1
|
Barrios Steed D, Koundakjian D, Harris AD, Rosato AE, Konstantinidis KT, Woodworth MH. Leveraging strain competition to address antimicrobial resistance with microbiota therapies. Gut Microbes 2025; 17:2488046. [PMID: 40195644 PMCID: PMC11988218 DOI: 10.1080/19490976.2025.2488046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Revised: 12/28/2024] [Accepted: 03/28/2025] [Indexed: 04/09/2025] Open
Abstract
The enteric microbiota is an established reservoir for multidrug-resistant organisms that present urgent clinical and public health threats. Observational data and small interventional studies suggest that microbiome interventions, such as fecal microbiota products and characterized live biotherapeutic bacterial strains, could be an effective antibiotic-sparing prevention approach to address these threats. However, bacterial colonization is a complex ecological phenomenon that remains understudied in the context of the human gut. Antibiotic resistance is one among many adaptative strategies that impact long-term colonization. Here we review and synthesize evidence of how bacterial competition and differential fitness in the context of the gut present opportunities to improve mechanistic understanding of colonization resistance, therapeutic development, patient care, and ultimately public health.
Collapse
Affiliation(s)
- Danielle Barrios Steed
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| | | | - Anthony D. Harris
- Department of Epidemiology & Public Health, University of Maryland School of Medicine, Baltimore, MD, USA
- Institute for Healthcare Computing, University of Maryland, Baltimore, MD, USA
| | - Adriana E Rosato
- Center for Molecular Medicine, MaineHealth Institute for Research, Scarborough, ME, USA
| | | | - Michael H Woodworth
- Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
2
|
Fields BD, Pascal DG, Rando OK, Huddleston ME, Ingram K, Hopton R, Grogg MW, Nelson MT, Voigt CA. Design of a Continuous GAA-Producing Probiotic as a Potential Mitigator of the Effects of Sleep Deprivation. ACS Synth Biol 2025. [PMID: 40378286 DOI: 10.1021/acssynbio.4c00690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2025]
Abstract
Creatine is a popular athletic supplement that has also been shown to improve cognitive performance upon sleep deprivation. However, it is rapidly cleared from the gastrointestinal tract a few hours after consumption. Toward providing a persistent creatine dose, we engineered the human probiotic Escherichia coli Nissle (EcN) to produce guanidinoacetic acid (GAA), which is converted to creatine in the liver. We find GAA-producing enzymes present in the human microbiome and compare their activities to known enzymes. Three copies of arginine:glycine amidinotransferase (AGAT) from Actinokineospora terrae are expressed from the genome, and native gcvP, argR, and argA are edited or deleted to improve substrate availability without negatively impacting cell viability. A standard EcN dose (1012 cells) produces 41 ± 7 mg GAA per hour under laboratory conditions. This work demonstrates that a probiotic bacterium can be engineered to produce sustained GAA titers known to impact cognitive performance.
Collapse
Affiliation(s)
- Brandon D Fields
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Daniel G Pascal
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Olivia K Rando
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| | - Mary E Huddleston
- Blue Halo Inc., 4401 Dayton-Xenia Rd, Dayton, Ohio 45432, United States
| | - Katherine Ingram
- Blue Halo Inc., 4401 Dayton-Xenia Rd, Dayton, Ohio 45432, United States
| | - Rachel Hopton
- Blue Halo Inc., 4401 Dayton-Xenia Rd, Dayton, Ohio 45432, United States
| | - Matthew W Grogg
- United States Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| | - M Tyler Nelson
- United States Air Force Research Laboratory, 711th Human Performance Wing, Wright-Patterson AFB, Ohio 45433, United States
| | - Christopher A Voigt
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
3
|
Chemla Y, Sweeney CJ, Wozniak CA, Voigt CA. Design and regulation of engineered bacteria for environmental release. Nat Microbiol 2025; 10:281-300. [PMID: 39905169 DOI: 10.1038/s41564-024-01918-0] [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: 09/22/2023] [Accepted: 12/04/2024] [Indexed: 02/06/2025]
Abstract
Emerging products of biotechnology involve the release of living genetically modified microbes (GMMs) into the environment. However, regulatory challenges limit their use. So far, GMMs have mainly been tested in agriculture and environmental cleanup, with few approved for commercial purposes. Current government regulations do not sufficiently address modern genetic engineering and limit the potential of new applications, including living therapeutics, engineered living materials, self-healing infrastructure, anticorrosion coatings and consumer products. Here, based on 47 global studies on soil-released GMMs and laboratory microcosm experiments, we discuss the environmental behaviour of released bacteria and offer engineering strategies to help improve performance, control persistence and reduce risk. Furthermore, advanced technologies that improve GMM function and control, but lead to increases in regulatory scrutiny, are reviewed. Finally, we propose a new regulatory framework informed by recent data to maximize the benefits of GMMs and address risks.
Collapse
Affiliation(s)
- Yonatan Chemla
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Connor J Sweeney
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Christopher A Voigt
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
4
|
Singh A, Mazumder A, Das S, Kanda A, Tyagi PK, Chaitanya MVNL. Harnessing the Power of Probiotics: Boosting Immunity and Safeguarding against Various Diseases and Infections. RECENT ADVANCES IN ANTI-INFECTIVE DRUG DISCOVERY 2025; 20:5-29. [PMID: 40302548 DOI: 10.2174/0127724344308638240530065552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 04/20/2024] [Accepted: 04/25/2024] [Indexed: 05/02/2025]
Abstract
The human microbiome, a diverse microorganism community, crucially defends against pathogens. Probiotics, postbiotics, and paraprobiotics alone and in combination are potent in countering fungal and waterborne infections, particularly against viral threats. This review focuses on the mechanisms of the microbiome against viral infections, emphasizing probiotic interventions. Certain Lactic Acid Bacteria (LAB) strains effectively eliminate toxic aflatoxin B1 (AFB1) from microfungi-produced mycotoxins. LAB binding to AFB1 persists post-gastric digestion, and pre-incubation with mycotoxins reduces probiotic adhesion to mucus. Oral probiotic administration in animals increases mycotoxin excretion, reducing associated health risks. Bifidobacterium longum and Lactobacillus rhamnosus show exceptional efficacy in removing cyanobacterial toxin microcystin-LR from drinking water. Engineered probiotics promise advanced therapeutic applications for metabolic disorders, Alzheimer's, and type 1 diabetes, serving as diagnostic tools for detecting pathogens and inflammation markers. In antimicrobial peptide production, genetically modified probiotics producing human β-defensin 2 (HBD2) treat Crohn's disease with implemented biocontainment strategies preventing unintended environmental impacts.
Collapse
Affiliation(s)
- Archna Singh
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Greater Noida, 201306, U.P., India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Greater Noida, 201306, U.P., India
| | - Saumya Das
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Greater Noida, 201306, U.P., India
| | - Anmol Kanda
- Noida Institute of Engineering and Technology (Pharmacy Institute), 19 Knowledge Park-II, Greater Noida, 201306, U.P., India
| | - Pankaj Kumar Tyagi
- Department of Biotechnology, Noida Institute of Engineering and Technology, 19 Knowledge Park-II, Greater Noida, 201306, U.P., India
| | - M V N L Chaitanya
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, 144001, Punjab, India
| |
Collapse
|
5
|
Kim SY, Randall JR, Gu R, Nguyen QD, Davies BW. Antibacterial action, proteolytic immunity, and in vivo activity of a Vibrio cholerae microcin. Cell Host Microbe 2024; 32:1959-1971.e6. [PMID: 39260372 PMCID: PMC11563924 DOI: 10.1016/j.chom.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Revised: 06/24/2024] [Accepted: 08/16/2024] [Indexed: 09/13/2024]
Abstract
Microcins are small antibacterial proteins that mediate interbacterial competition. Their narrow-spectrum activity provides opportunities to discover microbiome-sparing treatments. However, microcins have been found almost exclusively in Enterobacteriaceae. Their broader existence and potential implications in other pathogens remain unclear. Here, we identify and characterize a microcin active against pathogenic Vibrio cholerae: MvcC. We show that MvcC is reliant on the outer membrane porin OmpT to cross the outer membrane. MvcC then binds the periplasmic protein OppA to reach and disrupt the cytoplasmic membrane. We demonstrate that MvcC's cognate immunity protein is a protease, which precisely cleaves MvcC to neutralize its activity. Importantly, we show that MvcC is active against diverse cholera isolates and in a mouse model of V. cholerae colonization. Our results provide a detailed analysis of a microcin outside of Enterobacteriaceae and its potential to influence V. cholerae infection.
