1
|
Yüksel E, Kort R, Voragen AGJ. Structure and degradation dynamics of dietary pectin. Crit Rev Food Sci Nutr 2024:1-20. [PMID: 39681562 DOI: 10.1080/10408398.2024.2437573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2024]
Abstract
Pectin, a complex dietary fiber, constitutes a key structural component of the cell walls of numerous edible plant products. It is resistant to digestion by human enzymes and undergoes depolymerization and saccharification in the gastrointestinal tract through the action of carbohydrate-active enzymes (CAZymes) produced by gut microbiota. This enzymatic breakdown generates intermediate structural fragments, which are subsequently converted into pectin oligosaccharides (POS) and monosaccharides. POS exhibit prebiotic properties and have demonstrated potential health benefits, including anti-carcinogenic effects, mucoadhesive capabilities, and the promotion of beneficial gut bacterial growth. However, the current understanding of the molecular structure of pectin and its degradation dynamics remains fragmented within the literature, impeding progress in dietary fiber intervention research and the development of personalized nutrition approaches. This review aims to provide a comprehensive overview of the structural features of pectin and the intricate breakdown mechanisms orchestrated by CAZymes. It underscores the complex architecture of pectin that influences its breakdown dynamics and specifies the enzymatic requirements for the cleavage of its diverse structural components. These insights complement our accompanying review on the structure-function relationships between pectin and the human gut microbiota, previously published in this journal.
Collapse
Affiliation(s)
- Ecem Yüksel
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije University Amsterdam, Amsterdam, The Netherlands
| | - Remco Kort
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije University Amsterdam, Amsterdam, The Netherlands
- ARTIS-Micropia, Amsterdam, The Netherlands
| | - Alphons G J Voragen
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
- Keep Food Simple, Driebergen, The Netherlands
| |
Collapse
|
2
|
da Silva TF, Glória RDA, Americo MF, Freitas ADS, de Jesus LCL, Barroso FAL, Laguna JG, Coelho-Rocha ND, Tavares LM, le Loir Y, Jan G, Guédon É, Azevedo VADC. Unlocking the Potential of Probiotics: A Comprehensive Review on Research, Production, and Regulation of Probiotics. Probiotics Antimicrob Proteins 2024; 16:1687-1723. [PMID: 38539008 DOI: 10.1007/s12602-024-10247-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/12/2024] [Indexed: 10/02/2024]
Abstract
This review provides a comprehensive overview of the current state of probiotic research, covering a wide range of topics, including strain identification, functional characterization, preclinical and clinical evaluations, mechanisms of action, therapeutic applications, manufacturing considerations, and future directions. The screening process for potential probiotics involves phenotypic and genomic analysis to identify strains with health-promoting properties while excluding those with any factor that could be harmful to the host. In vitro assays for evaluating probiotic traits such as acid tolerance, bile metabolism, adhesion properties, and antimicrobial effects are described. The review highlights promising findings from in vivo studies on probiotic mitigation of inflammatory bowel diseases, chemotherapy-induced mucositis, dysbiosis, obesity, diabetes, and bone health, primarily through immunomodulation and modulation of the local microbiota in human and animal models. Clinical studies demonstrating beneficial modulation of metabolic diseases and human central nervous system function are also presented. Manufacturing processes significantly impact the growth, viability, and properties of probiotics, and the composition of the product matrix and supplementation with prebiotics or other strains can modify their effects. The lack of regulatory oversight raises concerns about the quality, safety, and labeling accuracy of commercial probiotics, particularly for vulnerable populations. Advancements in multi-omics approaches, especially probiogenomics, will provide a deeper understanding of the mechanisms behind probiotic functionality, allowing for personalized and targeted probiotic therapies. However, it is crucial to simultaneously focus on improving manufacturing practices, implementing quality control standards, and establishing regulatory oversight to ensure the safety and efficacy of probiotic products in the face of increasing therapeutic applications.
Collapse
Affiliation(s)
- Tales Fernando da Silva
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Rafael de Assis Glória
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Monique Ferrary Americo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Andria Dos Santos Freitas
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Luis Claudio Lima de Jesus
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Fernanda Alvarenga Lima Barroso
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Juliana Guimarães Laguna
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Nina Dias Coelho-Rocha
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Laisa Macedo Tavares
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Yves le Loir
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Gwénaël Jan
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Éric Guédon
- UMR1253, INRAE, L'Institut Agro Rennes Angers, STLO, Rennes, France
| | - Vasco Ariston de Carvalho Azevedo
- Institute of Biological Sciences, Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
3
|
Yüksel E, Voragen AGJ, Kort R. The pectin metabolizing capacity of the human gut microbiota. Crit Rev Food Sci Nutr 2024:1-23. [PMID: 39264366 DOI: 10.1080/10408398.2024.2400235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
The human gastrointestinal microbiota, densely populated with a diverse array of microorganisms primarily from the bacterial phyla Bacteroidota, Bacillota, and Actinomycetota, is crucial for maintaining health and physiological functions. Dietary fibers, particularly pectin, significantly influence the composition and metabolic activity of the gut microbiome. Pectin is fermented by gut bacteria using carbohydrate-active enzymes (CAZymes), resulting in the production of short-chain fatty acids (SCFAs) such as acetate, propionate, and butyrate, which provide various health benefits. The gastrointestinal microbiota has evolved to produce CAZymes that target different pectin components, facilitating cross-feeding within the microbial community. This review explores the fermentation of pectin by various gut bacteria, focusing on the involved transport systems, CAZyme families, SCFA synthesis capacity, and effects on microbial ecology in the gut. It addresses the complexities of the gut microbiome's response to pectin and highlights the importance of microbial cross-feeding in maintaining a balanced and diverse gut ecosystem. Through a systematic analysis of pectinolytic CAZyme production, this review provides insights into the enzymatic mechanisms underlying pectin degradation and their broader implications for human health, paving the way for more targeted and personalized dietary strategies.
Collapse
Affiliation(s)
- Ecem Yüksel
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Alphons G J Voragen
- Keep Food Simple, Driebergen, The Netherlands
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, The Netherlands
| | - Remco Kort
- Amsterdam Institute for Life and Environment (A-LIFE), Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
- ARTIS-Micropia, Amsterdam, The Netherlands
| |
Collapse
|
4
|
Song Z, Ge Y, Yu X, Liu R, Liu C, Cheng K, Guo L, Yao S. Development of a single nucleotide polymorphism-based strain-identified method for Streptococcus thermophilus CICC 6038 and Lactobacillus delbrueckii ssp. bulgaricus CICC 6047 using pan-genomics analysis. J Dairy Sci 2024; 107:4248-4258. [PMID: 38246550 DOI: 10.3168/jds.2023-23655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024]
Abstract
The health benefits conferred by probiotics is specific to individual probiotic strains, highlighting the importance of identifying specific strains for research and production purposes. Streptococcus thermophilus CICC 6038 and Lactobacillus delbrueckii ssp. bulgaricus CICC 6047 are exceedingly valuable for commercial use with an excellent mixed-culture fermentation. To differentiate these 2 strains from other S. thermophilus and L. delbrueckii ssp. bulgaricus, a specific, sensitive, accurate, rapid, convenient, and cost-effective method is required. In this study, we conducted a pan-genome analysis of S. thermophilus and L. delbrueckii ssp. bulgaricus to identify species-specific core genes, along with strain-specific SNPs. These genes were used to develop suitable PCR primers, and the conformity of sequence length and unique SNPs was confirmed by sequencing for qualitative identification at the strain level. The results demonstrated that SNPs analysis of PCR products derived from these primers could distinguish CICC 6038 and CICC 6047 accurately and reproducibly from the other strains of S. thermophilus and L. delbrueckii ssp. bulgaricus, respectively. The strain-specific PCR method based on SNPs herein is universally applicable for probiotics identification. It offers valuable insights into identifying probiotics at the strain level that is fit-for-purpose in quality control and compliance assessment of commercial dairy products.
Collapse
Affiliation(s)
- Zhiquan Song
- China National Research Institute of Food and Fermentation Industries Co. Ltd., China Center of Industrial Culture Collection, Beijing, 100015, China
| | - Yuanyuan Ge
- China National Research Institute of Food and Fermentation Industries Co. Ltd., China Center of Industrial Culture Collection, Beijing, 100015, China; Beijing Forestry University, College of Biological Sciences and Biotechnology, Beijing, 100083, China
| | - Xuejian Yu
- China National Research Institute of Food and Fermentation Industries Co. Ltd., China Center of Industrial Culture Collection, Beijing, 100015, China
| | - Rui Liu
- China National Research Institute of Food and Fermentation Industries Co. Ltd., China Center of Industrial Culture Collection, Beijing, 100015, China
| | - Chong Liu
- China National Research Institute of Food and Fermentation Industries Co. Ltd., China Center of Industrial Culture Collection, Beijing, 100015, China
| | - Kun Cheng
- China National Research Institute of Food and Fermentation Industries Co. Ltd., China Center of Industrial Culture Collection, Beijing, 100015, China
| | - Lizheng Guo
- China National Research Institute of Food and Fermentation Industries Co. Ltd., China Center of Industrial Culture Collection, Beijing, 100015, China
| | - Su Yao
- China National Research Institute of Food and Fermentation Industries Co. Ltd., China Center of Industrial Culture Collection, Beijing, 100015, China.
