1
|
Alexander LM, Khalid S, Gallego-Lopez GM, Astmann TJ, Oh JH, Heggen M, Huss P, Fisher R, Mukherjee A, Raman S, Choi IY, Smith MN, Rogers CJ, Epperly MW, Knoll LJ, Greenberger JS, van Pijkeren JP. Development of a Limosilactobacillus reuteri therapeutic delivery platform with reduced colonization potential. Appl Environ Microbiol 2024:e0031224. [PMID: 39480094 DOI: 10.1128/aem.00312-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Accepted: 09/09/2024] [Indexed: 11/02/2024] Open
Abstract
Bacterial biotherapeutic delivery vehicles have the potential to treat a variety of diseases. This approach obviates the need to purify the recombinant effector molecule, allows delivery of therapeutics in situ via oral or intranasal administration, and protects the effector molecule during gastrointestinal transit. Lactic acid bacteria have been broadly developed as therapeutic delivery vehicles though risks associated with the colonization of a genetically modified microorganism have so-far not been addressed. Here, we present an engineered Limosilactobacillus reuteri strain with reduced colonization potential. We applied a dual-recombineering scheme for efficient barcoding and generated mutants in genes encoding five previously characterized and four uncharacterized putative adhesins. Compared with the wild type, none of the mutants were reduced in their ability to survive gastrointestinal transit in mice. CmbA was identified as a key protein in L. reuteri adhesion to HT-29 and enteroid cells. The nonuple mutant, a single strain with all nine genes encoding adhesins inactivated, had reduced capacity to adhere to enteroid monolayers. The nonuple mutant producing murine IFN-β was equally effective as its wild-type counterpart in mitigating radiation toxicity in mice. Thus, this work established a novel therapeutic delivery platform that lays a foundation for its application in other microbial therapeutic delivery candidates and furthers the progress of the L. reuteri delivery system towards human use.IMPORTANCEOne major advantage to leverage gut microbes that have co-evolved with the vertebrate host is that evolution already has taken care of the difficult task to optimize survival within a complex ecosystem. The availability of the ecological niche will support colonization. However, long-term colonization of a recombinant microbe may not be desirable. Therefore, strategies need to be developed to overcome this potential safety concern. In this work, we developed a single strain in which we inactivated the encoding sortase, and eight genes encoding characterized/putative adhesins. Each individual mutant was characterized for growth and adhesion to epithelial cells. On enteroid cells, the nonuple mutant has a reduced adhesion potential compared with the wild-type strain. In a model of total-body irradiation, the nonuple strain engineered to release murine interferon-β performed comparable to a derivative of the wild-type strain that releases interferon-β. This work is an important step toward the application of recombinant L. reuteri in humans.
Collapse
Affiliation(s)
- Laura M Alexander
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Saima Khalid
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Gina M Gallego-Lopez
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Morgridge Institute for Research, Madison, Wisconsin, USA
| | - Theresa J Astmann
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mark Heggen
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Phil Huss
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Renee Fisher
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Amitava Mukherjee
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Srivatsan Raman
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - In Young Choi
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Morgan N Smith
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Michael W Epperly
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Laura J Knoll
- Department of Medical Microbiology and Immunology, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Joel S Greenberger
- Department of Radiation Oncology, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | |
Collapse
|
2
|
She F, Anderson BW, Khana DB, Zhang S, Steinchen W, Fung DK, Lucas LN, Lesser NG, Stevenson DM, Astmann TJ, Bange G, van Pijkeren JP, Amador-Noguez D, Wang JD. Allosteric Regulation of Pyruvate Kinase Enables Efficient and Robust Gluconeogenesis by Preventing Metabolic Conflicts and Carbon Overflow. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.15.607825. [PMID: 39211278 PMCID: PMC11361145 DOI: 10.1101/2024.08.15.607825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Glycolysis and gluconeogenesis are reciprocal metabolic pathways that utilize different carbon sources. Pyruvate kinase catalyzes the irreversible final step of glycolysis, yet the physiological function of its regulation is poorly understood. Through metabolomics and enzyme kinetics studies, we discovered that pyruvate kinase activity is inhibited during gluconeogenesis in the soil bacterium Bacillus subtilis . This regulation involves an extra C-terminal domain (ECTD) of pyruvate kinase, which is essential for autoinhibition and regulation by metabolic effectors. Introducing a pyruvate kinase mutant lacking the ECTD into B. subtilis resulted in defects specifically under gluconeogenic conditions, including inefficient carbon utilization, slower growth, and decreased resistance to the herbicide glyphosate. These defects are not caused by the phosphoenolpyruvate-pyruvate-oxaloacetate futile cycle. Instead, we identified two significant metabolic consequences of pyruvate kinase dysregulation during gluconeogenesis: increased carbon overflow into the medium and failure to expand glycolytic intermediates such as phosphoenolpyruvate (PEP). In silico analysis revealed that in wild-type cells, an expanded PEP pool enabled by pyruvate kinase regulation is critical for the thermodynamic feasibility of gluconeogenesis. Our findings underscore the importance of allosteric regulation during gluconeogenesis in coordinating metabolic flux, efficient energy utilization, and antimicrobial resistance.
Collapse
|
3
|
Kardos G, Laczkó L, Kaszab E, Timmer B, Szarka K, Prépost E, Bányai K. Phylogenetic Analysis of the Genes in D-Ala-D-Lactate Synthesizing Glycopeptide Resistance Operons: The Different Origins of Functional and Regulatory Genes. Antibiotics (Basel) 2024; 13:573. [PMID: 39061255 PMCID: PMC11273654 DOI: 10.3390/antibiotics13070573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 05/27/2024] [Accepted: 05/29/2024] [Indexed: 07/28/2024] Open
Abstract
The phylogenetic relationships of glycopeptide resistance proteins were investigated. The amino acid sequences of vanA, vanB, vanR and vanS were used as queries to search against bacterial genomes in the NCBI RefSeq database. Hits with >60% amino acid identity and >90% query coverage were aligned, and phylogenetic trees were reconstructed. The ligase gene phylogenies were highly similar for both queries, revealing two major clusters. One contained [[vanA:vanM][vanB:vanD]vanF] and related proteins, with proteins from different Bacillaceae, mostly from Paenibacillus spp., in basal positions to all, except vanB. Ligases from streptomycetes formed the other cluster. The relative positions of vanH and vanX differed from those of the associated ligases, but the basal position of the Paenibacillus spp. and the separation of proteins of Streptomyces origin were similar. The accessory genes vanW, vanY and vanZ were associated with vanB, vanA/vanM and vanA, respectively; the basal branches were always proteins from different Bacillaceae but never from streptomycetes. Multiple homologs of the regulatory genes vanR and vanS were found in the genomes; those associated with the different ligases were unique to the ligases. Similarly to the accessory genes, vanRS from Bacillales and Clostridia, but never from streptomycetes, was found in the basal positions. In conclusion, the core genes vanA/B/D/F/M, vanH and vanX originate most probably from glycopeptide-producing streptomycetes, with Paenibacillus spp. (or other Bacillaceae) mediating the transfer, while the accessory genes and the regulatory apparatus probably originate from these Bacillaceae.
Collapse
Affiliation(s)
- Gábor Kardos
- Institute of Metagenomics, University of Debrecen, H-4032 Debrecen, Hungary; (B.T.); (K.S.)
- One Health Institute, Faculty of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary; (L.L.); (E.K.)
| | - Levente Laczkó
- One Health Institute, Faculty of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary; (L.L.); (E.K.)
- HUN-REN-UD Conservation Biology Research Group, H-4032 Debrecen, Hungary
| | - Eszter Kaszab
- One Health Institute, Faculty of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary; (L.L.); (E.K.)
- Department of Microbiology and Infectious Diseases, University of Veterinary Medicine, H-1078 Budapest, Hungary
| | - Bálint Timmer
- Institute of Metagenomics, University of Debrecen, H-4032 Debrecen, Hungary; (B.T.); (K.S.)
- Department of Medical Microbiology and Immunology, University of Pécs, H-7624 Pécs, Hungary
| | - Krisztina Szarka
- Institute of Metagenomics, University of Debrecen, H-4032 Debrecen, Hungary; (B.T.); (K.S.)
- One Health Institute, Faculty of Health Sciences, University of Debrecen, H-4032 Debrecen, Hungary; (L.L.); (E.K.)
| | - Eszter Prépost
- Department of Health Industry, University of Debrecen, H-4032 Debrecen, Hungary;
| | - Krisztián Bányai
- Pathogen Discovery Group, HUN-REN Veterinary Medical Research Institute, H-1143 Budapest, Hungary
- National Laboratory for Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, H-1143 Budapest, Hungary
- Department of Pharmacology and Toxicology, University of Veterinary Medicine, H-1078 Budapest, Hungary
| |
Collapse
|
4
|
Quaresma LS, Santos RCV, Gomes GC, Américo MF, Campos GM, Laguna JG, Barroso FAL, Azevedo V, de Jesus LCL. Multidrug resistance profile in Lactobacillus delbrueckii: a food industry species with probiotic properties. World J Microbiol Biotechnol 2024; 40:235. [PMID: 38850338 DOI: 10.1007/s11274-024-04046-3] [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: 04/10/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Lactobacillus delbrueckii, a widely used lactic acid bacterium in the food industry, has been studied for its probiotic properties and reservoir of antibiotic-resistant genes, raising safety concerns for probiotic formulations and fermented products. This review consolidates findings from 60 articles published between 2012 and 2023, focusing on the global antibiotic resistance profile and associated genetic factors in L. delbrueckii strains. Resistance to aminoglycosides, particularly streptomycin, kanamycin, and gentamicin, as well as resistance to glycopeptides (vancomycin), fluoroquinolones (ciprofloxacin), and tetracyclines was predominant. Notably, although resistance genes have been identified, they have not been linked to mobile genetic elements, reducing the risk of dissemination. However, a significant limitation is the insufficient exploration of responsible genes or mobile elements in 80% of studies, hindering safety assessments. Additionally, most articles originated from Asian and Middle Eastern countries, with strains often isolated from fermented dairy foods. Therefore, these findings underscore the necessity for comprehensive analyses of new strains of L. delbrueckii for potential industrial and biotherapeutic applications and in combating the rise of antibiotic-resistant pathogens.
Collapse
Affiliation(s)
- Ludmila Silva Quaresma
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | - Gabriel Camargos Gomes
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Monique Ferrary Américo
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Gabriela Munis Campos
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | - Juliana Guimarães Laguna
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil
| | | | - Vasco Azevedo
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| | - Luís Cláudio Lima de Jesus
- Department of Genetics, Ecology, and Evolution, Federal University of Minas Gerais, Belo Horizonte, 31270-901, Brazil.
| |
Collapse
|
5
|
Barreto Pinilla CM, Brandelli A, Ataíde Isaia H, Guzman F, Sundfeld da Gama MA, Spadoti LM, Torres Silva E Alves A. Probiotic Potential and Application of Indigenous Non-Starter Lactic Acid Bacteria in Ripened Short-Aged Cheese. Curr Microbiol 2024; 81:202. [PMID: 38829392 DOI: 10.1007/s00284-024-03729-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Accepted: 05/04/2024] [Indexed: 06/05/2024]
Abstract
There are massive sources of lactic acid bacteria (LAB) in traditional dairy products. Some of these indigenous strains could be novel probiotics with applications in human health and supply the growing needs of the probiotic industry. In this work, were analyzed the probiotic and technological properties of three Lactobacilli strains isolated from traditional Brazilian cheeses. In vitro tests showed that the three strains are safe and have probiotic features. They presented antimicrobial activity against pathogenic bacteria, auto-aggregation values around 60%, high biofilm formation properties, and a survivor of more than 65% to simulated acid conditions and more than 100% to bile salts. The three strains were used as adjunct cultures separately in a pilot-scale production of Prato cheese. After 45 days of ripening, the lactobacilli counts in the cheeses were close to 8 Log CFU/g, and was observed a reduction in the lactococci counts (around -3 Log CFU/g) in a strain-dependent manner. Cheese primary and secondary proteolysis were unaffected by the probiotic candidates during the ripening, and the strains showed no lipolytic effect, as no changes in the fatty acid profile of cheeses were observed. Thus, our findings suggest that the three strains evaluated have probiotic properties and have potential as adjunct non-starter lactic acid bacteria (NSLAB) to improve the quality and functionality of short-aged cheeses.
Collapse
Affiliation(s)
| | - Adriano Brandelli
- Laboratory of Applied Microbiology and Biochemistry, Institute of Food Science and Technology (ICTA), Federal University of Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil
| | - Henrique Ataíde Isaia
- Laboratory of Applied Microbiology and Biochemistry, Institute of Food Science and Technology (ICTA), Federal University of Rio Grande Do Sul (UFRGS), Porto Alegre, Brazil
| | - Frank Guzman
- Grupo de Investigación en Epidemiología y Diseminación de la Resistencia a Antimicrobianos - "One Health", Universidad Científica del Sur, Lima, Perú
| | | | - Leila Maria Spadoti
- Dairy Technology Center (TECNOLAT) of the Food Technology Institute (ITAL), Campinas, São Paulo, Brazil
| | | |
Collapse
|
6
|
Tian S, Jiang Y, Han Q, Meng C, Ji F, Zhou B, Ye M. Putative Probiotic Ligilactobacillus salivarius Strains Isolated from the Intestines of Meat-Type Pigeon Squabs. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10289-1. [PMID: 38805143 DOI: 10.1007/s12602-024-10289-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/11/2024] [Indexed: 05/29/2024]
Abstract
This study aims to screen for potential probiotic lactic acid bacteria from the intestines of meat-type pigeon squabs. Ligilactobacillus salivarius YZU37 was identified as the best comprehensive performed strain. Being acid- and bile salt-tolerant, it displayed growth-inhibition activities against Staphylococcus aureus ATCC25923, Escherichia coli ATCC25922, and Salmonella typhimurium SL1344, exhibited sensitivity to 6 commonly used antibiotics, and endowed with good cell surface hydrophobicity, auto-aggregation property, and anti-oxidant activities. Results of in vitro experiments indicated that the bacteriostatic effects of this strain were related to the production of proteinaceous substances that depend on acidic conditions. Whole-genome sequencing of L. salivarius YZU37 was performed to elucidate the genetic basis underlying its probiotic potential. Pangenome analysis of L. salivarius YZU37 and other 212 L. salivarius strains available on NCBI database revealed a pigeon-unique gene coding choloylglycine hydrolase (CGH), which had higher enzyme-substrate binding affinity than that of the common CGH shared by L. salivarius strains of other sources. Annotation of the functional genes in the genome of L. salivarius YZU37 revealed genes involved in responses to acid, bile salt, heat, cold, heavy metal, and oxidative stresses. The whole genome analysis also revealed the absence of virulence and toxin genes and the presence of 65 genes distributed under 4 CAZymes classes, 2 CRISPR-cas regions, and 3 enterolysin A clusters which may confer the acid-dependent antimicrobial potential of L. salivarius YZU37. Altogether, our results highlighted the probiotic potential of L. salivarius YZU37. Further in vivo investigations are required to elucidate its beneficial effects on pigeons.