Collapse
Affiliation(s)
- Sun-Young Kim
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Justin R Randall
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Richard Gu
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Quoc D Nguyen
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA
| | - Bryan W Davies
- Department of Molecular Biosciences, The University of Texas at Austin, Austin, TX, USA; John Ring LaMontagne Center for Infectious Diseases, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
6
|
Gajendran VP, Rajamani S. Recent Advancements in Harnessing Lactic Acid Bacterial Metabolites for Fruits and Vegetables Preservation. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10392-3. [PMID: 39514163 DOI: 10.1007/s12602-024-10392-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Postharvest losses in fruits and vegetables exert substantial economic and environmental repercussions. Chemical interventions are being widely utilized for the past six decades which may lead to significant health complications. Bioprotection of fruits and vegetables is the need of the hour in which use of lactic acid bacteria (LAB) with GRAS status predominantly stands out. Incorporation of LAB in postharvest fruits and vegetables suppresses the growth of spoilage organisms by synthesizing various antimicrobial compounds such as bacteriocins, organic acids, hydrogen peroxide (H2O2), exopolysaccharides (EPS), and BLIS. For example, Pediococcus acidilactici, Lactobacillus plantarum, and Limosilactobacillus fermentum convert natural sugars in fruits and vegetables to lactic acid and create an acidic environment that do not favour spoilage organisms. LAB can improve the bioavailability of vitamins and minerals and enrich the phenolic profile and bioactivity components. LAB has remarkable physiological characteristics like resistance towards bacteriophage, proteolytic activity, and polysaccharide production which adds to the safety of foods. They modify the sensory properties and preserve the nutritional quality of fruits and vegetables. They can also perform therapeutic role in the intestinal tract as they tolerate low pH, high salt concentration. Thus application of LAB, whether independently or in conjunction with stabilizing agents as edible coatings, is regarded as an exceptionally promising methodology for ensuring safer consumption of fruits and vegetables. This review addresses the most recent research findings that harness the antagonistic property of lactic acid bacterial metabolites, formulations and coatings containing their bioactive compounds for extended shelf life of fruits and vegetables.
Collapse
Affiliation(s)
- Vaishnavi Pratha Gajendran
- Department of Agricultural Microbiology, Tamil Nadu Agricultural University, Coimbatore, 641003, Tamil Nadu, India
| | - Subhashini Rajamani
- Department of Agricultural Microbiology, Tamil Nadu Agricultural University, Coimbatore, 641003, Tamil Nadu, India.
| |
Collapse
|
7
|
Srivastava R, Lesser CF. Living Engineered Bacterial Therapeutics: Emerging Affordable Precision Interventions. Microb Biotechnol 2024; 17:e70057. [PMID: 39579048 PMCID: PMC11584976 DOI: 10.1111/1751-7915.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/24/2024] [Accepted: 11/07/2024] [Indexed: 11/25/2024] Open
Abstract
Live biotherapeutic products (LBPs), including engineered bacteria, are rapidly emerging as potential therapeutic interventions. These innovative therapies can serve as live in situ drug delivery platforms for the direct deposition of therapeutic payloads, including complex biologics, at sites of disease. This approach offers a platform likely to enhance therapeutic efficacy and decrease off-target side effects. LBPs also can likely be distributed at a relatively low price point, as their production can be economically scaled up. LBPs represent an exciting new means for ensuring healthy lives and promoting well-being for all ages, aligning with the World Health Organization's sustainable development goal 3.
Collapse
Affiliation(s)
- Rajkamal Srivastava
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Department of MicrobiologyBlavatnik Institute, Harvard Medical SchoolBostonMassachusettsUSA
| | - Cammie F. Lesser
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of MedicineMassachusetts General HospitalBostonMassachusettsUSA
- Department of MicrobiologyBlavatnik Institute, Harvard Medical SchoolBostonMassachusettsUSA
- Broad Institute of MIT and HarvardCambridgeMassachusettsUSA
- Ragon Institute of Harvard and MITCambridgeMassachusettsUSA
| |
Collapse
|
8
|
Kim TH, Cho BK, Lee DH. Synthetic Biology-Driven Microbial Therapeutics for Disease Treatment. J Microbiol Biotechnol 2024; 34:1947-1958. [PMID: 39233526 PMCID: PMC11540606 DOI: 10.4014/jmb.2407.07004] [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: 07/03/2024] [Revised: 08/05/2024] [Accepted: 08/06/2024] [Indexed: 09/06/2024]
Abstract
The human microbiome, consisting of microorganisms that coexist symbiotically with the body, impacts health from birth. Alterations in gut microbiota driven by factors such as diet and medication can contribute to diseases beyond the gut. Synthetic biology has paved the way for engineered microbial therapeutics, presenting promising treatments for a variety of conditions. Using genetically encoded biosensors and dynamic regulatory tools, engineered microbes can produce and deliver therapeutic agents, detect biomarkers, and manage diseases. This review organizes engineered microbial therapeutics by disease type, emphasizing innovative strategies and recent advancements. The scope of diseases includes gastrointestinal disorders, cancers, metabolic diseases, infections, and other ailments. Synthetic biology facilitates precise targeting and regulation, improving the efficacy and safety of these therapies. With promising results in animal models, engineered microbial therapeutics provide a novel alternative to traditional treatments, heralding a transformative era in diagnostics and treatment for numerous diseases.
Collapse
Affiliation(s)
- Tae Hyun Kim
- Synthetic Biology Research Center and the K-Biofoundry, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Byung Kwan Cho
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Republic of Korea
- KAIST Institutes for the BioCentury, KAIST, Daejeon 34141, Republic of Korea
- Graduate School of Engineering Biology, KAIST, Daejeon 34141, Republic of Korea
| | - Dae-Hee Lee
- Synthetic Biology Research Center and the K-Biofoundry, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
- Department of Biosystems and Bioengineering, KRIBB School of Biotechnology, University of Science and Technology (UST), Daejeon 34113, Republic of Korea
- Graduate School of Engineering Biology, KAIST, Daejeon 34141, Republic of Korea
- Department of Integrative Biotechnology, College of Biotechnology and Bioengineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
9
|
Singh A, Mazumder A, Das S, Tyagi PK, Chaitanya MVNL. Probiotics in Action: Enhancing Immunity and Combatting Diseases for Optimal Health. JOURNAL OF NATURAL REMEDIES 2024:1153-1167. [DOI: 10.18311/jnr/2024/35894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Accepted: 04/22/2024] [Indexed: 01/03/2025]
Abstract
This review offers an in-depth examination of the mechanisms underlying the microbiome's defense against viral infections, with a specific focus on probiotic interventions. Mycotoxins, secondary compounds produced by microfungi, pose significant health risks. Yet, certain strains of Lactic Acid Bacteria (LAB) have exhibited remarkable efficacy in eliminating aflatoxin B1 (AFB1), the most toxic member of the aflatoxin family. Experimental setups demonstrated AFB1 binding to specific LAB strains, persisting even after gastric digestion. Laboratory studies revealed a potential protective mechanism wherein pre-incubation of probiotics with mycotoxins reduced their adhesion to mucus. Animal trials further underscored the benefits of oral probiotic administration, showcasing increased fecal excretion of mycotoxins and mitigation of associated health risks. Cyanobacteria-generated microcystins in drinking water pose a significant threat to human health. Probiotic bacteria, particularly strains like Bifidobacterium longum and Lactobacillus rhamnosus, have demonstrated exceptional efficacy in removing the cyanobacterial peptide toxin microcystin-LR. Optimized conditions resulted in rapid toxin elimination, highlighting the potential of probiotics in water purification. Engineered probiotics represent a cutting-edge approach to tailor microorganisms for specific therapeutic applications, exhibiting promise in treating metabolic disorders, Alzheimer's disease, and type 1 diabetes. Additionally, they serve as innovative diagnostic tools, capable of detecting pathogens and inflammation markers within the body. In the realm of antimicrobial peptide production, probiotics offer a promising platform, with genetically modified strains engineered to produce human β-defensin 2 (HBD2) for treating Crohn's disease, showcasing their potential in targeted theurapetic delivery. Biocontainment strategies have been implemented to prevent unintended environmental impacts.