| |
Collapse
|
5
|
Li H, Leng C, Chen N, Ding Q, Yuan Y, Zheng Y, Zhu G, Chen C, Xu L, Shuai J, Jiang Q, Ren D, Wang H. Lactic acid bacteria reduce bacterial diarrhea in rabbits via enhancing immune function and restoring intestinal microbiota homeostasis. BMC Vet Res 2024; 20:151. [PMID: 38643127 PMCID: PMC11031951 DOI: 10.1186/s12917-024-03981-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Accepted: 03/17/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Numerous previous reports have demonstrated the efficacy of Lactic acid bacteria (LAB) in promoting growth and preventing disease in animals. In this study, Enterococcus faecium ZJUIDS-R1 and Ligilactobaciiius animalis ZJUIDS-R2 were isolated from the feces of healthy rabbits, and both strains showed good probiotic properties in vitro. Two strains (108CFU/ml/kg/day) were fed to weaned rabbits for 21 days, after which specific bacterial infection was induced to investigate the effects of the strains on bacterial diarrhea in the rabbits. RESULTS Our data showed that Enterococcus faecium ZJUIDS-R1 and Ligilactobaciiius animalis ZJUIDS-R2 interventions reduced the incidence of diarrhea and systemic inflammatory response, alleviated intestinal damage and increased antibody levels in animals. In addition, Enterococcus faecium ZJUIDS-R1 restored the flora abundance of Ruminococcaceae1. Ligilactobaciiius animalis ZJUIDS-R2 up-regulated the flora abundance of Adlercreutzia and Candidatus Saccharimonas. Both down-regulated the flora abundance of Shuttleworthia and Barnesiella to restore intestinal flora balance, thereby increasing intestinal short-chain fatty acid content. CONCLUSIONS These findings suggest that Enterococcus faecium ZJUIDS-R1 and Ligilactobaciiius animalis ZJUIDS-R2 were able to improve intestinal immunity, produce organic acids and regulate the balance of intestinal flora to enhance disease resistance and alleviate diarrhea-related diseases in weanling rabbits.
Collapse
Affiliation(s)
- Huimin Li
- College of Animal Sciences, Zhejiang University, 310058, Hangzhou, PR China
- Henan Provincial Engineering and Technology Center of Animal Disease Diagnosis and Integrated Control, Henan Key Laboratory of Insect Biology in Funiu Mountain, Nanyang Normal University, 473061, Nanyang, PR China
- Zhejiang Jinuo Saibur Biotechnology Co., LTD, 310010, Hangzhou, PR China
| | - Chaoliang Leng
- Henan Provincial Engineering and Technology Center of Animal Disease Diagnosis and Integrated Control, Henan Key Laboratory of Insect Biology in Funiu Mountain, Nanyang Normal University, 473061, Nanyang, PR China
| | - Nan Chen
- College of Animal Sciences, Zhejiang University, 310058, Hangzhou, PR China
| | - Qinchao Ding
- College of Animal Sciences, Zhejiang University, 310058, Hangzhou, PR China
| | - Yizhao Yuan
- College of Animal Sciences, Zhejiang University, 310058, Hangzhou, PR China
| | - Yilei Zheng
- College of Animal Sciences, Zhejiang University, 310058, Hangzhou, PR China
| | - Ge Zhu
- College of Animal Sciences, Zhejiang University, 310058, Hangzhou, PR China
| | - Chen Chen
- College of Animal Sciences, Zhejiang University, 310058, Hangzhou, PR China
| | - Lichang Xu
- College of Animal Sciences, Zhejiang University, 310058, Hangzhou, PR China
| | - Jiangbing Shuai
- Zhejiang Academy of Science & Technology for Inspection & Quarantine, 310016, Hangzhou, PR China
| | - Qinting Jiang
- Zhejiang Academy of Science & Technology for Inspection & Quarantine, 310016, Hangzhou, PR China
| | - Daxi Ren
- College of Animal Sciences, Zhejiang University, 310058, Hangzhou, PR China
| | - Huanan Wang
- College of Animal Sciences, Zhejiang University, 310058, Hangzhou, PR China.
- , Room 515 E Building, 866 Yuhangtang Rd, 310058, Hangzhou, Zhejiang, China.
| |
Collapse
|
6
|
Quintieri L, Fanelli F, Monaci L, Fusco V. Milk and Its Derivatives as Sources of Components and Microorganisms with Health-Promoting Properties: Probiotics and Bioactive Peptides. Foods 2024; 13:601. [PMID: 38397577 PMCID: PMC10888271 DOI: 10.3390/foods13040601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 01/31/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
Milk is a source of many valuable nutrients, including minerals, vitamins and proteins, with an important role in adult health. Milk and dairy products naturally containing or with added probiotics have healthy functional food properties. Indeed, probiotic microorganisms, which beneficially affect the host by improving the intestinal microbial balance, are recognized to affect the immune response and other important biological functions. In addition to macronutrients and micronutrients, biologically active peptides (BPAs) have been identified within the amino acid sequences of native milk proteins; hydrolytic reactions, such as those catalyzed by digestive enzymes, result in their release. BPAs directly influence numerous biological pathways evoking behavioral, gastrointestinal, hormonal, immunological, neurological, and nutritional responses. The addition of BPAs to food products or application in drug development could improve consumer health and provide therapeutic strategies for the treatment or prevention of diseases. Herein, we review the scientific literature on probiotics, BPAs in milk and dairy products, with special attention to milk from minor species (buffalo, sheep, camel, yak, donkey, etc.); safety assessment will be also taken into consideration. Finally, recent advances in foodomics to unveil the probiotic role in human health and discover novel active peptide sequences will also be provided.
Collapse
Affiliation(s)
| | - Francesca Fanelli
- National Research Council of Italy, Institute of Sciences of Food Production (CNR-ISPA), 70126 Bari, Italy; (L.Q.); (L.M.); (V.F.)
| | | | | |
Collapse
|
7
|
Fusco V, Fanelli F, Chieffi D. Recent and Advanced DNA-Based Technologies for the Authentication of Probiotic, Protected Designation of Origin (PDO) and Protected Geographical Indication (PGI) Fermented Foods and Beverages. Foods 2023; 12:3782. [PMID: 37893675 PMCID: PMC10606304 DOI: 10.3390/foods12203782] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/26/2023] [Accepted: 10/10/2023] [Indexed: 10/29/2023] Open
Abstract
The authenticity of probiotic products and fermented foods and beverages that have the status of protected designation of origin (PDO) or geographical indication (PGI) can be assessed via numerous methods. DNA-based technologies have emerged in recent decades as valuable tools to achieve food authentication, and advanced DNA-based methods and platforms are being developed. The present review focuses on the recent and advanced DNA-based techniques for the authentication of probiotic, PDO and PGI fermented foods and beverages. Moreover, the most promising DNA-based detection tools are presented. Strain- and species-specific DNA-based markers of microorganisms used as starter cultures or (probiotic) adjuncts for the production of probiotic and fermented food and beverages have been exploited for valuable authentication in several detection methods. Among the available technologies, propidium monoazide (PMA) real-time polymerase chain reaction (PCR)-based technologies allow for the on-time quantitative detection of viable microbes. DNA-based lab-on-a-chips are promising devices that can be used for the on-site and on-time quantitative detection of microorganisms. PCR-DGGE and metagenomics, even combined with the use of PMA, are valuable tools allowing for the fingerprinting of the microbial communities, which characterize PDO and PGI fermented foods and beverages, and they are necessary for authentication besides permitting the detection of extra or mislabeled species in probiotic products. These methods, in relation to the authentication of probiotic foods and beverages, need to be used in combination with PMA, culturomics or flow cytometry to allow for the enumeration of viable microorganisms.
Collapse
Affiliation(s)
- Vincenzina Fusco
- Institute of Sciences of Food Production, National Research Council of Italy (CNR-ISPA), 70126 Bari, Italy; (F.F.); (D.C.)
| | | | | |
Collapse
|
8
|
Salvador PBU, Dalmacio LMM, Kim SH, Kang DK, Balolong MP. Immunomodulatory potential of four candidate probiotic Lactobacillus strains from plant and animal origin using comparative genomic analysis. Access Microbiol 2022; 3:000299. [PMID: 35024559 PMCID: PMC8749136 DOI: 10.1099/acmi.0.000299] [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: 10/20/2020] [Accepted: 11/02/2021] [Indexed: 12/11/2022] Open
Abstract
Probiotic strains from different origins have shown promise in recent decades for their health benefits, for example in promoting and regulating the immune system. The immunomodulatory potential of four Lactobacillus strains from animal and plant origins was evaluated in this paper based on their genomic information. Comparative genomic analysis was performed through genome alignment, average nucleotide identity (ANI) analysis and gene mining for putative immunomodulatory genes. The genomes of the four Lactobacillus strains show relative similarities in multiple regions, as observed in the genome alignment. However, ANI analysis showed that L. mucosae LM1 and L. fermentum SK152 are the most similar when considering their nucleotide sequences alone. Gene mining of putative immunomodulatory genes studied from L. plantarum WCFS1 yielded multiple results in the four potential probiotic strains, with L. plantarum SK151 showing the largest number of genes at around 74 hits, followed by L. johnsonii PF01 at 41 genes when adjusted for matches with at least 30 % identity. Looking at the immunomodulatory genes in each strain, L. plantarum SK151 and L. johnsonii PF01 may have wider activity, covering both immune activation and immune suppression, as compared to L. mucosae LM1 and L. fermentum SK152, which could be more effective in activating immune cells and the pro-inflammatory cascade rather than suppressing it. The similarities and differences between the four Lactobacillus species showed that there is no definitive trend based on the origin of isolation alone. Moreover, higher percentage identities between genomes do not directly correlate with higher similarities in potential activity, such as in immunomodulation. The immunomodulatory function of each of the four Lactobacillus strains should be observed and verified experimentally in the future, since some the activity of some genes may be strain-specific, which would not be identified through comparative genomics alone.
