide and carried helveticin J and acidocin A bacteriocins. These results suggest that they can be employed as a new vaginal probiotic formulation and bio‐therapeutic preparation against urogenital infections. Further, in vivo studies are needed to evaluate human health benefits in clinical situations.
Collapse
Affiliation(s)
- Marina Scillato
- Department of Biomedical and Biotechnological Sciences, Microbiology Section, University of Catania, Catania, Italy
| | - Ambra Spitale
- Department of Biomedical and Biotechnological Sciences, Microbiology Section, University of Catania, Catania, Italy
| | - Gino Mongelli
- Department of Biomedical and Biotechnological Sciences, Microbiology Section, University of Catania, Catania, Italy
| | - Grete Francesca Privitera
- Department of Biomedical and Biotechnological Sciences, Microbiology Section, University of Catania, Catania, Italy
| | - Katia Mangano
- Department of Biomedical and Biotechnological Sciences, Oncologic, Clinical, and General Pathology Section, University of Catania, Catania, Italy
| | - Antonio Cianci
- Department of General Surgery and Medical-Surgical Specialties, University of Catania, Catania, Italy
| | - Stefania Stefani
- Department of Biomedical and Biotechnological Sciences, Microbiology Section, University of Catania, Catania, Italy
| | - Maria Santagati
- Department of Biomedical and Biotechnological Sciences, Microbiology Section, University of Catania, Catania, Italy
| |
Collapse
|
45
|
Crossing Kingdoms: How the Mycobiota and Fungal-Bacterial Interactions Impact Host Health and Disease. Infect Immun 2021; 89:IAI.00648-20. [PMID: 33526565 PMCID: PMC8090948 DOI: 10.1128/iai.00648-20] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The term “microbiota” invokes images of mucosal surfaces densely populated with bacteria. These surfaces and the luminal compartments they form indeed predominantly harbor bacteria. The term “microbiota” invokes images of mucosal surfaces densely populated with bacteria. These surfaces and the luminal compartments they form indeed predominantly harbor bacteria. However, research from this past decade has started to complete the picture by focusing on important but largely neglected constituents of the microbiota: fungi, viruses, and archaea. The community of commensal fungi, also called the mycobiota, interacts with commensal bacteria and the host. It is thus not surprising that changes in the mycobiota have significant impact on host health and are associated with pathological conditions such as inflammatory bowel disease (IBD). In this review we will give an overview of why the mycobiota is an important research area and different mycobiota research tools. We will specifically focus on distinguishing transient and actively colonizing fungi of the oral and gut mycobiota and their roles in health and disease. In addition to correlative and observational studies, we will discuss mechanistic studies on specific cross-kingdom interactions of fungi, bacteria, and the host.
Collapse
|
46
|
The role of lactobacilli in inhibiting skin pathogens. Biochem Soc Trans 2021; 49:617-627. [PMID: 33704415 DOI: 10.1042/bst20200329] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 02/12/2021] [Accepted: 02/17/2021] [Indexed: 12/25/2022]
Abstract
The human skin microbiota forms a key barrier against skin pathogens and is important in modulating immune responses. Recent studies identify lactobacilli as endogenous inhabitants of healthy skin, while inflammatory skin conditions are often associated with a disturbed skin microbiome. Consequently, lactobacilli-based probiotics are explored as a novel treatment of inflammatory skin conditions through their topical skin application. This review focuses on the potential beneficial role of lactobacilli (family Lactobacillaceae) in the skin habitat, where they can exert multifactorial local mechanisms of action against pathogens and inflammation. On one hand, lactobacilli have been shown to directly compete with skin pathogens through adhesion inhibition, production of antimicrobial metabolites, and by influencing pathogen metabolism. The competitive anti-pathogenic action of lactobacilli has already been described mechanistically for common different skin pathogens, such as Staphylococcus aureus, Cutibacterium acnes, and Candida albicans. On the other hand, lactobacilli also have an immunomodulatory capacity associated with a reduction in excessive skin inflammation. Their influence on the immune system is mediated by bacterial metabolites and cell wall-associated or excreted microbe-associated molecular patterns (MAMPs). In addition, lactobacilli can also enhance the skin barrier function, which is often disrupted as a result of infection or in inflammatory skin diseases. Some clinical trials have already translated these mechanistic insights into beneficial clinical outcomes, showing that topically applied lactobacilli can temporarily colonize the skin and promote skin health, but more and larger clinical trials are required to generate in vivo mechanistic insights and in-depth skin microbiome analysis.