Collapse
|
10
|
Baquero F, Beis K, Craik DJ, Li Y, Link AJ, Rebuffat S, Salomón R, Severinov K, Zirah S, Hegemann JD. The pearl jubilee of microcin J25: thirty years of research on an exceptional lasso peptide. Nat Prod Rep 2024; 41:469-511. [PMID: 38164764 DOI: 10.1039/d3np00046j] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2024]
Abstract
Covering: 1992 up to 2023Since their discovery, lasso peptides went from peculiarities to be recognized as a major family of ribosomally synthesized and post-translationally modified peptide (RiPP) natural products that were shown to be spread throughout the bacterial kingdom. Microcin J25 was first described in 1992, making it one of the earliest known lasso peptides. No other lasso peptide has since then been studied to such an extent as microcin J25, yet, previous review articles merely skimmed over all the research done on this exceptional lasso peptide. Therefore, to commemorate the 30th anniversary of its first report, we give a comprehensive overview of all literature related to microcin J25. This review article spans the early work towards the discovery of microcin J25, its biosynthetic gene cluster, and the elucidation of its three-dimensional, threaded lasso structure. Furthermore, the current knowledge about the biosynthesis of microcin J25 and lasso peptides in general is summarized and a detailed overview is given on the biological activities associated with microcin J25, including means of self-immunity, uptake into target bacteria, inhibition of the Gram-negative RNA polymerase, and the effects of microcin J25 on mitochondria. The in vitro and in vivo models used to study the potential utility of microcin J25 in a (veterinary) medicine context are discussed and the efforts that went into employing the microcin J25 scaffold in bioengineering contexts are summed up.
Collapse
Affiliation(s)
- Fernando Baquero
- Department of Microbiology, Ramón y Cajal University Hospital and Ramón y Cajal Institute for Health Research (IRYCIS), Madrid, Spain
- Network Center for Research in Epidemiology and Public Health (CIBER-ESP), Madrid, Spain
| | - Konstantinos Beis
- Department of Life Sciences, Imperial College London, London, SW7 2AZ, UK
- Rutherford Appleton Laboratory, Research Complex at Harwell, Didcot, Oxfordshire OX11 0FA, UK
| | - David J Craik
- Institute for Molecular Bioscience, Australian Research Council Centre of Excellence for Innovations in Peptide and Protein Science, The University of Queensland, 4072 Brisbane, Queensland, Australia
| | - Yanyan Li
- Laboratoire Molécules de Communication et Adaptation des Microorganismes (MCAM), UMR 7245, Muséum National d'Histoire Naturelle (MNHN), Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - A James Link
- Departments of Chemical and Biological Engineering, Chemistry, and Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Sylvie Rebuffat
- Laboratoire Molécules de Communication et Adaptation des Microorganismes (MCAM), UMR 7245, Muséum National d'Histoire Naturelle (MNHN), Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Raúl Salomón
- Instituto de Química Biológica "Dr Bernabé Bloj", Facultad de Bioquímica, Química y Farmacia, Instituto Superior de Investigaciones Biológicas (INSIBIO), CONICET-UNT, San Miguel de Tucumán, Argentina
| | - Konstantin Severinov
- Waksman Institute for Microbiology, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Séverine Zirah
- Laboratoire Molécules de Communication et Adaptation des Microorganismes (MCAM), UMR 7245, Muséum National d'Histoire Naturelle (MNHN), Centre National de la Recherche Scientifique (CNRS), Paris, France
| | - Julian D Hegemann
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS), Helmholtz Centre for Infection Research (HZI), Saarland University Campus, 66123 Saarbrücken, Germany.
- Department of Pharmacy, Campus E8 1, Saarland University, 66123 Saarbrücken, Germany
| |
Collapse
|
11
|
Mishra B, Mishra AK, Mohanta YK, Yadavalli R, Agrawal DC, Reddy HP, Gorrepati R, Reddy CN, Mandal SK, Shamim MZ, Panda J. Postbiotics: the new horizons of microbial functional bioactive compounds in food preservation and security. FOOD PRODUCTION, PROCESSING AND NUTRITION 2024; 6:28. [DOI: 10.1186/s43014-023-00200-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 11/05/2023] [Indexed: 01/05/2025]
Abstract
AbstractIn recent decades, consumers, manufacturers, and researchers have been more interested in functional foods, which include probiotics, prebiotics, and postbiotics. Probiotics are live microbes that, when regulated in enough quantities, provide health benefits on the host, while the prebiotics are substrates that host microorganisms selectively use. Postbiotics are metabolites and cell-wall components that are beneficial to the host and are released by living bacteria or after lysis. Postbiotic dietary supplements are more stable than probiotics and prebiotics. Many bioactivities of postbiotics are unknown or poorly understood. Hence, this study aims to present a synopsis of the regular elements and new developments of the postbiotics including health-promoting effects, production, conceptualization of terms, bioactivities, and applications in the field of food safety and preservation. Postbiotics aid in bio preservation and the reduction of biofilm development in food due to their organic acids, bacteriocins, and other antibacterial activities. The present study examines the production of postbiotic metabolites in situ in food and the effects of external and internal food components. The antimicrobial roles, removal of biofilms, and its applications in preservation and food safety have also been discussed. This paper also explored the various aspects like manipulation of postbiotic composition in the food system and its safety measures.
Graphical Abstract
Collapse
|
12
|
Dong S, Li L, Hao F, Fang Z, Zhong R, Wu J, Fang X. Improving quality of poultry and its meat products with probiotics, prebiotics, and phytoextracts. Poult Sci 2024; 103:103287. [PMID: 38104412 PMCID: PMC10966786 DOI: 10.1016/j.psj.2023.103287] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 11/08/2023] [Accepted: 11/13/2023] [Indexed: 12/19/2023] Open
Abstract
Remarkable changes have occurred in poultry farming and meat processing in recent years, driven by advancements in breeding technology, feed processing technology, farming conditions, and management practices. The incorporation of probiotics, prebiotics, and phytoextracts has made significant contributions to the development of poultry meat products that promote both health and functionality throughout the growth phase and during meat processing. Poultry fed with these substances improve meat quality, while incorporating probiotics, prebiotics, and phytoextracts in poultry processing, as additives or supplements, inhibits pathogens and offers health benefits to consumers. However, it is vital to assess the safety of functional fermented meat products containing these compounds and their potential effects on consumer health. Currently, there's still uncertainty in these aspects. Additionally, research on utilizing next-generation probiotic strains and synergistic combinations of probiotics and prebiotics in poultry meat products is in its early stages. Therefore, further investigation is required to gain a comprehensive understanding of the beneficial effects and safety considerations of these substances in poultry meat products in the future. This review offered a comprehensive overview of the applications of probiotics and prebiotics in poultry farming, focusing on their effects on nutrient utilization, growth efficiency, and gut health. Furthermore, potential of probiotics, prebiotics, and phytoextracts in enhancing poultry meat production was explored for improved health benefits and functionality, and possible issues associated with the use of these substances were discussed. Moreover, the conclusions drawn from this review and potential future perspectives in this field are presented.