Collapse
Affiliation(s)
- Paul Benedic U Salvador
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Ermita, Manila 1000, Philippines
| | - Leslie Michelle M Dalmacio
- Department of Biochemistry and Molecular Biology, College of Medicine, University of the Philippines Manila, Ermita, Manila 1000, Philippines
| | - Sang Hoon Kim
- Department of Animal Resources Science, College of Biotechnology and Bioengineering, Dankook University, Republic of Korea
| | - Dae-Kyung Kang
- Department of Animal Resources Science, College of Biotechnology and Bioengineering, Dankook University, Republic of Korea
| | - Marilen P Balolong
- Department of Biology, College of Arts and Sciences, University of the Philippines Manila, Ermita, Manila 1000, Philippines
| |
Collapse
|
9
|
Lactobacillus rhamnosus induces CYP3A and changes the pharmacokinetics of verapamil in rats. Toxicol Lett 2021; 352:46-53. [PMID: 34600097 DOI: 10.1016/j.toxlet.2021.09.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/16/2021] [Accepted: 09/27/2021] [Indexed: 12/19/2022]
Abstract
Verapamil, a calcium channel blocker, has been approved as the first-line drug for treatment of angina pectoris, hypertension and supraventricular tachycardia. Lactobacillus rhamnosus, one of the normal strains in human intestinal tract, is very popular in the probiotic market for conferring a health benefit on the host. This report investigated the potential of gut microbiota-drug interactions between lactobacillus rhamnosus and verapamil via using wild type (WT) and Cyp3a1/2 knockout (KO) rats. In WT rats, administration of Lactobacillus rhamnosus for 14 days decreased systemic exposure of verapamil and increased its metabolite norverapamil in vivo, and resulted in gut microbiota-drug interactions. In Cyp3a1/2 KO rats, however, this interaction disappeared. Further studies found that Lactobacillus rhamnosus induced CYP3A activity and expression, and changed the composition of gut microbiota, thus changing the pharmacokinetics of verapamil. These results demonstrated the interaction between lactobacillus rhamnosus and verapamil, and indicated that the effect of gut microbiota on metabolic enzymes cannot be ignored.
Collapse
|
10
|
Abramov VM, Kosarev IV, Priputnevich TV, Machulin AV, Abashina TN, Chikileva IO, Donetskova AD, Takada K, Melnikov VG, Vasilenko RN, Khlebnikov VS, Samoilenko VA, Nikonov IN, Sukhikh GT, Uversky VN, Karlyshev AV. S-layer protein 2 of vaginal Lactobacillus crispatus 2029 enhances growth, differentiation, VEGF production and barrier functions in intestinal epithelial cell line Caco-2. Int J Biol Macromol 2021; 189:410-419. [PMID: 34437917 DOI: 10.1016/j.ijbiomac.2021.08.150] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/11/2021] [Accepted: 08/18/2021] [Indexed: 12/15/2022]
Abstract
We have previously demonstrated the ability of the human vaginal strain Lactobacillus crispatus 2029 (LC2029) for strong adhesion to cervicovaginal epithelial cells, expression of the surface layer protein 2 (Slp2), and antagonistic activity against urogenital pathogens. Slp2 forms regular two-dimensional structure around the LC2029 cells,which is secreted into the medium and inhibits intestinal pathogen-induced activation of caspase-9 and caspase-3 in the human intestinal Caco-2 cells. Here, we elucidated the effects of soluble Slp2 on adhesion of proteobacteria pathogens inducing necrotizing enterocolitis (NEC), such as Escherichia coli ATCC E 2348/69, E. coli ATCC 31705, Salmonella Enteritidis ATCC 13076, Campylobacter jejuni ATCC 29428, and Pseudomonas aeruginosa ATCC 27853 to Caco-2 cells, as well as on growth promotion, differentiation, vascular endothelial growth factor (VEGF) production, and intestinal barrier function of Caco-2 cell monolayers. Slp2 acts as anti-adhesion agent for NEC-inducing proteobacteria, promotes growth of immature Caco-2 cells and their differentiation, and enhances expression and functional activity of sucrase, lactase, and alkaline phosphatase. Slp2 stimulates VEGF production, decreases paracellular permeability, and increases transepithelial electrical resistance, strengthening barrier function of Caco-2 cell monolayers. These data support the important role of Slp2 in the early postnatal development of the human small intestine enterocytes.
Collapse
Affiliation(s)
- Vyacheslav M Abramov
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Igor V Kosarev
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Tatiana V Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Andrey V Machulin
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Tatiana N Abashina
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Irina O Chikileva
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia; Laboratory of Cell Immunity, Blokhin National Research, Center of Oncology Ministry of Health RF, Moscow 115478, Russia
| | | | - Kazuhide Takada
- Division of Microbiology, Department of Pathology and Microbiology, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Vyacheslav G Melnikov
- Gabrichevsky Moscow Research Institute of Epidemiology and Microbiology, Federal Service for Supervision of Consumer Rights Protection and Human Welfare, Moscow 152212, Russia
| | - Raisa N Vasilenko
- Institute of Immunological Engineering, Lyubuchany 142380, Moscow Region, Russia
| | | | - Vladimir A Samoilenko
- Skryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", Pushchino 142290, Moscow Region, Russia
| | - Ilya N Nikonov
- Federal State Education Institution of Higher Professional Education Moscow State Academy of Veterinary Medicine and Biotechnology named after K.I. Skryabin, Moscow 109472, Russia
| | - Gennady T Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, Moscow 117997, Russia
| | - Vladimir N Uversky
- Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Andrey V Karlyshev
- Department of Science, Engineering and Computing, Kingston University London, Kingston upon Thames KT1 2EE, UK
| |
Collapse
|
11
|
Dutra-Silva L, Matteoli FP, Arisi ACM. Distribution of Genes Related to Probiotic Effects Across Lacticaseibacillus rhamnosus Revealed by Population Structure. Probiotics Antimicrob Proteins 2021; 15:548-557. [PMID: 34699013 DOI: 10.1007/s12602-021-09868-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/19/2021] [Indexed: 11/24/2022]
Abstract
The Gram-positive Lacticaseibacillus rhamnosus has been broadly reported as capable of exerting beneficial health effects. Bacterial genomic diversity may promote niche specialization, thus creating subpatterns within populations. As L. rhamnosus advantageous effects have been widely reported at strain level and few is known regarding the distribution of beneficial genes among L. rhamnosus strains, we investigated all publicly available genomes of Lactobacillus and Lacticaseibacillus genera to study the pangenome and general population structure of L. rhamnosus. Core genome multilocus sequence typing detected eight L. rhamnosus phylogroups (PG1 to PG8). L. rhamnosus harbors an open pangenome; PG1, PG3, PG4, and PG5 exhibited highly conserved gene distribution patterns. Genes significantly associated to the PG1, which comprises L. rhamnosus GG, are mainly phage-related. The adhesion operon spaCBA-srtC1 was found in 44 (24.7%) genomes; however, considering only the PG1, the prevalence was of 65%. In PG2 the spaCBA-srtC1 prevalence was of 43%. Nevertheless, both human and milk-derived strains harbored this operon. Further, two main types of bacteriocin clusters were found (Bact1 and Bact2). Bact1 predictions indicate the presence of garQ, encoding the class II bacteriocin garvieacin Q, that is mainly present in the closely related PG8A and a PG2 subcluster. PG2 harbors two distinct subclusters, harboring either spaCBA-srtC1 or Bact1. Our findings provide novel insights on the distribution of biotechnological relevant genes across L. rhamnosus population, uncovering intra-species patterns that may bring forth the development of more efficient probiotic products.
Collapse
Affiliation(s)
- Lorena Dutra-Silva
- Food Science and Technology Department, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Filipe P Matteoli
- Department of Soil Science, Luiz de Queiroz College of Agriculture, Piracicaba, SP, Brazil.
| | | |
Collapse
|
12
|
Klotz C, Goh YJ, O'Flaherty S, Barrangou R. S-layer associated proteins contribute to the adhesive and immunomodulatory properties of Lactobacillus acidophilus NCFM. BMC Microbiol 2020; 20:248. [PMID: 32787778 PMCID: PMC7425073 DOI: 10.1186/s12866-020-01908-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Accepted: 07/16/2020] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Surface layers (S-layers) are two-dimensional crystalline arrays of repeating proteinaceous subunits that form the outermost layer of many bacterial cell envelopes. Within the Lactobacillus genus, S-layer presence is frequently associated with probiotic-relevant properties such as improved adherence to host epithelial cells and modulation of the immune response. However, recent studies have demonstrated that certain S-layer functions may be supplemented by a novel subset of proteins embedded within its lattice, termed S-layer associated proteins (SLAPs). In the following study, four Lactobacillus acidophilus NCFM SLAPs (LBA0046, LBA0864, LBA1426, and LBA1539) were selected for in silico and phenotypic assessment. RESULTS Despite lacking any sequence similarity or catalytic domains that may indicate function, the genes encoding the four proteins of interest were shown to be unique to S-layer-forming, host-adapted lactobacilli species. Likewise, their corresponding deletion mutants exhibited broad, host-relevant phenotypes including decreased inflammatory profiles and reduced adherence to Caco-2 intestinal cells, extracellular matrices, and mucin in vitro. CONCLUSIONS Overall, the data presented in this study collectively links several previously uncharacterized extracellular proteins to roles in the underlying host adaptive mechanisms of L. acidophilus.