Collapse
Affiliation(s)
- Shaoqi Tian
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, 225009, China
| | - Yinhong Jiang
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, 225009, China
| | - Qiannan Han
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, 225009, China
| | - Chuang Meng
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, 225009, China
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China
| | - Feng Ji
- Institute of Animal Husbandry and Veterinary Medicine, Beijing Academy of Agriculture and Forestry Sciences, Beijing, 100089, China
| | - Bin Zhou
- College of Animal Science and Technology, Yangzhou University, Yangzhou, 225009, China
| | - Manhong Ye
- College of Bioscience and Biotechnology, Yangzhou University, Yangzhou, 225009, China.
- Jiangsu Key Laboratory of Zoonosis, Yangzhou University, Yangzhou, 225009, China.
| |
Collapse
|
7
|
Singh K, Park S. Construction of prophage-free and highly-transformable Limosilactobacillus reuteri strains and their use for production of 1,3-propanediol. Biotechnol Bioeng 2024; 121:317-328. [PMID: 37747698 DOI: 10.1002/bit.28559] [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: 02/21/2023] [Revised: 08/28/2023] [Accepted: 09/07/2023] [Indexed: 09/26/2023]
Abstract
The lactic acid bacterium Limosilactobacillus reuteri (formerly Lactobacillus reuteri) is a desirable host for the production of 1,3-propanediol (1,3-PDO) from glycerol when 1,3-PDO is used in the food or cosmetic industry. However, the production is hindered by strain instability, causing cell lysis, and difficult gene manipulation. This study reveals that the stability of L. reuteri DSM 20016 and its 1,3-PDO production, especially in the alcohol dehydrogenases (ADHs)-deletion mutants, are greatly enhanced after the deletion of two prophages (Φ3 and Φ4) present in the L. reuteri's chromosome. The resulting phage-free and ADHs-deletion mutant could produce >825 mM 1,3-PDO in 48 h without cell lysis at the theoretical maximum yield on glucose of ~2 mol/mol. Compared to the wild-type strain, the mutant exhibited a 45.2% increase in 1,3-PDO production titer and a 2.1-fold increase in yield. In addition, this study reports that the transformation efficiency of L. reuteri Δadh2Δadh6 mutant strains were greatly enhanced by >300-fold after the deletion of prophage Φ3, probably due to the removal of a restriction-modification (RM) system which resides in the phage genome. With improved stability and higher transformation efficiency, recombinant L. reuteri DSM 20016 Δadh2Δadh6ΔΦ3ΔΦ4 can be a more reliable and amenable host for industrial applications.
Collapse
Affiliation(s)
- Kalpana Singh
- School of Energy and Chemical Engineering, UNIST, Ulsan, Republic of Korea
| | - Sunghoon Park
- School of Energy and Chemical Engineering, UNIST, Ulsan, Republic of Korea
| |
Collapse
|
8
|
Guo Q, Yan Y, Zhang Z, Xu B, Bangash HL, Sui X, Yang Y, Zhou Z, Zhao S, Peng N. Developing the Limosilactobacillus reuteri Chassis through an Endogenous Programmable Endonuclease-Based Genome Editing Tool. ACS Synth Biol 2023; 12:3487-3496. [PMID: 37934952 DOI: 10.1021/acssynbio.3c00450] [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] [Indexed: 11/09/2023]
Abstract
Using genetically tractable probiotics to engineer live biotherapeutic products (LBPs) for disease treatment is urgently needed. Limosilactobacillus reuteri is an important vertebrate gut symbiont, which has great potential for developing LBPs. However, in L. reuteri, synthetic biology work is largely limited by the long editing cycle. In this study, we identified a subtype II-A CRISPR-Cas9 system in L. reuteri 03 and found the endogenous Cas9 (LrCas9) recognizing a broad protospacer-adjacent motif (PAM) sequence (3'-NDR; N = A, G, T, C; D = A, G, T; R = A, G). We reprogrammed the LrCas9 for efficient gene deletion (95.46%), point mutation (86.36%), large fragment deletion (40 kb), and gene integration (1743 bp, 73.9%), which uncovered the function of the repeated conserved domains in mucus-binding protein. Moreover, we analyzed the distribution of endogenous endonucleases in 304 strains of L. reuteri and found the existence of programmable endonucleases in 98.36% of L. reuteri strains suggesting the potential to reprogram endogenous endonucleases for genetic manipulation in the majority of L. reuteri strains. In conclusion, this study highlights the development of a new probiotic chassis based on endogenous endonucleases in L. reuteri 03, which paves the way for the development of genome editing tools for functional genetic studies in other L. reuteri. We believe that the development of an endogenous endonuclease-based genetic tool will greatly facilitate the construction of LBPs.
Collapse
Affiliation(s)
- Qiujin Guo
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei, P.R. China
| | - Yiting Yan
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei, P.R. China
| | - Zhenting Zhang
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei, P.R. China
- Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang, 550025 Guizhou, P.R. China
| | - Boya Xu
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei, P.R. China
| | - Hina Lqbal Bangash
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei, P.R. China
| | - Xin Sui
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei, P.R. China
| | - Yalin Yang
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, P.R. China
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, P.R. China
| | - Zhigang Zhou
- Sino-Norway Joint Lab on Fish Gut Microbiota, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, P.R. China
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, P.R. China
| | - Shumiao Zhao
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei, P.R. China
| | - Nan Peng
- National Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, College of Life Science and Technology, Huazhong Agricultural University, Wuhan, 430070 Hubei, P.R. China
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Institute of Feed Research, Chinese Academy of Agricultural Sciences, Beijing 100081, P.R. China
| |
Collapse
|
9
|
Qiao N, Gaur G, Modesto M, Chinnici F, Scarafile D, Borruso L, Marin AC, Spiezio C, Valente D, Sandri C, Gänzle MG, Mattarelli P. Physiological and genomic characterization of Lactiplantibacillus plantarum isolated from Indri indri in Madagascar. J Appl Microbiol 2023; 134:lxad255. [PMID: 37934609 DOI: 10.1093/jambio/lxad255] [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: 04/25/2023] [Revised: 07/21/2023] [Accepted: 11/01/2023] [Indexed: 11/09/2023]
Abstract
AIMS Indri indri is a lemur of Madagascar which is critically endangered. The analysis of the microbial ecology of the intestine offers tools to improve conservation efforts. This study aimed to achieve a functional genomic analysis of three Lactiplantibacillus plantarum isolates from indris. METHODS AND RESULTS Samples were obtained from 18 indri; 3 isolates of Lp. plantarum were obtained from two individuals. The three isolates were closely related to each other, with <10 single nucleotide polymorphisms, suggesting that the two individuals shared diet-associated microbes. The genomes of the three isolates were compared to 96 reference strains of Lp. plantarum. The three isolates of Lp. plantarum were not phenotypically resistant to antibiotics but shared all 17 genes related to antimicrobial resistance that are part of the core genome of Lp. plantarum. The genomes of the three indri isolates of Lp. plantarum also encoded for the 6 core genome genes coding for enzymes related to metabolism of hydroxybenzoic and hydroxycinnamic acids. The phenotype for metabolism of hydroxycinnamic acids by indri isolates of Lp. plantarum matched the genotype. CONCLUSIONS Multiple antimicrobial resistance genes and gene coding for metabolism of phenolic compounds were identified in the genomes of the indri isolates, suggesting that Lp. plantarum maintains antimicrobial resistance in defense of antimicrobial plant secondary pathogens and that their metabolism by intestinal bacteria aids digestion of plant material by primate hosts.
Collapse
Affiliation(s)
- Nanzhen Qiao
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Gautam Gaur
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Monica Modesto
- Department of Agricultural and Food Sciences, University of Bologna, Bologna 40126, Italy
| | - Fabio Chinnici
- Department of Agricultural and Food Sciences, University of Bologna, Bologna 40126, Italy
| | - Donatella Scarafile
- Department of Agricultural and Food Sciences, University of Bologna, Bologna 40126, Italy
| | - Luigimaria Borruso
- Faculty of Agricultural, Environmental and Food Sciences, Free University of Bolzano-Bozen, Bolzano 39100, Italy
| | - Antonio Castro Marin
- Department of Agricultural and Food Sciences, University of Bologna, Bologna 40126, Italy
| | - Caterina Spiezio
- Department of Animal Health Care and Management, Parco Natura Viva, Bussolengo (Verona) 37012, Italy
| | - Daria Valente
- Department of Life Sciences and Systems Biology, University of Torino, Turin 10124, Italy
| | - Camillo Sandri
- Department of Agricultural and Food Sciences, University of Bologna, Bologna 40126, Italy
- Department of Animal Health Care and Management, Parco Natura Viva, Bussolengo (Verona) 37012, Italy
| | - Michael G Gänzle
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Paola Mattarelli
- Department of Agricultural and Food Sciences, University of Bologna, Bologna 40126, Italy
| |
Collapse
|
10
|
Choi IY, Oh JH, Wang Z, van Pijkeren JP. Bioluminescent monitoring of recombinant lactic acid bacteria and their products. mBio 2023; 14:e0119723. [PMID: 37668408 PMCID: PMC10653940 DOI: 10.1128/mbio.01197-23] [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: 05/09/2023] [Accepted: 06/29/2023] [Indexed: 09/06/2023] Open
Abstract
IMPORTANCE Lactic acid bacteria constitute a genetically diverse group of microorganisms with significant roles in the food industry, biotechnology, agriculture, and medicine. A core understanding of bacterial physiology in diverse environments is crucial to select and develop bacteria for industrial and medical applications. However, there is a lack of versatile tools to track (recombinant) protein production in lactic acid bacteria. In this study, we adapted a peptide-based bioluminescent tagging system that is functional across multiple genera and species. This system enables tracking of tagged proteins both in vitro and in situ, while it also can be used to enumerate recombinant bacteria from the mouse gastrointestinal tract with accuracy comparable to that of conventional plate counts. Our work expands the lactic acid bacteria genetic toolbox and will facilitate researchers in industry and academia with opportunities to monitor microbes and proteins under different physiologically relevant conditions.
Collapse
Affiliation(s)
- In Young Choi
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Zhiying Wang
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | |
Collapse
|
11
|
Liu Y, Armbrister SA, Okeugo B, Mills TW, Daniel RC, Oh JH, van Pijkeren JP, Park ES, Saleh ZM, Lahiri S, Roos S, Rhoads JM. Probiotic-Derived Ecto-5'-Nucleotidase Produces Anti-Inflammatory Adenosine Metabolites in Treg-Deficient Scurfy Mice. Probiotics Antimicrob Proteins 2023; 15:1001-1013. [PMID: 37178405 PMCID: PMC10926147 DOI: 10.1007/s12602-023-10089-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2023] [Indexed: 05/15/2023]
Abstract
Probiotic Limosilactobacillus reuteri DSM 17938 (DSM 17938) prolongs the survival of Treg-deficient scurfy (SF) mice and reduces multiorgan inflammation by a process requiring adenosine receptor 2A (A2A) on T cells. We hypothesized that L. reuteri-derived ecto-5'-nucleotidase (ecto-5'NT) activity acts to generate adenosine, which may be a central mediator for L. reuteri protection in SF mice. We evaluated DSM 17938-5'NT activity and the associated adenosine and inosine levels in plasma, gut, and liver of SF mice. We examined orally fed DSM 17938, DSM 17938Δ5NT (with a deleted 5'NT gene), and DSM 32846 (BG-R46) (a naturally selected strain derived from DSM 17938). Results showed that DSM 17938 and BG-R46 produced adenosine while "exhausting" AMP, whereas DSM 17938∆5NT did not generate adenosine in culture. Plasma 5'NT activity was increased by DSM 17938 or BG-R46, but not by DSM 17938Δ5NT in SF mice. BG-R46 increased both adenosine and inosine levels in the cecum of SF mice. DSM 17938 increased adenosine levels, whereas BG-R46 increased inosine levels in the liver. DSM 17938Δ5NT did not significantly change the levels of adenosine or inosine in the GI tract or the liver of SF mice. Although regulatory CD73+CD8+ T cells were decreased in spleen and blood of SF mice, these regulatory T cells could be increased by orally feeding DSM 17938 or BG-R46, but not DSM 17938Δ5NT. In conclusion, probiotic-5'NT may be a central mediator of DSM 17938 protection against autoimmunity. Optimal 5'NT activity from various probiotic strains could be beneficial in treating Treg-associated immune disorders in humans.
Collapse
Affiliation(s)
- Yuying Liu
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Shabba A Armbrister
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Beanna Okeugo
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Tingting W Mills
- Department of Biochemistry & Molecular Biology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Rhea C Daniel
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, 53706, USA
| | | | - Evelyn S Park
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Zeina M Saleh
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Sharmistha Lahiri
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| | - Stefan Roos
- Department of Molecular Sciences, Uppsala BioCenter, Swedish University of Agricultural Sciences, Uppsala, Sweden
- BioGaia AB, Stockholm, Sweden
| | - JMarc Rhoads
- Department of Pediatrics, Division of Gastroenterology, McGovern Medical School, the University of Texas Health Science Center at Houston, Houston, TX, 77030, USA
| |
Collapse
|
12
|
Duche RT, Singh A, Wandhare AG, Sangwan V, Sihag MK, Nwagu TNT, Panwar H, Ezeogu LI. Antibiotic resistance in potential probiotic lactic acid bacteria of fermented foods and human origin from Nigeria. BMC Microbiol 2023; 23:142. [PMID: 37208603 DOI: 10.1186/s12866-023-02883-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 05/06/2023] [Indexed: 05/21/2023] Open
Abstract
INTRODUCTION Probiotic lactobacilli are generally recognized as safe (GRAS) and are being used in several food and pharma formulations. However, growing concern of antibiotic resistance in bacterial strains of food origin and its possible transmission via functional foods is increasingly being emphasized. OBJECTIVES This study screened potential probiotic lactic acid bacteria (LAB) strains for their phenotypic and genotypic antibiotic resistance profiles. METHODS Susceptibility to different antibiotics was assayed by the Kirby Bauer standard disc diffusion protocol. Both conventional and SYBR-RTq-PCR were used for detection of resistance coding genes. RESULTS A variable susceptibility pattern was documented against different antibiotic classes. LAB strains irrespective of origin displayed marked phenotypic resistance against cephalosporins, aminoglycosides, quinolones, glycopeptides; and methicillin among beta-lactams with few exceptions. In contrast, high sensitivity was recorded against macrolides, sulphonamides and carbapenems sub-group of beta-lactams with some variations. parC, associated with ciprofloxacin resistance was detected in 76.5% of the strains. Other prevalent resistant determinants observed were aac(6?)Ii (42.1%), ermB, ermC (29.4%), and tetM (20.5%). Six (?17.6%) of the isolates were free from genetic resistance determinants screened in this study. CONCLUSION Study revealed presence of antibiotic resistance determinants among lactobacilli from both fermented foods and human sources.