Collapse
Affiliation(s)
- Sashuang Dong
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510630, PR China; Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan 512000, PR China
| | - Lanyin Li
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510630, PR China
| | - Fanyu Hao
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510630, PR China
| | - Ziying Fang
- Weiran Food Biotechnology (Shenzhen) Co., Ltd., Shenzhen 518000, PR China
| | - Ruimin Zhong
- Guangdong Provincial Key Laboratory of Utilization and Conservation of Food and Medicinal Resources in Northern Region, Shaoguan University, Shaoguan 512000, PR China
| | - Jianfeng Wu
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510630, PR China.
| | - Xiang Fang
- College of Food Science, South China Agricultural University, Guangzhou, Guangdong 510630, PR China.
| |
Collapse
|
13
|
Lamichhane B, Mawad AMM, Saleh M, Kelley WG, Harrington PJ, Lovestad CW, Amezcua J, Sarhan MM, El Zowalaty ME, Ramadan H, Morgan M, Helmy YA. Salmonellosis: An Overview of Epidemiology, Pathogenesis, and Innovative Approaches to Mitigate the Antimicrobial Resistant Infections. Antibiotics (Basel) 2024; 13:76. [PMID: 38247636 PMCID: PMC10812683 DOI: 10.3390/antibiotics13010076] [Citation(s) in RCA: 54] [Impact Index Per Article: 54.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 12/24/2023] [Accepted: 01/10/2024] [Indexed: 01/23/2024] Open
Abstract
Salmonella is a major foodborne pathogen and a leading cause of gastroenteritis in humans and animals. Salmonella is highly pathogenic and encompasses more than 2600 characterized serovars. The transmission of Salmonella to humans occurs through the farm-to-fork continuum and is commonly linked to the consumption of animal-derived food products. Among these sources, poultry and poultry products are primary contributors, followed by beef, pork, fish, and non-animal-derived food such as fruits and vegetables. While antibiotics constitute the primary treatment for salmonellosis, the emergence of antibiotic resistance and the rise of multidrug-resistant (MDR) Salmonella strains have highlighted the urgency of developing antibiotic alternatives. Effective infection management necessitates a comprehensive understanding of the pathogen's epidemiology and transmission dynamics. Therefore, this comprehensive review focuses on the epidemiology, sources of infection, risk factors, transmission dynamics, and the host range of Salmonella serotypes. This review also investigates the disease characteristics observed in both humans and animals, antibiotic resistance, pathogenesis, and potential strategies for treatment and control of salmonellosis, emphasizing the most recent antibiotic-alternative approaches for infection control.
Collapse
Affiliation(s)
- Bibek Lamichhane
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Asmaa M. M. Mawad
- Botany and Microbiology Department, Faculty of Science, Assiut University, Assiut 71516, Egypt
| | - Mohamed Saleh
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - William G. Kelley
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Patrick J. Harrington
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Cayenne W. Lovestad
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Jessica Amezcua
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Mohamed M. Sarhan
- Faculty of Pharmacy, King Salman International University (KSIU), Ras Sudr 8744304, Egypt
| | - Mohamed E. El Zowalaty
- Veterinary Medicine and Food Security Research Group, Medical Laboratory Sciences Program, Faculty of Health Sciences, Abu Dhabi Women’s Campus, Higher Colleges of Technology, Abu Dhabi 41012, United Arab Emirates
| | - Hazem Ramadan
- Hygiene and Zoonoses Department, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Melissa Morgan
- Department of Animal and Food Sciences, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| | - Yosra A. Helmy
- Department of Veterinary Science, Martin-Gatton College of Agriculture, Food and Environment, University of Kentucky, Lexington, KY 40546, USA
| |
Collapse
|
14
|
Lebovich M, Lora MA, Gracia-David J, Andrews LB. Genetic Circuits for Feedback Control of Gamma-Aminobutyric Acid Biosynthesis in Probiotic Escherichia coli Nissle 1917. Metabolites 2024; 14:44. [PMID: 38248847 PMCID: PMC10819706 DOI: 10.3390/metabo14010044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/23/2024] Open
Abstract
Engineered microorganisms such as the probiotic strain Escherichia coli Nissle 1917 (EcN) offer a strategy to sense and modulate the concentration of metabolites or therapeutics in the gastrointestinal tract. Here, we present an approach to regulate the production of the depression-associated metabolite gamma-aminobutyric acid (GABA) in EcN using genetic circuits that implement negative feedback. We engineered EcN to produce GABA by overexpressing glutamate decarboxylase and applied an intracellular GABA biosensor to identify growth conditions that improve GABA biosynthesis. We next employed characterized genetically encoded NOT gates to construct genetic circuits with layered feedback to control the rate of GABA biosynthesis and the concentration of GABA produced. Looking ahead, this approach may be utilized to design feedback control of microbial metabolite biosynthesis to achieve designable smart microbes that act as living therapeutics.
Collapse
Affiliation(s)
- Matthew Lebovich
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Biotechnology Training Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Marcos A. Lora
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
| | - Jared Gracia-David
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Department of Biology, Amherst College, Amherst, MA 01002, USA
| | - Lauren B. Andrews
- Department of Chemical Engineering, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Biotechnology Training Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts Amherst, Amherst, MA 01003, USA
| |
Collapse
|
15
|
Hu M, Zhang T, Miao M, Li K, Luan Q, Sun G. Expectations for employing Escherichia coli Nissle 1917 in food science and nutrition. Crit Rev Food Sci Nutr 2024; 65:1802-1810. [PMID: 38189668 DOI: 10.1080/10408398.2023.2301416] [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: 01/09/2024]
Abstract
As a promising probiotic strain, Escherichia coli Nissle 1917 (EcN) has been demonstrated to confer beneficial effects on intestinal health, immune function, and pathogen prevention. Additionally, EcN has also been widely studied due to its clear genomic information, tractable gene regulation, and simple growth conditions. This review summarizes the various applications potential of EcN in food science and nutrition, including inflammation prevention, tumor-targeting therapy, antibacterial agents for food, and nutrient production with a focus on specific case studies. Moreover, we highlight the major challenges of employing EcN in food science and nutrition, including regulatory approval, stability during food processing, and consumer acceptance. Finally, we conclude with a discussion on perspectives related to employing EcN in food science and nutrition.
Collapse
Affiliation(s)
- Miaomiao Hu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
| | - Tao Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Ming Miao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi, Jiangsu, China
- International Joint Laboratory on Food Safety, Jiangnan University, Wuxi, Jiangsu, China
| | - Kewen Li
- Baolingbao Biology Co., Ltd, Yucheng, Shandong, China
| | - Qingmin Luan
- Baolingbao Biology Co., Ltd, Yucheng, Shandong, China
| | - Guilian Sun
- Baolingbao Biology Co., Ltd, Yucheng, Shandong, China
| |
Collapse
|
16
|
Fishbein SRS, Mahmud B, Dantas G. Antibiotic perturbations to the gut microbiome. Nat Rev Microbiol 2023; 21:772-788. [PMID: 37491458 DOI: 10.1038/s41579-023-00933-y] [Citation(s) in RCA: 127] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/15/2023] [Indexed: 07/27/2023]
Abstract
Antibiotic-mediated perturbation of the gut microbiome is associated with numerous infectious and autoimmune diseases of the gastrointestinal tract. Yet, as the gut microbiome is a complex ecological network of microorganisms, the effects of antibiotics can be highly variable. With the advent of multi-omic approaches for systems-level profiling of microbial communities, we are beginning to identify microbiome-intrinsic and microbiome-extrinsic factors that affect microbiome dynamics during antibiotic exposure and subsequent recovery. In this Review, we discuss factors that influence restructuring of the gut microbiome on antibiotic exposure. We present an overview of the currently complex picture of treatment-induced changes to the microbial community and highlight essential considerations for future investigations of antibiotic-specific outcomes. Finally, we provide a synopsis of available strategies to minimize antibiotic-induced damage or to restore the pretreatment architectures of the gut microbial community.
Collapse
Affiliation(s)
- Skye R S Fishbein
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Bejan Mahmud
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA.
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA.