Collapse
Affiliation(s)
- Courtney Klotz
- Genomic Sciences Graduate Program North Carolina State University, Raleigh, NC, USA.,Department of Food, Bioprocessing & Nutrition Sciences, North Carolina State University, Raleigh, NC, USA
| | - Yong Jun Goh
- Department of Food, Bioprocessing & Nutrition Sciences, North Carolina State University, Raleigh, NC, USA
| | - Sarah O'Flaherty
- Department of Food, Bioprocessing & Nutrition Sciences, North Carolina State University, Raleigh, NC, USA
| | - Rodolphe Barrangou
- Genomic Sciences Graduate Program North Carolina State University, Raleigh, NC, USA. .,Department of Food, Bioprocessing & Nutrition Sciences, North Carolina State University, Raleigh, NC, USA.
| |
Collapse
|
13
|
Klotz C, Goh YJ, O'Flaherty S, Johnson B, Barrangou R. Deletion of S-Layer Associated Ig-Like Domain Protein Disrupts the Lactobacillus acidophilus Cell Surface. Front Microbiol 2020; 11:345. [PMID: 32256464 PMCID: PMC7090030 DOI: 10.3389/fmicb.2020.00345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 02/17/2020] [Indexed: 01/18/2023] Open
Abstract
Bacterial surface-layers (S-layers) are crystalline arrays of repeating proteinaceous subunits that coat the exterior of many cell envelopes. S-layers have demonstrated diverse functions in growth and survival, maintenance of cell integrity, and mediation of host interactions. Additionally, S-layers can act as scaffolds for the outward display of auxiliary proteins and glycoproteins. These non-covalently bound S-layer associated proteins (SLAPs) have characterized roles in cell division, adherence to intestinal cells, and modulation of the host immune response. Recently, IgdA (LBA0695), a Lactobacillus acidophilus SLAP that possesses a Group 3 immunoglobulin (Ig)-like domain and GW (Gly-Tryp) dipeptide surface anchor, was recognized for its high conservation among S-layer-forming lactobacilli, constitutive expression, and surface localization. These findings prompted its selection for examination within the present study. Although IgdA and corresponding orthologs were shown to be unique to host-adapted lactobacilli, the Ig domain itself was specific to vertebrate-adapted species suggesting a role in vertebrate adaptation. Using a counterselective gene replacement system, igdA was deleted from the L. acidophilus NCFM chromosome. The resultant mutant, NCK2532, exhibited a visibly disrupted cell surface which likely contributed to its higher salt sensitivity, severely reduced adhesive capacity, and altered immunogenicity profile. Transcriptomic analyses revealed the induction of several stress response genes and secondary surface proteins. Due to the broad impact of IgdA on the cellular physiology and probiotic attributes of L. acidophilus, identification of similar proteins in alternative bacterial species may help pinpoint next-generation host-adapted probiotic candidates.
Collapse
Affiliation(s)
- Courtney Klotz
- Genomic Sciences Graduate Program, North Carolina State University, Raleigh, NC, United States.,Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC, United States
| | - Yong Jun Goh
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC, United States
| | - Sarah O'Flaherty
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC, United States
| | - Brant Johnson
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC, United States.,Microbiology Graduate Program, North Carolina State University, Raleigh, NC, United States
| | - Rodolphe Barrangou
- Genomic Sciences Graduate Program, North Carolina State University, Raleigh, NC, United States.,Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, Raleigh, NC, United States.,Microbiology Graduate Program, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
14
|
Abramov VM, Kosarev IV, Priputnevich TV, Machulin AV, Khlebnikov VS, Pchelintsev SY, Vasilenko RN, Sakulin VK, Suzina NE, Chikileva IO, Derysheva EI, Melnikov VG, Nikonov IN, Samoilenko VA, Svetoch EE, Sukhikh GT, Uversky VN, Karlyshev AV. S-layer protein 2 of Lactobacillus crispatus 2029, its structural and immunomodulatory characteristics and roles in protective potential of the whole bacteria against foodborne pathogens. Int J Biol Macromol 2020; 150:400-412. [PMID: 32045605 DOI: 10.1016/j.ijbiomac.2020.02.065] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2020] [Revised: 02/07/2020] [Accepted: 02/07/2020] [Indexed: 01/11/2023]
Abstract
We have previously demonstrated that human vaginal Lactobacillus crispatus 2029 (LC2029) strain is highly adhesive to cervicovaginal epithelial cells, exhibits antagonistic activity against genitourinary pathogens and expresses surface-layer protein (Slp). The aims of the present study were elucidation of Slp structural and immunomodulatory characteristics and its roles in protective properties of the whole vaginal LC2029 bacteria against foodborne pathogens. Enteric Caco-2 and colon HT-29 cell lines were used as the in vitro models of the human intestinal epithelial layer. LC2029 strain has two homologous surface-layer (S-layer) genes, slp1 and slp2. Whilst we found no evidence for the expression of slp1 under the growth conditions used, a very high level of expression of the slp2 gene was detected. C-terminal part of the amino sequence of Slp2 protein was found to be highly similar to that of the conserved C-terminal region of SlpA protein of L. crispatus Zj001 isolated from pig intestines and CbsA protein of L. crispatus JCM5810 isolated from chicken intestines, and was substantially variable at the N-terminal and middle regions. The amino acid sequence identity between SlpA and CbsA was as high as 84%, whilst the identity levels of these sequences with that of Slp2 were only 49% and 50% (respectively). LC2029 strain was found to be both acid and bile tolerant. Survival in simulated gastric and intestinal juices of LC2029 cells unable to produce Slp2 was reduced by 2-3 logs. Vaginal L. crispatus 1385 (LC1385) strain not expressing Slp was also very sensitive to gastric and intestinal stresses. Slp2 was found to be non-covalently bound to the surface of the bacterium, acting as an adhesin and facilitating interaction of LC2029 lactobacilli with the host immature or fully differentiated Caco-2 cells, as well as HT-29 cells. No toxicity to or damage of Caco-2 or HT-29 epithelial cells were detected after 24 h of colonization by LC2029 lactobacilli. Both Slp2 protein and LC2029 cells induced NF-kB activation in Caco-2 and HT-29 cells, but did not induce expression of innate immunity mediators Il-8, Il-1β, and TNF-α. Slp2 and LC2029 inhibited Il-8 production in Caco-2 and HT-29 cells induced by MALP-2 and increased production of anti-inflammatory cytokine Il-6. Slp2 inhibited production of CXCL1 and RANTES by Caco-2 cells during differentiation and maturation process within 15 days. Culturing Caco-2 and HT-29 cells in the presence of Slp2 increased adhesion of bifidobacteria BLI-2780 to these enterocytes. Upon binding to Caco-2 and HT-29 cells, Slp2 protein and LC2029 lactobacilli were recognized by toll-like receptors (TLR) 2/6. It was shown that LC2029 strain is a strong co-aggregator of foodborne pathogens Campylobacter jejuni, Salmonella enteritidis, and Escherichia coli O157:H used in this study. The Slp2 was responsible for the ability of LC2029 to co-aggregate these enteropathogens. Slp2 and intact LC2029 lactobacilli inhibited foodborne pathogen-induced activation of caspase-9 and caspase-3 as apoptotic biomarkers in Caco-2 and HT-29 cells. In addition, Slp2 and Slp2-positive LC2029 strain reduced adhesion of tested pathogenic bacteria to Caco-2 and HT-29 cells. Slp2-positive LC2029 strain but not Slp2 alone provided bactericidal effect on foodborne pathogens. These results suggest a range of mechanisms involved in inhibition of growth, viability, and cell-adhesion properties of pathogenic Proteobacteria by the Slp2 producing LC2029, which may be useful in treatment of necrotizing enterocolitis (NEC) in newborns and foodborne infectious diseases in children and adults, increasing the colonization resistance and maintaining the intestinal homeostasis.