Collapse
Affiliation(s)
- Rachael T Duche
- Department of Dairy Microbiology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
- Department of Microbiology, Federal University of Agriculture Makurdi-Nigeria, Makurdi, Nigeria
- Department of Microbiology, University of Nigeria Nsukka, Nsukka, Nigeria
- UNESCO International Centre for Biotechnology, Nsukka, Nigeria
| | - Anamika Singh
- Department of Dairy Microbiology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - Arundhati Ganesh Wandhare
- Department of Dairy Microbiology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - Vikas Sangwan
- Department of Dairy Microbiology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - Manvesh Kumar Sihag
- Department of Dairy Chemistry, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India
| | - Tochukwu N T Nwagu
- Department of Microbiology, University of Nigeria Nsukka, Nsukka, Nigeria
| | - Harsh Panwar
- Department of Dairy Microbiology, Guru Angad Dev Veterinary and Animal Sciences University, Ludhiana, Punjab, India.
| | - Lewis I Ezeogu
- Department of Microbiology, University of Nigeria Nsukka, Nsukka, Nigeria.
- UNESCO International Centre for Biotechnology, Nsukka, Nigeria.
| |
Collapse
|
13
|
Gaur G, Damm S, Passon M, Lo HK, Schieber A, Gänzle MG. Conversion of hydroxycinnamic acids by Furfurilactobacillus milii in sorghum fermentations: Impact on profile of phenolic compounds in sorghum and on ecological fitness of Ff. milii. Food Microbiol 2023; 111:104206. [PMID: 36681402 DOI: 10.1016/j.fm.2022.104206] [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: 09/10/2022] [Revised: 11/08/2022] [Accepted: 12/07/2022] [Indexed: 12/14/2022]
Abstract
The conversion of phenolic compounds by lactobacilli in food fermentations contributes to food quality. The metabolism of phenolics by lactobacilli has been elucidated in the past years but information on the contribution of specific enzymes in food fermentations remains scarce. This study aimed to address this gap by disruption of genes coding for the hydroxycimmanic acid reductase Par1, the hydroxycinnamic acid decarboxylase Pad, the hydrocinnamic esterase EstR, and strains with disruption of all three genes in Furfurilactobacillus milii FUA3583. The conversion of phenolics by Ff. milii and its isogenic mutants in sorghum fermentations was studied by LC-UV and LC-UV-MS/MS analyses. Ff. milii FUA3583 converted hydroxycinnamic acids predominantly with Par1. Vinylphenols were detected only in mutants lacking par1. A phenotype for the estR defective mutant was not identified. The formation of pyrano-3-deoxyanthocyanidins was observed only after fermentation with strains expressing Pad. Specifically, formation of these compounds was low with Ff. milii FUA3583, substantially increased in the Par1 mutant and abolished in all mutants with disrupted pad. Competition experiments with Ff. milii FUA3583 and its isogenic mutants demonstrated that expression of one of the two metabolic pathways for hydroxycinnamic acids increases the ecological fitness of the strain. Disruption of EstR in a Δpar1Δpar2Δpad background improved ecological fitness, indirectly demonstrating a phenotype of the esterase in Ff. milii. The documentation of the functionality of genes coding for conversion of hydroxycinnamic acids may support the selection of starter cultures for improved quality of fermented cereal products.
Collapse
Affiliation(s)
- Gautam Gaur
- University of Alberta, Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada
| | - Sandra Damm
- University of Bonn, Department of Nutritional and Food Sciences, Molecular Food Technology, Bonn, Germany
| | - Maike Passon
- University of Bonn, Department of Nutritional and Food Sciences, Molecular Food Technology, Bonn, Germany
| | - Hiu Kwan Lo
- University of Alberta, Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada
| | - Andreas Schieber
- University of Bonn, Department of Nutritional and Food Sciences, Molecular Food Technology, Bonn, Germany
| | - Michael G Gänzle
- University of Alberta, Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada.
| |
Collapse
|
14
|
Keddar K, Ziar H, Belmadani N, Monnoye M, Gérard P, Riazi A. Probiotic Bacteria from Human Milk Can Alleviate Oral Bovine Casein Sensitization in Juvenile Wistar Rats. Microorganisms 2023; 11:microorganisms11041030. [PMID: 37110453 PMCID: PMC10140966 DOI: 10.3390/microorganisms11041030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
This study aims to see if probiotic bacteria from human milk could ameliorate oral cow's milk sensitization. The probiotic potential of the SL42 strain isolated from the milk of a healthy young mother was first determined. Rats were then randomly gavaged with cow's milk casein without an adjuvant or assigned to the control group. Each group was further subdivided into three groups, with each receiving only Limosilactobacillus reuteri DSM 17938, SL42, or a phosphate-buffered saline solution. Body weight, temperature, eosinophils, serum milk casein-specific IgE (CAS-IgE), histamine, and serum S100A8/A9 and inflammatory cytokine concentrations were measured. The animals were sacrificed after 59 days; histological sections were prepared, and the spleen or thymus weights, as well as the diversity of the gut microbiota, were measured. On days 1 and 59, SL42 abridged systemic allergic responses to casein by dropping histamine levels (25.7%), CAS-specific IgE levels (53.6%), eosinophil numbers (17%), S100A8/9 (18.7%), and cytokine concentrations (25.4-48.5%). Analyses of histological sections of the jejunum confirmed the protective effect of probiotic bacteria in the CAS-challenged groups. Lactic acid bacteria and Clostridia species were also increased in all probiotic-treated groups. These findings suggest that probiotics derived from human milk could be used to alleviate cow's milk casein allergy.
Collapse
Affiliation(s)
- Kawtar Keddar
- Laboratoire des Micro-Organismes Bénéfiques, des Aliments Fonctionnels et de la Santé (LMBAFS), Abdelhamid Ibn Badis University, Hocine Hamadou Street, Mostaganem 27000, Algeria
- Laboratoire de Bio-Economie, Sécurité Alimentaire et Santé, Abdelhamid Ibn Badis University, Hocine Hamadou Street, Mostaganem 27000, Algeria
| | - Hasnia Ziar
- Laboratoire des Micro-Organismes Bénéfiques, des Aliments Fonctionnels et de la Santé (LMBAFS), Abdelhamid Ibn Badis University, Hocine Hamadou Street, Mostaganem 27000, Algeria
- Micalis Institute, INRAE, AgroParisTech, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - Noussaiba Belmadani
- Laboratoire des Micro-Organismes Bénéfiques, des Aliments Fonctionnels et de la Santé (LMBAFS), Abdelhamid Ibn Badis University, Hocine Hamadou Street, Mostaganem 27000, Algeria
- Laboratoire de Bio-Economie, Sécurité Alimentaire et Santé, Abdelhamid Ibn Badis University, Hocine Hamadou Street, Mostaganem 27000, Algeria
| | - Magali Monnoye
- Micalis Institute, INRAE, AgroParisTech, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - Philippe Gérard
- Micalis Institute, INRAE, AgroParisTech, Paris-Saclay University, 78350 Jouy-en-Josas, France
| | - Ali Riazi
- Laboratoire des Micro-Organismes Bénéfiques, des Aliments Fonctionnels et de la Santé (LMBAFS), Abdelhamid Ibn Badis University, Hocine Hamadou Street, Mostaganem 27000, Algeria
| |
Collapse
|
15
|
Liu Y, Armbrister SA, Okeugo B, Mills TW, Daniel RC, Oh JH, Pijkeren JP, Park ES, Saleh ZM, Lahiri S, Roos S, Rhoads JM. Probiotic-derived ecto-5'-nucleotidase produces anti-inflammatory adenosine metabolites in Treg-deficient scurfy mice. RESEARCH SQUARE 2023:rs.3.rs-2781715. [PMID: 37066419 PMCID: PMC10104250 DOI: 10.21203/rs.3.rs-2781715/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/18/2023]
Abstract
Probiotic Limosilactobacillus reuteri DSM 17938 (DSM 17938) prolonges the survival of Treg-deficient scurfy (SF) mice and reduces multiorgan inflammation by a process requiring adenosine receptor 2A (A 2A ) on T cells. We hypothesized that L. reuteri -derived ecto-5'-nucleotidase (ecto-5'NT) activity acts to generate adenosine, which may be a central mediator for L. reuteri protection in SF mice. We evaluated DSM 17938-5'NT activity and the associated adenosine and inosine levels in plasma, gut and liver of SF mice. We examined orally fed DSM 17938, DSM 17938Δ5NT (with a deleted 5'NT gene), and DSM 32846 (BG-R46) (a naturally selected strain derived from DSM 17938). Results showed that DSM 17938 and BG-R46 produced adenosine while "exhausting" AMP, whereas DSM 17938∆5NT did not generate adenosine in culture. Plasma 5'NT activity was increased by DSM 17938 or BG-R46, but not by DSM 17938Δ5NT in SF mice. BG-R46 increased both adenosine and inosine levels in the cecum of SF mice. DSM 17938 increased adenosine levels, whereas BG-R46 increased inosine levels in the liver. DSM 17938Δ5NT did not significantly change the levels of adenosine or inosine in the GI tract or the liver of SF mice. Although regulatory CD73 + CD8 + T cells were decreased in spleen and blood of SF mice, these regulatory T cells could be increased by orally feeding DSM 17938 or BG-R46, but not DSM 17938Δ5NT. In conclusion, probiotic-5'NT may be a central mediator of DSM 17938 protection against autoimmunity. Optimal 5'NT activity from various probiotic strains could be beneficial in treating Treg-associated immune disorders in humans.
Collapse
|
16
|
Gaur G, Chen C, Gänzle MG. Characterization of isogenic mutants with single or double deletions of four phenolic acid esterases in Lactiplantibacillus plantarum TMW1.460. Int J Food Microbiol 2023; 388:110100. [PMID: 36706579 DOI: 10.1016/j.ijfoodmicro.2023.110100] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Revised: 11/05/2022] [Accepted: 01/15/2023] [Indexed: 01/22/2023]
Abstract
In plants, hydroxycinnamic and hydroxybenzoic acids occur mainly as esters. This study aimed to determine the contribution of individual phenolic acid esterases in Lp. plantarum TMW1.460, which encodes for four esterases: TanA, Lp_0796, Est_1092 and a homolog of Lj0536 and Lj1228 that was termed HceP. To determine which of the phenolic acid esterases present in Lp plantarum TMW1.460 are responsible for esterase activity, mutants with deletions in lp_0796, est_1092, tanB, hceP, or hceP and est_1092 were constructed. The phenotype of wild type strain and mutants was determined with esters of hydroxycinnamic acids (chlorogenic acid and ethyl ferulate) and of hydroxybenzoic acids (methyl gallate, tannic acid and epigallocatechin-3-gallate). Lp. plantarum TMW1.460 hydrolysed chlorogenic acid, methyl ferulate and methyl gallate but not tannic acid or epigallocatechin gallate. The phenotype of mutant strains during growth in mMRS differed from the wild type as follows: Lp. plantarum TMW1.460ΔhceP did not hydrolyse esters of hydroxycinnamic acids; Lp. plantarum TMW1.460ΔtanB did not hydrolyse esters of hydroxybenzoic acids; disruption of est_1092 or Lp_0796 did not alter the phenotype. The phenotype of Lp. plantarum TMW1.460ΔΔhceP/est_1092 was identical to Lp. plantarum TMW1.460ΔhceP. The metabolism of phenolic acids during growth of the mutant strains in broccoli puree and wheat sourdough did not differ from metabolism of the wild type strain. In conclusion, esters of hydroxycinnamic and hydroxybenzoic acids each are hydrolysed by dedicated enzymes. The hydroxycinnamic acid esterase HceP is not expressed, or not active during growth of Lp. plantarum TMW1.460 in all food substrates.
Collapse
Affiliation(s)
- Gautam Gaur
- University of Alberta, Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada
| | - Chen Chen
- University of Alberta, Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada; Shanghai Institute of Technology, School of Perfume and Aroma Technology, Shanghai, PR China
| | - Michael G Gänzle
- University of Alberta, Department of Agricultural, Food and Nutritional Science, Edmonton, Alberta, Canada.
| |
Collapse
|
17
|
Heng YC, Menon N, Chen B, Loo BZL, Wong GWJ, Lim ACH, Silvaraju S, Kittelmann S. Ligilactobacillus ubinensis sp. nov., a novel species isolated from the wild ferment of a durian fruit ( Durio zibethinus). Int J Syst Evol Microbiol 2023; 73. [PMID: 36920986 DOI: 10.1099/ijsem.0.005733] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2023] Open
Abstract
A Gram-stain-positive, rod-shaped, non-spore-forming, catalase-negative, urease-negative, homofermentative and facultatively anaerobic strain, named WILCCON 0076T, was isolated from a wild ferment of pieces of a 'Kampung' durian fruit collected on the island of Ubin (Pulau Ubin), Singapore. The durian had fallen to the ground from a durian tree (Durio zibethinus), on which a group of long-tailed macaques had been observed picking and eating the fruits. Comparative analyses of 16S rRNA gene sequences indicated that WILCCON 0076T potentially represented a novel species within the genus Ligilactobacillus, with the most closely related type strain being Ligilactobacillus agilis DSM 20509T (16S rRNA gene sequence similarity of 97.2 %). Average nucleotide identity and digital DNA-DNA hybridization prediction values were only 86.0% and 18.9 %, respectively. On the basis of the results of a polyphasic approach that included phylogenomic, chemotaxonomic and morphological analyses, we propose a novel species with the name Ligilactobacillus ubinensis sp. nov. (type strain WILCCON 0076T=DSM 114293T=LMG 32698T).
