1
|
Komura T, Nishikawa Y. Caenorhabditis elegans as a model host to study effects of lactic acid bacteria and functional food factors. Biosci Biotechnol Biochem 2025; 89:324-331. [PMID: 39577859 DOI: 10.1093/bbb/zbae172] [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: 09/12/2024] [Accepted: 11/15/2024] [Indexed: 11/24/2024]
Abstract
Several approaches for regulating health and disease by intestinal bacteria, bacteria isolated from food products, and food factors have been investigated; however, the molecular mechanisms underlying the interactions between these bacteria and their hosts are still unknown. Caenorhabditis elegans is a crucial model for exploring various biological phenomena and has been used in studying intestinal bacteria, mainly in the field of anti-aging, anti-amyloid β activity in Alzheimer's disease, and its interaction to hosts. The nematode model is expected to be increasingly utilized for elucidating the interactions of beneficial bacteria or food factors with hosts. Herein, we review anti-aging and suppression of amyloid β expression in a C. elegans model fed lactic acid bacteria, and our developed methods for orally administering food factors and assessing advanced glycation end products as an aging indicator.
Collapse
Affiliation(s)
- Tomomi Komura
- Graduate School of Human Science and Environment, University of Hyogo, Himeji, Hyogo, Japan
- Research Institute for Food and Nutritional Sciences, University of Hyogo, Himeji, Hyogo, Japan
| | - Yoshikazu Nishikawa
- Faculty of Food and Nutrition Science, Tezukayama Gakuin University, Sakai, Osaka, Japan
| |
Collapse
|
2
|
Grollemund PM, Poupet C, Comte É, Bonnet M, Veisseire P, Bornes S. A clustering-based survival comparison procedure designed to study the Caenorhabditis elegans model. Sci Rep 2024; 14:28257. [PMID: 39550445 PMCID: PMC11569119 DOI: 10.1038/s41598-024-79913-y] [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: 05/06/2024] [Accepted: 11/13/2024] [Indexed: 11/18/2024] Open
Abstract
Caenorhabditis elegans is highly important in current research, serving as a pivotal model organism that has greatly advanced the understanding of fundamental biological processes such as development, cellular biology, and neurobiology, helping to promote major advances in various fields of science. In this context, the survival of a nematode under various conditions is commonly investigated via statistical survival analysis, which is typically based on hypothesis testing, providing valuable insights into the factors influencing its longevity and response to various environmental factors. The extensive reliance on hypothesis testing is acknowledged as a concern in the scientific analysis process, emphasizing the need for a comprehensive evaluation of alternative statistical approaches to ensure a rigorous and unbiased interpretation of research findings. In this work, we propose an alternative method to hypothesis testing for evaluating differences in nematode survival. Our approach relies on a clustering technique that takes into account the complete structure of survival curves, enabling a more comprehensive assessment of survival dynamics. The proposed methodology helps to identify complex effects on nematode survival and enables us to derive the probability that treatment induces a specific effect. To highlight the application and benefits of the proposed methodology, it is applied to two different datasets, one simple and one more complex.
Collapse
Affiliation(s)
- Paul-Marie Grollemund
- University of Clermont Auvergne, CNRS, LMBP, Clermont-Ferrand, France.
- University of Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France.
| | - Cyril Poupet
- University of Tours, INRAE, ISP, 37000, Tours, France.
| | - Élise Comte
- University of Clermont Auvergne, CNRS, LMBP, Clermont-Ferrand, France
| | - Muriel Bonnet
- University of Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| | - Philippe Veisseire
- University of Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| | - Stéphanie Bornes
- University of Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| |
Collapse
|
3
|
Gharieb MM, Rizk A, Elfeky N. Anticandidal activity of a wild Bacillus subtilis NAM against clinical isolates of pathogenic Candida albicans. ANN MICROBIOL 2024; 74:23. [DOI: 10.1186/s13213-024-01764-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/13/2024] [Indexed: 01/03/2025] Open
Abstract
Abstract
Background
Resistance to antifungal medications poses a significant obstacle in combating fungal infections. The development of novel therapeutics for Candida albicans is necessary due to the increasing resistance of candidiasis to the existing medications. The utilization of biological control is seen as a more advantageous and less hazardous strategy therefore the objective of this study is to identify the antifungal properties of Bacillus subtilis against pathogenic C. albicans.
Results
We conducted a study to evaluate the antifungal properties of three bacterial isolates against the human pathogen Candida albicans. One of the bacterial isolates exhibited a potent antifungal activity against this fungal pathogen. This bacterium was identified as Bacillus subtilis based on the 16Sr RNA gene sequence. It exhibited inhibitory efficacy ranging from 33.5 to 44.4% against 15 Candida isolates. The optimal incubation duration for achieving the maximum antifungal activity was determined to be 48 h, resulting in a mean inhibition zone diameter of 29 ± 0.39 mm. The Potato Dextrose agar (PDA) medium was the best medium for the most effective antifungal activity. Incubation temperature of 25oC and medium pH value of 8.0 were the most favorable conditions for maximum antagonistic activity that resulted fungal growth inhibition of 40 ± 0.16 and 36 ± 0.94 mm respectively. Furthermore, the addition of 10.5 mg/ml of bacterial filtrate to C. albicans colonies resulted in 86.51%. decrease in the number of germinated cells. The fungal cell ultrastructural responses due to exposure to B. subtilis filtrate after 48 h were investigated using transmission electron microscopy (TEM). It revealed primary a drastic abnormality that lead to cellular disintegration including folding and lysis of the cell wall, total collapse of the yeast cells, and malformed germ tube following the exposure to the filtrate. However, the control culture treatment had a characteristic morphology of the normal fungal cells featuring a consistently dense central region, a well-organized nucleus, and a cytoplasm containing several components of the endomembrane system. The cells were surrounded by a uniform and intact cell wall.
Conclusion
The current study demonstrates a notable antifungal properties of B. subtilis against C. albicans as a result of production of bioactive components of the bacterial exudate. This finding could be a promising natural antifungal agent that could be utilized to combat C. albicans.
Collapse
|
4
|
Xu Z, Li Y, Xu A, Xue L, Soteyome T, Yuan L, Ma Q, Seneviratne G, Hong W, Mao Y, Kjellerup BV, Liu J. Differential alteration in Lactiplantibacillus plantarum subsp. plantarum quorum-sensing systems and reduced Candida albicans yeast survival and virulence gene expression in dual-species interaction. Microbiol Spectr 2024; 12:e0035324. [PMID: 38717160 PMCID: PMC11237386 DOI: 10.1128/spectrum.00353-24] [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: 02/08/2024] [Accepted: 04/15/2024] [Indexed: 06/06/2024] Open
Abstract
Candida albicans (C. albicans) and Lactiplantibacillus plantarum subsp. plantarum (L. plantarum) are frequently identified in various niches, but their dual-species interaction, especially with C. albicans in yeast form, remains unclear. This study aimed to investigate the dual-species interaction of L. plantarum and C. albicans, including proliferation, morphology, and transcriptomes examined by selective agar plate counting, microscopy, and polymicrobial RNA-seq, respectively. Maintaining a stable and unchanged growth rate, L. plantarum inhibited C. albicans yeast cell proliferation but not hyphal growth. Combining optical microscopy and atomic force microscopy, cell-to-cell direct contact and co-aggregation with L. plantarum cells surrounding C. albicans yeast cells were observed during dual-species interaction. Reduced C. albicans yeast cell proliferation in mixed culture was partially due to L. plantarum cell-free culture supernatant but not the acidic environment. Upon polymicrobial transcriptomics analysis, interesting changes were identified in both L. plantarum and C. albicans gene expression. First, two L. plantarum quorum-sensing systems showed contrary changes, with the activation of lamBDCA and repression of luxS. Second, the upregulation of stress response-related genes and downregulation of cell cycle, cell survival, and cell integrity-related pathways were identified in C. albicans, possibly connected to the stress posed by L. plantarum and the reduced yeast cell proliferation. Third, a large scale of pathogenesis and virulence factors were downregulated in C. albicans, indicating the potential interruption of pathogenic activities by L. plantarum. Fourth, partial metabolism and transport pathways were changed in L. plantarum and C. albicans. The information in this study might aid in understanding the behavior of L. plantarum and C. albicans in dual-species interaction.IMPORTANCEThe anti-Candida albicans activity of Lactiplantibacillus plantarum has been explored in the past decades. However, the importance of C. albicans yeast form and the effect of C. albicans on L. plantarum had also been omitted. In this study, the dual-species interaction of L. plantarum and C. albicans was investigated with a focus on the transcriptomes. Cell-to-cell direct contact and co-aggregation with L. plantarum cells surrounding C. albicans yeast cells were observed. Upon polymicrobial transcriptomics analysis, interesting changes were identified, including contrary changes in two L. plantarum quorum-sensing systems and reduced cell survival-related pathways and pathogenesis determinants in C. albicans.
Collapse
Affiliation(s)
- Zhenbo Xu
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Department of Laboratory Medicine, the Second Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong, China
| | - Yaqin Li
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
| | - Aijuan Xu
- Guangzhou Hybribio Medical Laboratory, Guangzhou, China
| | - Liang Xue
- Guangdong Province Key Laboratory for Green Processing of Natural Products and Product Safety, Engineering Research Center of Starch and Vegetable Protein Processing Ministry of Education, School of Food Science and Engineering, South China University of Technology, Guangzhou, China
- Guangdong Provincial Key Laboratory of Microbial Safety and Health, State Key Laboratory of Applied Microbiology Southern China, Institute of Microbiology, Guangdong Academy of Sciences, China, Guangzhou, Guangdong
- Key Laboratory of Agricultural Microbiomics and Precision Application, Ministry of Agriculture and Rural Affairs, Guangdong Academy of Sciences, Guangzhou, Guangdong, China
| | - Thanapop Soteyome
- Home Economics Technology, Rajamangala University of Technology Phra Nakhon, Bangkok, Thailand
| | - Lei Yuan
- School of Food Science and Engineering, Yangzhou University, Yangzhou, Jiangsu, China
| | - Qin Ma
- Key Laboratory of Functional Foods, Ministry of Agriculture, Guangdong Key Laboratory of Agricultural Products Processing, Sericultural and Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Guangzhou, China
| | | | - Wei Hong
- GMU-GIBH Joint School of Life Sciences, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yuzhu Mao
- Department of Civil and Environmental Engineering, University of Maryland, College Park, Maryland, USA
| | - Birthe V. Kjellerup
- Department of Civil and Environmental Engineering, University of Maryland, College Park, Maryland, USA
| | - Junyan Liu
- Guangdong Provincial Key Laboratory of Lingnan Specialty Food Science and Technology, College of Light Industry and Food Science, Academy of Contemporary Agricultural Engineering Innovations, Zhongkai University of Agriculture and Engineering, Guangzhou, China
- Key Laboratory of Green Processing and Intelligent Manufacturing of Lingnan Specialty Food, Ministry of Agriculture, Guangzhou, China
| |
Collapse
|
5
|
Garcia MT, do Carmo PHF, Figueiredo-Godoi LMA, Gonçalves NI, de Lima PMN, Ramos LDP, de Oliveira LD, Borges ALS, Shukla A, Junqueira JC. Gellan-Based Hydrogel as a Drug Delivery System for Caffeic Acid Phenethyl Ester in the Treatment of Oral Candida albicans Infections. Pharmaceutics 2024; 16:298. [PMID: 38543192 PMCID: PMC10975514 DOI: 10.3390/pharmaceutics16030298] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 02/05/2024] [Accepted: 02/10/2024] [Indexed: 02/13/2025] Open
Abstract
Candida albicans can cause various types of oral infections, mainly associated with denture stomatitis. Conventional therapy has been linked to high recurrence, toxicity, and fungal resistance, necessitating the search for new drugs and delivery systems. In this study, caffeic acid phenethyl ester (CAPE) and gellan gum (GG) were studied as an antifungal agent and carrier system, respectively. First, we observed that different GG formulations (0.6 to 1.0% wt/vol) were able to incorporate and release CAPE, reaching a controlled and prolonged release over 180 min at 1.0% of GG. CAPE-GG formulations exhibited antifungal activity at CAPE concentrations ranging from 128 to >512 µg/mL. Furthermore, CAPE-GG formulations significantly decreased the fungal viability of C. albicans biofilms at short times (12 h), mainly at 1.0% of GG (p < 0.001). C. albicans protease activity was also reduced after 12 h of treatment with CAPE-GG formulations (p < 0.001). Importantly, CAPE was not cytotoxic to human keratinocytes, and CAPE-GG formulations at 1.0% decreased the fungal burden (p = 0.0087) and suppressed inflammation in a rat model of denture stomatitis. Altogether, these results indicate that GG is a promising delivery system for CAPE, showing effective activity against C. albicans and potential to be used in the treatment of denture stomatitis.