Collapse
Affiliation(s)
- Vyacheslav M Abramov
- Institute of Immunological Engineering, 142380 Lyubuchany, Moscow Region, Russia
| | - Igor V Kosarev
- Institute of Immunological Engineering, 142380 Lyubuchany, Moscow Region, Russia
| | - Tatiana V Priputnevich
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, 117997 Moscow, Russia
| | - Andrey V Machulin
- Scryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", 142290 Pushchino, Moscow Region, Russia
| | | | | | - Raisa N Vasilenko
- Institute of Immunological Engineering, 142380 Lyubuchany, Moscow Region, Russia
| | - Vadim K Sakulin
- Institute of Immunological Engineering, 142380 Lyubuchany, Moscow Region, Russia
| | - Natalia E Suzina
- Scryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", 142290 Pushchino, Moscow Region, Russia
| | - Irina O Chikileva
- Institute of Immunological Engineering, 142380 Lyubuchany, Moscow Region, Russia; Laboratory of Cell Immunity, Blokhin National Research, Center of Oncology Ministry of Health RF, 115478 Moscow, Russia
| | - Evgenia I Derysheva
- Institute for Biological Instrumentation, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", 142290, Pushchino, Moscow Region, Russia
| | - Vyacheslav G Melnikov
- Gabrichevsky Moscow Research Institute of Epidemiology and Microbiology, Federal Service for Supervision of Consumer Rights Protection and Human Welfare, 152212 Moscow, Russia
| | - Ilya N Nikonov
- Federal Research Center "All-Russian Research and Technological Institute of Poultry" of the Russian Academy of Science, 141311 Sergiev Posad, Moscow Region, Russia
| | - Vladimir A Samoilenko
- Scryabin Institute of Biochemistry and Physiology of Microorganisms, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", 142290 Pushchino, Moscow Region, Russia
| | - Eduard E Svetoch
- State Research Center for Applied Microbiology and Biotechnology, Obolensk, Moscow Region, Russia
| | - Gennady T Sukhikh
- Kulakov National Medical Research Center for Obstetrics, Gynecology and Perinatology of the Ministry of Health, 117997 Moscow, Russia
| | - Vladimir N Uversky
- Institute for Biological Instrumentation, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Science", 142290, Pushchino, Moscow Region, Russia; Department of Molecular Medicine, Morsani College of Medicine, University of South Florida, Tampa, FL 33612, USA.
| | - Andrey V Karlyshev
- Department of Science, Engineering and Computing, Kingston University London, Kingston upon Thames KT1 2EE, UK
| |
Collapse
|
15
|
Abstract
Probiotic bacteria have been used as a health-promoting factor for a very long time. Nowadays, products containing probiotic bacteria are becoming more and more popular on the market. The term probiotics refers to the products belonging to the following groups: probiotic drugs (medicinal products – live biotherapeutic products for human use), medical devices, probiotic foods (e.g. foods, food ingredients, dietary supplements or food for special medical purposes), directly fed microorganisms (for animal use) and designer probiotics (genetically modified probiotics). Safety assessment of bacterial strains used as probiotics should be carefully studied. Even though probiotic bacteria have the generally recognized as safe (GRAS status), there are several reports about side effects triggered by the presence of these organisms. Microorganisms used as probiotics may cause systemic infections, stimulate the immune system, disturb metabolism and participate in horizontal gene transfer.
Collapse
Affiliation(s)
- Anna Zawistowska-Rojek
- Department of Antibiotics and Microbiology, National Medicines Institute, Warsaw, Poland ; Department of Pharmaceutical Microbiology, Medical University of Warsaw, Warsaw, Poland
| | - Stefan Tyski
- Department of Antibiotics and Microbiology, National Medicines Institute, Warsaw, Poland ; Department of Pharmaceutical Microbiology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
16
|
|
17
|
Lugli GA, Mangifesta M, Mancabelli L, Milani C, Turroni F, Viappiani A, van Sinderen D, Ventura M. Compositional assessment of bacterial communities in probiotic supplements by means of metagenomic techniques. Int J Food Microbiol 2019; 294:1-9. [DOI: 10.1016/j.ijfoodmicro.2019.01.011] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2018] [Revised: 10/31/2018] [Accepted: 01/20/2019] [Indexed: 10/27/2022]
|
18
|
Abstract
The Klaenhammer group at North Carolina State University pioneered genomic applications in food microbiology and beneficial lactic acid bacteria used as starter cultures and probiotics. Dr. Todd Klaenhammer was honored to be the first food scientist elected to the National Academy of Sciences (2001). The program was recognized with the highest research awards presented by the American Dairy Science Association (Borden Award 1996), the Institute of Food Technologists (Nicholas Appert Medal, 2007), and the International Dairy Federation (Eli Metchnikoff Award in Biotechnology, 2010) as well as with the Outstanding Achievement Award from the University of Minnesota (2001) and the Oliver Max Gardner Award (2009) for outstanding research across the 16-campus University of North Carolina system. Dr. Klaenhammer is a fellow of the American Association for the Advancement of Science, the American Dairy Science Association, and the Institute of Food Technology. Over his career, six of his PhD graduate students were awarded the annual Kenneth Keller award for the outstanding PhD dissertation that year in the College of Agriculture and Life Sciences. He championed the use of basic microbiology and genomic approaches to set a platform for translational applications of beneficial microbes in foods and their use in food preservation and probiotics and as oral delivery vehicles for vaccines and biotherapeutics. Dr. Klaenhammer was also a founding and co-chief editor of the Annual Review of Food Science and Technology.
Collapse
Affiliation(s)
- Todd Robert Klaenhammer
- Department of Food, Bioprocessing & Nutrition Sciences, North Carolina State University, Raleigh, North Carolina 27695, USA;
| |
Collapse
|
19
|
Elbanna K, El Hadad S, Assaeedi A, Aldahlawi A, Khider M, Alhebshi A. In vitro and in vivo evidences for innate immune stimulators lactic acid bacterial starters isolated from fermented camel dairy products. Sci Rep 2018; 8:12553. [PMID: 30135492 PMCID: PMC6105719 DOI: 10.1038/s41598-018-31006-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Accepted: 08/02/2018] [Indexed: 02/07/2023] Open
Abstract
Probiotics are commensals with special characteristics that are essential for the development of the immune system, and may protect mucosal surfaces against pathogens. In this study, a total of 40 lactic acid bacteria (LAB) were isolated from different raw and fermented camel's milk samples collected from Saudi Arabia (Makkah area) and Egypt (Fayoum), and tested for the probiotic properties. Among them, Pro 4 and Pro 7 isolates exhibited excellent probiotic potential including bile salt (0.2-0.6%), phenol tolerance (0.2-0.4%) and salt tolerance (0.0-10%). Furthermore, both strains exhibited antimicrobial activity against wide range of food-borne pathogens and Dermatophytes with average zone inhibition of 37.5, 35.5, 34.5, 27.5, 25 and 23.5 mm for Staphylococcus aureus, Trichophyton mentagrophytes, Escherichia coli, Listeria monocytogens, Candida albicans and Salmonella typhi, respectively. Furthermore, the in vivo study indicated that these strains significantly improved the mucosal immune responses through an increase in expression of TLR2 and IFNγ mRNA in mice intestine as well as increased the synthesis of polyclonal IgG, IgM and IgA in mice blood sera. Accordingly, due to these unique probiotic properties, both selected strains could be potentially used as probiotic starter cultures for fermented dairy foods as well as functional food and health products.
Collapse
Affiliation(s)
- Khaled Elbanna
- Deptartment of Agricultural Microbiology, Faculty of Agriculture, Fayoum University, Fayoum, Egypt.
- Department of Biology, Faculty of Applied Science, Umm Al-Qura University, Makkah, Saudi Arabia.
| | - Sahar El Hadad
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- ResearchCenter of Genetic Engineering and Bioinformatics, VACSERA, Cairo, Egypt
| | - Abdelrahaman Assaeedi
- Department of Biology, Faculty of Applied Science, Umm Al-Qura University, Makkah, Saudi Arabia
| | - Alia Aldahlawi
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
- Immunology Unit, King Fahad for medical research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Manal Khider
- Department of Dairy Science, Faculty of Agriculture, Fayoum University, Fayoum, Egypt
| | - Alawiah Alhebshi
- Department of Biological Science, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
20
|
Cazorla SI, Maldonado-Galdeano C, Weill R, De Paula J, Perdigón GDV. Oral Administration of Probiotics Increases Paneth Cells and Intestinal Antimicrobial Activity. Front Microbiol 2018; 9:736. [PMID: 29713315 PMCID: PMC5911494 DOI: 10.3389/fmicb.2018.00736] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2018] [Accepted: 03/28/2018] [Indexed: 12/12/2022] Open
Abstract
The huge amount of intestinal bacteria represents a continuing threat to the intestinal barrier. To meet this challenge, gut epithelial cells produce antimicrobial peptides (AMP) that act at the forefront of innate immunity. We explore whether this antimicrobial activity and Paneth cells, the main intestinal cell responsible of AMP production, are influenced by probiotics administration, to avoid the imbalance of intestinal microbiota and preserve intestinal barrier. Administration of Lactobacillus casei CRL 431 (Lc 431) and L. paracasei CNCM I-1518 (Lp 1518) to 42 days old mice, increases the number of Paneth cells on small intestine, and the antimicrobial activity against the pathogens Staphylococcus aureus and Salmonella Typhimurium in the intestinal fluids. Specifically, strong damage of the bacterial cell with leakage of cytoplasmic content, and cellular fragmentation were observed in S. Typhimurium and S. aureus. Even more important, probiotics increase the antimicrobial activity of the intestinal fluids at the different ages, from weaning (21 days old) to old age (180 days old). Intestinal antimicrobial activity stimulated by oral probiotics, do not influence significantly the composition of total anaerobic bacteria, lactobacilli and enterobacteria in the large intestine, at any age analyzed. This result, together with the antimicrobial activity observed against the same probiotic bacteria; endorse the regular consumption of probiotics without adverse effect on the intestinal homeostasis in healthy individuals. We demonstrate that oral probiotics increase intestinal antimicrobial activity and Paneth cells in order to strengthen epithelial barrier against pathogens. This effect would be another important mechanism by which probiotics protect the host mainly against infectious diseases.