Collapse
Affiliation(s)
- Yu Chyuan Heng
- Wilmar International Limited, WIL@NUS Corporate Laboratory, Centre for Translational Medicine, National University of Singapore, Singapore, Singapore
| | - Nandita Menon
- Wilmar International Limited, WIL@NUS Corporate Laboratory, Centre for Translational Medicine, National University of Singapore, Singapore, Singapore
| | - Binbin Chen
- Wilmar International Limited, WIL@NUS Corporate Laboratory, Centre for Translational Medicine, National University of Singapore, Singapore, Singapore
| | - Bryan Zong Lin Loo
- Wilmar International Limited, WIL@NUS Corporate Laboratory, Centre for Translational Medicine, National University of Singapore, Singapore, Singapore
| | - Garrett Wei Jie Wong
- Wilmar International Limited, WIL@NUS Corporate Laboratory, Centre for Translational Medicine, National University of Singapore, Singapore, Singapore
| | - Amber Ching Han Lim
- Wilmar International Limited, WIL@NUS Corporate Laboratory, Centre for Translational Medicine, National University of Singapore, Singapore, Singapore
| | - Shaktheeshwari Silvaraju
- Wilmar International Limited, WIL@NUS Corporate Laboratory, Centre for Translational Medicine, National University of Singapore, Singapore, Singapore
| | - Sandra Kittelmann
- Wilmar International Limited, WIL@NUS Corporate Laboratory, Centre for Translational Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
18
|
Wang L, Ying R, Liu Y, Sun Q, Sha W. Metabolic Profiles of Clinical Isolates of Drug-Susceptible and Multidrug-Resistant Mycobacterium tuberculosis: A Metabolomics-Based Study. Infect Drug Resist 2023; 16:2667-2680. [PMID: 37163145 PMCID: PMC10164396 DOI: 10.2147/idr.s405987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Accepted: 04/20/2023] [Indexed: 05/11/2023] Open
Abstract
Background Mycobacterium tuberculosis (MTB) is a global and highly deleterious pathogen that creates an enormous pressure on global public health. Although several effective drugs have been used to treat tuberculosis, the emergence of multidrug-resistant Mycobacterium tuberculosis (MDR-MTB) has further increased the public health burden. The aim of this study was to describe in depth the metabolic changes in clinical isolates of drug-susceptible Mycobacterium tuberculosis (DS-MTB) and MDR-MTB and to provide clues to the mechanisms of drug resistance based on metabolic pathways. Methods Based on the minimum inhibition concentration (MIC) of multiple anti-tuberculosis drugs, two clinical isolates were selected, one DS-MTB isolate (isoniazid MIC=0.06 mg/L, rifampin MIC=0.25 mg/L) and one MDR-MTB isolate (isoniazid MIC=4 mg/L, rifampin MIC=8 mg/L). Through high-throughput metabolomics, the metabolic profiles of the DS-MTB isolate and the MDR-MTB isolate and their cultured supernatants were revealed. Results Compared with the DS-MTB isolate, 128 metabolites were significantly altered in the MDR-MTB isolate and 66 metabolites were significantly altered in the cultured supernatant. The differential metabolites were significantly enriched in pyrimidine metabolism, purine metabolism, nicotinate and nicotinamide metabolism, arginine acid metabolism, and phenylalanine metabolism. Furthermore, metabolomics analysis of the bacterial cultured supernatants showed a significant increase in 10 amino acids (L-citrulline, L-glutamic acid, L-aspartic acid, L-norleucine, L-phenylalanine, L-methionine, L-tyrosine, D-tryptophan, valylproline, and D-methionine) and a significant decrease in 2 amino acids (L-lysine and L-arginine) in MDR-MTB isolate. Conclusion The present study provided a metabolite alteration profile as well as a cultured supernatant metabolite alteration profile of MDR-MTB clinical isolate, providing clues to the potential metabolic pathways and mechanisms of multidrug resistance.
Collapse
Affiliation(s)
- Li Wang
- Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Ruoyan Ying
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Yidian Liu
- Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
| | - Qin Sun
- Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
| | - Wei Sha
- Clinic and Research Center of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Department of Tuberculosis, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, People’s Republic of China
- Shanghai Key Laboratory of Tuberculosis, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, People’s Republic of China
- Correspondence: Wei Sha; Qin Sun, Email ;
| |
Collapse
|
19
|
Johansson P, Säde E, Hultman J, Auvinen P, Björkroth J. Pangenome and genomic taxonomy analyses of Leuconostoc gelidum and Leuconostoc gasicomitatum. BMC Genomics 2022; 23:818. [PMID: 36494615 PMCID: PMC9733070 DOI: 10.1186/s12864-022-09032-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Accepted: 11/21/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Leuconostoc gelidum and Leuconostoc gasicomitatum have dual roles in foods. They may spoil cold-stored packaged foods but can also be beneficial in kimchi fermentation. The impact in food science as well as the limited number of publicly available genomes prompted us to create pangenomes and perform genomic taxonomy analyses starting from de novo sequencing of the genomes of 37 L. gelidum/L. gasicomitatum strains from our culture collection. Our aim was also to evaluate the recently proposed change in taxonomy as well as to study the genomes of strains with different lifestyles in foods. METHODS We selected as diverse a set of strains as possible in terms of sources, previous genotyping results and geographical distribution, and included also 10 publicly available genomes in our analyses. We studied genomic taxonomy using pairwise average nucleotide identity (ANI) and calculation of digital DNA-DNA hybridisation (dDDH) scores. Phylogeny analyses were done using the core gene set of 1141 single-copy genes and a set of housekeeping genes commonly used for lactic acid bacteria. In addition, the pangenome and core genome sizes as well as some properties, such as acquired antimicrobial resistance (AMR), important due to the growth in foods, were analysed. RESULTS Genome relatedness indices and phylogenetic analyses supported the recently suggested classification that restores the taxonomic position of L. gelidum subsp. gasicomitatum back to the species level as L. gasicomitatum. Genome properties, such as size and coding potential, revealed limited intraspecies variation and showed no attribution to the source of isolation. The distribution of the unique genes between species and subspecies was not associated with the previously documented lifestyle in foods. None of the strains carried any acquired AMR genes or genes associated with any known form of virulence. CONCLUSION Genome-wide examination of strains confirms that the proposition to restore the taxonomic position of L. gasicomitatum is justified. It further confirms that the distribution and lifestyle of L. gelidum and L. gasicomitatum in foods have not been driven by the evolution of functional and phylogenetic diversification detectable at the genome level.
Collapse
Affiliation(s)
- Per Johansson
- grid.7737.40000 0004 0410 2071Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland
| | - Elina Säde
- grid.7737.40000 0004 0410 2071Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland
| | - Jenni Hultman
- grid.7737.40000 0004 0410 2071Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland
| | - Petri Auvinen
- grid.7737.40000 0004 0410 2071Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Johanna Björkroth
- grid.7737.40000 0004 0410 2071Department of Food Hygiene and Environmental Health, University of Helsinki, Helsinki, Finland
| |
Collapse
|
20
|
Oh YJ, Kim SA, Yang SH, Kim DH, Cheng YY, Kang JI, Lee SY, Han NS. Integrated genome-based assessment of safety and probiotic characteristics of Lactiplantibacillus plantarum PMO 08 isolated from kimchi. PLoS One 2022; 17:e0273986. [PMID: 36190947 PMCID: PMC9529155 DOI: 10.1371/journal.pone.0273986] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 08/19/2022] [Indexed: 11/18/2022] Open
Abstract
Lactiplantibacillus plantarum PMO 08 has been used as a probiotic starter culture for plant-based fermented beverages, with various health-promoting effects such as cholesterol-lowering and anti-inflammatory activities. This study aimed to analyze the genome sequence of Lp. plantarum PMO 08 and identify its safety and probiotic characteristics at the genomic level. For this, complete genome sequencing was conducted to investigate the genes associated with risk and probiotic characteristics by using Pacbio combined with Illumina HiSeq. This bacterial strain has one circular chromosome of 3,247,789 bp with 44.5% G + C content and two plasmids of 50,296 bp with 39.0% G + C content and 19,592 bp with 40.5% G + C content. Orthologous average nucleotide identity analysis showed that PMO 08 belongs to the Lp. plantarum group with 99.14% similarity to Lp. plantarum WCFS1. No deleterious genes were determined in the virulence factor analysis, and no hemolysin activity or secondary bile salt synthesis were detected in vitro test. In the case of antibiotic resistance analysis, PMO 08 was resistant to ampicillin in vitro test, but these genes were not transferable. In addition, the strain showed same carbohydrate utilization with Lp. plantarum WCFS1, except for mannopyranoside, which only our strain can metabolize. The strain also harbors a gene for inositol monophosphatase family protein related with phytate hydrolysis and have several genes for metabolizing various carbohydrate which were rich in plant environment. Furthermore, in probiotic characteristics several genes involved in phenotypes such as acid/bile tolerance, adhesion ability, and oxidative stress response were detected in genome analysis. This study demonstrates that Lp. plantarum PMO 08 harbors several probiotic-related genes (with no deleterious genes) and is a suitable probiotic starter for plant-based fermentation.
Collapse
Affiliation(s)
| | - Seul-Ah Kim
- Brain Korea 21 Center for Bio-Health Industry, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Soo Hwi Yang
- Brain Korea 21 Center for Bio-Health Industry, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Da Hye Kim
- Brain Korea 21 Center for Bio-Health Industry, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Ya-Yun Cheng
- Brain Korea 21 Center for Bio-Health Industry, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | | | | | - Nam Soo Han
- Brain Korea 21 Center for Bio-Health Industry, Division of Animal, Horticultural, and Food Sciences, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
- * E-mail:
| |
Collapse
|
21
|
AI-2/LuxS Quorum Sensing System Promotes Biofilm Formation of Lactobacillus rhamnosus GG and Enhances the Resistance to Enterotoxigenic Escherichia coli in Germ-Free Zebrafish. Microbiol Spectr 2022; 10:e0061022. [PMID: 35700135 PMCID: PMC9430243 DOI: 10.1128/spectrum.00610-22] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
The LuxS enzyme plays a key role in both quorum sensing (QS) and the regulation of bacterial growth. It catalyzes the production of autoinducer-2 (AI-2) signaling molecule, which is a component of the methyl cycle and methionine metabolism. This study aimed at investigating the differences between the Lactobacillus rhamnosus GG (LGG) wild-type strain (WT) and its luxS mutant (ΔluxS) during biofilm formation and when resisting to inflammation caused by Enterotoxigenic Escherichia coli (ETEC) in germ-free zebrafish. Our results suggest that in the absence of luxS when LGG was knocked out, biofilm formation, extracellular polysaccharide secretion and adhesion were all compromised. Addition of synthetic AI-2 indeed rescued, at least partially, the deficiencies observed in the mutant strain. The colonizing and immunomodulatory function in WT versus ΔluxS mutants were further studied in a germ-free zebrafish model. The concentration of AI-2 signaling molecules decreased sharply in zebrafish infected with the ΔluxS. At the same time, compared with the ΔluxS, the wild-type strain could colonize the germ-free zebrafish more effectively. Our transcriptome results suggest that genes involved in immunity, signal transduction, and cell adhesion were downregulated in zebrafish infected with ΔluxS and WT. In the WT, the immune system of germ-free zebrafish was activated more effectively through the MAPK and NF-κB pathway, and its ability to fight the infection against ETEC was increased. Together, our results demonstrate that the AI-2/LuxS system plays an important role in biofilm formation to improve LGG and alleviate inflammation caused by ETEC in germ-free zebrafish. IMPORTANCELactobacillus rhamnosus GG is a widely used probiotic to improve host intestinal health, promote growth, reduce diarrhea, and modulate immunity. In recent years, the bacterial quorum sensing system has attracted much attention; however, there has not been much research on the effect of the LuxS/AI-2 quorum sensing system of Lactobacillus on bacteriostasis, microbial ecology balance, and immune regulation in intestine. In this study, we used germ-free zebrafish as an animal model to compare the differences between wild-type and luxS mutant strains. We showed how AI-2/LuxS QS affects the release of AI-2 and how QS regulates the colonization, EPS synthesis and biofilm formation of LGG. This study provides an idea for the targeted regulation of animal intestinal health with probiotics by controlling bacteria quorum sensing system.
Collapse
|
22
|
Özçam M, Oh JH, Tocmo R, Acharya D, Zhang S, Astmann TJ, Heggen M, Ruiz-Ramírez S, Li F, Cheng CC, Vivas E, Rey FE, Claesen J, Bugni TS, Walter J, van Pijkeren JP. A secondary metabolite drives intraspecies antagonism in a gut symbiont that is inhibited by cell-wall acetylation. Cell Host Microbe 2022; 30:824-835.e6. [PMID: 35443156 DOI: 10.1016/j.chom.2022.03.033] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 12/16/2021] [Accepted: 03/25/2022] [Indexed: 11/03/2022]
Abstract
The mammalian microbiome encodes numerous secondary metabolite biosynthetic gene clusters; yet, their role in microbe-microbe interactions is unclear. Here, we characterized two polyketide synthase gene clusters (fun and pks) in the gut symbiont Limosilactobacillus reuteri. The pks, but not the fun, cluster encodes antimicrobial activity. Forty-one of 51 L. reuteri strains tested are sensitive to Pks products; this finding was independent of strains' host origin. Sensitivity to Pks was also established in intraspecies competition experiments in gnotobiotic mice. Comparative genome analyses between Pks-resistant and -sensitive strains identified an acyltransferase gene (act) unique to Pks-resistant strains. Subsequent cell-wall analysis of wild-type and act mutant strains showed that Act acetylates cell-wall components, providing resistance to Pks-mediated killing. Additionally, pks mutants lost their competitive advantage, while act mutants lost their Pks resistance in in vivo competition assays. These findings provide insight into how closely related gut symbionts can compete and co-exist in the gastrointestinal tract.