Collapse
Affiliation(s)
- Maíra Terra Garcia
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12245-000, SP, Brazil
| | - Paulo Henrique Fonseca do Carmo
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12245-000, SP, Brazil
| | - Lívia Mara Alves Figueiredo-Godoi
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12245-000, SP, Brazil
| | - Natália Inês Gonçalves
- Department of Dental Materials and Prosthodontics, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12245-000, SP, Brazil
| | - Patrícia Michelle Nagai de Lima
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12245-000, SP, Brazil
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI 02912, USA
| | - Lucas de Paula Ramos
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12245-000, SP, Brazil
| | - Luciane Dias de Oliveira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12245-000, SP, Brazil
| | - Alexandre Luiz Souto Borges
- Department of Dental Materials and Prosthodontics, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12245-000, SP, Brazil
| | - Anita Shukla
- Center for Biomedical Engineering, School of Engineering, Brown University, Providence, RI 02912, USA
| | - Juliana Campos Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University (UNESP), São José dos Campos 12245-000, SP, Brazil
| |
Collapse
|
6
|
Dong J, Wang S, Li M, Liu J, Sun Z, Mandlaa, Chen Z. Application of a Chitosan-based Active Packaging Film Prepared with Cell-free Supernatant of Lacticaseibacillus paracasei ALAC-4 in Mongolian Cheese Preservation. J Food Prot 2023; 86:100158. [PMID: 37699510 DOI: 10.1016/j.jfp.2023.100158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/13/2023] [Accepted: 09/06/2023] [Indexed: 09/14/2023]
Abstract
Fungal spoilage of food is a worldwide concern prompting the development of many antimicrobial agents and applications. In this study, the cell-free supernatant (CFS) of Lacticaseibacillus paracasei ALAC-4 had a significant inhibition effect on fungi. The CFS with antifungal activities were combined with chitosan (CS) matrix to prepare an active packaging CS-CFS films by using a solvent casting method and used for the packaging of Mongolian cheese for 15 days during storage at 4 ± 1℃. The optimized formulation of the film were 1.25% (w/v) chitosan, 1.75% (w/v) gelatin, 0.3% (v/v) glycerol, and 9.6% (w/v) CFS. It was found that CS-CFS films exhibited strong antifungal activities against molds and yeasts, especially Candida albicans, and also had excellent mechanical properties. Additionally, FTIR spectroscopy indicated that hydrogen bonds between the CFS and CS formed, and there was a smooth surface, compact cross-section observed in SEM morphologies of CS-CFS films. Furthermore, CS-CFS film also displayed a strong antifungal effect against molds and yeasts on cheese surface. These results suggest that the chitosan-based CS-CFS film has a promising application for Mongolian cheese and food preservation.
Collapse
Affiliation(s)
- Jing Dong
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Shuai Wang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Minyu Li
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Jin Liu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Ziyu Sun
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China
| | - Mandlaa
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China.
| | - Zhongjun Chen
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China.
| |
Collapse
|
7
|
Kumaree KK, Prasanth MI, Sivamaruthi BS, Kesika P, Tencomnao T, Chaiyasut C, Prasansuklab A. Lactobacillus paracasei HII01 enhances lifespan and promotes neuroprotection in Caenorhabditis elegans. Sci Rep 2023; 13:16707. [PMID: 37794096 PMCID: PMC10550917 DOI: 10.1038/s41598-023-43846-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Accepted: 09/28/2023] [Indexed: 10/06/2023] Open
Abstract
Achieving healthy aging and providing protection from aging-related diseases is a major global concern. Probiotics, are a safer and more natural alternative. Moreover, identifying novel probiotics can help develop a new therapeutic approach and may help in personalized probiotic-formulations for individual's unique gut microbiome. In this study, we evaluated the benefits of our novel probiotic strains in promoting healthy aging and whether they protect against Amyloid β toxicity of Alzheimer's disease. Henceforth, we analyzed the impact of four different probiotics (Lactobacillus paracasei HII01, L. rhamnosus, L. reuteri, L. salivarius) on the lifespan extension of Caenorhabditis elegans model. Our results determine that L. paracasei HII01 provided the most positive effect on longevity and antiaging effects on C. elegans. The qPCR data and mutant-based studies indicated that L. paracasei HII01-mediated lifespan extension could be modulated by DAF-16 mediated pathway. The probiotic strains also protected the worms from the toxicity induced by β-Amyloid-expressing (Aβ) transgenic C. elegans strains, and L. paracasei HII01 provided the most significant protection. Overall, identifying novel probiotics is an important area of research that can improve health outcomes. Our study showed that L. paracasei HII01 could be considered a dietary supplement for providing healthy aging and preventing aging-related diseases.
Collapse
Affiliation(s)
- Kishoree K Kumaree
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- College of Public Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Bhagavathi Sundaram Sivamaruthi
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Periyanaina Kesika
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Chaiyavat Chaiyasut
- Innovation Center for Holistic Health, Nutraceuticals, and Cosmeceuticals, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, 50200, Thailand.
| | - Anchalee Prasansuklab
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, 10330, Thailand.
- College of Public Health Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
8
|
Fusco-Almeida AM, de Matos Silva S, dos Santos KS, de Lima Gualque MW, Vaso CO, Carvalho AR, Medina-Alarcón KP, Pires ACMDS, Belizario JA, de Souza Fernandes L, Moroz A, Martinez LR, Ruiz OH, González Á, Mendes-Giannini MJS. Alternative Non-Mammalian Animal and Cellular Methods for the Study of Host-Fungal Interactions. J Fungi (Basel) 2023; 9:943. [PMID: 37755051 PMCID: PMC10533014 DOI: 10.3390/jof9090943] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/28/2023] [Accepted: 08/30/2023] [Indexed: 09/28/2023] Open
Abstract
In the study of fungal pathogenesis, alternative methods have gained prominence due to recent global legislation restricting the use of mammalian animals in research. The principle of the 3 Rs (replacement, reduction, and refinement) is integrated into regulations and guidelines governing animal experimentation in nearly all countries. This principle advocates substituting vertebrate animals with other invertebrate organisms, embryos, microorganisms, or cell cultures. This review addresses host-fungus interactions by employing three-dimensional (3D) cultures, which offer more faithful replication of the in vivo environment, and by utilizing alternative animal models to replace traditional mammals. Among these alternative models, species like Caenorhabditis elegans and Danio rerio share approximately 75% of their genes with humans. Furthermore, models such as Galleria mellonella and Tenebrio molitor demonstrate similarities in their innate immune systems as well as anatomical and physiological barriers, resembling those found in mammalian organisms.
Collapse
Affiliation(s)
- Ana Marisa Fusco-Almeida
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Samanta de Matos Silva
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
- Basic and Applied Microbiology Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellin 050010, Colombia; (O.H.R.); (Á.G.)
| | - Kelvin Sousa dos Santos
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Marcos William de Lima Gualque
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Carolina Orlando Vaso
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Angélica Romão Carvalho
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Kaila Petrolina Medina-Alarcón
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Ana Carolina Moreira da Silva Pires
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Jenyffie Araújo Belizario
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Lígia de Souza Fernandes
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Andrei Moroz
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| | - Luis R. Martinez
- Department of Oral Biology, College of Dentistry, University of Florida, Gainesville, FL 32610, USA;
- Emerging Pathogens Institute, University of Florida, Gainesville, FL 32610, USA
- Center for Immunology and Transplantation, University of Florida, Gainesville, FL 32610, USA
- Center for Translational Research in Neurodegenerative Disease, University of Florida, Gainesville, FL 32610, USA
| | - Orville Hernandez Ruiz
- Basic and Applied Microbiology Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellin 050010, Colombia; (O.H.R.); (Á.G.)
- Cellular and Molecular Biology Group University of Antioquia, Corporation for Biological Research, Medellin 050010, Colombia
| | - Ángel González
- Basic and Applied Microbiology Group (MICROBA), School of Microbiology, Universidad de Antioquia, Medellin 050010, Colombia; (O.H.R.); (Á.G.)
| | - Maria José Soares Mendes-Giannini
- Department of Clinical Analysis, School of Pharmaceutical Science, Universidade Estadual Paulista (UNESP), Araraquara 14800-903, SP, Brazil; (A.M.F.-A.); (S.d.M.S.); (K.S.d.S.); (M.W.d.L.G.); (C.O.V.); (A.R.C.); (K.P.M.-A.); (A.C.M.d.S.P.); (J.A.B.); (L.d.S.F.); (A.M.)