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
17
|
Bartram E, Asai M, Gabant P, Wigneshweraraj S. Enhancing the antibacterial function of probiotic Escherichia coli Nissle: when less is more. Appl Environ Microbiol 2023; 89:e0097523. [PMID: 37930328 PMCID: PMC10686094 DOI: 10.1128/aem.00975-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 09/11/2023] [Indexed: 11/07/2023] Open
Abstract
Probiotic bacteria confer multiple health benefits, including preventing the growth, colonization, or carriage of harmful bacteria in the gut. Bacteriocins are antibacterial peptides produced by diverse bacteria, and their production is tightly regulated and coordinated at the transcriptional level. A popular strategy for enhancing the antibacterial properties of probiotic bacteria is to retrofit them with the ability to overproduce heterologous bacteriocins. This is often achieved from non-native constitutive promoters or in response to host or pathogen signal from synthetic promoters. How the dysregulated overproduction of heterologous bacteriocins affects the fitness and antibacterial efficacy of the retrofitted probiotic bacteria is often overlooked. We have conferred the prototypical probiotic Escherichia coli strain Nissle (EcN) the ability to produce microcin C (McC) from the wild-type promoter and two mutant promoters that allow, relative to the wild-type promoter, high and low amounts of McC production. This was done by introducing specific changes to the sequence of the wild-type promoter driving transcription of the McC operon while ensuring that the modified promoters respond to native regulation. By studying the transcriptomic responses and antibacterial efficacy of the retrofitted EcN bacteria in a Galleria mellonella infection model of enterohemorrhagic E. coli, we show that EcN bacteria that produce the lowest amount of McC display the highest antibacterial efficacy with little-to-none undesired collateral impact on their fitness. The results highlight considerations researchers may take into account when retrofitting probiotic bacteria with heterogenous gene products for therapeutic, prophylactic, or diagnostic applications. Bacteria that resist killing by antibiotics are a major risk to modern medicine. The use of beneficial "probiotic" bacteria to make antibiotic-like compounds at the site of infection in the body is emerging as a popular alternative to the use of conventional antibiotics. A potential drawback of engineering probiotic bacteria in this way is that producing antibiotic-like compounds could impart undesired side effects on the performance of such bacteria, thereby compromising their intended use. This study highlights considerations researchers may take into account when engineering probiotic bacteria for therapeutic, prophylactic, or diagnostic applications.
Collapse
Affiliation(s)
- Emma Bartram
- Section of Molecular Microbiology, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| | - Masanori Asai
- Section of Molecular Microbiology, Imperial College London, London, United Kingdom
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, United Kingdom
| | | | - Sivaramesh Wigneshweraraj
- Section of Molecular Microbiology, Imperial College London, London, United Kingdom
- Centre for Bacterial Resistance Biology, Imperial College London, London, United Kingdom
| |
Collapse
|
18
|
Choudhury A, Ortiz PS, Young M, Mahmud MT, Stoffel RT, Greathouse KL, Kearney CM. Control of Helicobacter pylori with engineered probiotics secreting selective guided antimicrobial peptides. Microbiol Spectr 2023; 11:e0201423. [PMID: 37712669 PMCID: PMC10580918 DOI: 10.1128/spectrum.02014-23] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 07/26/2023] [Indexed: 09/16/2023] Open
Abstract
Helicobacter pylori is the primary cause of 78% of gastric cancer cases, providing an opportunity to prevent cancer by controlling a single bacterial pathogen within the complex gastric microbiota. We developed highly selective antimicrobial agents against H. pylori by fusing an H. pylori-binding guide peptide (MM1) to broad-spectrum antimicrobial peptides. The common dairy probiotic Lactococcus lactis was then engineered to secrete these guided antimicrobial peptides (gAMPs). When co-cultured in vitro with H. pylori, the gAMP probiotics lost no toxicity compared to unguided AMP probiotics against the target, H. pylori, while losing >90% of their toxicity against two tested off-target bacteria. To test binding to H. pylori, the MM1 guide was fused to green fluorescent protein (GFP), resulting in enhanced binding compared to unguided GFP as measured by flow cytometry. In contrast, MM1-GFP showed no increased binding over GFP against five different off-target bacteria. These highly selective gAMP probiotics were then tested by oral gavage in mice infected with H. pylori. As a therapy, the probiotics outperformed antibiotic treatment, effectively eliminating H. pylori in just 5 days, and also protected mice from challenge infection as a prophylactic. As expected, the gAMP probiotics were as toxic against H. pylori as the unguided AMP probiotics. However, a strong rebound in gastric species diversity was found with both the selective gAMP probiotics and the non-selective AMP probiotics. Eliminating the extreme microbial dysbiosis caused by H. pylori appeared to be the major factor in diversity recovery. IMPORTANCE Alternatives to antibiotics in the control of Helicobacter pylori and the prevention of gastric cancer are needed. The high prevalence of H. pylori in the human population, the induction of microbial dysbiosis by antibiotics, and increasing antibiotic resistance call for a more sustainable approach. By selectively eliminating the pathogen and retaining the commensal community, H. pylori control may be achieved without adverse health outcomes. Antibiotics are typically used as a therapeutic post-infection, but a more targeted, less disruptive approach could be used as a long-term prophylactic against H. pylori or, by extension, against other gastrointestinal pathogens. Furthermore, the modular nature of the guided antimicrobial peptide (gAMP) technology allows for the substitution of different guides for different pathogens and the use of a cocktail of gAMPs to avoid the development of pathogen resistance.
Collapse
Affiliation(s)
| | | | - Mikaeel Young
- Department of Biology, Baylor University, Waco, Texas, USA
| | | | - Ryan T. Stoffel
- Baylor Sciences Building Vivarium, Baylor University, Waco, Texas, USA
| | - K. Leigh Greathouse
- Department of Biology, Baylor University, Waco, Texas, USA
- Robbins College of Health and Human Sciences, Baylor University, Waco, Texas, USA
| | | |
Collapse
|
19
|
Khalifa A, Ibrahim HIM, Sheikh A. Bacillus subtilis PM5 from Camel Milk Boosts Chicken Immunity and Abrogates Salmonella entertitidis Infections. Microorganisms 2023; 11:1719. [PMID: 37512891 PMCID: PMC10385966 DOI: 10.3390/microorganisms11071719] [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: 05/05/2023] [Revised: 06/15/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
With the practice of a successful livestock industry using antibiotics, which has continued for more than five decades, researchers have long been interested in finding alternatives to antibiotics for poultry production. Probiotics can potentially reduce enteric diseases in livestock and enhance their productivity. The aim of this study was to isolate putative probiotics from camel milk and test them against Salmonella infection as well as host immune development. Thirteen different isolates were obtained from six different camel milk samples from dairy farms in Saudi Arabia. Three of the six isolates (PM1, PM2, PM3, PM4, PM5, and PM6) that showed Gram-positive characters reacted negatively to catalase and hemolytic assays. PM1, PM5, and PM6 showed significant nonpolar surface properties (>51% hydrophobic) and potent antimicrobial activities against avian pathogens, namely S. enterica, S. typhi, S. aureus, and E. coli. PM5 exhibited substantial probiotic traits; therefore, further focus was given to it. PM5 was identified as Bacillus subtilis OQ913924 by the 16S rRNA sequencing method and showed similarity matrix > 99%. An in vivo chicken model was used to access the health benefits of probiotics. After salmonella infection, the mucosal immune response was significantly increased (p < 0.01), and none of the challenge protocols caused mortality or clinical symptoms after infection in intestinal contents. S. enterica organ infiltration in the spleen, thymus, and small intestine was significantly reduced in the B. subtilis PM5-fed chickens. The S. enterica load in chicken feces was reduced from CFU 7.2 to 5.2 in oral-fed B. subtilis PM5-fed chickens. Probiotic-fed chickens showed buffered intestinal content and positively regulated the level of butyric acid (p < 0.05), and intestinal interleukin 1 beta (IL1-β), C-reactive protein (CRP), and interferon gamma (IFN-γ) levels were reduced (p < 0.05). In addition, B. subtilis PM5 showed significant binding to peritoneal macrophages cells and inhibited S. enterica surface adhesion, indicating co-aggregation of B. subtilis PM5 in macrophage cells. It could be concluded that supplementation with probiotics can improve the growth performance of broilers and the quality of broiler chickens against enteric pathogens. The introduction of this probiotic into the commercial poultry feed market in the near future may assist in narrowing the gap that now exists between chicken breeding and consumer demand.