Collapse
Affiliation(s)
- Silvia I Cazorla
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina.,Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán, Argentina
| | - Carolina Maldonado-Galdeano
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina.,Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán, Argentina
| | - Ricardo Weill
- Departamento de Investigación y Desarrollo, DANONE, Buenos Aires, Argentina
| | - Juan De Paula
- Servicio de Gastroenterología, Hospital Italiano de Buenos Aires, Buenos Aires, Argentina
| | - Gabriela D V Perdigón
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina.,Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán, Argentina
| |
Collapse
|
21
|
Characterization of Lactobacillus amylolyticus L6 as potential probiotics based on genome sequence and corresponding phenotypes. Lebensm Wiss Technol 2018. [DOI: 10.1016/j.lwt.2017.12.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
22
|
Xin Y, Guo T, Mu Y, Kong J. Coupling the recombineering to Cre-lox system enables simplified large-scale genome deletion in Lactobacillus casei. Microb Cell Fact 2018; 17:21. [PMID: 29433512 PMCID: PMC5808424 DOI: 10.1186/s12934-018-0872-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2017] [Accepted: 02/09/2018] [Indexed: 12/22/2022] Open
Abstract
Background Lactobacillus casei is widely used in the dairy and pharmaceutical industries and a promising candidate for use as cell factories. Recently, genome sequencing and functional genomics provide the possibility for reducing L. casei genome. However, it was still limited by the inefficient and laborious genome deletion methods. Results Here, we proposed a genome minimization strategy based on LCABL_13040-50-60 recombineering and Cre-lox site-specific recombination system in L. casei. The LCABL_13040-50-60 recombineering system was used to introduce two lox sites (lox66 and lox71) into 5′ and 3′ ends of the targeted region. Subsequently, the targeted region was excised by Cre recombinase. The robustness of the strategy was demonstrated by single-deletion of a nonessential ~ 39.3 kb or an important ~ 12.8 kb region and simultaneous deletion of two non-continuous genome regions (5.2 and 6.6 kb) with 100% efficiency. Furthermore, a cyclical application of this strategy generated a double-deletion mutant of which 1.68% of the chromosome was sequentially excised. Moreover, biological features (including growth rate, electroporation efficiency, cell morphology or heterologous protein productivity) of these mutants were characterized. Conclusions To our knowledge, this strategy is the first instance of sequential deletion of large-scale genome regions in L. casei. We expected this efficient and inexpensive tool can help for rapid genome streamlining and generation restructured L. casei strains used as cell factories. Electronic supplementary material The online version of this article (10.1186/s12934-018-0872-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yongping Xin
- State Key Laboratory of Microbial Technology, Shandong University, 27 Shanda Nanlu, Jinan, 250100, People's Republic of China
| | - Tingting Guo
- State Key Laboratory of Microbial Technology, Shandong University, 27 Shanda Nanlu, Jinan, 250100, People's Republic of China
| | - Yingli Mu
- State Key Laboratory of Microbial Technology, Shandong University, 27 Shanda Nanlu, Jinan, 250100, People's Republic of China
| | - Jian Kong
- State Key Laboratory of Microbial Technology, Shandong University, 27 Shanda Nanlu, Jinan, 250100, People's Republic of China.
| |
Collapse
|
23
|
Takahashi J, Rindfleisch JA. Prescribing Probiotics. Integr Med (Encinitas) 2018. [DOI: 10.1016/b978-0-323-35868-2.00105-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
24
|
Johnson BR, O'Flaherty S, Goh YJ, Carroll I, Barrangou R, Klaenhammer TR. The S-layer Associated Serine Protease Homolog PrtX Impacts Cell Surface-Mediated Microbe-Host Interactions of Lactobacillus acidophilus NCFM. Front Microbiol 2017; 8:1185. [PMID: 28713337 PMCID: PMC5491966 DOI: 10.3389/fmicb.2017.01185] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2017] [Accepted: 06/12/2017] [Indexed: 01/21/2023] Open
Abstract
Health-promoting aspects attributed to probiotic microorganisms, including adhesion to intestinal epithelia and modulation of the host mucosal immune system, are mediated by proteins found on the bacterial cell surface. Notably, certain probiotic and commensal bacteria contain a surface (S-) layer as the outermost stratum of the cell wall. S-layers are non-covalently bound semi-porous, crystalline arrays of self-assembling, proteinaceous subunits called S-layer proteins (SLPs). Recent evidence has shown that multiple proteins are non-covalently co-localized within the S-layer, designated S-layer associated proteins (SLAPs). In Lactobacillus acidophilus NCFM, SLP and SLAPs have been implicated in both mucosal immunomodulation and adhesion to the host intestinal epithelium. In this study, a S-layer associated serine protease homolog, PrtX (prtX, lba1578), was deleted from the chromosome of L. acidophilus NCFM. Compared to the parent strain, the PrtX-deficient strain (ΔprtX) demonstrated increased autoaggregation, an altered cellular morphology, and pleiotropic increases in adhesion to mucin and fibronectin, in vitro. Furthermore, ΔprtX demonstrated increased in vitro immune stimulation of IL-6, IL-12, and IL-10 compared to wild-type, when exposed to mouse dendritic cells. Finally, in vivo colonization of germ-free mice with ΔprtX led to an increase in epithelial barrier integrity. The absence of PrtX within the exoproteome of a ΔprtX strain caused morphological changes, resulting in a pleiotropic increase of the organisms’ immunomodulatory properties and interactions with some intestinal epithelial cell components.
Collapse
Affiliation(s)
- Brant R Johnson
- Graduate Program in Microbiology, College of Agriculture and Life Sciences, North Carolina State University, RaleighNC, United States.,Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, RaleighNC, United States
| | - Sarah O'Flaherty
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, RaleighNC, United States
| | - Yong Jun Goh
- Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, RaleighNC, United States
| | - Ian Carroll
- Department of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel HillChapel Hill, NC, United States.,Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel HillChapel Hill, NC, United States
| | - Rodolphe Barrangou
- Graduate Program in Microbiology, College of Agriculture and Life Sciences, North Carolina State University, RaleighNC, United States.,Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, RaleighNC, United States
| | - Todd R Klaenhammer
- Graduate Program in Microbiology, College of Agriculture and Life Sciences, North Carolina State University, RaleighNC, United States.,Department of Food, Bioprocessing and Nutrition Sciences, North Carolina State University, RaleighNC, United States.,Center for Gastrointestinal Biology and Disease, University of North Carolina at Chapel HillChapel Hill, NC, United States
| |
Collapse
|
25
|
Carvalho RDDO, do Carmo FLR, de Oliveira Junior A, Langella P, Chatel JM, Bermúdez-Humarán LG, Azevedo V, de Azevedo MS. Use of Wild Type or Recombinant Lactic Acid Bacteria as an Alternative Treatment for Gastrointestinal Inflammatory Diseases: A Focus on Inflammatory Bowel Diseases and Mucositis. Front Microbiol 2017; 8:800. [PMID: 28536562 PMCID: PMC5422521 DOI: 10.3389/fmicb.2017.00800] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Accepted: 04/19/2017] [Indexed: 12/26/2022] Open
Abstract
The human gastrointestinal tract (GIT) is highly colonized by bacterial communities, which live in a symbiotic relationship with the host in normal conditions. It has been shown that a dysfunctional interaction between the intestinal microbiota and the host immune system, known as dysbiosis, is a very important factor responsible for the development of different inflammatory conditions of the GIT, such as the idiopathic inflammatory bowel diseases (IBD), a complex and multifactorial disorder of the GIT. Dysbiosis has also been implicated in the pathogenesis of other GIT inflammatory diseases such as mucositis usually caused as an adverse effect of chemotherapy. As both diseases have become a great clinical problem, many research groups have been focusing on developing new strategies for the treatment of IBD and mucositis. In this review, we show that lactic acid bacteria (LAB) have been capable in preventing and treating both disorders in animal models, suggesting they may be ready for clinical trials. In addition, we present the most current studies on the use of wild type or genetically engineered LAB strains designed to express anti-inflammatory proteins as a promising strategy in the treatment of IBD and mucositis.