| |
Collapse
|
9
|
Sala A, Ardizzoni A, Spaggiari L, Vaidya N, van der Schaaf J, Rizzato C, Cermelli C, Mogavero S, Krüger T, Himmel M, Kniemeyer O, Brakhage AA, King BL, Lupetti A, Comar M, de Seta F, Tavanti A, Blasi E, Wheeler RT, Pericolini E. A New Phenotype in Candida-Epithelial Cell Interaction Distinguishes Colonization- versus Vulvovaginal Candidiasis-Associated Strains. mBio 2023; 14:e0010723. [PMID: 36856418 PMCID: PMC10128025 DOI: 10.1128/mbio.00107-23] [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: 01/18/2023] [Accepted: 02/09/2023] [Indexed: 03/02/2023] Open
Abstract
Vulvovaginal candidiasis (VVC) affects nearly 3/4 of women during their lifetime, and its symptoms seriously reduce quality of life. Although Candida albicans is a common commensal, it is unknown if VVC results from a switch from a commensal to pathogenic state, if only some strains can cause VVC, and/or if there is displacement of commensal strains with more pathogenic strains. We studied a set of VVC and colonizing C. albicans strains to identify consistent in vitro phenotypes associated with one group or the other. We find that the strains do not differ in overall genetic profile or behavior in culture media (i.e., multilocus sequence type [MLST] profile, rate of growth, and filamentation), but they show strikingly different behaviors during their interactions with vaginal epithelial cells. Epithelial infections with VVC-derived strains yielded stronger fungal proliferation and shedding of fungi and epithelial cells. Transcriptome sequencing (RNA-seq) analysis of representative epithelial cell infections with selected pathogenic or commensal isolates identified several differentially activated epithelial signaling pathways, including the integrin, ferroptosis, and type I interferon pathways; the latter has been implicated in damage protection. Strikingly, inhibition of type I interferon signaling selectively increases fungal shedding of strains in the colonizing cohort, suggesting that increased shedding correlates with lower interferon pathway activation. These data suggest that VVC strains may intrinsically have enhanced pathogenic potential via differential elicitation of epithelial responses, including the type I interferon pathway. Therefore, it may eventually be possible to evaluate pathogenic potential in vitro to refine VVC diagnosis. IMPORTANCE Despite a high incidence of VVC, we still have a poor understanding of this female-specific disease whose negative impact on women's quality of life has become a public health issue. It is not yet possible to determine by genotype or laboratory phenotype if a given Candida albicans strain is more or less likely to cause VVC. Here, we show that Candida strains causing VVC induce more fungal shedding from epithelial cells than strains from healthy women. This effect is also accompanied by increased epithelial cell detachment and differential activation of the type I interferon pathway. These distinguishing phenotypes suggest it may be possible to evaluate the VVC pathogenic potential of fungal isolates. This would permit more targeted antifungal treatments to spare commensals and could allow for displacement of pathogenic strains with nonpathogenic colonizers. We expect these new assays to provide a more targeted tool for identifying fungal virulence factors and epithelial responses that control fungal vaginitis.
Collapse
Affiliation(s)
- Arianna Sala
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Andrea Ardizzoni
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Luca Spaggiari
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Nikhil Vaidya
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Jane van der Schaaf
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, Maine, USA
| | - Cosmeri Rizzato
- Department of Translational Research and of New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Claudio Cermelli
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Selene Mogavero
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Thomas Krüger
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Maximilian Himmel
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Olaf Kniemeyer
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Axel A. Brakhage
- Department of Molecular and Applied Microbiology, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Benjamin L. King
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, Maine, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| | - Antonella Lupetti
- Department of Translational Research and of New Technologies in Medicine and Surgery, University of Pisa, Pisa, Italy
| | - Manola Comar
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | - Francesco de Seta
- Institute for Maternal and Child Health—IRCCS Burlo Garofolo, Trieste, Italy
- Department of Medical, Surgical and Health Sciences, University of Trieste, Trieste, Italy
| | | | - Elisabetta Blasi
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| | - Robert T. Wheeler
- Department of Molecular and Biomedical Sciences, University of Maine, Orono, Maine, USA
- Graduate School of Biomedical Sciences and Engineering, University of Maine, Orono, Maine, USA
| | - Eva Pericolini
- Department of Surgical, Medical, Dental and Morphological Sciences with Interest in Transplant, Oncological and Regenerative Medicine, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
10
|
Poupet C, Rifa É, Theil S, Bonnet M, Veisseire P, Cardin G, Guéret É, Rialle S, Chassard C, Nivoliez A, Bornes S. In vivo investigation of Lcr35 ® anti-candidiasis properties in Caenorhabditis elegans reveals the involvement of highly conserved immune pathways. Front Microbiol 2022; 13:1062113. [PMID: 36620055 PMCID: PMC9816150 DOI: 10.3389/fmicb.2022.1062113] [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: 10/05/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022] Open
Abstract
Lactic acid bacteria, including the microorganisms formerly designated as Lactobacillus, are the major representatives of Live Biotherapeutic Microorganisms (LBM) when used for therapeutic purposes. However, in most cases, the mechanisms of action remain unknown. The antifungal potential of LBM has already been demonstrated using preclinical models (cell cultures, laboratory animals). Understanding their mechanisms of action is strategic for the development of new therapeutics for humans. Here, Caenorhabditis elegans was used as an in vivo model to analyze pro-longevity, anti-aging and anti-candidiasis effects of the LBM Lacticaseibacillus rhamnosus (formerly Lactobacillus rhamnosus) Lcr35®. A high-throughput transcriptomic analysis revealed a specific response of C. elegans depending on whether it is in the presence of the LBM L. rhamnosus Lcr35® (structural response), the yeast Candida albicans (metabolic response) or both (structural and metabolic responses) in a preventive and a curative conditions. Studies on C. elegans mutants demonstrated that the p38 MAPK (sek-1, skn-1) and the insulin-like (daf-2, daf-16) signaling pathways were involved in the extended lifespan provided by L. rhamnosus Lcr35® strain whereas the JNK pathway was not involved (jnk-1). In addition, the anti C. albicans effect of the bacterium requires the daf-16 and sek-1 genes while it is independent of daf-2 and skn-1. Moreover, the anti-aging effect of Lcr35®, linked to the extension of longevity, is not due to protection against oxidative stress (H2O2). Taken together, these results formally show the involvement of the p38 MAP kinase and insulin-like signaling pathways for the longevity extension and anti-Candida albicans properties of Lcr35® with, however, differences in the genes involved. Overall, these findings provide new insight for understanding the mechanisms of action of a probiotic strain with antimicrobial potential.
Collapse
Affiliation(s)
- Cyril Poupet
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France,*Correspondence: Cyril Poupet,
| | - Étienne Rifa
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| | - Sébastien Theil
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| | - Muriel Bonnet
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| | - Philippe Veisseire
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| | - Guillaume Cardin
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| | - Élise Guéret
- MGX, Univ Montpellier, CNRS, INSERM, Montpellier, France
| | | | | | | | - Stéphanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| |
Collapse
|
11
|
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
|
12
|
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: 48] [Impact Index Per Article: 16.0] [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
|
13
|
García-Gamboa R, Domínguez-Simi MÁ, Gradilla-Hernández MS, Bravo-Madrigal J, Moya A, González-Avila M. Antimicrobial and Antibiofilm Effect of Inulin-Type Fructans, Used in Synbiotic Combination with Lactobacillus spp. Against Candida albicans. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2022; 77:212-219. [PMID: 35461373 DOI: 10.1007/s11130-022-00966-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 04/07/2022] [Indexed: 06/14/2023]
Abstract
There is great interest in the search for new alternatives to antimicrobial drugs, and the use of prebiotics and probiotics is a promising approach to this problem. This study aimed to assess the effect of inulin-type fructans, used in synbiotic combinations with Lactobacillus paracasei or Lactobacillus plantarum, on the production of short-chain fatty acids and antimicrobial activity against Candida albicans. The inhibition assay using the L. paracasei and L. plantarum supernatants resulting from the metabolization of inulin-type fructans displayed growth inhibition and antibiofilm formation against C. albicans. Inhibition occurred at concentrations of 12.5, 25, and 50% of the L. paracasei supernatant and at a concentration of 50% of the L. plantarum supernatant. The analysis of short-chain fatty acids by gas chromatography showed that lactic acid was the dominating produced metabolite. However, acetic, propionic, and butyric acids were also detected in supernatants from both probiotics. Therefore, the synbiotic formulation of L. paracasei or L. plantarum in the presence of inulin-type fructans constitutes with anticandidal effect is a possible option to produce antifungal drugs or antimicrobial compounds.
Collapse
Affiliation(s)
- Ricardo García-Gamboa
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco A.C., Av. Normalistas No. 800, col Colinas de la Normal, C.P. 44270, Guadalajara, Jalisco, Mexico
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Av. General Ramón Corona No. 2514, Col. Nuevo México, Jalisco, C.P. 45138, Zapopan, Mexico
| | - Miguel Ángel Domínguez-Simi
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco A.C., Av. Normalistas No. 800, col Colinas de la Normal, C.P. 44270, Guadalajara, Jalisco, Mexico
| | - Misael Sebastián Gradilla-Hernández
- Tecnologico de Monterrey, Escuela de Ingenieria y Ciencias, Av. General Ramón Corona No. 2514, Col. Nuevo México, Jalisco, C.P. 45138, Zapopan, Mexico
| | - Jorge Bravo-Madrigal
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco A.C., Av. Normalistas No. 800, col Colinas de la Normal, C.P. 44270, Guadalajara, Jalisco, Mexico
| | - Andrés Moya
- Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunitat Valenciana (FISABIO), Av. de Cataluña 21, 46020, València, Spain
| | - Marisela González-Avila
- Centro de Investigación y Asistencia en Tecnología y Diseño del Estado de Jalisco A.C., Av. Normalistas No. 800, col Colinas de la Normal, C.P. 44270, Guadalajara, Jalisco, Mexico.
| |
Collapse
|
14
|
Combined impacts of various plant derivative extracts and lactic acid bacteria on yeasts to develop a nutritional bar with antifungal properties. FOOD BIOSCI 2022. [DOI: 10.1016/j.fbio.2022.101718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
15
|
Hu R, Zhang Y, Qian W, Leng Y, Long Y, Liu X, Li J, Wan X, Wei X. Pediococcus acidilactici Promotes the Longevity of C. elegans by Regulating the Insulin/IGF-1 and JNK/MAPK Signaling, Fat Accumulation and Chloride Ion. Front Nutr 2022; 9:821685. [PMID: 35433778 PMCID: PMC9010657 DOI: 10.3389/fnut.2022.821685] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 02/21/2022] [Indexed: 12/12/2022] Open
Abstract
Probiotics are known to contribute to the anti-oxidation, immunoregulation, and aging delay. Here, we investigated the extension of lifespan by fermented pickles-origin Pediococcus acidilactici (PA) in Caenorhabditis elegans (C. elegans), and found that PA promoted a significantly extended longevity of wild-type C. elegans. The further results revealed that PA regulated the longevity via promoting the insulin/IGF-1 signaling, JNK/MAPK signaling but not TOR signaling in C. elegans, and that PA reduced the reactive oxygen species (ROS) levels and modulated expression of genes involved in fatty acids uptake and lipolysis, thus reducing the fat accumulation in C. elegans. Moreover, this study identified the nrfl-1 as the key regulator of the PA-mediated longevity, and the nrfl-1/daf-18 signaling might be activated. Further, we highlighted the roles of one chloride ion exchanger gene sulp-6 in the survival of C. elegans and other two chloride ion channel genes clh-1 and clh-4 in the prolonged lifespan by PA-feeding through the modulating expression of genes involved in inflammation. Therefore, these findings reveal the detailed and novel molecular mechanisms on the longevity of C. elegans promoted by PA.