Collapse
Affiliation(s)
- Ashraf Khalifa
- Biological Science Department, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia
- Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef 62511, Egypt
| | - Hairul-Islam Mohamed Ibrahim
- Biological Science Department, College of Science, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia
- Molecular Biology Division, Pondicherry Centre for Biological Sciences and Educational Trust, Pondicherry 605004, India
| | - Abdullah Sheikh
- Camel Research Center, King Faisal University, P.O. Box 400, Al-Ahsa 31982, Saudi Arabia
| |
Collapse
|
20
|
Lebovich M, Andrews LB. Genetic circuits for feedback control of gamma-aminobutyric acid biosynthesis in probiotic Escherichia coli Nissle 1917. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.09.544351. [PMID: 37333167 PMCID: PMC10274909 DOI: 10.1101/2023.06.09.544351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Engineered microorganisms such as the probiotic strain Escherichia coli Nissle 1917 (EcN) offer a strategy to sense and modulate the concentration of metabolites or therapeutics in the gastrointestinal tract. Here, we present an approach to regulate production of the depression-associated metabolite gamma-aminobutyric acid (GABA) in EcN using genetic circuits that implement negative feedback. We engineered EcN to produce GABA by overexpressing glutamate decarboxylase (GadB) from E. coli and applied an intracellular GABA biosensor to identify growth conditions that improve GABA biosynthesis. We next employed characterized genetically-encoded NOT gates to construct genetic circuits with layered feedback to control the rate of GABA biosynthesis and the concentration of GABA produced. Looking ahead, this approach may be utilized to design feedback control of microbial metabolite biosynthesis to achieve designable smart microbes that act as living therapeutics.
Collapse
Affiliation(s)
- Matthew Lebovich
- University of Massachusetts Amherst, Department of Chemical Engineering, Amherst, MA, USA
- University of Massachusetts Amherst, Biotechnology Training Program, Amherst, MA
| | - Lauren B. Andrews
- University of Massachusetts Amherst, Department of Chemical Engineering, Amherst, MA, USA
- University of Massachusetts Amherst, Biotechnology Training Program, Amherst, MA
- University of Massachusetts Amherst, Molecular and Cellular Biology Graduate Program, Amherst, MA
| |
Collapse
|
21
|
Zhou K, Sun L, Zhang X, Xu X, Mi K, Ma W, Zhang L, Huang L. Salmonella antimicrobials inherited and the non-inherited resistance: mechanisms and alternative therapeutic strategies. Front Microbiol 2023; 14:1176317. [PMID: 37303797 PMCID: PMC10249997 DOI: 10.3389/fmicb.2023.1176317] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 04/24/2023] [Indexed: 06/13/2023] Open
Abstract
Salmonella spp. is one of the most important foodborne pathogens. Typhoid fever and enteritis caused by Salmonella enterica are associated with 16-33 million infections and 500,000 to 600,000 deaths annually worldwide. The eradication of Salmonella is becoming increasingly difficult because of its remarkable capacity to counter antimicrobial agents. In addition to the intrinsic and acquired resistance of Salmonella, increasing studies indicated that its non-inherited resistance, which commonly mentioned as biofilms and persister cells, plays a critical role in refractory infections and resistance evolution. These remind the urgent demand for new therapeutic strategies against Salmonella. This review starts with escape mechanisms of Salmonella against antimicrobial agents, with particular emphasis on the roles of the non-inherited resistance in antibiotic failure and resistance evolution. Then, drug design or therapeutic strategies that show impressive effects in overcoming Salmonella resistance and tolerance are summarized completely, such as overcoming the barrier of outer membrane by targeting MlaABC system, reducing persister cells by limiting hydrogen sulfide, and applying probiotics or predatory bacteria. Meanwhile, according to the clinical practice, the advantages and disadvantages of above strategies are discussed. Finally, we further analyze how to deal with this tricky problems, thus can promote above novel strategies to be applied in the clinic as soon as possible. We believed that this review will be helpful in understanding the relationships between tolerance phenotype and resistance of Salmonella as well as the efficient control of antibiotic resistance.
Collapse
Affiliation(s)
- Kaixiang Zhou
- Department of Veterinary Medicine Science, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan, Hubei, China
| | - Lei Sun
- Department of Veterinary Medicine Science, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan, Hubei, China
| | - Xuehua Zhang
- Department of Veterinary Medicine Science, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan, Hubei, China
| | - Xiangyue Xu
- Department of Veterinary Medicine Science, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan, Hubei, China
| | - Kun Mi
- Department of Veterinary Medicine Science, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan, Hubei, China
| | - Wenjin Ma
- Department of Veterinary Medicine Science, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan, Hubei, China
| | - Lan Zhang
- Department of Veterinary Medicine Science, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan, Hubei, China
| | - Lingli Huang
- Department of Veterinary Medicine Science, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
- National Reference Laboratory of Veterinary Drug Residues (HZAU), Wuhan, Hubei, China
- MOA Key Laboratory for Detection of Veterinary Drug Residues, Wuhan, Hubei, China
| |
Collapse
|
22
|
Cesaro A, Lin S, Pardi N, de la Fuente-Nunez C. Advanced delivery systems for peptide antibiotics. Adv Drug Deliv Rev 2023; 196:114733. [PMID: 36804008 PMCID: PMC10771258 DOI: 10.1016/j.addr.2023.114733] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Revised: 02/07/2023] [Accepted: 02/14/2023] [Indexed: 02/19/2023]
Abstract
Antimicrobial peptides (AMPs) hold promise as alternatives to traditional antibiotics for preventing and treating multidrug-resistant infections. Although they have potent antimicrobial efficacy, AMPs are mainly limited by their susceptibility to proteases and potential off-site cytotoxicity. Designing the right delivery system for peptides can help to overcome such limitations, thus improving the pharmacokinetic and pharmacodynamic profiles of these drugs. The versatility of peptides and their genetically encodable structure make them suitable for both conventional and nucleoside-based formulations. In this review, we describe the main drug delivery procedures developed so far for peptide antibiotics: lipid nanoparticles, polymeric nanoparticles, hydrogels, functionalized surfaces, and DNA- and RNA-based delivery systems.
Collapse
Affiliation(s)
- Angela Cesaro
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Shuangzhe Lin
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States
| | - Norbert Pardi
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, United States; Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, United States; Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, United States.
| |
Collapse
|
23
|
Cirilo E, Junior NR, Andrade T, Souza C, Kaufmann C, Kohler T, Datsch L, Vieira B, Junior J, Carvalho P, Eyng C, Nunes R. Effects of probiotics on blood metabolites, enterocytes, growth, and carcass characteristics of broilers challenged with Salmonella Serovar Heidelberg. Livest Sci 2023. [DOI: 10.1016/j.livsci.2023.105188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/22/2023]
|
24
|
Kim SY, Parker JK, Gonzalez-Magaldi M, Telford MS, Leahy DJ, Davies BW. Export of diverse and bioactive peptides through a type I secretion system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.26.525739. [PMID: 36747863 PMCID: PMC9900886 DOI: 10.1101/2023.01.26.525739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Microcins are peptide antibiotics secreted by Gram-negative bacteria that inhibit the growth of neighboring microbes. They are exported from the cytosol to the environment in a one-step process through a specific type I secretion system (T1SS). While the rules governing export of natural or non-native substrates have been resolved for T1SSs that secrete large proteins, relatively little is known about substrate requirements for peptides exported through T1SSs that secrete microcins. Here, we investigate the prototypic microcin V T1SS from Escherichia coli and show it can export a remarkably wide range of natural and synthetic peptides. We demonstrate that secretion through this system is not affected by peptide charge or hydrophobicity and appears only constrained by peptide length. A varied range of bioactive peptides, including an antibacterial peptide, a microbial signaling factor, a protease inhibitor, and a human hormone, can all be secreted and elicit their intended biological effect. Secretion through this system is not limited to E. coli , and we demonstrate its function in additional Gram-negative species that can inhabit the gastrointestinal tract. Our findings uncover the highly promiscuous nature of peptide export thorough the microcin V T1SS, which has implications for native cargo capacity and use of Gram-negative bacteria for peptide research and delivery. Importance Microcin type I secretion systems in Gram-negative bacteria transport antibacterial peptides from the cytoplasm to the extracellular environment in single step. In nature, each microcin secretion system is generally paired with a specific peptide. We know little about the export capacity of these transporters and how peptide sequence influences secretion. Here, we investigate the microcin V type I secretion system. Remarkably, our studies show this system can export diverse peptides and is only limited by peptide length. Furthermore, we demonstrate that various bioactive peptides can be secreted, and this system can be used in Gram-negative species that colonize the gastrointestinal tract. These finding expand our understanding of secretion through type I systems and their potential uses in peptide applications.