Collapse
Affiliation(s)
- Mustafa Özçam
- Department of Food Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Restituto Tocmo
- Department of Food Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Deepa Acharya
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Shenwei Zhang
- Department of Food Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Theresa J Astmann
- Department of Food Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Mark Heggen
- Department of Food Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | | | - Fuyong Li
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Christopher C Cheng
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | - Eugenio Vivas
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Federico E Rey
- Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jan Claesen
- Department of Cardiovascular and Metabolic Sciences and Center for Microbiome and Human Health, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Tim S Bugni
- Pharmaceutical Sciences Division, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Jens Walter
- Department of Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2P5, Canada; Department of Medicine and APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland; School of Microbiology, University College Cork, Cork T12 YT20, Ireland
| | - Jan-Peter van Pijkeren
- Department of Food Science, University of Wisconsin-Madison, Madison, WI 53706, USA; Food Research Institute, University of Wisconsin-Madison, Madison, WI 53706, USA.
| |
Collapse
|
23
|
Mayank, Sidhu JS, Joshi G, Sindhu J, Kaur N, Singh N. Structural Diversity of D‐Alanine: D‐Alanine Ligase and Its Exploration in Development of Antibacterial Agents Against the Multi‐Variant Bacterial Infections. ChemistrySelect 2022. [DOI: 10.1002/slct.202104373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Mayank
- Department of Chemistry Indian Institute of Technology Ropar Punjab 140001 India
- School of Pharmaceutical Sciences Lovely Professional University Phagwara India
| | - Jagpreet Singh Sidhu
- Department of Pharmaceutical Sciences and Natural Products School of Health Science Central University of Punjab Bathinda 151 001 India
| | - Gaurav Joshi
- School of Pharmacy Graphic Era Hill University Dehradun Uttarakhand India
| | - Jayant Sindhu
- Department of Chemistry COBS&H CCS Haryana Agricultural University Hisar 125004 India
| | - Navneet Kaur
- Department of Chemistry Panjab University Chandigarh 160014 India
| | - Narinder Singh
- Department of Chemistry Indian Institute of Technology Ropar Punjab 140001 India
| |
Collapse
|
24
|
Gallus M, Vogel RF, Ehrmann MA. Optimization of a cultivation procedure to selectively isolate lactic acid bacteria from insects. J Appl Microbiol 2021; 132:3001-3016. [PMID: 34957661 DOI: 10.1111/jam.15427] [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: 09/06/2021] [Revised: 12/17/2021] [Accepted: 12/21/2021] [Indexed: 11/26/2022]
Abstract
AIMS Natural niches and transmission routes of lactic acid bacteria (LAB) are highly versatile. Proposed routes of transmission to food fermentations are from plant material via insects or vice versa. This study aimed to establish a method for the selective isolation of LAB from insects. METHODS AND RESULTS Varied parameters that influence growth and selectivity are temperature, type of carbohydrate and atmosphere. Additionally, effects of antibiotics to suppress non-LAB species were evaluated. A model consortium consisting of 12 species representing different lifestyles was inoculated in growth medium to identify conditions for highest diversity and recovery rate. The method was applied to isolate LAB from Drosophila melanogaster, Sitotroga cerealella, Tribolium castaneum and Tenebrio molitor. Isolated species were Leuconostoc mesenteroides, Paucilactobacillus vaccinostercus and Lactiplantibacillus plantarum from D. melanogaster and L. mesenteroides, Pediococcus pentosaceus and Latilactobacillus curvatus from T. molitor. No LAB could be isolated from T. castaneum and S. cerealella. 16S rDNA amplicon sequencing of DNA obtained from insects corroborated part of our results. CONCLUSION A combination of different enrichment conditions ensures a high probability to isolate LAB species from insects and can be helpful above already known non-cultivation methods. SIGNIFICANCE AND IMPACT OF THE STUDY The novel method allows to selectively isolate LAB from insects and the strategy of the method is of interest to study other niches.
Collapse
Affiliation(s)
- Marion Gallus
- Lehrstuhl für Technische Mikrobiologie, Technische Universität München, Gregor-Mendel-Str. 4, 85354, Freising, Germany
| | - Rudi F Vogel
- Lehrstuhl für Technische Mikrobiologie, Technische Universität München, Gregor-Mendel-Str. 4, 85354, Freising, Germany
| | - Matthias A Ehrmann
- Lehrstuhl für Technische Mikrobiologie, Technische Universität München, Gregor-Mendel-Str. 4, 85354, Freising, Germany
| |
Collapse
|
25
|
Liu HY, Giraud A, Seignez C, Ahl D, Guo F, Sedin J, Walden T, Oh JH, van Pijkeren JP, Holm L, Roos S, Bertilsson S, Phillipson M. Distinct B cell subsets in Peyer's patches convey probiotic effects by Limosilactobacillus reuteri. MICROBIOME 2021; 9:198. [PMID: 34602091 PMCID: PMC8487498 DOI: 10.1186/s40168-021-01128-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 07/01/2021] [Indexed: 05/02/2023]
Abstract
BACKGROUND Intestinal Peyer's patches (PPs) form unique niches for bacteria-immune cell interactions that direct host immunity and shape the microbiome. Here we investigate how peroral administration of probiotic bacterium Limosilactobacillus reuteri R2LC affects B lymphocytes and IgA induction in the PPs, as well as the downstream consequences on intestinal microbiota and susceptibility to inflammation. RESULTS The B cells of PPs were separated by size to circumvent activation-dependent cell identification biases due to dynamic expression of markers, which resulted in two phenotypically, transcriptionally, and spatially distinct subsets: small IgD+/GL7-/S1PR1+/Bcl6, CCR6-expressing pre-germinal center (GC)-like B cells with innate-like functions located subepithelially, and large GL7+/S1PR1-/Ki67+/Bcl6, CD69-expressing B cells with strong metabolic activity found in the GC. Peroral L. reuteri administration expanded both B cell subsets and enhanced the innate-like properties of pre-GC-like B cells while retaining them in the sub-epithelial compartment by increased sphingosine-1-phosphate/S1PR1 signaling. Furthermore, L. reuteri promoted GC-like B cell differentiation, which involved expansion of the GC area and autocrine TGFβ-1 activation. Consequently, PD-1-T follicular helper cell-dependent IgA induction and production was increased by L. reuteri, which shifted the intestinal microbiome and protected against dextran-sulfate-sodium induced colitis and dysbiosis. CONCLUSIONS The Peyer's patches sense, enhance and transmit probiotic signals by increasing the numbers and effector functions of distinct B cell subsets, resulting in increased IgA production, altered intestinal microbiota, and protection against inflammation. Video abstract.
Collapse
Affiliation(s)
- Hao-Yu Liu
- Laboratory of Animal Physiology and Molecular Nutrition, College of Animal Science and Technology, Yangzhou University, 225009 Yangzhou, PR China
- Department of Medical Cell Biology, Uppsala University, PO box 571, 75123 Uppsala, Sweden
| | - Antoine Giraud
- Department of Medical Cell Biology, Uppsala University, PO box 571, 75123 Uppsala, Sweden
| | - Cedric Seignez
- Department of Medical Cell Biology, Uppsala University, PO box 571, 75123 Uppsala, Sweden
| | - David Ahl
- Department of Medical Cell Biology, Uppsala University, PO box 571, 75123 Uppsala, Sweden
| | - Feilong Guo
- Department of Medical Cell Biology, Uppsala University, PO box 571, 75123 Uppsala, Sweden
| | - John Sedin
- Department of Medical Cell Biology, Uppsala University, PO box 571, 75123 Uppsala, Sweden
| | - Tomas Walden
- Department of Medical Cell Biology, Uppsala University, PO box 571, 75123 Uppsala, Sweden
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, WI 53706 USA
| | | | - Lena Holm
- Department of Medical Cell Biology, Uppsala University, PO box 571, 75123 Uppsala, Sweden
| | - Stefan Roos
- Department of Molecular Sciences, Swedish University of Agricultural Sciences, 75007 Uppsala, Sweden
| | - Stefan Bertilsson
- Department of Aquatic Sciences and Assessment, Swedish University of Agricultural Sciences, 75651 Uppsala, Sweden
- Science for Life Laboratory, 75237 Uppsala, Sweden
| | - Mia Phillipson
- Department of Medical Cell Biology, Uppsala University, PO box 571, 75123 Uppsala, Sweden
- Science for Life Laboratory, 75237 Uppsala, Sweden
| |
Collapse
|
26
|
Singh K, Ainala SK, Park S. Metabolic engineering of Lactobacillus reuteri DSM 20,016 for improved 1,3-propanediol production from glycerol. BIORESOURCE TECHNOLOGY 2021; 338:125590. [PMID: 34298333 DOI: 10.1016/j.biortech.2021.125590] [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] [Received: 05/26/2021] [Revised: 07/11/2021] [Accepted: 07/13/2021] [Indexed: 06/13/2023]
Abstract
The production of 1,3-propanediol (1,3-PDO) from glycerol was studied by GRAS and native 1,3-PDO producer, Lactobacillus reuteri DSM 20016. This strain ferments glucose with production of lactate, acetate, ethanol, and converts glycerol to 1,3-PDO using NADH generated by glucose metabolism. To improve 1,3-PDO production, alcohol dehydrogenases (ADH) were disrupted and 1,3-PDO oxidoreductases (PDOR) were overexpressed. Deletion of ADH (adh2) enhanced 1,3-PDO production yield on glucose by reducing ethanol synthesis, and overexpression of PDOR (pduQ) elevated 1,3-PDO production rate and cell growth rate. The strain with simultaneous adh2 deletion, pduQ overexpression (Δadh2pduQ++) could produce 687 mM 1,3-PDO with the yield of 1.2 ± 0.08 mol 1,3-PDO/mol glucose by fed-batch bioreactor cultivation in 48 h. However, the 1,3-PDO production rate was greatly reduced in the late period of bioreactor culture, mainly due to high lactate accumulation. This is the first report on rational metabolic engineering of L. reuteri for improved 1,3-PDO production.
Collapse
Affiliation(s)
- Kalpana Singh
- School of Energy and Chemical Engineering, UNIST, Ulsan 44919, Republic of Korea
| | - Satish Kumar Ainala
- NOROO Bio R&D Center, NOROO Holdings Co., Ltd, Gyeonggi-do 16229, Republic of Korea
| | - Sunghoon Park
- School of Energy and Chemical Engineering, UNIST, Ulsan 44919, Republic of Korea.
| |
Collapse
|
27
|
Fugaban JII, Vazquez Bucheli JE, Kim B, Holzapfel WH, Todorov SD. Safety and beneficial properties of bacteriocinogenic Pediococcus acidilactici and Pediococcus pentosaceus isolated from silage. Lett Appl Microbiol 2021; 73:725-734. [PMID: 34549812 DOI: 10.1111/lam.13562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/28/2021] [Accepted: 09/15/2021] [Indexed: 10/20/2022]
Abstract
The control of Listeria monocytogenes is a relevant goal for dairy products, a process that begins from the supply of feed and management of animals' health. In the present study, we evaluated the safety of two bacteriocinogenic Pediococcus strains and show that both can be considered as safe, based on their haemolytic activity, biogenic amine production and antibiotic resistance, all evaluated through phenotypical and biomolecular approaches. Both strains have shown potential as a producer of γ-aminobutiric acid (GABA) and carry an incomplete set of genes related to folate biosynthesis; both strains were able to adhere to Caco-2 cell lines with adhesion rates of 6·59% ± 3·73 and 0·84% ± 0·48. Laboratory prepared clover silage, inoculated with each bacteriocinogenic Pediococcus strain and contaminated with L. monocytogenes, proved the hypothesis for bioprotective effect of the tested strains, with the tested pathogen eliminated in the first 24 h of the experiment. These results indicate that evaluated strains can be potential beneficial candidates for application in silage production.
Collapse
Affiliation(s)
- J I I Fugaban
- ProBacLab, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, Republic of Korea
| | - J E Vazquez Bucheli
- ProBacLab, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, Republic of Korea
| | - B Kim
- HEM Inc., Pohang, Gyungbuk, Republic of Korea
| | - W H Holzapfel
- ProBacLab, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, Republic of Korea
| | - S D Todorov
- ProBacLab, Department of Advanced Convergence, Handong Global University, Pohang, Gyeongbuk, Republic of Korea
| |
Collapse
|
28
|
Baker LM, Davies TS, Masetti G, Hughes TR, Marchesi JR, Jack AA, Joyce TSC, Allen MD, Plummer SF, Michael DR, Ramanathan G, Del Sol R, Facey PD. A genome guided evaluation of the Lab4 probiotic consortium. Genomics 2021; 113:4028-4038. [PMID: 34391865 DOI: 10.1016/j.ygeno.2021.08.007] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/16/2021] [Accepted: 08/10/2021] [Indexed: 12/01/2022]
Abstract
In this study, we present the draft genome sequences of the Lab4 probiotic consortium using whole genome sequencing. Draft genome sequences were retrieved and deposited for each of the organisms; PRJNA559984 for B. bifidum CUL20, PRJNA482335 for Lactobacillus acidophilus CUL60, PRJNA482434 for Lactobacillus acid. Probiogenomic in silico analyses confirmed existing taxonomies and identified the presence putative gene sequences that were functionally related to the performance of each organism during in vitro assessments of bile and acid tolerability, adherence to enterocytes and susceptibility to antibiotics. Predictions of genomic stability identified no significant risk of horizontal gene transfer in any of the Lab4 strains and the absence of both antibiotic resistance and virulence genes. These observations were supported by the outcomes of acute phase and repeat dose tolerability studies in Wistar rats where challenge with high doses of Lab4 did not result in any mortalities, clinical/histopathological abnormalities nor indications of systemic toxicity. Detection of increased numbers of lactobacilli and bifidobacteria in the faeces of supplemented rats implied an ability to survive transit through the gastrointestinal tract and/or impact upon the intestinal microbiota composition. In summary, this study provides in silico, in vitro and in vivo support for probiotic functionality and the safety of the Lab4 consortium.