Collapse
|
16
|
Ahamefule CS, Ezeuduji BC, Ogbonna JC, Moneke AN, Ike AC, Jin C, Wang B, Fang W. Caenorhabditis elegans as an Infection Model for Pathogenic Mold and Dimorphic Fungi: Applications and Challenges. Front Cell Infect Microbiol 2021; 11:751947. [PMID: 34722339 PMCID: PMC8554291 DOI: 10.3389/fcimb.2021.751947] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Accepted: 09/28/2021] [Indexed: 11/13/2022] Open
Abstract
The threat burden from pathogenic fungi is universal and increasing with alarming high mortality and morbidity rates from invasive fungal infections. Understanding the virulence factors of these fungi, screening effective antifungal agents and exploring appropriate treatment approaches in in vivo modeling organisms are vital research projects for controlling mycoses. Caenorhabditis elegans has been proven to be a valuable tool in studies of most clinically relevant dimorphic fungi, helping to identify a number of virulence factors and immune-regulators and screen effective antifungal agents without cytotoxic effects. However, little has been achieved and reported with regard to pathogenic filamentous fungi (molds) in the nematode model. In this review, we have summarized the enormous breakthrough of applying a C. elegans infection model for dimorphic fungi studies and the very few reports for filamentous fungi. We have also identified and discussed the challenges in C. elegans-mold modeling applications as well as the possible approaches to conquer these challenges from our practical knowledge in C. elegans-Aspergillus fumigatus model.
Collapse
Affiliation(s)
- Chukwuemeka Samson Ahamefule
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, Nanning, China.,College of Life Science and Technology, Guangxi University, Nanning, China.,Department of Microbiology, University of Nigeria, Nsukka, Nigeria
| | | | - James C Ogbonna
- Department of Microbiology, University of Nigeria, Nsukka, Nigeria
| | - Anene N Moneke
- Department of Microbiology, University of Nigeria, Nsukka, Nigeria
| | - Anthony C Ike
- Department of Microbiology, University of Nigeria, Nsukka, Nigeria
| | - Cheng Jin
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, Nanning, China.,College of Life Science and Technology, Guangxi University, Nanning, China
| | - Bin Wang
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, Nanning, China.,State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning, China
| | - Wenxia Fang
- National Engineering Research Center for Non-Food Biorefinery, Guangxi Academy of Sciences, Nanning, China.,State Key Laboratory of Non-Food Biomass and Enzyme Technology, Guangxi Academy of Sciences, Nanning, China
| |
Collapse
|
17
|
Li S, Su B, He QS, Wu H, Zhang T. Alterations in the oral microbiome in HIV infection: causes, effects and potential interventions. Chin Med J (Engl) 2021; 134:2788-2798. [PMID: 34670249 PMCID: PMC8667981 DOI: 10.1097/cm9.0000000000001825] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Indexed: 12/02/2022] Open
Abstract
ABSTRACT A massive depletion of CD4+ T lymphocytes has been described in early and acute human immunodeficiency virus (HIV) infection, leading to an imbalance between the human microbiome and immune responses. In recent years, a growing interest in the alterations in gut microbiota in HIV infection has led to many studies; however, only few studies have been conducted to explore the importance of oral microbiome in HIV-infected individuals. Evidence has indicated the dysbiosis of oral microbiota in people living with HIV (PLWH). Potential mechanisms might be related to the immunodeficiency in the oral cavity of HIV-infected individuals, including changes in secretory components such as reduced levels of enzymes and proteins in saliva and altered cellular components involved in the reduction and dysfunction of innate and adaptive immune cells. As a result, disrupted oral immunity in HIV-infected individuals leads to an imbalance between the oral microbiome and local immune responses, which may contribute to the development of HIV-related diseases and HIV-associated non-acquired immunodeficiency syndrome comorbidities. Although the introduction of antiretroviral therapy (ART) has led to a significant decrease in occurrence of the opportunistic oral infections in HIV-infected individuals, the dysbiosis in oral microbiome persists. Furthermore, several studies with the aim to investigate the ability of probiotics to regulate the dysbiosis of oral microbiota in HIV-infected individuals are ongoing. However, the effects of ART and probiotics on oral microbiome in HIV-infected individuals remain unclear. In this article, we review the composition of the oral microbiome in healthy and HIV-infected individuals and the possible effect of oral microbiome on HIV-associated oral diseases. We also discuss how ART and probiotics influence the oral microbiome in HIV infection. We believe that a deeper understanding of composition and function of the oral microbiome is critical for the development of effective preventive and therapeutic strategies for HIV infection.
Collapse
Affiliation(s)
- Shuang Li
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Bin Su
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Qiu-Shui He
- Institute of Biomedicine, Research Center for Infections and Immunity, University of Turku, Turku 20520, Finland
| | - Hao Wu
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| | - Tong Zhang
- Beijing Key Laboratory for HIV/AIDS Research, Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing 100069, China
| |
Collapse
|
18
|
Zhang L, Zhan H, Xu W, Yan S, Ng SC. The role of gut mycobiome in health and diseases. Therap Adv Gastroenterol 2021; 14:17562848211047130. [PMID: 34589139 PMCID: PMC8474302 DOI: 10.1177/17562848211047130] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 08/31/2021] [Indexed: 02/04/2023] Open
Abstract
The gut microbiome comprised of microbes from multiple kingdoms, including bacteria, fungi, and viruses. Emerging evidence suggests that the intestinal fungi (the gut "mycobiome") play an important role in host immunity and inflammation. Advances in next generation sequencing methods to study the fungi in fecal samples and mucosa tissues have expanded our understanding of gut fungi in intestinal homeostasis and systemic immunity in health and their contribution to different human diseases. In this review, the current status of gut mycobiome in health, early life, and different diseases including inflammatory bowel disease, colorectal cancer, and metabolic diseases were summarized.
Collapse
Affiliation(s)
| | | | - Wenye Xu
- Center for Gut Microbiota Research, Faculty of
Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong,
China,Li Ka Shing Institute of Health Science, The
Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive disease,
Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin,
Hong Kong, China,Department of Medicine and Therapeutics,
Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong,
China
| | - Shuai Yan
- Center for Gut Microbiota Research, Faculty of
Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong,
China,Li Ka Shing Institute of Health Science, The
Chinese University of Hong Kong, Shatin, Hong Kong, China,State Key Laboratory for Digestive disease,
Institute of Digestive Disease, The Chinese University of Hong Kong, Shatin,
Hong Kong, China,Department of Anaesthesia and Intensive Care
and Peter Hung Pain Research Institute, The Chinese University of Hong Kong,
Shatin, Hong Kong, China
| | | |
Collapse
|
19
|
Possamai Rossatto FC, Tharmalingam N, Escobar IE, d’Azevedo PA, Zimmer KR, Mylonakis E. Antifungal Activity of the Phenolic Compounds Ellagic Acid (EA) and Caffeic Acid Phenethyl Ester (CAPE) against Drug-Resistant Candida auris. J Fungi (Basel) 2021; 7:jof7090763. [PMID: 34575801 PMCID: PMC8466507 DOI: 10.3390/jof7090763] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2021] [Revised: 09/06/2021] [Accepted: 09/06/2021] [Indexed: 12/17/2022] Open
Abstract
Candida auris is an emerging healthcare-associated fungal pathogen that has become a serious global health threat. Current treatment options are limited due to drug resistance. New therapeutic strategies are required to target this organism and its pathogenicity. Plant polyphenols are structurally diverse compounds that present a vast range of biological properties. In the present study, plant-derived molecules ellagic acid (EA) and caffeic acid phenethyl ester (CAPE) were investigated for their antifungal and antivirulence activities against Candida auris. We also tested against C. albicans. The minimum inhibitory concentration (MIC) for EA ranged from 0.125 to 0.25 µg/mL and for CAPE ranged from 1 to 64 µg/mL against drug-resistant C. auris strains. Killing kinetics determined that after 4 h treatment with CAPE, there was a complete reduction of viable C. auris cells compared to fluconazole. Both compounds might act by modifying the fungal cell wall. CAPE significantly reduced the biomass and the metabolic activity of C. auris biofilm and impaired C. auris adhesion to cultured human epithelial cells. Furthermore, both compounds prolonged the survival rate of Galleria mellonella infected by C. auris (p = 0.0088 for EA at 32 mg/kg and p = 0.0028 for CAPE at 4 mg/kg). In addition, EA at 4 μg/mL prolonged the survival of C. albicans-infected Caenorhabditis elegans (p < 0.0001). CAPE was not able to prolong the survival of C. albicans-infected C. elegans. These findings highlight the antifungal and antivirulence effects of EA and CAPE against C. auris, and warrant further investigation as novel antifungal agents against drug-resistant infections.
Collapse
Affiliation(s)
- Fernanda Cristina Possamai Rossatto
- Laboratory of Biofilms and Alternative Models, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil; (F.C.P.R.); (P.A.d.); (K.R.Z.)
| | - Nagendran Tharmalingam
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, 593 Eddy Street, P.O. Box 328/330, Providence, RI 02903, USA; (N.T.); (I.E.E.)
| | - Iliana E. Escobar
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, 593 Eddy Street, P.O. Box 328/330, Providence, RI 02903, USA; (N.T.); (I.E.E.)
| | - Pedro Alves d’Azevedo
- Laboratory of Biofilms and Alternative Models, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil; (F.C.P.R.); (P.A.d.); (K.R.Z.)
| | - Karine Rigon Zimmer
- Laboratory of Biofilms and Alternative Models, Federal University of Health Sciences of Porto Alegre, Porto Alegre 90050-170, RS, Brazil; (F.C.P.R.); (P.A.d.); (K.R.Z.)
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, 593 Eddy Street, P.O. Box 328/330, Providence, RI 02903, USA; (N.T.); (I.E.E.)
- Correspondence: ; Tel.: +1-401-444-7845; Fax: +1-401-444-8179
| |
Collapse
|
20
|
Andrade JC, Kumar S, Kumar A, Černáková L, Rodrigues CF. Application of probiotics in candidiasis management. Crit Rev Food Sci Nutr 2021; 62:8249-8264. [PMID: 34024191 DOI: 10.1080/10408398.2021.1926905] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Candidiasis (e.g., oral, gastrointestinal, vaginal, urinary tract, systemic) is a worldwide growing problem, since antifungal resistance and immunosuppression states are rising. To address this problem, very few drugs are available for the treatment of Candida spp. infections. Therefore, novel therapeutic strategies are urgently required. Probiotics have been proposed for the prevention and treatment of bacterial infections due to their safety record and efficacy, however, little is still known about their potential role regarding fungal infections. The purpose of this review is to present an updated summary of the evidence of the antifungal effects of probiotics along with a discussion of their potential use as an alternative/complementary therapy against Candida spp. infections. Thus, we performed a literature search using appropriate keywords ("Probiotic + Candida", "Candidiasis treatment", and "Probiotic + candidiasis") to retrieve relevant studies (both preclinical and clinical) with special emphasis on the works published in the last 5 years. An increasing amount of evidence has shown the potential usefulness of probiotics in the management of oral and vulvovaginal candidiasis in recent years. Among other results, we found that, as for bacterial infections, Lactobacillus, Bifidobacterium, and Saccharomyces are the most studied and effective genus for this purpose. However, in other areas, particularly in skincandidiaisis, studies are low or lacking. Thus, further investigation is necessary including in vitro and in vivo studies to establish the usefulness of probiotics in the management of candidiasis.