Collapse
|
47
|
Singh RV, Sambyal K, Negi A, Sonwani S, Mahajan R. Chitinases production: A robust enzyme and its industrial applications. BIOCATAL BIOTRANSFOR 2021. [DOI: 10.1080/10242422.2021.1883004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Affiliation(s)
| | - Krishika Sambyal
- University Institute of Biotechnology, Chandigarh University, Gharuan, India
| | - Anjali Negi
- University Institute of Biotechnology, Chandigarh University, Gharuan, India
| | - Shubham Sonwani
- Department of Biosciences, Christian Eminent College, Indore, India
| | - Ritika Mahajan
- Department of Microbiology, School of Sciences, JAIN (Deemed-to-be University), Bengaluru, India
| |
Collapse
|
48
|
Vij R, Hube B, Brunke S. Uncharted territories in the discovery of antifungal and antivirulence natural products from bacteria. Comput Struct Biotechnol J 2021; 19:1244-1252. [PMID: 33680363 PMCID: PMC7905183 DOI: 10.1016/j.csbj.2021.02.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 02/02/2021] [Accepted: 02/02/2021] [Indexed: 12/26/2022] Open
Abstract
Many fungi can cause deadly diseases in humans, and nearly every human will suffer from some kind of fungal infection in their lives. Only few antifungals are available, and some of these fail to treat intrinsically resistant species and the ever-increasing number of fungal strains that have acquired resistance. In nature, bacteria and fungi display versatile interactions that range from friendly co-existence to predation. The first antifungal drugs, nystatin and amphotericin B, were discovered in bacteria as mediators of such interactions, and bacteria continue to be an important source of antifungals. To learn more about the ecological bacterial-fungal interactions that drive the evolution of natural products and exploit them, we need to identify environments where such interactions are pronounced, and diverse. Here, we systematically analyze historic and recent developments in this field to identify potentially under-investigated niches and resources. We also discuss alternative strategies to treat fungal infections by utilizing the antagonistic potential of bacteria to target fungal stress pathways and virulence factors, and thereby suppress the evolution of antifungal resistance.
Collapse
Affiliation(s)
- Raghav Vij
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knoell Institute Jena (HKI), Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knoell Institute Jena (HKI), Germany
- Institute of Microbiology, Friedrich Schiller University, Jena, Germany
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology – Hans Knoell Institute Jena (HKI), Germany
| |
Collapse
|
49
|
Chee WJY, Chew SY, Than LTL. Vaginal microbiota and the potential of Lactobacillus derivatives in maintaining vaginal health. Microb Cell Fact 2020; 19:203. [PMID: 33160356 PMCID: PMC7648308 DOI: 10.1186/s12934-020-01464-4] [Citation(s) in RCA: 257] [Impact Index Per Article: 51.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/28/2020] [Indexed: 12/15/2022] Open
Abstract
Human vagina is colonised by a diverse array of microorganisms that make up the normal microbiota and mycobiota. Lactobacillus is the most frequently isolated microorganism from the healthy human vagina, this includes Lactobacillus crispatus, Lactobacillus gasseri, Lactobacillus iners, and Lactobacillus jensenii. These vaginal lactobacilli have been touted to prevent invasion of pathogens by keeping their population in check. However, the disruption of vaginal ecosystem contributes to the overgrowth of pathogens which causes complicated vaginal infections such as bacterial vaginosis (BV), sexually transmitted infections (STIs), and vulvovaginal candidiasis (VVC). Predisposing factors such as menses, pregnancy, sexual practice, uncontrolled usage of antibiotics, and vaginal douching can alter the microbial community. Therefore, the composition of vaginal microbiota serves an important role in determining vagina health. Owing to their Generally Recognised as Safe (GRAS) status, lactobacilli have been widely utilised as one of the alternatives besides conventional antimicrobial treatment against vaginal pathogens for the prevention of chronic vaginitis and the restoration of vaginal ecosystem. In addition, the effectiveness of Lactobacillus as prophylaxis has also been well-founded in long-term administration. This review aimed to highlight the beneficial effects of lactobacilli derivatives (i.e. surface-active molecules) with anti-biofilm, antioxidant, pathogen-inhibition, and immunomodulation activities in developing remedies for vaginal infections. We also discuss the current challenges in the implementation of the use of lactobacilli derivatives in promotion of human health. In the current review, we intend to provide insights for the development of lactobacilli derivatives as a complementary or alternative medicine to conventional probiotic therapy in vaginal health.