Collapse
|
25
|
Khalifa A, Ibrahim HIM. Enterococcus faecium from chicken feces improves chicken immune response and alleviates Salmonella infections: a pilot study. J Anim Sci 2023; 101:6991176. [PMID: 36651637 PMCID: PMC10011332 DOI: 10.1093/jas/skad016] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 01/09/2023] [Indexed: 01/19/2023] Open
Abstract
Probiotics reduce the emergence of antibiotic resistance in the livestock industry. Chicken feces are reservoirs of beneficial microbiomes. The aim of this study was to isolate putative probiotics from the intestinal contents of broiler chickens. Five fecal samples were collected from two poultry farms in Al-Ahsa, Saudi Arabia. Of the 11 morphologically distinct isolates from chicken feces (CF) samples, five isolates displayed positive reactions to Gram staining, catalase, and oxidase tests, and reacted negatively to a hemolytic assay. The isolates CF1, CF2, CF3, CF8, and CF11 were selected for further analysis of probiotic characterization, gastric survival capacity, antibiotic susceptibility, and antimicrobial activity against poultry infected with Salmonella enterica. CF2 and CF11 showed the highest hydrophobic values (> 51% hydrophobic nature). CF1, CF2, and CF11 showed potent antimicrobial activities. The active isolate CF2 was identified as Enterococcus faecium by 16s rRNA sequencing and showed a genetic similarity of 99.1%. An in vivo study was conducted using a chicken model. Enterococcus faecium-fed chickens showed an improved body weight and a lower mortality rate (17-34%). Salmonella enterica colony-forming unit (CFU) invasion in the spleen and thymus was significantly reduced in the E. faecium-fed chickens. The fecal S. enterica load was reduced from CFU 6.8 to 3.9/g in oral-administered E. faecium-fed chickens. Enterococcus faecium-fed chickens showed increased levels (P < 0.01) of butyric acid and reduced levels (P < 0.01) of intestinal interleukin 1 beta, C-reactive protein, and interferon gamma levels compared to those in the S. enterica-infected chicken group. In addition, E. faecium showed significant binding to Caco-2 epithelial cells in vitro and inhibited S. enterica colonization, indicating co-aggregation of E. faecium in epithelial cells. These results revealed that an E. faecium formulation could prevent bacterial infection and improve the quality of broiler chickens.
Collapse
Affiliation(s)
- Ashraf Khalifa
- Biological Sciences Department, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia.,Botany and Microbiology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| | - Hairul Islam Mohamed Ibrahim
- Biological Sciences Department, College of Science, King Faisal University, Al-Ahsa, Saudi Arabia.,Molecular Biology Division, Pondicherry Centre for Biological Sciences and Educational Trust, Pondicherry, India
| |
Collapse
|
26
|
In Vitro and In Vivo Evaluation of Lacticaseibacillus rhamnosus GG and Bifidobacterium lactis Bb12 Against Avian Pathogenic Escherichia coli and Identification of Novel Probiotic-Derived Bioactive Peptides. Probiotics Antimicrob Proteins 2022; 14:1012-1028. [PMID: 34458959 DOI: 10.1007/s12602-021-09840-1] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2021] [Indexed: 12/24/2022]
Abstract
Avian pathogenic E. coli (APEC), an extra-intestinal pathogenic E. coli (ExPEC), causes colibacillosis in poultry and is also a potential foodborne zoonotic pathogen. Currently, APEC infections in poultry are controlled by antibiotic medication; however, the emergence of multi-drug-resistant APEC strains and increased restrictions on the use of antibiotics in food-producing animals necessitate the development of new antibiotic alternative therapies. Here, we tested the anti-APEC activity of multiple commensal and probiotic bacteria in an agar-well diffusion assay and identified Lacticaseibacillus rhamnosus GG and Bifidobacterium lactis Bb12 producing strong zone of inhibition against APEC. In co-culture assay, L. rhamnosus GG and B. lactis Bb12 completely inhibited the APEC growth by 24 h. Further investigation revealed that antibacterial product(s) in the culture supernatants of L. rhamnosus GG and B. lactis Bb12 were responsible for the anti-APEC activity. The analysis of culture supernatants using LC-MS/MS identified multiple novel bioactive peptides (VQAAQAGDTKPIEV, AFDNTDTSLDSTFKSA, VTDTSGKAGTTKISNV, and AESSDTNLVNAKAA) in addition to the production of lactic acid. The oral administration (108 CFU/chicken) of L. rhamnosus GG significantly (P < 0.001) reduced the colonization (~ 1.6 logs) of APEC in the cecum of chickens. Cecal microbiota analysis revealed that L. rhamnosus GG moderated the APEC-induced alterations of the microbial community in the cecum of chickens. Further, L. rhamnosus GG decreased (P < 0.05) the abundance of phylum Proteobacteria, particularly those belonging to Enterobacteriaceae (Escherichia-Shigella) family. These studies indicate that L. rhamnosus GG is a promising probiotic to control APEC infections in chickens. Further studies are needed to optimize the delivery of L. rhamnosus GG in feed or water and in conditions simulating the field to facilitate its development for commercial applications.
Collapse
|
27
|
Abd El-Hack ME, Alagawany M, El-Shall NA, Shehata AM, Abdel-Moneim AME, Naiel MAE. Probiotics in Poultry Nutrition as a Natural Alternative for Antibiotics. ANTIBIOTIC ALTERNATIVES IN POULTRY AND FISH FEED 2022:137-159. [DOI: 10.2174/9789815049015122010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Since the early 1950s, antibiotics have been used in poultry for improving
feed efficiency and growth performance. Nevertheless, various side effects have
appeared, such as antibiotic resistance, antibiotic residues in eggs and meat, and
imbalance of beneficial intestinal bacteria. Consequently, it is essential to find other
alternatives that include probiotics that improve poultry production. Probiotics are live
microorganisms administered in adequate doses and improve host health. Probiotics are
available to be used as feed additives, increasing the availability of the nutrients for
enhanced growth by digesting the feed properly. Immunity and meat and egg quality
can be improved by supplementation of probiotics in poultry feed. Furthermore, the
major reason for using probiotics as feed additives is that they can compete with
various infectious diseases causing pathogens in poultry's gastrointestinal tract. Hence,
this chapter focuses on the types and mechanisms of action of probiotics and their
benefits, by feed supplementation, for poultry health and production.