Collapse
Affiliation(s)
- José Carlos Andrade
- CESPU, Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra, Gandra PRD, Portugal
| | - Sunil Kumar
- Faculty of Biosciences, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Barabanki, Uttar Pradesh, India
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur, Chhattisgarh, India
| | - Lucia Černáková
- Department of Microbiology and Virology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| | - Célia F Rodrigues
- LEPABE - Laboratory for Process Engineering, Environment, Biotechnology and Energy, Faculty of Engineering, University of Porto, Porto, Portugal
| |
Collapse
|
21
|
Sá NPD, Barros PPD, Junqueira JC, Valério AD, Lino CI, Oliveira RBD, Rosa CA, Johann S. Antivirulence activity and in vivo efficacy of a thiazole derivative against candidiasis. J Mycol Med 2021; 31:101134. [PMID: 33862540 DOI: 10.1016/j.mycmed.2021.101134] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022]
Abstract
Candida albicans is a pathogen equipped with a variety of commensal and virulence traits that help it colonize the microbiota and invade host tissue during infection. In this study, we investigated the potential anticandidal activity of 3-[2-(4-(4-methoxyphenyl)thiazol-2-yl)hydrazino)]butan-1-ol (MT), a thiazolylhydrazone compound synthesized by our group, and identified it as a promising antifungal agent. The activity of MT was evaluated in vitro and in vivo against C. albicans as well as its ability to inhibit virulence factors. For this, the ability of MT to inhibit the adhesion of C. albicans to human buccal epithelial cells and biofilm formation and filamentation was tested. In addition, the potential in vivo activity of MT was evaluated in murine models of oral candidiasis. Our results confirmed the antifungal activity of MT, with a minimal inhibitory concentration range of 0.5-2 µg/mL. Indeed, MT treatment in vitro decreased the expression of C. albicans genes involved in biofilm formation and morphogenesis and encoding hydrolytic enzymes, which was also confirmed through phenotypic observations. In addition, MT promoted a decrease in the colony forming units recovered from the tongues of mice with oral candidiasis. In this work, we present a potent antivirulence compound that shows potential for candidiasis therapy, especially for topical use.
Collapse
Affiliation(s)
- Nívea Pereira de Sá
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, PO Box 486, 31270-901 Belo Horizonte, Minas Gerais, Brazil; Department of Microbiology and Immunology, Stony Brook University, Stony Brook, NY, United States
| | - Patrícia Pimentel de Barros
- Departamento de Biociências e Diagnóstico Bucal, Instituto de Ciência e Tecnologia, Universidade Estadual Paulista "Júlio de Mesquita Filho", São José dos Campos, São Paulo, Brazil; Postgraduate Program in Nursing, University of Guarulhos, São Paulo, Brazil
| | - Juliana Campos Junqueira
- Departamento de Biociências e Diagnóstico Bucal, Instituto de Ciência e Tecnologia, Universidade Estadual Paulista "Júlio de Mesquita Filho", São José dos Campos, São Paulo, Brazil
| | - Aline Dias Valério
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, PO Box 486, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Cleudiomar Inácio Lino
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Renata Barbosa de Oliveira
- Departamento de Produtos Farmacêuticos, Faculdade de Farmácia, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Carlos Augusto Rosa
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, PO Box 486, 31270-901 Belo Horizonte, Minas Gerais, Brazil
| | - Susana Johann
- Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, PO Box 486, 31270-901 Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
22
|
Scorzoni L, Alves de Paula e Silva AC, de Oliveira HC, Tavares dos Santos C, de Lacorte Singulani J, Akemi Assato P, Maria Marcos C, Teodoro Oliveira L, Ferreira Fregonezi N, Rossi DCP, Buffoni Roque da Silva L, Pelleschi Taborda C, Fusco-Almeida AM, Soares Mendes-Giannini MJ. In Vitro and In Vivo Effect of Peptides Derived from 14-3-3 Paracoccidioides spp. Protein. J Fungi (Basel) 2021; 7:jof7010052. [PMID: 33451062 PMCID: PMC7828505 DOI: 10.3390/jof7010052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 12/28/2020] [Accepted: 01/05/2021] [Indexed: 02/06/2023] Open
Abstract
Background: Paracoccidioidomycosis (PCM) is a chronic disease that causes sequelae and requires prolonged treatment; therefore, new therapeutic approaches are necessary. In view of this, three peptides from Paracoccidioides brasiliensis 14-3-3 protein were selected based on its immunogenicity and therapeutic potential. Methods: The in vitro antifungal activity and cytotoxicity of the 14-3-3 peptides were evaluated. The influence of the peptides in immunological and survival aspects was evaluated in vivo, using Galleria mellonella and the expression of antimicrobial peptide genes in Caenorhabditis elegans. Results: None of the peptides were toxic to HaCaT (skin keratinocyte), MRC-5 (lung fibroblast), and A549 (pneumocyte) cell lines, and only P1 exhibited antifungal activity against Paracoccidioides spp. The peptides could induce an immune response in G. mellonella. Moreover, the peptides caused a delay in the death of Paracoccidioides spp. infected larvae. Regarding C. elegans, the three peptides were able to increase the expression of the antimicrobial peptides. These peptides had essential effects on different aspects of Paracoccidioides spp. infection showing potential for a therapeutic vaccine. Future studies using mammalian methods are necessary to validate our findings.
Collapse
Affiliation(s)
- Liliana Scorzoni
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil; (L.S.); (A.C.A.d.P.eS.); (H.C.d.O.); (C.T.d.S.); (J.d.L.S.); (P.A.A.); (C.M.M.); (L.T.O.); (N.F.F.); (A.M.F.-A.)
| | - Ana Carolina Alves de Paula e Silva
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil; (L.S.); (A.C.A.d.P.eS.); (H.C.d.O.); (C.T.d.S.); (J.d.L.S.); (P.A.A.); (C.M.M.); (L.T.O.); (N.F.F.); (A.M.F.-A.)
| | - Haroldo Cesar de Oliveira
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil; (L.S.); (A.C.A.d.P.eS.); (H.C.d.O.); (C.T.d.S.); (J.d.L.S.); (P.A.A.); (C.M.M.); (L.T.O.); (N.F.F.); (A.M.F.-A.)
| | - Claudia Tavares dos Santos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil; (L.S.); (A.C.A.d.P.eS.); (H.C.d.O.); (C.T.d.S.); (J.d.L.S.); (P.A.A.); (C.M.M.); (L.T.O.); (N.F.F.); (A.M.F.-A.)
| | - Junya de Lacorte Singulani
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil; (L.S.); (A.C.A.d.P.eS.); (H.C.d.O.); (C.T.d.S.); (J.d.L.S.); (P.A.A.); (C.M.M.); (L.T.O.); (N.F.F.); (A.M.F.-A.)
| | - Patricia Akemi Assato
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil; (L.S.); (A.C.A.d.P.eS.); (H.C.d.O.); (C.T.d.S.); (J.d.L.S.); (P.A.A.); (C.M.M.); (L.T.O.); (N.F.F.); (A.M.F.-A.)
| | - Caroline Maria Marcos
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil; (L.S.); (A.C.A.d.P.eS.); (H.C.d.O.); (C.T.d.S.); (J.d.L.S.); (P.A.A.); (C.M.M.); (L.T.O.); (N.F.F.); (A.M.F.-A.)
| | - Lariane Teodoro Oliveira
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil; (L.S.); (A.C.A.d.P.eS.); (H.C.d.O.); (C.T.d.S.); (J.d.L.S.); (P.A.A.); (C.M.M.); (L.T.O.); (N.F.F.); (A.M.F.-A.)
| | - Nathália Ferreira Fregonezi
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil; (L.S.); (A.C.A.d.P.eS.); (H.C.d.O.); (C.T.d.S.); (J.d.L.S.); (P.A.A.); (C.M.M.); (L.T.O.); (N.F.F.); (A.M.F.-A.)
| | - Diego Conrado Pereira Rossi
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (D.C.P.R.); (L.B.R.d.S.); (C.P.T.)
| | - Leandro Buffoni Roque da Silva
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (D.C.P.R.); (L.B.R.d.S.); (C.P.T.)
| | - Carlos Pelleschi Taborda
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo 05508-000, Brazil; (D.C.P.R.); (L.B.R.d.S.); (C.P.T.)
| | - Ana Marisa Fusco-Almeida
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil; (L.S.); (A.C.A.d.P.eS.); (H.C.d.O.); (C.T.d.S.); (J.d.L.S.); (P.A.A.); (C.M.M.); (L.T.O.); (N.F.F.); (A.M.F.-A.)
| | - Maria José Soares Mendes-Giannini
- School of Pharmaceutical Sciences, São Paulo State University (UNESP), Araraquara, São Paulo 14800-903, Brazil; (L.S.); (A.C.A.d.P.eS.); (H.C.d.O.); (C.T.d.S.); (J.d.L.S.); (P.A.A.); (C.M.M.); (L.T.O.); (N.F.F.); (A.M.F.-A.)
- Correspondence:
| |
Collapse
|
23
|
de Alvarenga JA, de Barros PP, de Camargo Ribeiro F, Rossoni RD, Garcia MT, Dos Santos Velloso M, Shukla S, Fuchs BB, Shukla A, Mylonakis E, Junqueira JC. Probiotic Effects of Lactobacillus paracasei 28.4 to Inhibit Streptococcus mutans in a Gellan-Based Formulation. Probiotics Antimicrob Proteins 2020; 13:506-517. [PMID: 32980974 DOI: 10.1007/s12602-020-09712-0] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/20/2020] [Indexed: 12/21/2022]
Abstract
Streptococcus mutans is considered to be a major bacterium involved in dental caries, and the control of virulence mechanisms is fundamental to prevent disease. Probiotics present a promising preventive method; however, the use of probiotics requires its incorporation into delivery materials to facilitate oral colonization. Thus, we performed a comprehensive study examining preventive effects of Lactobacillus paracasei 28.4-enriched gellan hydrogel materials to inhibit S. mutans in planktonic and biofilm states, addressing its influence in the production of extracellular polysaccharides (EPS) and altered gene expression of several cariogenic virulence factors. L. paracasei 28.4, a strain isolated from the oral cavity of a caries-free individual, was incorporated in three gellan hydrogels (0.5%, 0.75%, and 1% w/v). The pretreatment with probiotic-gellan formulations provided a release of L. paracasei cells over 24 h that was sufficient to inhibit the planktonic growth of S. mutans, independent of the gellan concentrations and pH variations. This pretreatment also had inhibitory activity against S. mutans biofilms, exhibiting a reduction of 0.57 to 1.54 log10 in CFU/mL (p < 0.0001) and a decrease of 68.8 to 71.3% in total biomass (p < 0.0001) compared with the control group. These inhibitory effects were associated with the decreased production of EPS by 80% (p < 0.0001) and the downregulation of luxS, brpA, gbpB, and gtfB genes. The gellan formulation containing L. paracasei 28.4 exhibited probiotic effects for preventing S. mutans growth, biofilm formation, and production of cariogenic factors to suggest possible use in tooth decay prevention.