Collapse
Affiliation(s)
| | - Fillipe L R do Carmo
- Federal University of Minas Gerais - Instituto de Ciências BiológicasBelo Horizonte, Brazil
| | | | - Philippe Langella
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Jean-Marc Chatel
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Luis G Bermúdez-Humarán
- Micalis Institute, Institut National de la Recherche Agronomique, AgroParisTech, Université Paris-SaclayJouy-en-Josas, France
| | - Vasco Azevedo
- Federal University of Minas Gerais - Instituto de Ciências BiológicasBelo Horizonte, Brazil
| | - Marcela S de Azevedo
- Federal University of Minas Gerais - Instituto de Ciências BiológicasBelo Horizonte, Brazil
| |
Collapse
|
26
|
Bosma EF, Forster J, Nielsen AT. Lactobacilli and pediococci as versatile cell factories - Evaluation of strain properties and genetic tools. Biotechnol Adv 2017; 35:419-442. [PMID: 28396124 DOI: 10.1016/j.biotechadv.2017.04.002] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/29/2017] [Accepted: 04/03/2017] [Indexed: 12/14/2022]
Abstract
This review discusses opportunities and bottlenecks for cell factory development of Lactic Acid Bacteria (LAB), with an emphasis on lactobacilli and pediococci, their metabolism and genetic tools. In order to enable economically feasible bio-based production of chemicals and fuels in a biorefinery, the choice of product, substrate and production organism is important. Currently, the most frequently used production hosts include Escherichia coli and Saccharomyces cerevisiae, but promising examples are available of alternative hosts such as LAB. Particularly lactobacilli and pediococci can offer benefits such as thermotolerance, an extended substrate range and increased tolerance to stresses such as low pH or high alcohol concentrations. This review will evaluate the properties and metabolism of these organisms, and provide an overview of their current biotechnological applications and metabolic engineering. We substantiate the review by including experimental results from screening various lactobacilli and pediococci for transformability, growth temperature range and ability to grow under biotechnologically relevant stress conditions. Since availability of efficient genetic engineering tools is a crucial prerequisite for industrial strain development, genetic tool development is extensively discussed. A range of genetic tools exist for Lactococcus lactis, but for other species of LAB like lactobacilli and pediococci such tools are less well developed. Whereas lactobacilli and pediococci have a long history of use in food and beverage fermentation, their use as platform organisms for production purposes is rather new. By harnessing their properties such as thermotolerance and stress resistance, and by using emerging high-throughput genetic tools, these organisms are very promising as versatile cell factories for biorefinery applications.
Collapse
Affiliation(s)
- Elleke F Bosma
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet B220, 2800 Kgs. Lyngby, Denmark
| | - Jochen Forster
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet B220, 2800 Kgs. Lyngby, Denmark
| | - Alex Toftgaard Nielsen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet B220, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
27
|
Walsh AM, Crispie F, Claesson MJ, Cotter PD. Translating Omics to Food Microbiology. Annu Rev Food Sci Technol 2017; 8:113-134. [DOI: 10.1146/annurev-food-030216-025729] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Aaron M. Walsh
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Fiona Crispie
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
| | - Marcus J. Claesson
- APC Microbiome Institute, University College Cork, Cork, Ireland
- School of Microbiology, University College Cork, Cork, Ireland
| | - Paul D. Cotter
- Teagasc Food Research Centre, Moorepark, Fermoy, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|
28
|
Stout E, Klaenhammer T, Barrangou R. CRISPR-Cas Technologies and Applications in Food Bacteria. Annu Rev Food Sci Technol 2017; 8:413-437. [DOI: 10.1146/annurev-food-072816-024723] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Clustered regularly interspaced short palindromic repeats (CRISPRs) and CRISPR-associated (Cas) proteins form adaptive immune systems that occur in many bacteria and most archaea. In addition to protecting bacteria from phages and other invasive mobile genetic elements, CRISPR-Cas molecular machines can be repurposed as tool kits for applications relevant to the food industry. A primary concern of the food industry has long been the proper management of food-related bacteria, with a focus on both enhancing the outcomes of beneficial microorganisms such as starter cultures and probiotics and limiting the presence of detrimental organisms such as pathogens and spoilage microorganisms. This review introduces CRISPR-Cas as a novel set of technologies to manage food bacteria and offers insights into CRISPR-Cas biology. It primarily focuses on the applications of CRISPR-Cas systems and tools in starter cultures and probiotics, encompassing strain-typing, phage resistance, plasmid vaccination, genome editing, and antimicrobial activity.
Collapse
Affiliation(s)
- Emily Stout
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University, Raleigh, North Carolina 27695
| | - Todd Klaenhammer
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University, Raleigh, North Carolina 27695
| | - Rodolphe Barrangou
- Department of Food, Bioprocessing, and Nutrition Sciences, North Carolina State University, Raleigh, North Carolina 27695
| |
Collapse
|
29
|
Goldstein EJC, Johnson SJ, Maziade PJ, Evans CT, Sniffen JC, Millette M, McFarland LV. Probiotics and prevention of Clostridium difficile infection. Anaerobe 2016; 45:114-119. [PMID: 27988389 DOI: 10.1016/j.anaerobe.2016.12.007] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 12/04/2016] [Accepted: 12/07/2016] [Indexed: 12/16/2022]
Abstract
The role of probiotics as adjunctive measures in the prevention of Clostridium difficile infection (CDI) has been controversial. However, a growing body of evidence has suggested that they have a role in primary prevention of CDI. Elements of this controversy are reviewed and the proposed mechanisms of action, the value and cost effectiveness of probiotics are addressed with a focus on three agents, Saccharomyces boulardii, Lactobacillus rhamnosus GG and the combination of Lactobacillus acidophilus CL1285, Lactobacillus casei LBC80R, Lactobacillus rhamnosus CLR2 (Bio-K+).
Collapse
Affiliation(s)
- E J C Goldstein
- R M Alden Research Laboratory, Santa Monica, CA, USA; UCLA School of Medicine, Los Angeles, CA, USA.
| | | | | | - C T Evans
- Department of Preventive Medicine and Center for Healthcare Studies, Northwestern University, Chicago, IL, USA; Edward Hines Jr VA Hospital, Chicago, IL, USA
| | | | - M Millette
- Bio-K Plus International Inc., Laval, Canada
| | - L V McFarland
- Department of Medicinal Chemistry, University of Washington, Seattle, WA, USA
| |
Collapse
|
30
|
Miljkovic M, Bertani I, Fira D, Jovcic B, Novovic K, Venturi V, Kojic M. Shortening of the Lactobacillus paracasei subsp. paracasei BGNJ1-64 AggLb Protein Switches Its Activity from Auto-aggregation to Biofilm Formation. Front Microbiol 2016; 7:1422. [PMID: 27660628 PMCID: PMC5014864 DOI: 10.3389/fmicb.2016.01422] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Accepted: 08/26/2016] [Indexed: 12/27/2022] Open
Abstract
AggLb is the largest (318.6 kDa) aggregation-promoting protein of Lactobacillus paracasei subsp. paracasei BGNJ1-64 responsible for forming large cell aggregates, which causes auto-aggregation, collagen binding and pathogen exclusion in vitro. It contains an N-terminus leader peptide, followed by six successive collagen binding domains, 20 successive repeats (CnaB-like domains) and an LPXTG sorting signal at the C-terminus for cell wall anchoring. Experimental information about the roles of the domains of AggLb is currently unknown. To define the domain that confers cell aggregation and the key domains for interactions of specific affinity between AggLb and components of the extracellular matrix, we constructed a series of variants of the aggLb gene and expressed them in Lactococcus lactis subsp. lactis BGKP1-20 using a lactococcal promoter. All of the variants contained a leader peptide, an inter collagen binding-CnaB domain region (used to raise an anti-AggLb antibody), an anchor domain and a different number of collagen binding and CnaB-like domains. The role of the collagen binding repeats of the N-terminus in auto-aggregation and binding to collagen and fibronectin was confirmed. Deletion of the collagen binding repeats II, III, and IV resulted in a loss of the strong auto-aggregation, collagen and fibronectin binding abilities whereas the biofilm formation capability was increased. The strong auto-aggregation, collagen and fibronectin binding abilities of AggLb were negatively correlated to biofilm formation.
Collapse
Affiliation(s)
- Marija Miljkovic
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade Belgrade, Serbia
| | - Iris Bertani
- Bacteriology Group, International Centre for Genetic Engineering and Biotechnology, Area Science Park Trieste, Italy
| | - Djordje Fira
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of BelgradeBelgrade, Serbia; Department of Biochemistry, Faculty of Biology, University of BelgradeBelgrade, Serbia
| | - Branko Jovcic
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of BelgradeBelgrade, Serbia; Department of Biochemistry, Faculty of Biology, University of BelgradeBelgrade, Serbia
| | - Katarina Novovic
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade Belgrade, Serbia
| | - Vittorio Venturi
- Bacteriology Group, International Centre for Genetic Engineering and Biotechnology, Area Science Park Trieste, Italy
| | - Milan Kojic
- Laboratory for Molecular Microbiology, Institute of Molecular Genetics and Genetic Engineering, University of Belgrade Belgrade, Serbia
| |
Collapse
|
31
|
AcmB Is an S-Layer-Associated β-N-Acetylglucosaminidase and Functional Autolysin in Lactobacillus acidophilus NCFM. Appl Environ Microbiol 2016; 82:5687-97. [PMID: 27422832 PMCID: PMC5007774 DOI: 10.1128/aem.02025-16] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 12/23/2022] Open
Abstract
Autolysins, also known as peptidoglycan hydrolases, are enzymes that hydrolyze specific bonds within bacterial cell wall peptidoglycan during cell division and daughter cell separation. Within the genome of Lactobacillus acidophilus NCFM, there are 11 genes encoding proteins with peptidoglycan hydrolase catalytic domains, 9 of which are predicted to be functional. Notably, 5 of the 9 putative autolysins in L. acidophilus NCFM are S-layer-associated proteins (SLAPs) noncovalently colocalized along with the surface (S)-layer at the cell surface. One of these SLAPs, AcmB, a β-N-acetylglucosaminidase encoded by the gene lba0176 (acmB), was selected for functional analysis. In silico analysis revealed that acmB orthologs are found exclusively in S-layer- forming species of Lactobacillus. Chromosomal deletion of acmB resulted in aberrant cell division, autolysis, and autoaggregation. Complementation of acmB in the ΔacmB mutant restored the wild-type phenotype, confirming the role of this SLAP in cell division. The absence of AcmB within the exoproteome had a pleiotropic effect on the extracellular proteins covalently and noncovalently bound to the peptidoglycan, which likely led to the observed decrease in the binding capacity of the ΔacmB strain for mucin and extracellular matrices fibronectin, laminin, and collagen in vitro. These data suggest a functional association between the S-layer and the multiple autolysins noncovalently colocalized at the cell surface of L. acidophilus NCFM and other S-layer-producing Lactobacillus species. IMPORTANCELactobacillus acidophilus is one of the most widely used probiotic microbes incorporated in many dairy foods and dietary supplements. This organism produces a surface (S)-layer, which is a self-assembling crystalline array found as the outermost layer of the cell wall. The S-layer, along with colocalized associated proteins, is an important mediator of probiotic activity through intestinal adhesion and modulation of the mucosal immune system. However, there is still a dearth of information regarding the basic cellular and evolutionary function of S-layers. Here, we demonstrate that multiple autolysins, responsible for breaking down the cell wall during cell division, are associated with the S-layer. Deletion of the gene encoding one of these S-layer-associated autolysins confirmed its autolytic role and resulted in reduced binding capacity to mucin and intestinal extracellular matrices. These data suggest a functional association between the S-layer and autolytic activity through the extracellular presentation of autolysins.