Collapse
Affiliation(s)
- L M Baker
- Swansea University Medical School, Swansea University, Singleton Park Campus, Swansea SA2 8PP, United Kingdom
| | - T S Davies
- Cultech Limited, Unit 2 Christchurch Road, Baglan Industrial Park, Port Talbot SA12 7BZ, United Kingdom
| | - G Masetti
- Cultech Limited, Unit 2 Christchurch Road, Baglan Industrial Park, Port Talbot SA12 7BZ, United Kingdom
| | - T R Hughes
- Systems Immunity Research Institute, Henry Welcome Building, Cardiff University, CF14 4XN, United Kingdom
| | - J R Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - A A Jack
- Cultech Limited, Unit 2 Christchurch Road, Baglan Industrial Park, Port Talbot SA12 7BZ, United Kingdom
| | - T S C Joyce
- Cultech Limited, Unit 2 Christchurch Road, Baglan Industrial Park, Port Talbot SA12 7BZ, United Kingdom
| | - M D Allen
- Cultech Limited, Unit 2 Christchurch Road, Baglan Industrial Park, Port Talbot SA12 7BZ, United Kingdom
| | - S F Plummer
- Cultech Limited, Unit 2 Christchurch Road, Baglan Industrial Park, Port Talbot SA12 7BZ, United Kingdom
| | - D R Michael
- Cultech Limited, Unit 2 Christchurch Road, Baglan Industrial Park, Port Talbot SA12 7BZ, United Kingdom
| | - G Ramanathan
- Pharmacology based Clinical Trials, Pennington Biomedical Research Center, 6400 Perkins Rd, Baton Rouge, LA 70808, USA
| | - R Del Sol
- Swansea University Medical School, Swansea University, Singleton Park Campus, Swansea SA2 8PP, United Kingdom
| | - P D Facey
- Swansea University Medical School, Swansea University, Singleton Park Campus, Swansea SA2 8PP, United Kingdom.
| |
Collapse
|
29
|
Qin Y, Xu L, Teng Y, Wang Y, Ma P. Discovery of novel antibacterial agents: Recent developments in D-alanyl-D-alanine ligase inhibitors. Chem Biol Drug Des 2021; 98:305-322. [PMID: 34047462 DOI: 10.1111/cbdd.13899] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/09/2021] [Accepted: 05/23/2021] [Indexed: 01/14/2023]
Abstract
Bacterial infections can cause serious problems that threaten public health over a long period of time. Moreover, the continuous emergence of drug-resistant bacteria necessitates the development of novel antibacterial agents. D-alanyl-D-alanine ligase (Ddl) is an indispensable adenosine triphosphate-dependent bacterial enzyme involved in the biosynthesis of peptidoglycan precursor, which catalyzes the ligation of two D-alanine molecules into one D-alanyl-D-alanine dipeptide. This dipeptide is an essential component of the intracellular peptidoglycan precursor, uridine diphospho-N-acetylmuramic acid (UDP-MurNAc)-pentapeptide, that maintains the integrity of the bacterial cell wall by cross-linking the peptidoglycan chain, and is crucial for the survival of pathogens. Consequently, Ddl is expected to be a promising target for the development of antibacterial agents. In this review, we present a brief introduction regarding the structure and function of Ddl, as well as an overview of the various Ddl inhibitors currently being used as antibacterial agents, specifically highlighting their inhibitory activities, structure-activity relationships and mechanisms of action.
Collapse
Affiliation(s)
- Yinhui Qin
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, China
| | - Linlin Xu
- Department of Pharmacy, Taian City Central Hospital, Taian, China
| | - Yuetai Teng
- Department of Pharmacy, Jinan Vocational College of Nursing, Jinan, China
| | - Yinhu Wang
- School of Pharmaceutical Sciences, Liaocheng University, Liaocheng, China
| | - Peizhi Ma
- Department of Pharmacy, Henan Provincial People's Hospital, Zhengzhou University People's Hospital, Henan University People's Hospital, Zhengzhou, China
| |
Collapse
|
30
|
Guan Y, Cui Y, Qu X, Jing K. Safety and robustness aspects analysis of Lactobacillus delbrueckii ssp. bulgaricus LDB-C1 based on the genome analysis and biological tests. Arch Microbiol 2021; 203:3955-3964. [PMID: 34021387 DOI: 10.1007/s00203-021-02383-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 04/21/2021] [Accepted: 05/12/2021] [Indexed: 12/26/2022]
Abstract
Lactobacillus delbrueckii subsp. bulgaricus (L. bulgaricus) is a microaerophylic anaerobe, which is widely used in the production of yogurt, cheese, and other fermented dairy products. L. bulgaricus and its partner Streptococcus thermophilus were used as starter cultures of yogurt in the world for thousands of years. In our previous study, L. bulgaricus LDB-C1 was obtained from traditional fermented milk, and possessed some characteristics like high exopolysaccharide yield and good fermentation performance. The analysis of its CRISPR-Cas system, antibiotic resistance, virulence factors, and mobile elements, was performed to reveal the stability of the strain LDB-C1. It was found that LDB-C1 contains a plenty of spacers in the CRISPR region, indicating it might have better performance against the infection of phages and plasmids. Furthermore, the acquired or transmittable antibiotic resistance/virulence factor genes were absent in the tested L. bulgaricus strain LDB-C1.
Collapse
Affiliation(s)
- Yuxuan Guan
- Department of Food Science and Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150090, People's Republic of China
| | - Yanhua Cui
- Department of Food Science and Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150090, People's Republic of China.
| | - Xiaojun Qu
- Institute of Microbiology, Heilongjiang Academy of Sciences, Harbin, 150010, People's Republic of China
| | - Kai Jing
- Department of Food Science and Engineering, School of Chemistry and Chemical Engineering, Harbin Institute of Technology, Harbin, 150090, People's Republic of China
| |
Collapse
|
31
|
Tuyarum C, Songsang A, Lertworapreecha M. In vitro evaluation of the probiotic potential of Lactobacillus isolated from native swine manure. Vet World 2021; 14:1133-1142. [PMID: 34220114 PMCID: PMC8243659 DOI: 10.14202/vetworld.2021.1133-1142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/22/2021] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: Using antimicrobials as a feed additive in swine production is prohibited because it is a major cause of the emergence of antimicrobial-resistant bacteria. Probiotics such as Lactobacillus spp. are an attractive alternative to reduce antimicrobial resistance and promote swine growth. This study aimed to evaluate the in vitro probiotic properties of Lactobacillus isolated from indigenous swine manure. Materials and Methods: A total of 30 fecal samples from healthy individual indigenous pigs were collected and isolated on de Man, Rogosa, and Sharpe agar. The preliminary screen identified candidates with antibacterial activity against six pathogens and >50% survival and tolerance to acid (pH 3.0) and 1% bile salt. Isolates that passed the initial screen will be tested for other probiotic properties. Results: Of the 314 isolates from 30 pig manure samples, 17 isolates satisfied all initial conditions for probiotic properties. Each isolate has unique, distinctive properties. Isolates B4, B5, B8, B17, B87, and B144 formed thick biofilms, whereas isolates B5, B8, and 27 adhered well to the intestinal wall and exhibited strong autoaggregation properties. Isolate B4 aggregated with Enterohemorrhagic Escherichia coli and Enteropathogenic E. coli. Tests in pH-adjusted cell-free medium indicated that the antibacterial activity resulted from bacterial acidification rather than bacteriocin formation. Sequence analysis (16S rRNA) revealed 16 of the isolates were Lactobacillus plantarum, and only one isolate was Lactobacillus salivarius. Conclusion: We isolated 17 Lactobacillus from swine manure and demonstrated that their probiotic properties might be useful as a probiotic cocktail for swine feed.
Collapse
Affiliation(s)
- Chiraprapha Tuyarum
- Microbiology Program, Department of Biology, Faculty of Science, Thaksin University, Phatthalung, 93210, Thailand
| | - Aporn Songsang
- Faculty of Technology and Community Development, Thaksin University, Phatthalung, 93210, Thailand
| | - Monthon Lertworapreecha
- Microbiology Program, Department of Biology, Faculty of Science, Thaksin University, Phatthalung, 93210, Thailand
| |
Collapse
|
32
|
Zhang Z, Wang K, Oh JH, Zhang S, van Pijkeren JP, Cheng CC, Ren D, Wei H, Gänzle MG, Walter J. A Phylogenetic View on the Role of Glycerol for Growth Enhancement and Reuterin Formation in Limosilactobacillus reuteri. Front Microbiol 2021; 11:601422. [PMID: 33408707 PMCID: PMC7779471 DOI: 10.3389/fmicb.2020.601422] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Accepted: 12/02/2020] [Indexed: 11/13/2022] Open
Abstract
Lineages within the species Limosilactobacillus reuteri have specialized to various hosts and their genomes reflect these adaptations. The pdu-cbi-cob-hem gene cluster is conserved in most human and poultry isolates but is infrequent in rodent and porcine isolates. This gene cluster confers the transformation of glycerol into 3-hydroxy-propionaldehyde (reuterin), which can either be secreted and function as precursor of the antimicrobial compound acrolein or serve as an electron acceptor that enhances the organisms’ growth rate. However, it remains unclear which of these two functions is more relevant for L. reuteri evolution and ecology. Here we characterized the effect of glycerol on growth rate and reuterin formation in L. reuteri strains across different phylogenetic lineages during growth on ecologically relevant carbohydrates. We further evaluated the innate reuterin resistance among these strains to infer a possible role of reuterin in the evolution of strains. Results revealed that the poultry/human lineage VI strain, L. reuteri DSM 17938 shows more growth enhancement through glycerol and greater capacity for reuterin production on glucose and maltose as compared to human lineage II strains. Interestingly, reuterin production in lineage II strains was significantly elevated on raffinose and lactose, reaching levels similar to DSM 17938. On all carbohydrates tested, reuterin production occurred during the exponential growth phase and became undetectable during the stationary growth phase. The amount of reuterin produced was sufficient to inhibit E. coli, suggesting that it could be ecologically relevant, but the resistance towards reuterin among L. reuteri strains was highly variable and, for the most part, unrelated to the strain’s capacity for reuterin production. Overall, the findings suggest differences in the substrate-specific regulation of the pdu cluster in L. reuteri lineages that might be reflective of their ecological niches, e.g., chicken foregut versus human infant and adult large intestine. Such information can inform future studies on the ecology of L. reuteri and guide the development of synbiotic applications to improve the therapeutic use of this species.
Collapse
Affiliation(s)
- Zhihong Zhang
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China.,Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Kaiming Wang
- Department of Physiology, CEGIIR, University of Alberta, Edmonton, AB, Canada
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, United States
| | - Shenwei Zhang
- Department of Food Science, University of Wisconsin-Madison, Madison, WI, United States
| | | | - Christopher C Cheng
- Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Dayong Ren
- Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada.,College of Food Science and Engineering, Jilin Agricultural University, Changchun, China
| | - Hua Wei
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Michael G Gänzle
- Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Jens Walter
- Agricultural, Food and Nutritional Science, University of Alberta, Edmonton, AB, Canada.,Department of Physiology, CEGIIR, University of Alberta, Edmonton, AB, Canada.,Department of Biological Sciences, University of Alberta, Edmonton, AB, Canada.,APC Microbiome Ireland, School of Microbiology, University College Cork, Cork, Ireland.,Department of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
33
|
Oh JH, Schueler KL, Stapleton DS, Alexander LM, Yen CLE, Keller MP, Attie AD, van Pijkeren JP. Secretion of Recombinant Interleukin-22 by Engineered Lactobacillus reuteri Reduces Fatty Liver Disease in a Mouse Model of Diet-Induced Obesity. mSphere 2020; 5:e00183-20. [PMID: 32581074 PMCID: PMC7316485 DOI: 10.1128/msphere.00183-20] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/08/2020] [Indexed: 02/06/2023] Open
Abstract
The incidence of metabolic syndrome continues to rise globally. In mice, intravenous administration of interleukin-22 (IL-22) ameliorates various disease phenotypes associated with diet-induced metabolic syndrome. In patients, oral treatment is favored over intravenous treatment, but methodologies to deliver IL-22 via the oral route are nonexistent. The goal of this study was to assess to what extent engineered Lactobacillus reuteri secreting IL-22 could ameliorate nonalcoholic fatty liver disease. We used a mouse model of diet-induced obesity and assessed various markers of metabolic syndrome following treatment with L. reuteri and a recombinant derivative. Mice that received an 8-week treatment of wild-type probiotic gained less weight and had a smaller fat pad than the control group, but these phenotypes were not further enhanced by recombinant L. reuteri However, L. reuteri secreting IL-22 significantly reduced liver weight and triglycerides at levels that exceeded those of the probiotic wild-type treatment group. Our findings are interesting in light of the observed phenotypes associated with reduced nonalcoholic liver disease, in humans the most prevalent chronic liver disease, following treatment of a next-generation probiotic that is administered orally. Once biological and environmental containment strategies are in place, therapeutic applications of recombinant Lactobacillus reuteri are on the horizon.IMPORTANCE In humans, nonalcoholic fatty liver disease (NAFLD) is the most prevalent liver disease due to the increased prevalence of obesity. While treatment of NAFLD is often geared toward lifestyle changes, such as diet and exercise, the use of dietary supplements such as probiotics is underinvestigated. Here, we report that probiotic Lactobacillus reuteri reduces fatty liver in a mouse model of diet-induced obesity. This phenotype was further enhanced upon delivery of recombinant interleukin-22 by engineered Lactobacillus reuteri These observations pave the road to a better understanding of probiotic mechanisms driving the reduction of diet-induced steatosis and to development of next-generation probiotics for use in the clinic. Ultimately, these studies may lead to rational selection of (engineered) probiotics to ameliorate fatty liver disease.
Collapse
Affiliation(s)
- Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kathryn L Schueler
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Donnie S Stapleton
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Laura M Alexander
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Chi-Liang Eric Yen
- Department of Nutritional Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | |
Collapse
|
34
|
Ortiz-Velez L, Goodwin A, Schaefer L, Britton RA. Challenges and Pitfalls in the Engineering of Human Interleukin 22 (hIL-22) Secreting Lactobacillus reuteri. Front Bioeng Biotechnol 2020; 8:543. [PMID: 32582668 PMCID: PMC7289926 DOI: 10.3389/fbioe.2020.00543] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 05/05/2020] [Indexed: 12/12/2022] Open
Abstract
Engineered microbes for the delivery of intestinally directed therapeutics is a promising avenue for the treatment of various intestinal diseases including inflammatory bowel disease (IBD) and intestinal graft vs. host disease (GVHD). This modality of treatment would allow for the targeted delivery of therapeutics to the site of inflammation or disease while minimizing the systemic side effects that often accompany treatment of these pathologies. Here, we show the challenges encountered and overcome in successfully engineering Lactobacillus reuteri to secrete high levels of biologically active human interleukin 22 (hIL-22). Initial hIL-22 constructs secreted high levels of hIL-22, however we found the majority of hIL-22 was cleaved and not biologically active. Several strategies were explored to improve the production of intact hIL-22, with the optimization of the signal sequence for peptide secretion having the most impact of production of intact hIL-22. This resulted in L. reuteri secreting high concentrations (up to 700 ng/mL) of hIL-22. Bioactivity of hIL-22 was confirmed by the secretion of interleukin 10 (IL-10) from the colon cancer derived epithelial cell line Colo205 and the secretion of Regenerating islet-derived protein 3 alpha (Reg3α) from human jejunal enteroids. The secretion of bioactive hIL-22 imposed a significant cost for L. reuteri as bacterial growth was significantly impaired upon induction. Future challenges and optimization strategies for the delivery of hIL-22 to the human intestinal tract are discussed.