Collapse
Affiliation(s)
- Janaína Araújo de Alvarenga
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| | - Patrícia Pimentel de Barros
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil.
| | - Felipe de Camargo Ribeiro
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| | - Rodnei Dennis Rossoni
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| | - Maíra Terra Garcia
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| | - Marisol Dos Santos Velloso
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| | - Shashank Shukla
- School of Engineering, Brown University, Providence, RI, USA
| | - Beth Burgwyn Fuchs
- Rhode Island Hospital, Alpert Medical School & Brown University, Providence, RI, USA
| | - Anita Shukla
- School of Engineering, Brown University, Providence, RI, USA
| | - Eleftherios Mylonakis
- Rhode Island Hospital, Alpert Medical School & Brown University, Providence, RI, USA
| | - Juliana Campos Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, Av. Francisco José Longo 777, São Dimas, São José dos Campos, SP, 12245-000, Brazil
| |
Collapse
|
24
|
Poupet C, Chassard C, Nivoliez A, Bornes S. Caenorhabditis elegans, a Host to Investigate the Probiotic Properties of Beneficial Microorganisms. Front Nutr 2020; 7:135. [PMID: 33425969 PMCID: PMC7786404 DOI: 10.3389/fnut.2020.00135] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 07/14/2020] [Indexed: 12/11/2022] Open
Abstract
Caenorhabditis elegans, a non-parasitic nematode emerges as a relevant and powerful candidate as an in vivo model for microorganisms-microorganisms and microorganisms-host interactions studies. Experiments have demonstrated the probiotic potential of bacteria since they can provide to the worm a longer lifespan, an increased resistance to pathogens and to oxidative or heat stresses. Probiotics are used to prevent or treat microbiota dysbiosis and associated pathologies but the molecular mechanisms underlying their capacities are still unknown. Beyond safety and healthy aspects of probiotics, C. elegans represents a powerful way to design large-scale studies to explore transkingdom interactions and to solve questioning about the molecular aspect of these interactions. Future challenges and opportunities would be to validate C. elegans as an in vivo tool for high-throughput screening of microorganisms for their potential probiotic use on human health and to enlarge the panels of microorganisms studied as well as the human diseases investigated.
Collapse
Affiliation(s)
- Cyril Poupet
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| | | | | | - Stéphanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, Aurillac, France
| |
Collapse
|
25
|
Rossoni RD, de Barros PP, Mendonça IDC, Medina RP, Silva DHS, Fuchs BB, Junqueira JC, Mylonakis E. The Postbiotic Activity of Lactobacillus paracasei 28.4 Against Candida auris. Front Cell Infect Microbiol 2020; 10:397. [PMID: 32850495 PMCID: PMC7417517 DOI: 10.3389/fcimb.2020.00397] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 06/29/2020] [Indexed: 12/11/2022] Open
Abstract
Candida auris has emerged as a medically important pathogen with considerable resistance to antifungal agents. The ability to produce biofilms is an important pathogenicity feature of this species that aids escape of host immune responses and antimicrobial agents. The objective of this study was to verify antifungal action using in vitro and in vivo models of the Lactobacillus paracasei 28.4 probiotic cells and postbiotic activity of crude extract (LPCE) and fraction 1 (LPF1), derived from L. paracasei 28.4 supernatant. Both live cells and cells free supernatant of L. paracasei 28.4 inhibited C. auris suggesting probiotic and postbiotic effects. The minimum inhibitory concentration (MIC) for LPCE was 15 mg/mL and ranges from 3.75 to 7.5 mg/mL for LPF1. Killing kinetics determined that after 24 h treatment with LPCE or LPF1 there was a complete reduction of viable C. auris cells compared to fluconazole, which decreased the initial inoculum by 1-logCFU during the same time period. LPCE and LPF1 significantly reduced the biomass (p = 0.0001) and the metabolic activity (p = 0.0001) of C. auris biofilm. There was also a total reduction (~108 CFU/mL) in viability of persister C. auris cells after treatment with postbiotic elements (p < 0.0001). In an in vivo study, injection of LPCE and LPF1 into G. mellonella larvae infected with C. auris prolonged survival of these insects compared to a control group (p < 0.05) and elicited immune responses by increasing the number of circulating hemocytes and gene expression of antimicrobial peptide galiomicin. We concluded that the L. paracasei 28.4 cells and postbiotic elements (LPCE and LPF1) have antifungal activity against planktonic cells, biofilms, and persister cells of C. auris. Postbiotic supplementation derived from L. paracasei 28.4 protected G. mellonella infected with C. auris and enhanced its immune status indicating a dual function in modulating a host immune response.
Collapse
Affiliation(s)
- Rodnei Dennis Rossoni
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, São José dos Campos, Brazil.,Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, Providence, RI, United States
| | - Patrícia Pimentel de Barros
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, São José dos Campos, Brazil.,Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, Providence, RI, United States
| | - Iatã do Carmo Mendonça
- Department of Organic Chemistry, Center for Bioassays, Biosynthesis and Ecophysiology of Natural Products, Institute of Chemistry, São Paulo State University, UNESP, Araraquara, Brazil
| | - Rebeca Previate Medina
- Department of Organic Chemistry, Center for Bioassays, Biosynthesis and Ecophysiology of Natural Products, Institute of Chemistry, São Paulo State University, UNESP, Araraquara, Brazil
| | - Dulce Helena Siqueira Silva
- Department of Organic Chemistry, Center for Bioassays, Biosynthesis and Ecophysiology of Natural Products, Institute of Chemistry, São Paulo State University, UNESP, Araraquara, Brazil
| | - Beth Burgwyn Fuchs
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, Providence, RI, United States
| | - Juliana Campos Junqueira
- Department of Biosciences and Oral Diagnosis, Institute of Science and Technology, São Paulo State University/UNESP, São José dos Campos, Brazil
| | - Eleftherios Mylonakis
- Division of Infectious Diseases, Rhode Island Hospital, Warren Alpert Medical School at Brown University, Providence, RI, United States
| |
Collapse
|
26
|
Veisseire P, Bonnet M, Saraoui T, Poupet C, Camarès O, Gachinat M, Callon C, Febvre G, Chassard C, Bornes S. Investigation into In Vitro and In Vivo Caenorhabditis elegans Models to Select Cheese Yeasts as Probiotic Candidates for their Preventive Effects against Salmonella Typhimurium. Microorganisms 2020; 8:microorganisms8060922. [PMID: 32570901 PMCID: PMC7356738 DOI: 10.3390/microorganisms8060922] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 06/12/2020] [Accepted: 06/17/2020] [Indexed: 12/17/2022] Open
Abstract
The design of multiscale strategies integrating in vitro and in vivo models is necessary for the selection of new probiotics. In this regard, we developed a screening assay based on the investigation of the potential of yeasts from cheese as probiotics against the pathogen Salmonella Typhimurium UPsm1 (ST). Two yeasts isolated from raw-milk cheese (Saccharomyces cerevisiae 16, Sc16; Debaryomyces hansenii 25, Dh25), as well as S. cerevisiae subspecies boulardii (CNCM I-1079, Sb1079), were tested against ST by applying in vitro and in vivo tests. Adherence measurements to Caco-2 and HT29-MTX intestinal cells indicated that the two tested cheese yeasts presented a better adhesion than the probiotic Sb1079 as the control strain. Further, the Dh25 was the cheese yeast most likely to survive in the gastrointestinal tract. What is more, the modulation of the TransEpithelial Electrical Resistance (TEER) of differentiated Caco-2 cell monolayers showed the ability of Dh25 to delay the deleterious effects of ST. The influence of microorganisms on the in vivo model Caenorhabditis elegans was evaluated by measuring the longevity of the worm. This in vivo approach revealed that this yeast increased the worm’s lifespan and protected it against ST infection, confirming that this in vivo model can be useful for screening probiotic cheese yeasts.
Collapse
Affiliation(s)
- Philippe Veisseire
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
- Correspondence: ; Tel.: +33-(0)4-43-79-11-28
| | - Muriel Bonnet
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Taous Saraoui
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Cyril Poupet
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Olivier Camarès
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Marylise Gachinat
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Cécile Callon
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Guy Febvre
- Université Clermont Auvergne, Laboratoire Météorologie Physique, CNRS, F-15000 Aurillac, France;
| | - Christophe Chassard
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| | - Stéphanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, F-15000 Aurillac, France; (M.B.); (T.S.); (C.P.); (O.C.); (M.G.); (C.C.); (C.C.); (S.B.)
| |
Collapse
|
27
|
Development of Probiotic Formulations for Oral Candidiasis Prevention: Gellan Gum as a Carrier To Deliver Lactobacillus paracasei 28.4. Antimicrob Agents Chemother 2020; 64:AAC.02323-19. [PMID: 32253208 DOI: 10.1128/aac.02323-19] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Accepted: 03/29/2020] [Indexed: 01/12/2023] Open
Abstract
Probiotics might provide an alternative approach for the control of oral candidiasis. However, studies on the antifungal activity of probiotics in the oral cavity are based on the consumption of yogurt or other dietary products, and it is necessary to use appropriate biomaterials and specific strains to obtain probiotic formulations targeted for local oral administration. In this study, we impregnated gellan gum, a natural biopolymer used as a food additive, with a probiotic and investigated its antifungal activity against Candida albicans Lactobacillus paracasei 28.4, a strain recently isolated from the oral cavity of a caries-free individual, was incorporated in several concentrations of gellan gum (0.6% to 1% [wt/vol]). All tested concentrations could incorporate L. paracasei cells while maintaining bacterial viability. Probiotic-gellan gum formulations were stable for 7 days when stored at room temperature or 4°C. Long-term storage of bacterium-impregnated gellan gum was achieved when L. paracasei 28.4 was lyophilized. The probiotic-gellan gum formulations provided a release of L. paracasei cells over 24 h that was sufficient to inhibit the growth of C. albicans, with effects dependent on the cell concentrations incorporated into gellan gum. The probiotic-gellan gum formulations also had inhibitory activity against Candida sp. biofilms by reducing the number of Candida sp. cells (P < 0.0001), decreasing the total biomass (P = 0.0003), and impairing hyphae formation (P = 0.0002), compared to the control group which received no treatment. Interestingly, a probiotic formulation of 1% (wt/vol) gellan gum provided an oral colonization of L. paracasei in mice with approximately 6 log CFU/ml after 10 days. This formulation inhibited C. albicans growth (P < 0.0001), prevented the development of candidiasis lesions (P = 0.0013), and suppressed inflammation (P = 0.0006) compared to the mice not treated in the microscopic analysis of the tongue dorsum. These results indicate that gellan gum is a promising biomaterial and can be used as a carrier system to promote oral colonization for probiotics that prevent oral candidiasis.
Collapse
|
28
|
Li D, She X, Calderone R. The antifungal pipeline: the need is established. Are there new compounds? FEMS Yeast Res 2020; 20:5827531. [DOI: 10.1093/femsyr/foaa023] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 04/28/2020] [Indexed: 12/19/2022] Open
Abstract
ABSTRACT
Our review summarizes and compares the temporal development (eras) of antifungal drug discovery as well as antibacterial ventures. The innovation gap that occurred in antibacterial discovery from 1960 to 2000 was likely due to tailoring of existing compounds to have better activity than predecessors. Antifungal discovery also faced innovation gaps. The semi-synthetic antibiotic era was followed closely by the resistance era and the heightened need for new compounds and targets. With the immense contribution of comparative genomics, antifungal targets became part of the discovery focus. These targets by definition are absolutely required to be fungal- or even lineage (clade) specific. Importantly, targets need to be essential for growth and/or have important roles in disease and pathogenesis. Two types of antifungals are discussed that are mostly in the FDA phase I–III clinical trials. New antifungals are either modified to increase bioavailability and stability for instance, or are new compounds that inhibit new targets. One of the important developments in incentivizing new antifungal discovery has been the prolific number of publications of global and country-specific incidence. International efforts that champion global antimicrobial drug discovery are discussed. Still, interventions are needed. The current pipeline of antifungals and alternatives to antifungals are discussed including vaccines.