Collapse
Affiliation(s)
- Wallace Jeng Yang Chee
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Shu Yih Chew
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| | - Leslie Thian Lung Than
- Department of Medical Microbiology, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor Malaysia
| |
Collapse
|
50
|
Dausset C, Bornes S, Miquel S, Kondjoyan N, Angenieux M, Nakusi L, Veisseire P, Alaterre E, Bermúdez-Humarán LG, Langella P, Engel E, Forestier C, Nivoliez A. Identification of sulfur components enhancing the anti-Candida effect of Lactobacillus rhamnosus Lcr35. Sci Rep 2020; 10:17074. [PMID: 33051479 PMCID: PMC7553951 DOI: 10.1038/s41598-020-74027-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 07/30/2020] [Indexed: 01/09/2023] Open
Abstract
GYNOPHILUS (Lcr REGENERANS) is a live biotherapeutic product (LBP) aimed at restoring the vaginal microbiome and contains the live biotherapeutic microorganism Lactobacillus rhamnosus Lcr35. In this study, the LBP formulation and manufacturing process significantly enhanced the anti-Candida activity of L. rhamnosus Lcr35, with a complete loss of viability of the yeast after 48 h of coincubation. Sodium thiosulfate (STS), one excipient of the product, was used as a potentiator of the anti-Candida spp. activity of Lactobacilli. This contact-independent phenomenon induced fungal cell disturbances, as observed by electron microscopy observations. Nonverbal sensory experiments showed clear odor dissimilarities between cocultures of L. rhamnosus Lcr35 and C. albicans in the presence and absence of STS, suggesting an impact of odor-active metabolites. A volatolomic approach allowed the identification of six odor-active compounds, including one sulfur compound that was identified as S-methyl thioacetate (MTA). MTA was associated with the antifungal effect of Lcr35, and its functional link was established in vitro. We show for the first time that the LBP GYNOPHILUS, which is a highly active product in the reduction of vulvovaginal candidiasis, requires the presence of a sulfur compound to fully achieve its antifungal effect.
Collapse
Affiliation(s)
- Caroline Dausset
- Research and Development Department, BIOSE, 24 avenue Georges Pompidou, 15000, Aurillac, France. .,Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France. .,Université Clermont Auvergne, CNRS, Laboratoire Microorganismes : Génome et Environnement, 63000, Clermont-Ferrand, France.
| | - Stéphanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000, Aurillac, France
| | - Sylvie Miquel
- Université Clermont Auvergne, CNRS, Laboratoire Microorganismes : Génome et Environnement, 63000, Clermont-Ferrand, France
| | - Nathalie Kondjoyan
- INRAE, UR370 QuaPA, Microcontaminants, Aroma & Separation Science Group (MASS), 63123, Saint-Genès-Champanelle, France
| | - Magaly Angenieux
- INRAE, UR370 QuaPA, Microcontaminants, Aroma & Separation Science Group (MASS), 63123, Saint-Genès-Champanelle, France
| | - Laurence Nakusi
- Université Clermont Auvergne, CNRS, Laboratoire Microorganismes : Génome et Environnement, 63000, Clermont-Ferrand, France
| | - Philippe Veisseire
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000, Aurillac, France
| | - Elina Alaterre
- Research and Development Department, BIOSE, 24 avenue Georges Pompidou, 15000, Aurillac, France.,HORIBA ABX SAS, Parc Euromédecine, Rue du Caducée, BP 7290, 34184, Montpellier Cedex 4, France
| | - Luis G Bermúdez-Humarán
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Philippe Langella
- Micalis Institute, INRAE, AgroParisTech, Université Paris-Saclay, 78350, Jouy-en-Josas, France
| | - Erwan Engel
- INRAE, UR370 QuaPA, Microcontaminants, Aroma & Separation Science Group (MASS), 63123, Saint-Genès-Champanelle, France
| | - Christiane Forestier
- Université Clermont Auvergne, CNRS, Laboratoire Microorganismes : Génome et Environnement, 63000, Clermont-Ferrand, France
| | - Adrien Nivoliez
- Research and Development Department, BIOSE, 24 avenue Georges Pompidou, 15000, Aurillac, France
| |
Collapse
|