Collapse
Affiliation(s)
- Laura Ortiz-Velez
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Annie Goodwin
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
- Section of Pediatric Gastroenterology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, Houston, TX, United States
| | - Laura Schaefer
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
| | - Robert A. Britton
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, United States
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, United States
| |
Collapse
|
35
|
Cheng CC, Duar RM, Lin X, Perez-Munoz ME, Tollenaar S, Oh JH, van Pijkeren JP, Li F, van Sinderen D, Gänzle MG, Walter J. Ecological Importance of Cross-Feeding of the Intermediate Metabolite 1,2-Propanediol between Bacterial Gut Symbionts. Appl Environ Microbiol 2020; 86:e00190-20. [PMID: 32276972 PMCID: PMC7237793 DOI: 10.1128/aem.00190-20] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2020] [Accepted: 03/27/2020] [Indexed: 12/20/2022] Open
Abstract
Cross-feeding based on the metabolite 1,2-propanediol has been proposed to have an important role in the establishment of trophic interactions among gut symbionts, but its ecological importance has not been empirically established. Here, we show that in vitro growth of Lactobacillus reuteri (syn. Limosilactobacillus reuteri) ATCC PTA 6475 is enhanced through 1,2-propanediol produced by Bifidobacterium breve UCC2003 and Escherichia coli MG1655 from the metabolization of fucose and rhamnose, respectively. Work with isogenic mutants showed that the trophic interaction is dependent on the pduCDE operon in L. reuteri, which encodes the ability to use 1,2-propanediol, and the l-fucose permease (fucP) gene in B. breve, which is required for 1,2-propanediol formation from fucose. Experiments in gnotobiotic mice revealed that, although the pduCDE operon bestows a fitness burden on L. reuteri ATCC PTA 6475 in the mouse digestive tract, the ecological performance of the strain was enhanced in the presence of B. breve UCC2003 and the mucus-degrading species Bifidobacterium bifidum The use of the respective pduCDE and fucP mutants of L. reuteri and B. breve in the mouse experiments indicated that the trophic interaction was specifically based on 1,2-propanediol. Overall, our work established the ecological importance of cross-feeding relationships based on 1,2-propanediol for the fitness of a bacterial symbiont in the vertebrate gut.IMPORTANCE Through experiments in gnotobiotic mice that employed isogenic mutants of bacterial strains that produce (Bifidobacterium breve) and utilize (Lactobacillus reuteri) 1,2-propanediol, this study provides mechanistic insight into the ecological ramifications of a trophic interaction between gut symbionts. The findings improve our understanding on how cross-feeding influences the competitive fitness of L. reuteri in the vertebrate gut and revealed a putative selective force that shaped the evolution of the species. The findings are relevant since they provide a basis to design rational microbial-based strategies to modulate gut ecosystems, which could employ mixtures of bacterial strains that establish trophic interactions or a personalized approach based on the ability of a resident microbiota to provide resources for the incoming microbe.
Collapse
Affiliation(s)
| | - Rebbeca M Duar
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada
- Evolve BioSystems, Inc., Davis, California, USA
| | - Xiaoxi Lin
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada
| | - Maria Elisa Perez-Munoz
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada
| | - Stephanie Tollenaar
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | | - Fuyong Li
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada
| | - Douwe van Sinderen
- School of Microbiology and APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Michael G Gänzle
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada
| | - Jens Walter
- Department of Biological Sciences, University of Alberta, Edmonton, Canada
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, Edmonton, Canada
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
- School of Microbiology and APC Microbiome Institute, University College Cork, Cork, Ireland
- Department of Medicine, University College Cork, Cork, Ireland
| |
Collapse
|
36
|
Zheng J, Wittouck S, Salvetti E, Franz CMAP, Harris HMB, Mattarelli P, O'Toole PW, Pot B, Vandamme P, Walter J, Watanabe K, Wuyts S, Felis GE, Gänzle MG, Lebeer S. A taxonomic note on the genus Lactobacillus: Description of 23 novel genera, emended description of the genus Lactobacillus Beijerinck 1901, and union of Lactobacillaceae and Leuconostocaceae. Int J Syst Evol Microbiol 2020; 70:2782-2858. [PMID: 32293557 DOI: 10.1099/ijsem.0.004107] [Citation(s) in RCA: 1570] [Impact Index Per Article: 392.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The genus Lactobacillus comprises 261 species (at March 2020) that are extremely diverse at phenotypic, ecological and genotypic levels. This study evaluated the taxonomy of Lactobacillaceae and Leuconostocaceae on the basis of whole genome sequences. Parameters that were evaluated included core genome phylogeny, (conserved) pairwise average amino acid identity, clade-specific signature genes, physiological criteria and the ecology of the organisms. Based on this polyphasic approach, we propose reclassification of the genus Lactobacillus into 25 genera including the emended genus Lactobacillus, which includes host-adapted organisms that have been referred to as the Lactobacillus delbrueckii group, Paralactobacillus and 23 novel genera for which the names Holzapfelia, Amylolactobacillus, Bombilactobacillus, Companilactobacillus, Lapidilactobacillus, Agrilactobacillus, Schleiferilactobacillus, Loigolactobacilus, Lacticaseibacillus, Latilactobacillus, Dellaglioa, Liquorilactobacillus, Ligilactobacillus, Lactiplantibacillus, Furfurilactobacillus, Paucilactobacillus, Limosilactobacillus, Fructilactobacillus, Acetilactobacillus, Apilactobacillus, Levilactobacillus, Secundilactobacillus and Lentilactobacillus are proposed. We also propose to emend the description of the family Lactobacillaceae to include all genera that were previously included in families Lactobacillaceae and Leuconostocaceae. The generic term 'lactobacilli' will remain useful to designate all organisms that were classified as Lactobacillaceae until 2020. This reclassification reflects the phylogenetic position of the micro-organisms, and groups lactobacilli into robust clades with shared ecological and metabolic properties, as exemplified for the emended genus Lactobacillus encompassing species adapted to vertebrates (such as Lactobacillus delbrueckii, Lactobacillus iners, Lactobacillus crispatus, Lactobacillus jensensii, Lactobacillus johnsonii and Lactobacillus acidophilus) or invertebrates (such as Lactobacillus apis and Lactobacillus bombicola).
Collapse
Affiliation(s)
- Jinshui Zheng
- Huazhong Agricultural University, State Key Laboratory of Agricultural Microbiology, Hubei Key Laboratory of Agricultural Bioinformatics, Wuhan, Hubei, PR China
| | - Stijn Wittouck
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | - Elisa Salvetti
- Dept. of Biotechnology, University of Verona, Verona, Italy
| | - Charles M A P Franz
- Max Rubner-Institut, Department of Microbiology and Biotechnology, Kiel, Germany
| | - Hugh M B Harris
- School of Microbiology & APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
| | - Paola Mattarelli
- University of Bologna, Dept. of Agricultural and Food Sciences, Bologna, Italy
| | - Paul W O'Toole
- School of Microbiology & APC Microbiome Ireland, University College Cork, Co. Cork, Ireland
| | - Bruno Pot
- Research Group of Industrial Microbiology and Food Biotechnology (IMDO), Vrije Universiteit Brussel, Brussels, Belgium
| | - Peter Vandamme
- Laboratory of Microbiology, Department of Biochemistry and Microbiology, Ghent University, Ghent, Belgium
| | - Jens Walter
- Department of Biological Sciences, University of Alberta, Edmonton, Canada.,Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Canada
| | - Koichi Watanabe
- Food Industry Research and Development Institute, Bioresource Collection and Research Center, Hsinchu, Taiwan, ROC.,National Taiwan University, Dept. of Animal Science and Technology, Taipei, Taiwan, ROC
| | - Sander Wuyts
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| | | | - Michael G Gänzle
- Hubei University of Technology, College of Bioengineering and Food Science, Wuhan, Hubei, PR China.,Department of Agricultural, Food & Nutritional Science, University of Alberta, Edmonton, Canada
| | - Sarah Lebeer
- Research Group Environmental Ecology and Applied Microbiology, Department of Bioscience Engineering, University of Antwerp, Antwerp, Belgium
| |
Collapse
|
37
|
Genetic Determinants of Hydroxycinnamic Acid Metabolism in Heterofermentative Lactobacilli. Appl Environ Microbiol 2020; 86:AEM.02461-19. [PMID: 31862715 DOI: 10.1128/aem.02461-19] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2019] [Accepted: 12/16/2019] [Indexed: 02/06/2023] Open
Abstract
Phenolic acids are among the most abundant phenolic compounds in edible parts of plants. Lactic acid bacteria (LAB) metabolize phenolic acids, but the enzyme responsible for reducing hydroxycinnamic acids to phenylpropionic acids (HcrB) was only recently characterized in Lactobacillus plantarum In this study, heterofermentative LAB species were screened for their hydroxycinnamic acid metabolism. Data on strain-specific metabolism in combination with comparative genomic analyses identified homologs of HcrB as putative phenolic acid reductases. Par1 and HcrF both encode putative multidomain proteins with 25% and 63% amino acid identity to HcrB, respectively. Of these genes, par1 in L. rossiae and hcrF in L. fermentum were overexpressed in response to hydroxycinnamic acids. The deletion of par1 in L. rossiae led to the loss of phenolic acid metabolism. The strain-specific metabolism of phenolic acids was congruent with the genotype of lactobacilli; however, phenolic acid reductases were not identified in strains of Weissella cibaria that reduced hydroxycinnamic acids to phenylpropionic acids. Phylogenetic analysis of major genes involved in hydroxycinnamic acid metabolism in strains of the genus Lactobacillus revealed that Par1 was found to be the most widely distributed phenolic acid reductase, while HcrB was the least abundant, present in less than 9% of Lactobacillus spp. In conclusion, this study increased the knowledge on the genetic determinants of hydroxycinnamic acid metabolism, explaining the species- and strain-specific metabolic variations in lactobacilli and providing evidence of additional enzymes involved in hydroxycinnamic acid metabolism of lactobacilli.IMPORTANCE The metabolism of secondary plant metabolites, including phenolic compounds, by food-fermenting lactobacilli is a significant contributor to the safety, quality, and nutritional quality of fermented foods. The enzymes mediating hydrolysis, reduction, and decarboxylation of phenolic acid esters and phenolic acids in lactobacilli, however, are not fully characterized. The genomic analyses presented here provide evidence for three novel putative phenolic acid reductases. Matching comparative genomic analyses with phenotypic analysis and quantification of gene expression indicates that two of the three putative phenolic acid reductases, Par1 and HcrF, are involved in reduction of hydroxycinnamic acids to phenylpropionic acids; however, the activity of Par2 may be unrelated to phenolic acids and recognizes other secondary plant metabolites. These findings expand our knowledge on the metabolic potential of lactobacilli and facilitate future studies on activity and substrate specificity of enzymes involved in metabolism of phenolic compounds.
Collapse
|
38
|
Rozman V, Mohar Lorbeg P, Accetto T, Bogovič Matijašić B. Characterization of antimicrobial resistance in lactobacilli and bifidobacteria used as probiotics or starter cultures based on integration of phenotypic and in silico data. Int J Food Microbiol 2020; 314:108388. [DOI: 10.1016/j.ijfoodmicro.2019.108388] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2019] [Revised: 10/02/2019] [Accepted: 10/11/2019] [Indexed: 12/16/2022]
|
39
|
Towards high-throughput genome engineering in lactic acid bacteria. Curr Opin Biotechnol 2020; 61:181-188. [DOI: 10.1016/j.copbio.2019.12.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 12/07/2019] [Accepted: 12/17/2019] [Indexed: 11/22/2022]
|
40
|
Prophages in Lactobacillus reuteri Are Associated with Fitness Trade-Offs but Can Increase Competitiveness in the Gut Ecosystem. Appl Environ Microbiol 2019; 86:AEM.01922-19. [PMID: 31676478 PMCID: PMC6912086 DOI: 10.1128/aem.01922-19] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 10/24/2019] [Indexed: 12/20/2022] Open
Abstract
Bacteriophages derived from lysogens are abundant in gut microbiomes. Currently, mechanistic knowledge is lacking on the ecological ramifications of prophage carriage yet is essential to explain the abundance of lysogens in the gut. An extensive screen of the bacterial gut symbiont Lactobacillus reuteri revealed that biologically active prophages are widely distributed in this species. L. reuteri 6475 produces phages throughout the mouse intestinal tract, but phage production is associated with reduced fitness of the lysogen. However, phage production provides a competitive advantage in direct competition with a nonlysogenic strain of L. reuteri that is sensitive to these phages. This combination of increased competition with a fitness trade-off provides a potential explanation for the domination of lysogens in gut ecosystem and how lysogens can coexist with sensitive hosts. The gut microbiota harbors a diverse phage population that is largely derived from lysogens, which are bacteria that contain dormant phages in their genome. While the diversity of phages in gut ecosystems is getting increasingly well characterized, knowledge is limited on how phages contribute to the evolution and ecology of their host bacteria. Here, we show that biologically active prophages are widely distributed in phylogenetically diverse strains of the gut symbiont Lactobacillus reuteri. Nearly all human- and rodent-derived strains, but less than half of the tested strains of porcine origin, contain active prophages, suggesting different roles of phages in the evolution of host-specific lineages. To gain insight into the ecological role of L. reuteri phages, we developed L. reuteri strain 6475 as a model to study its phages. After administration to mice, L. reuteri 6475 produces active phages throughout the intestinal tract, with the highest number detected in the distal colon. Inactivation of recA abolished in vivo phage production, which suggests that activation of the SOS response drives phage production in the gut. In conventional mice, phage production reduces bacterial fitness as fewer wild-type bacteria survive gut transit compared to the mutant lacking prophages. However, in gnotobiotic mice, phage production provides L. reuteri with a competitive advantage over a sensitive host. Collectively, we uncovered that the presence of prophages, although associated with a fitness trade-off, can be advantageous for a gut symbiont by killing a competitor strain in its intestinal niche. IMPORTANCE Bacteriophages derived from lysogens are abundant in gut microbiomes. Currently, mechanistic knowledge is lacking on the ecological ramifications of prophage carriage yet is essential to explain the abundance of lysogens in the gut. An extensive screen of the bacterial gut symbiont Lactobacillus reuteri revealed that biologically active prophages are widely distributed in this species. L. reuteri 6475 produces phages throughout the mouse intestinal tract, but phage production is associated with reduced fitness of the lysogen. However, phage production provides a competitive advantage in direct competition with a nonlysogenic strain of L. reuteri that is sensitive to these phages. This combination of increased competition with a fitness trade-off provides a potential explanation for the domination of lysogens in gut ecosystem and how lysogens can coexist with sensitive hosts.