Collapse
Affiliation(s)
- Dongmei Li
- Department of Microbiology and Immunology, Georgetown University Medical Center, Georgetown University, NW 302 Med Dent Building, 3900 Reservoir Rd NW, Washington, DC 20057, USA
| | - Xiaodong She
- Jiangsu Key laboratory of Molecular Biology for Skin Disease and STIs, Institute of Dermatology, Chinese Academy of Medical Sciences (CAMS), Nanjing 210029, China
| | - Richard Calderone
- Department of Microbiology and Immunology, Georgetown University Medical Center, Georgetown University, NW 302 Med Dent Building, 3900 Reservoir Rd NW, Washington, DC 20057, USA
| |
Collapse
|
29
|
Poupet C, Veisseire P, Bonnet M, Camarès O, Gachinat M, Dausset C, Chassard C, Nivoliez A, Bornes S. Curative Treatment of Candidiasis by the Live Biotherapeutic Microorganism Lactobacillus rhamnosus Lcr35 ® in the Invertebrate Model Caenorhabditis elegans: First Mechanistic Insights. Microorganisms 2019; 8:microorganisms8010034. [PMID: 31878039 PMCID: PMC7022838 DOI: 10.3390/microorganisms8010034] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
The resistance of Candida albicans to conventional drug treatments, as well as the recurrence phenomena due to dysbiosis caused by antifungal treatments, have highlighted the need to implement new therapeutic methodologies. The antifungal potential of live biotherapeutic products (LBP) has already been demonstrated using preclinical models (cell cultures, laboratory animals). Understanding their mechanisms of action is strategic for the development of new therapeutics for humans. In this study, we investigated the curative anti-C. albicans properties of Lactobacillus rhamnosus Lcr35® using the in vitro Caco-2 cell and the in vivo Caenorhabditis elegans models. We showed that Lcr35® does inhibit neither the growth (p = 0.603) nor the biofilm formation (p = 0.869) of C. albicans in vitro. Lcr35® protects the animal from the fungal infection (+225% of survival, p < 2 × 10–16) even if the yeast is detectable in its intestine. In contrast, the Lcr35® cell-free supernatant does not appear to have any antipathogenic effect. At the mechanistic level, the DAF-16/Forkhead Box O transcription factor is activated by Lcr35® and genes of the p38 MAP Kinase signaling pathway and genes involved in the antifungal response are upregulated in presence of Lcr35® after C. albicans infection. These results suggest that the LBM strain acts by stimulating its host via DAF-16 and the p38 MAPK pathway.
Collapse
Affiliation(s)
- Cyril Poupet
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000 Aurillac, France
- Correspondence: ; Tel.: +33-(0)4-43-79-11-29
| | - Philippe Veisseire
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000 Aurillac, France
| | - Muriel Bonnet
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000 Aurillac, France
| | - Olivier Camarès
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000 Aurillac, France
| | - Marylise Gachinat
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000 Aurillac, France
| | - Caroline Dausset
- Biose Industrie, 24 avenue Georges Pompidou, 15000 Aurillac, France
| | - Christophe Chassard
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000 Aurillac, France
| | - Adrien Nivoliez
- Biose Industrie, 24 avenue Georges Pompidou, 15000 Aurillac, France
| | - Stéphanie Bornes
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000 Aurillac, France
| |
Collapse
|
30
|
Efficacy of Using Probiotics with Antagonistic Activity against Pathogens of Wound Infections: An Integrative Review of Literature. BIOMED RESEARCH INTERNATIONAL 2019; 2019:7585486. [PMID: 31915703 PMCID: PMC6930797 DOI: 10.1155/2019/7585486] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Accepted: 10/03/2019] [Indexed: 02/06/2023]
Abstract
The skin and its microbiota serve as physical barriers to prevent invasion of pathogens. Skin damage can be a consequence of illness, surgery, and burns. The most effective wound management strategy is to prevent infections, promote healing, and prevent excess scarring. It is well established that probiotics can aid in skin healing by stimulating the production of immune cells, and they also exhibit antagonistic effects against pathogens via competitive exclusion of pathogens. Our aim was to conduct a review of recent literature on the efficacy of using probiotics against pathogens that cause wound infections. In this integrative review, we searched through the literature published in the international following databases: PubMed, ScienceDirect, Web of Science, and Scopus using the search terms “probiotic” AND “wound infection.” During a comprehensive review and critique of the selected research, fourteen in vitro studies, 8 animal studies, and 19 clinical studies were found. Two of these in vitro studies also included animal studies, yielding a total of 39 articles for inclusion in the review. The most commonly used probiotics for all studies were well-known strains of the species Lactobacillus plantarum, Lactobacillus casei, Lactobacillus acidophilus, and Lactobacillus rhamnosus. All in vitro studies showed successful inhibition of chosen skin or wound pathogens by the selected probiotics. Within the animal studies on mice, rats, and rabbits, probiotics showed strong opportunities for counteracting wound infections. Most clinical studies showed slight or statistically significant lower incidence of surgical site infections, foot ulcer infection, or burn infections for patients using probiotics. Several of these studies also indicated a statistically significant wound healing effect for the probiotic groups. This review indicates that exogenous and oral application of probiotics has shown reduction in wound infections, especially when used as an adjuvant to antibiotic therapy, and therefore the potential use of probiotics in this field remains worthy of further studies, perhaps focused more on typical skin inhabitants as next-generation probiotics with high potential.
Collapse
|
31
|
Zangl I, Pap IJ, Aspöck C, Schüller C. The role of Lactobacillus species in the control of Candida via biotrophic interactions. MICROBIAL CELL 2019; 7:1-14. [PMID: 31921929 PMCID: PMC6946018 DOI: 10.15698/mic2020.01.702] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microbial communities have an important role in health and disease. Candida spp. are ubiquitous commensals and sometimes opportunistic fungal pathogens of humans, colonizing mucosal surfaces of the genital, urinary, respiratory and gastrointestinal tracts and the oral cavity. They mainly cause local mucosal infections in immune competent individuals. However, in the case of an ineffective immune defense, Candida infections may become a serious threat. Lactobacillus spp. are part of the human microbiome and are natural competitors of Candida in the vaginal environment. Lactic acid, low pH and other secreted metabolites are environmental signals sensed by fungal species present in the microbiome. This review briefly discusses the ternary interaction between host, Lactobacillus species and Candida with regard to fungal infections and the potential antifungal and fungistatic effect of Lactobacillus species. Our understanding of these interactions is incomplete due to the variability of the involved species and isolates and the complexity of the human host.
Collapse
Affiliation(s)
- Isabella Zangl
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Applied Genetics and Cell Biology (DAGZ), Tulln, Austria
| | - Ildiko-Julia Pap
- University Hospital of St. Pölten, Institute for Hygiene and Microbiology, St Pölten, Austria
| | - Christoph Aspöck
- University Hospital of St. Pölten, Institute for Hygiene and Microbiology, St Pölten, Austria
| | - Christoph Schüller
- University of Natural Resources and Life Sciences Vienna (BOKU), Department of Applied Genetics and Cell Biology (DAGZ), Tulln, Austria.,Bioactive Microbial Metabolites (BiMM), BOKU, Tulln, Austria
| |
Collapse
|
32
|
Ribeiro F, Rossoni R, Barros P, Santos J, Fugisaki L, Leão M, Junqueira J. Action mechanisms of probiotics on
Candida
spp. and candidiasis prevention: an update. J Appl Microbiol 2019; 129:175-185. [DOI: 10.1111/jam.14511] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Revised: 10/18/2019] [Accepted: 10/30/2019] [Indexed: 12/19/2022]
Affiliation(s)
- F.C. Ribeiro
- Department of Biosciences and Oral Diagnosis Institute of Science and Technology São Paulo State University/UNESP Sao Jose dos Campos Brazil
| | - R.D. Rossoni
- Department of Biosciences and Oral Diagnosis Institute of Science and Technology São Paulo State University/UNESP Sao Jose dos Campos Brazil
| | - P.P. Barros
- Department of Biosciences and Oral Diagnosis Institute of Science and Technology São Paulo State University/UNESP Sao Jose dos Campos Brazil
| | - J.D. Santos
- Department of Biosciences and Oral Diagnosis Institute of Science and Technology São Paulo State University/UNESP Sao Jose dos Campos Brazil
| | - L.R.O. Fugisaki
- Department of Biosciences and Oral Diagnosis Institute of Science and Technology São Paulo State University/UNESP Sao Jose dos Campos Brazil
| | - M.P.V. Leão
- Bioscience Basic Institute University of Taubaté Bom Conselho Taubaté SP Brazil
| | - J.C. Junqueira
- Department of Biosciences and Oral Diagnosis Institute of Science and Technology São Paulo State University/UNESP Sao Jose dos Campos Brazil
| |
Collapse
|
33
|
Poupet C, Saraoui T, Veisseire P, Bonnet M, Dausset C, Gachinat M, Camarès O, Chassard C, Nivoliez A, Bornes S. Lactobacillus rhamnosus Lcr35 as an effective treatment for preventing Candida albicans infection in the invertebrate model Caenorhabditis elegans: First mechanistic insights. PLoS One 2019; 14:e0216184. [PMID: 31693670 PMCID: PMC6834333 DOI: 10.1371/journal.pone.0216184] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2019] [Accepted: 10/22/2019] [Indexed: 12/26/2022] Open
Abstract
The increased recurrence of Candida albicans infections is associated with greater resistance to antifungal drugs. This involves the establishment of alternative therapeutic protocols, such as probiotic microorganisms whose antifungal potential has already been demonstrated using preclinical models (cell cultures, laboratory animals). Understanding the mechanisms of action of probiotic microorganisms has become a strategic need for the development of new therapeutics for humans. In this study, we investigated the prophylactic anti-C. albicans properties of Lactobacillus rhamnosus Lcr35® using the in vitro Caco-2 cell model and the in vivo Caenorhabditis elegans model. In Caco-2 cells, we showed that the strain Lcr35® significantly inhibited the growth (~2 log CFU.mL-1) and adhesion (150 to 6,300 times less) of the pathogen. Moreover, in addition to having a pro-longevity activity in the nematode (+42.9%, p = 3.56.10-6), Lcr35® protects the animal from the fungal infection (+267% of survival, p < 2.10-16) even if the yeast is still detectable in its intestine. At the mechanistic level, we noticed the repression of genes of the p38 MAPK signalling pathway and genes involved in the antifungal response induced by Lcr35®, suggesting that the pathogen no longer appears to be detected by the worm immune system. However, the DAF-16/FOXO transcription factor, implicated in the longevity and antipathogenic response of C. elegans, is activated by Lcr35®. These results suggest that the probiotic strain acts by stimulating its host via DAF-16 but also by suppressing the virulence of the pathogen.