Collapse
|
32
|
Soares SC, Geyik H, Ramos RT, de Sá PH, Barbosa EG, Baumbach J, Figueiredo HC, Miyoshi A, Tauch A, Silva A, Azevedo V. GIPSy: Genomic island prediction software. J Biotechnol 2016; 232:2-11. [DOI: 10.1016/j.jbiotec.2015.09.008] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 08/28/2015] [Accepted: 09/11/2015] [Indexed: 10/23/2022]
|
33
|
Chae JP, Pajarillo EAB, Oh JK, Kim H, Kang DK. Revealing the combined effects of lactulose and probiotic enterococci on the swine faecal microbiota using 454 pyrosequencing. Microb Biotechnol 2016; 9:486-95. [PMID: 27305897 PMCID: PMC4919990 DOI: 10.1111/1751-7915.12370] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Revised: 05/11/2016] [Accepted: 05/20/2016] [Indexed: 12/02/2022] Open
Abstract
Demand for the development of non‐antibiotic growth promoters in animal production has increased in recent years. This report compared the faecal microbiota of weaned piglets under the administration of a basal diet (CON) or that containing prebiotic lactulose (LAC), probiotic Enterococcus faecium NCIMB 11181 (PRO) or their synbiotic combination (SYN). At the phylum level, the Firmicutes to Bacteroidetes ratio increased in the treatment groups compared with the CON group, and the lowest proportion of Proteobacteria was observed in the LAC group. At the family level, Enterobacteriaceae decreased in all treatments; more than a 10‐fold reduction was observed in the LAC (0.99%) group compared with the CON group. At the genus level, the highest Oscillibacter proportion was detected in PRO, the highest Clostridium in LAC and the highest Lactobacillus in SYN; the abundance of Escherichia was lowest in the LAC group. Clustering in the discriminant analysis of principal components revealed distinct separation of the feeding groups (CON, LAC, PRO and SYN), showing different microbial compositions according to different feed additives or their combination. These results suggest that individual materials and their combination have unique actions and independent mechanisms for changes in the distal gut microbiota.
Collapse
Affiliation(s)
- Jong Pyo Chae
- Department of Animal Resources Science, Dankook University, Cheonan, 330-714, Korea
| | | | - Ju Kyoung Oh
- Department of Animal Resources Science, Dankook University, Cheonan, 330-714, Korea
| | - Heebal Kim
- Department of Agricultural Biotechnology, Seoul National University, Seoul, 151-921, Korea
| | - Dae-Kyung Kang
- Department of Animal Resources Science, Dankook University, Cheonan, 330-714, Korea
| |
Collapse
|
34
|
McFarland LV. From yaks to yogurt: the history, development, and current use of probiotics. Clin Infect Dis 2016; 60 Suppl 2:S85-90. [PMID: 25922406 DOI: 10.1093/cid/civ054] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
The development of probiotics, which are living bacteria or yeasts used to confer a health benefit on the host, has paralleled our research in food preservation, microbiologic identification techniques, and our understanding of how the complex interactions in microbiota impact the host's health and recovery from disease. This review briefly describes the history of probiotics, where probiotic strains were originally isolated, and the types of probiotic products currently available on the global market. In addition, the uses or indications for these probiotics are described, along with the types of clinical investigations that have been done. Continuing challenges persist for the proper probiotic strain identification, regulatory pathways, and how healthcare providers can choose a specific strain to recommend to their patients.
Collapse
Affiliation(s)
- Lynne V McFarland
- Department of Medicinal Chemistry, University of Washington, Seattle
| |
Collapse
|
35
|
Bertrand TF, Natalia BE, Gordon TN, BongsiysiGilake N, Irene AA, Samuel W. Probiotic properties of lactic acid bacteria isolated from fermented sap of palm tree (Elaeis guineensis). ACTA ACUST UNITED AC 2015. [DOI: 10.5897/jma2014.0353] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
|
36
|
Conserved S-Layer-Associated Proteins Revealed by Exoproteomic Survey of S-Layer-Forming Lactobacilli. Appl Environ Microbiol 2015; 82:134-45. [PMID: 26475115 PMCID: PMC4702614 DOI: 10.1128/aem.01968-15] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 10/13/2015] [Indexed: 11/20/2022] Open
Abstract
The Lactobacillus acidophilus homology group comprises Gram-positive species that include L. acidophilus, L. helveticus, L. crispatus, L. amylovorus, L. gallinarum, L. delbrueckii subsp. bulgaricus, L. gasseri, and L. johnsonii. While these bacteria are closely related, they have varied ecological lifestyles as dairy and food fermenters, allochthonous probiotics, or autochthonous commensals of the host gastrointestinal tract. Bacterial cell surface components play a critical role in the molecular dialogue between bacteria and interaction signaling with the intestinal mucosa. Notably, the L. acidophilus complex is distinguished in two clades by the presence or absence of S-layers, which are semiporous crystalline arrays of self-assembling proteinaceous subunits found as the outermost layer of the bacterial cell wall. In this study, S-layer-associated proteins (SLAPs) in the exoproteomes of various S-layer-forming Lactobacillus species were proteomically identified, genomically compared, and transcriptionally analyzed. Four gene regions encoding six putative SLAPs were conserved in the S-layer-forming Lactobacillus species but not identified in the extracts of the closely related progenitor, L. delbrueckii subsp. bulgaricus, which does not produce an S-layer. Therefore, the presence or absence of an S-layer has a clear impact on the exoproteomic composition of Lactobacillus species. This proteomic complexity and differences in the cell surface properties between S-layer- and non-S-layer-forming lactobacilli reveal the potential for SLAPs to mediate intimate probiotic interactions and signaling with the host intestinal mucosa.
Collapse
|
37
|
Nader-Macías MEF, Juárez Tomás MS. Profiles and technological requirements of urogenital probiotics. Adv Drug Deliv Rev 2015; 92:84-104. [PMID: 25858665 DOI: 10.1016/j.addr.2015.03.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 02/20/2015] [Accepted: 03/27/2015] [Indexed: 12/14/2022]
Abstract
Probiotics, defined as live microorganisms that, when administered in adequate amounts, confer a health benefit on the host, are considered a valid and novel alternative for the prevention and treatment of female urogenital tract infections. Lactobacilli, the predominant microorganisms of the healthy human vaginal microbiome, can be included as active pharmaceutical ingredients in probiotics products. Several requirements must be considered or criteria fulfilled during the development of a probiotic product or formula for the female urogenital tract. This review deals with the main selection criteria for urogenital probiotic microorganisms: host specificity, potential beneficial properties, functional specifications, technological characteristics and clinical trials used to test their effect on certain physiological and pathological conditions. Further studies are required to complement the current knowledge and support the clinical applications of probiotics in the urogenital tract. This therapy will allow the restoration of the ecological equilibrium of the urogenital tract microbiome as well as the recovery of the sexual and reproductive health of women.
Collapse
|
38
|
|
39
|
Abstract
Genome analysis using next generation sequencing technologies has revolutionized the characterization of lactic acid bacteria and complete genomes of all major groups are now available. Comparative genomics has provided new insights into the natural and laboratory evolution of lactic acid bacteria and their environmental interactions. Moreover, functional genomics approaches have been used to understand the response of lactic acid bacteria to their environment. The results have been instrumental in understanding the adaptation of lactic acid bacteria in artisanal and industrial food fermentations as well as their interactions with the human host. Collectively, this has led to a detailed analysis of genes involved in colonization, persistence, interaction and signaling towards to the human host and its health. Finally, massive parallel genome re-sequencing has provided new opportunities in applied genomics, specifically in the characterization of novel non-GMO strains that have potential to be used in the food industry. Here, we provide an overview of the state of the art of these functional genomics approaches and their impact in understanding, applying and designing lactic acid bacteria for food and health.
Collapse
|