Collapse
|
41
|
CRISPR/Cas9-Assisted Seamless Genome Editing in Lactobacillus plantarum and Its Application in N-Acetylglucosamine Production. Appl Environ Microbiol 2019; 85:AEM.01367-19. [PMID: 31444197 DOI: 10.1128/aem.01367-19] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2019] [Accepted: 08/14/2019] [Indexed: 12/30/2022] Open
Abstract
Lactobacillus plantarum is a potential starter and health-promoting probiotic bacterium. Effective, precise, and diverse genome editing of Lactobacillus plantarum without introducing exogenous genes or plasmids is of great importance. In this study, CRISPR/Cas9-assisted double-stranded DNA (dsDNA) and single-stranded DNA (ssDNA) recombineering was established in L. plantarum WCFS1 to seamlessly edit the genome, including gene knockouts, insertions, and point mutations. To optimize our editing method, phosphorothioate modification was used to improve the dsDNA insertion, and adenine-specific methyltransferase was used to improve the ssDNA recombination efficiency. These strategies were applied to engineer L. plantarum WCFS1 toward producing N-acetylglucosamine (GlcNAc). nagB was truncated to eliminate the reverse reaction of fructose-6-phosphate (F6P) to glucosamine 6-phosphate (GlcN-6P). Riboswitch replacement and point mutation in glmS1 were introduced to relieve feedback repression. The resulting strain produced 797.3 mg/liter GlcNAc without introducing exogenous genes or plasmids. This strategy may contribute to the available methods for precise and diverse genetic engineering in lactic acid bacteria and boost strain engineering for more applications.IMPORTANCE CRISPR/Cas9-assisted recombineering is restricted in lactic acid bacteria because of the lack of available antibiotics and vectors. In this study, a seamless genome editing method was carried out in Lactobacillus plantarum using CRISPR/Cas9-assisted double-stranded DNA (dsDNA) and single-stranded DNA (ssDNA) recombineering, and recombination efficiency was effectively improved by endogenous adenine-specific methyltransferase overexpression. L. plantarum WCFS1 produced 797.3 mg/liter N-acetylglucosamine (GlcNAc) through reinforcement of the GlcNAc pathway, without introducing exogenous genes or plasmids. This seamless editing strategy, combined with the potential exogenous GlcNAc-producing pathway, makes this strain an attractive candidate for industrial use in the future.
Collapse
|
42
|
Zuo F, Zeng Z, Hammarström L, Marcotte H. Inducible Plasmid Self-Destruction (IPSD) Assisted Genome Engineering in Lactobacilli and Bifidobacteria. ACS Synth Biol 2019; 8:1723-1729. [PMID: 31277549 DOI: 10.1021/acssynbio.9b00114] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Genome engineering is essential for application of synthetic biology in probiotics including lactobacilli and bifidobacteria. Several homologous recombination system-based mutagenesis tools have been developed for these bacteria, but still have many limitations in different species or strains. Here we developed a genome engineering method based on an inducible self-destruction plasmid delivering homologous DNA into bacteria. Excision of the replicon by induced recombinase facilitates selection of homologous recombination events. This new genome editing tool called inducible plasmid self-destruction (IPSD) was successfully used to perform gene knockout and knock-in in lactobacilli and bifidobacteria. Due to its simplicity and universality, the IPSD strategy may provide a general approach for genetic engineering of various bacterial species.
Collapse
Affiliation(s)
- Fanglei Zuo
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China
- Department of Laboratory Medicine, Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm SE-141 86, Sweden
| | - Zhu Zeng
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, PR China
- Department of Laboratory Medicine, Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm SE-141 86, Sweden
| | - Lennart Hammarström
- Department of Laboratory Medicine, Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm SE-141 86, Sweden
| | - Harold Marcotte
- Department of Laboratory Medicine, Division of Clinical Immunology and Transfusion Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm SE-141 86, Sweden
| |
Collapse
|
43
|
Alexander LM, Oh JH, Stapleton DS, Schueler KL, Keller MP, Attie AD, van Pijkeren JP. Exploiting Prophage-Mediated Lysis for Biotherapeutic Release by Lactobacillus reuteri. Appl Environ Microbiol 2019; 85:e02335-18. [PMID: 30683744 PMCID: PMC6498169 DOI: 10.1128/aem.02335-18] [Citation(s) in RCA: 17] [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: 10/03/2018] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Abstract
Lactobacillus reuteri has the potential to be developed as a microbial therapeutic delivery platform because of an established safety profile, health-promoting properties, and available genome editing tools. Here, we show that L. reuteri VPL1014 exhibits a low mutation rate compared to other Gram-positive bacteria, which we expect will contribute to the stability of genetically modified strains. VPL1014 encodes two biologically active prophages, which are induced during gastrointestinal transit. We hypothesized that intracellularly accumulated recombinant protein can be released following bacteriophage-mediated lysis. To test this, we engineered VPL1014 to accumulate leptin, our model protein, inside the cell. In vitro prophage induction of recombinant VPL1014 released leptin into the extracellular milieu, which corresponded to bacteriophage production. We also employed a plasmid system that does not require antibiotic in the growth medium for plasmid maintenance. Collectively, these data provide new avenues to exploit native prophages to deliver therapeutic molecules.IMPORTANCE Lactic acid bacteria (LAB) have been explored as potential biotherapeutic vehicles for the past 20 years. To secrete a therapeutic in the extracellular milieu, one typically relies on the bacterial secretion pathway, i.e., the Sec pathway. Overexpression of a secreted protein can overload the secretory pathway and impact the organism's fitness, and optimization of the signal peptide is also required to maximize the efficiency of the release of mature protein. Here, we describe a previously unexplored approach to release therapeutics from the probiotic Lactobacillus reuteri We demonstrate that an intracellularly accumulated recombinant protein is released following prophage activation. Since we recently demonstrated that prophages are activated during gastrointestinal transit, we propose that this method will provide a straightforward and efficient approach to deliver therapeutics in vivo.
Collapse
Affiliation(s)
- Laura M Alexander
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Donald S Stapleton
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Kathryn L Schueler
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | | |
Collapse
|
44
|
Gut Symbionts Lactobacillus reuteri R2lc and 2010 Encode a Polyketide Synthase Cluster That Activates the Mammalian Aryl Hydrocarbon Receptor. Appl Environ Microbiol 2019; 85:AEM.01661-18. [PMID: 30389766 PMCID: PMC6498181 DOI: 10.1128/aem.01661-18] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 10/24/2018] [Indexed: 12/20/2022] Open
Abstract
Temporary changes in the composition of the microbiota, for example, by oral administration of probiotics, can modulate the host immune system. However, the underlying mechanisms by which probiotics interact with the host are often unknown. Here, we show that Lactobacillus reuteri R2lc and 2010 harbor an orthologous PKS gene cluster that activates the aryl hydrocarbon receptor (AhR). AhR is a ligand-activated transcription factor that plays a key role in a variety of diseases, including amelioration of intestinal inflammation. Understanding the mechanism by which a bacterium modulates the immune system is critical for applying rational selection strategies for probiotic supplementation. Finally, heterologous and/or optimized expression of PKS is a logical next step toward the development of next-generation probiotics to prevent and treat disease. A mechanistic understanding of microbe-host interactions is critical to developing therapeutic strategies for targeted modulation of the host immune system. Different members of the gut symbiont species Lactobacillus reuteri modulate host health by, for example, reduction of intestinal inflammation. Previously, it was shown that L. reuteri activates the aryl hydrocarbon receptor (AhR), a ligand-activated transcription factor that plays an important role in the mucosal immune system, by the production of tryptophan catabolites. Here, we identified a novel pathway by which L. reuteri activates AhR, which is independent of tryptophan metabolism. We screened a library of 36 L. reuteri strains and determined that R2lc and 2010, strains with a pigmented phenotype, are potent AhR activators. By whole-genome sequencing and comparative genomics, we identified genes unique to R2lc and 2010. Our analyses demonstrated that R2lc harbors two genetically distinct polyketide synthase (PKS) clusters, functionally unknown (fun) and pks, each carried by a multicopy plasmid. Inactivation of pks, but not fun, abolished the ability of R2lc to activate AhR. L. reuteri 2010 has a gene cluster homologous to the pks cluster in R2lc with an identical gene organization, which is also responsible for AhR activation. In conclusion, we identified a novel PKS pathway in L. reuteri R2lc and 2010 that is responsible for AhR activation. IMPORTANCE Temporary changes in the composition of the microbiota, for example, by oral administration of probiotics, can modulate the host immune system. However, the underlying mechanisms by which probiotics interact with the host are often unknown. Here, we show that Lactobacillus reuteri R2lc and 2010 harbor an orthologous PKS gene cluster that activates the aryl hydrocarbon receptor (AhR). AhR is a ligand-activated transcription factor that plays a key role in a variety of diseases, including amelioration of intestinal inflammation. Understanding the mechanism by which a bacterium modulates the immune system is critical for applying rational selection strategies for probiotic supplementation. Finally, heterologous and/or optimized expression of PKS is a logical next step toward the development of next-generation probiotics to prevent and treat disease.
Collapse
|
45
|
In Vitro Probiotic Potential of Lactic Acid Bacteria Isolated from Aguamiel and Pulque and Antibacterial Activity Against Pathogens. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9030601] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Probiotics can act as a natural barrier against several pathogens, such Helicobacter pylori, a bacterium linked to stomach cancer. The aim of the present study was to isolate and identify lactic acid bacteria (LAB) from pulque and aguamiel, and evaluate their probiotic potential and antimicrobial effect on Escherichia coli, Staphylococcus aureus, and Helicobacter pylori. Ten isolates were selected and evaluated for in vitro resistance to antibiotics and gastrointestinal conditions, and antimicrobial activity against E. coli and S. aureus and the effect on H. pylori strains. 16S rRNA identification was performed. Ten potential probiotic isolates were confirmed as belonging to the genera Lactobacillus and Pediococcus. All the strains were susceptible to clinical antibiotics, except to vancomycin. Sixty percent of the isolates exhibited antimicrobial activity against E. coli and S. aureus. The growth of H. pylori ATCC 43504 was suppressed by all the LAB, and the urease activity from all the H. pylori strains was inhibited, which may decrease its chances for survival in the stomach. The results suggest that LAB isolated from pulque and aguamiel could be an option to establish a harmless relationship between the host and H. pylori, helping in their eradication therapy.
Collapse
|
46
|
Oh JH, Alexander LM, Pan M, Schueler KL, Keller MP, Attie AD, Walter J, van Pijkeren JP. Dietary Fructose and Microbiota-Derived Short-Chain Fatty Acids Promote Bacteriophage Production in the Gut Symbiont Lactobacillus reuteri. Cell Host Microbe 2019; 25:273-284.e6. [PMID: 30658906 DOI: 10.1016/j.chom.2018.11.016] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Revised: 09/10/2018] [Accepted: 11/28/2018] [Indexed: 01/14/2023]
Abstract
The mammalian intestinal tract contains a complex microbial ecosystem with many lysogens, which are bacteria containing dormant phages (prophages) inserted within their genomes. Approximately half of intestinal viruses are derived from lysogens, suggesting that these bacteria encounter triggers that promote phage production. We show that prophages of the gut symbiont Lactobacillus reuteri are activated during gastrointestinal transit and that phage production is further increased in response to a fructose-enriched diet. Fructose and exposure to short-chain fatty acids activate the Ack pathway, involved in generating acetic acid, which in turn triggers the bacterial stress response that promotes phage production. L. reuteri mutants of the Ack pathway or RecA, a stress response component, exhibit decreased phage production. Thus, prophages in a gut symbiont can be induced by diet and metabolites affected by diet, which provides a potential mechanistic explanation for the effects of diet on the intestinal phage community.
Collapse
Affiliation(s)
- Jee-Hwan Oh
- Department of Food Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Laura M Alexander
- Department of Food Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Meichen Pan
- Department of Food Science, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Kathryn L Schueler
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Mark P Keller
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Alan D Attie
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Jens Walter
- Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada; Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2P5, Canada
| | | |
Collapse
|
47
|
Salvetti E, Harris HMB, Felis GE, O'Toole PW. Comparative Genomics of the Genus Lactobacillus Reveals Robust Phylogroups That Provide the Basis for Reclassification. Appl Environ Microbiol 2018; 84:e00993-18. [PMID: 29915113 PMCID: PMC6102987 DOI: 10.1128/aem.00993-18] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2018] [Accepted: 06/12/2018] [Indexed: 12/12/2022] Open
Abstract
The genus Lactobacillus includes over 200 species that are widely used in fermented food preservation and biotechnology or that are explored for beneficial effects on health. Naming, classifying, and comparing lactobacilli have been challenging due to the high level of phenotypic and genotypic diversity that they display and because of the uncertain degree of relatedness between them and associated genera. The aim of this study was to investigate the feasibility of dividing the genus Lactobacillus into more homogeneous genera/clusters, exploiting genome-based data. The relatedness of 269 species belonging primarily to the families Lactobacillaceae and Leuconostocaceae was investigated through phylogenetic analysis (by the use of ribosomal proteins and housekeeping genes) and the assessment of the average amino acid identity (AAI) and the percentage of conserved proteins (POCP). For each subgeneric group that emerged, conserved signature genes were identified. Both distance-based and sequence-based metrics showed that the Lactobacillus genus was paraphyletic and revealed the presence of 10 methodologically consistent subclades, which were also characterized by a distinct distribution of conserved signature orthologues. We present two ways to reclassify lactobacilli: a conservative division into two subgeneric groups based on the presence/absence of a key carbohydrate utilization gene or a more radical subdivision into 10 groups that satisfy more stringent criteria for genomic relatedness.IMPORTANCE Lactobacilli have significant scientific and economic value, but their extraordinary diversity means that they are not robustly classified. The 10 homogeneous genera/subgeneric entities that we identify here are characterized by uniform patterns of the presence/absence of specific sets of genes which offer potential as discovery tools for understanding differential biological features. Reclassification/subdivision of the genus Lactobacillus into more uniform taxonomic nuclei will also provide accurate molecular markers that will be enabling for regulatory approval applications. Reclassification will facilitate scientific communication related to lactobacilli and prevent misidentification issues, which are still the major cause of mislabeling of probiotic and food products reported worldwide.
Collapse
Affiliation(s)
- Elisa Salvetti
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Hugh M B Harris
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| | - Giovanna E Felis
- Department of Biotechnology, University of Verona, Verona, Italy
| | - Paul W O'Toole
- School of Microbiology, University College Cork, Cork, Ireland
- APC Microbiome Institute, University College Cork, Cork, Ireland
| |
Collapse
|