Collapse
Affiliation(s)
- Cyril Poupet
- Université Clermont Auvergne, INRA, VetAgro Sup, Aurillac, France
| | - Taous Saraoui
- Université Clermont Auvergne, INRA, VetAgro Sup, Aurillac, France
| | | | - Muriel Bonnet
- Université Clermont Auvergne, INRA, VetAgro Sup, Aurillac, France
| | | | | | - Olivier Camarès
- Université Clermont Auvergne, INRA, VetAgro Sup, Aurillac, France
| | | | | | - Stéphanie Bornes
- Université Clermont Auvergne, INRA, VetAgro Sup, Aurillac, France
| |
Collapse
|
34
|
Caenorhabditis Elegans and Probiotics Interactions from a Prolongevity Perspective. Int J Mol Sci 2019; 20:ijms20205020. [PMID: 31658751 PMCID: PMC6834311 DOI: 10.3390/ijms20205020] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 02/08/2023] Open
Abstract
Probiotics exert beneficial effects on host health through different mechanisms of action, such as production of antimicrobial substances, competition with pathogens, enhancement of host mucosal barrier integrity and immunomodulation. In the context of ageing, which is characterized by several physiological alterations leading to a low grade inflammatory status called inflammageing, evidences suggest a potential prolongevity role of probiotics. Unraveling the mechanisms underlying anti-ageing effects requires the use of simple model systems. To this respect, the nematode Caenorhabditis elegans represents a suitable model organism for the study of both host-microbe interactions and for ageing studies, because of conserved signaling pathways and host defense mechanisms involved in the regulation of its lifespan. Therefore, this review analyses the impact of probiotics on C. elegans age-related parameters, with particular emphasis on oxidative stress, immunity, inflammation and protection from pathogen infections. The picture emerging from our analysis highlights that several probiotic strains are able to exert anti-ageing effects in nematodes by acting on common molecular pathways, such as insulin/insulin-like growth factor-1 (IIS) and p38 mitogen-activated protein kinase (p38 MAPK). In this perspective, C. elegans appears to be advantageous for shedding light on key mechanisms involved in host prolongevity in response to probiotics supplementation.
Collapse
|
35
|
Caenorhabditis elegans as a model animal for investigating fungal pathogenesis. Med Microbiol Immunol 2019; 209:1-13. [PMID: 31555911 DOI: 10.1007/s00430-019-00635-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 09/18/2019] [Indexed: 12/27/2022]
Abstract
The morbidity and mortality associated with systemic fungal infections in humans cannot be underestimated. The nematode Caenorhabditis elegans has become popular for the in vivo study of the pathogenesis of human fungal pathogens and as an antifungal drug-screening tool. C. elegans offers many advantages as a model organism for the study of human fungal diseases, including lack of ethics requirements, easy maintenance in the laboratory, fully sequenced genome, availability of genetic mutants, and the possibility of liquid assays for high-throughput antifungal screening. Its major drawbacks include the inability to grow at 37 °C and absence of an adaptive immune response. However, several virulence factors involved in the pathogenesis of medically important fungal pathogens have been identified using the C. elegans model, consequently providing new leads for drug discovery and potential drug targets. We review the use of C. elegans as a model animal to understand the pathogenesis of medically important human fungal pathogens and the discovery of novel antifungal compounds. The review makes a case for C. elegans as a suitable invertebrate model for a plethora of practical applications in the investigation of fungal pathogenesis as well as its amenability for liquid-based high-throughput screening of potential antifungal compounds.
Collapse
|
36
|
Graf K, Last A, Gratz R, Allert S, Linde S, Westermann M, Gröger M, Mosig AS, Gresnigt MS, Hube B. Keeping Candida commensal: how lactobacilli antagonize pathogenicity of Candida albicans in an in vitro gut model. Dis Model Mech 2019; 12:dmm.039719. [PMID: 31413153 PMCID: PMC6765188 DOI: 10.1242/dmm.039719] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 08/02/2019] [Indexed: 12/20/2022] Open
Abstract
The intestine is the primary reservoir of Candida albicans that can cause systemic infections in immunocompromised patients. In this reservoir, the fungus exists as a harmless commensal. However, antibiotic treatment can disturb the bacterial microbiota, facilitating fungal overgrowth and favoring pathogenicity. The current in vitro gut models that are used to study the pathogenesis of C. albicans investigate the state in which C. albicans behaves as a pathogen rather than as a commensal. We present a novel in vitro gut model in which the fungal pathogenicity is reduced to a minimum by increasing the biological complexity. In this model, enterocytes represent the epithelial barrier and goblet cells limit C. albicans adhesion and invasion. Significant protection against C. albicans-induced necrotic damage was achieved by the introduction of a microbiota of antagonistic lactobacilli. We demonstrated a time-, dose- and species-dependent protective effect against C. albicans-induced cytotoxicity. This required bacterial growth, which relied on the presence of host cells, but was not dependent on the competition for adhesion sites. Lactobacillus rhamnosus reduced hyphal elongation, a key virulence attribute. Furthermore, bacterial-driven shedding of hyphae from the epithelial surface, associated with apoptotic epithelial cells, was identified as a main and novel mechanism of damage protection. However, host cell apoptosis was not the driving mechanism behind shedding. Collectively, we established an in vitro gut model that can be used to experimentally dissect commensal-like interactions of C. albicans with a bacterial microbiota and the host epithelial barrier. We also discovered fungal shedding as a novel mechanism by which bacteria contribute to the protection of epithelial surfaces.This article has an associated First Person interview with the joint first authors of the paper.
Collapse
Affiliation(s)
- Katja Graf
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Antonia Last
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Rena Gratz
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Stefanie Allert
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Susanne Linde
- Center for Electron Microscopy Jena University Hospital, Ziegelmühlenweg 1, 07743 Jena, Germany
| | - Martin Westermann
- Center for Electron Microscopy Jena University Hospital, Ziegelmühlenweg 1, 07743 Jena, Germany
| | - Marko Gröger
- Center for Sepsis Control and Care (CSCC), University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany
| | - Alexander S Mosig
- Center for Sepsis Control and Care (CSCC), University Hospital Jena, Am Klinikum 1, 07747 Jena, Germany.,Institute of Biochemistry II, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany
| | - Mark S Gresnigt
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans-Knoell-Institute, Beutenbergstraße 11A, 07745 Jena, Germany .,Friedrich Schiller University, Fürstengraben 1, 07743 Jena, Germany
| |
Collapse
|
37
|
Vaginal microbiota of asymptomatic bacterial vaginosis and vulvovaginal candidiasis: Are they different from normal microbiota? Microb Pathog 2019; 134:103599. [DOI: 10.1016/j.micpath.2019.103599] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 03/29/2019] [Accepted: 06/11/2019] [Indexed: 12/22/2022]
|
38
|
Krüger W, Vielreicher S, Kapitan M, Jacobsen ID, Niemiec MJ. Fungal-Bacterial Interactions in Health and Disease. Pathogens 2019; 8:E70. [PMID: 31117285 PMCID: PMC6630686 DOI: 10.3390/pathogens8020070] [Citation(s) in RCA: 142] [Impact Index Per Article: 23.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 05/02/2019] [Accepted: 05/16/2019] [Indexed: 12/28/2022] Open
Abstract
Fungi and bacteria encounter each other in various niches of the human body. There, they interact directly with one another or indirectly via the host response. In both cases, interactions can affect host health and disease. In the present review, we summarized current knowledge on fungal-bacterial interactions during their commensal and pathogenic lifestyle. We focus on distinct mucosal niches: the oral cavity, lung, gut, and vagina. In addition, we describe interactions during bloodstream and wound infections and the possible consequences for the human host.
Collapse
Affiliation(s)
- Wibke Krüger
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
| | - Sarah Vielreicher
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
| | - Mario Kapitan
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
- Center for Sepsis Control and Care, Jena 07747, Germany.
| | - Ilse D Jacobsen
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
- Center for Sepsis Control and Care, Jena 07747, Germany.
- Institute of Microbiology, Friedrich Schiller University, Jena 07743, Germany.
| | - Maria Joanna Niemiec
- Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute, Jena 07745, Germany.
- Center for Sepsis Control and Care, Jena 07747, Germany.
| |
Collapse
|
39
|
Kosgey JC, Jia L, Fang Y, Yang J, Gao L, Wang J, Nyamao R, Cheteu M, Tong D, Wekesa V, Vasilyeva N, Zhang F. Probiotics as antifungal agents: Experimental confirmation and future prospects. J Microbiol Methods 2019; 162:28-37. [PMID: 31071354 DOI: 10.1016/j.mimet.2019.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Revised: 04/30/2019] [Accepted: 05/02/2019] [Indexed: 12/13/2022]
Abstract
Fungal burden throughout the world is very high and it keeps escalating due to increasing numbers of immunocompromised individuals. In contrast, the drugs used in management of fungal infections are so few some with high toxicity. Furthermore, highly resistant fungal pathogens are emerging for example Candida auris, Candida glabrata, Candida gullemondii and Aspergillus species among others. Thus now, more than ever, there is a need for combined efforts and an all round search for possible solutions to curb these problems. Therefore, the role of probiotics in management of fungal infections is indispensable. In fact, the antimicrobial activity of probiotics has been screened with promising results against microbial pathogens. Although, recent reports indicated that probiotics may also contribute to protect against fungal infections, the research done in checking antifungal activity of probiotics has used varied technology. This calls for harmonization of the methods used to screen and confirm the antimicrobial activity of probiotics and other candidate microorganisms. We therefore sought to address issues of disparity in probiotic research and their outcomes. Thus this paper is in order as it comprehensively reviews' publications, provides a summary of the methods and future prospects of probiotics as antifungal agents.
Collapse
Affiliation(s)
- Janet Cheruiyot Kosgey
- Department of Microbiology, Harbin Medical University, Harbin 150081, China; WU Lien-Teh Institute, Harbin Medical University, Harbin 150081, China; School of biological and life sciences, Technical University of Kenya, 52428-00200, Kenya
| | - Lina Jia
- Department of Microbiology, Harbin Medical University, Harbin 150081, China; WU Lien-Teh Institute, Harbin Medical University, Harbin 150081, China
| | - Yong Fang
- Department of Microbiology, Harbin Medical University, Harbin 150081, China; WU Lien-Teh Institute, Harbin Medical University, Harbin 150081, China
| | - Jianxun Yang
- WU Lien-Teh Institute, Harbin Medical University, Harbin 150081, China; Department of Dermatology, The 2nd Hospital of Harbin Medical University, Harbin 150081, China
| | - Lei Gao
- Department of Microbiology, Harbin Medical University, Harbin 150081, China; Electron Microscopy Center, Basic Medical Science College, Harbin Medical University, China
| | - Jielin Wang
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Rose Nyamao
- Department of Microbiology, Harbin Medical University, Harbin 150081, China; WU Lien-Teh Institute, Harbin Medical University, Harbin 150081, China
| | - Martin Cheteu
- Department of Microbiology, Harbin Medical University, Harbin 150081, China; WU Lien-Teh Institute, Harbin Medical University, Harbin 150081, China
| | - Dandan Tong
- Department of Microbiology, Harbin Medical University, Harbin 150081, China
| | - Vitalis Wekesa
- School of biological and life sciences, Technical University of Kenya, 52428-00200, Kenya; Flamingo Horticulture, Dudutech Division, P.O Box 1927, 20117, Naivasha, Kenya
| | - Natalia Vasilyeva
- Kashkin Research Institute of Medical Mycology, Department of Microbiology, North-Western State Medical University named after Machnikov, Saint Petersburg, Russia
| | - Fengmin Zhang
- Department of Microbiology, Harbin Medical University, Harbin 150081, China; WU Lien-Teh Institute, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|