Collapse
Affiliation(s)
| | | | - Nahed A. El-Shall
- Alexandria University,Department of poultry and fish diseases,Elbehira,Egypt
| | | | | | | |
Collapse
|
28
|
Mortzfeld BM, Palmer JD, Bhattarai SK, Dupre HL, Mercado-Lubio R, Silby MW, Bang C, McCormick BA, Bucci V. Microcin MccI47 selectively inhibits enteric bacteria and reduces carbapenem-resistant Klebsiella pneumoniae colonization in vivo when administered via an engineered live biotherapeutic. Gut Microbes 2022; 14:2127633. [PMID: 36175830 PMCID: PMC9542533 DOI: 10.1080/19490976.2022.2127633] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The gastrointestinal (GI) tract is the reservoir for multidrug resistant (MDR) pathogens, specifically carbapenem-resistant (CR) Klebsiella pneumoniae and other Enterobacteriaceae, which often lead to the spread of antimicrobial resistance genes, severe extraintestinal infections, and lethal outcomes. Selective GI decolonization has been proposed as a new strategy for preventing transmission to other body sites and minimizing spreading to susceptible individuals. Here, we purify the to-date uncharacterized class IIb microcin I47 (MccI47) and demonstrate potent inhibition of numerous Enterobacteriaceae, including multidrug-resistant clinical isolates, in vitro at concentrations resembling those of commonly prescribed antibiotics. We then genetically modify the probiotic bacterium Escherichia coli Nissle 1917 (EcN) to produce MccI47 from a stable multicopy plasmid by using MccI47 toxin production in a counterselection mechanism to engineer one of the native EcN plasmids, which renders provisions for inducible expression and plasmid selection unnecessary. We then test the clinical relevance of the MccI47-producing engineered EcN in a murine CR K. pneumoniae colonization model and demonstrate significant MccI47-dependent reduction of CR K. pneumoniae abundance after seven days of daily oral live biotherapeutic administration without disruption of the resident microbiota. This study provides the first demonstration of MccI47 as a potent antimicrobial against certain Enterobacteriaceae, and its ability to significantly reduce the abundance of CR K. pneumoniae in a preclinical animal model, when delivered from an engineered live biotherapeutic product. This study serves as the foundational step toward the use of engineered live biotherapeutic products aimed at the selective removal of MDR pathogens from the GI tract.
Collapse
Affiliation(s)
- Benedikt M. Mortzfeld
- Department of Microbiology and Physiological Systems, Universty of Massachusetts Chan Medical School, Worcester, MA, USA,Program in Microbiome Dynamics, Universty of Massachusetts Chan Medical School, Worcester, MA, USA,CONTACT Benedikt M. Mortzfeld Program in Microbiome Dynamics Universty of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Jacob D. Palmer
- Department of Zoology, University of Oxford, Oxford, UK,Department of Biochemistry, University of Oxford, Oxford, UK
| | - Shakti K. Bhattarai
- Department of Microbiology and Physiological Systems, Universty of Massachusetts Chan Medical School, Worcester, MA, USA,Program in Microbiome Dynamics, Universty of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Haley L. Dupre
- Department of Bioengineering, University of Massachusetts Dartmouth, North Dartmouth, MA, USA
| | - Regino Mercado-Lubio
- Department of Microbiology and Physiological Systems, Universty of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Mark W. Silby
- Department of Biology, University of Massachusetts Dartmouth, Dartmouth, MA, USA
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität Zu Kiel, Kiel, Germany
| | - Beth A. McCormick
- Department of Microbiology and Physiological Systems, Universty of Massachusetts Chan Medical School, Worcester, MA, USA,Program in Microbiome Dynamics, Universty of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Vanni Bucci
- Department of Microbiology and Physiological Systems, Universty of Massachusetts Chan Medical School, Worcester, MA, USA,Program in Microbiome Dynamics, Universty of Massachusetts Chan Medical School, Worcester, MA, USA,Program in Systems Biology, Universty of Massachusetts Chan Medical School, Worcester, MA, USA,Vanni Bucci Department of Microbiology and Physiological Systems, Universty of Massachusetts Chan Medical School, Worcester, MA, USA
| |
Collapse
|
29
|
Lynch JP, Goers L, Lesser CF. Emerging strategies for engineering Escherichia coli Nissle 1917-based therapeutics. Trends Pharmacol Sci 2022; 43:772-786. [PMID: 35232591 PMCID: PMC9378478 DOI: 10.1016/j.tips.2022.02.002] [Citation(s) in RCA: 79] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/02/2022] [Accepted: 02/04/2022] [Indexed: 12/11/2022]
Abstract
Engineered microbes are rapidly being developed for the delivery of therapeutic modalities to sites of disease. Escherichia coli Nissle 1917 (EcN), a genetically tractable probiotic with a well-established human safety record, is emerging as a favored chassis. Here, we summarize the latest progress in rationally engineered variants of EcN for the treatment of infectious diseases, metabolic disorders, and inflammatory bowel diseases (IBDs) when administered orally, as well as cancers when injected directly into tumors or the systemic circulation. We also discuss emerging studies that raise potential safety concerns regarding these EcN-based strains as therapeutics due to their secretion of a genotoxic colibactin that can promote the formation of DNA double-stranded breaks in mammalian DNA.
Collapse
Affiliation(s)
- Jason P Lynch
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, MA 02115, USA; Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Lisa Goers
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, MA 02115, USA; Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Cammie F Lesser
- Center for Bacterial Pathogenesis, Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, MA 02115, USA; Department of Microbiology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA.
| |
Collapse
|
30
|
Porter SB, Johnston BD, Kisiela D, Clabots C, Sokurenko EV, Johnson JR. Bacteriophage Cocktail and Microcin-Producing Probiotic Escherichia coli Protect Mice Against Gut Colonization With Multidrug-Resistant Escherichia coli Sequence Type 131. Front Microbiol 2022; 13:887799. [PMID: 35547133 PMCID: PMC9082999 DOI: 10.3389/fmicb.2022.887799] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/06/2022] [Indexed: 11/24/2022] Open
Abstract
Non-antibiotic measures are needed to reduce the rate of infections due to multidrug-resistant organisms (MDROs), including by eliminating the commensal reservoir that underlies such strains’ dissemination and leads to recurrent infections. Here, we tested a cocktail of pre-selected bacteriophages and an engineered microcin C7-producing probiotic Escherichia coli Nissle-1917 strain for their ability to reduce gut colonization by an E. coli strain from sequence type 131 (ST131)-H30R, which is the major clonal group of MDROs among extraintestinal clinical E. coli isolates. Although the bacteriophage cocktail was highly effective against ST131-H30R strains both in vitro and in a murine model of subcutaneous sepsis, it was only weakly and transiently effective against gut colonization by the target ST131-H30R strain (0.5 log10 decrease on d + 1: p < 0.001; no significant effect on d + 4 and beyond). The probiotic strain, while also highly active against ST131-H30R in vitro, was ineffective against ST131-H30R gut colonization despite its abundant presence in feces. Nonetheless, despite failing as decolonizing agents when administered separately, when co-administered the bacteriophage cocktail and probiotic strain exhibited striking synergy against ST131-H30R gut colonization. This combinatory effect was most pronounced on d + 1 (3.3 log10 target strain decrease: p < 0.001), and persisted until d + 7 (0.5 log10 decrease; p < 0.02.). Although by d + 10 the ST131-H30R load was fully restored, these findings provide proof of concept for combined bacteriophage-plus-probiotic administration to reduce or, possibly, to prevent gut colonization with MDROs in high-risk individuals.
Collapse
Affiliation(s)
- Stephen B Porter
- Minneapolis VA Health Care System, Veterans Health Administration, United States Department of Veterans Affairs, Minneapolis, MN, United States
| | - Brian D Johnston
- Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| | - Dagmara Kisiela
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - Connie Clabots
- Minneapolis VA Health Care System, Veterans Health Administration, United States Department of Veterans Affairs, Minneapolis, MN, United States
| | - Evgeni V Sokurenko
- Department of Microbiology, University of Washington, Seattle, WA, United States
| | - James R Johnson
- Minneapolis VA Health Care System, Veterans Health Administration, United States Department of Veterans Affairs, Minneapolis, MN, United States.,Department of Medicine, University of Minnesota, Minneapolis, MN, United States
| |
Collapse
|
31
|
Immobilization Techniques on Bioprocesses: Current Applications Regarding Enzymes, Microorganisms, and Essential Oils. FOOD BIOPROCESS TECH 2022. [DOI: 10.1007/s11947-022-02780-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
|
32
|
Yu H, Shang L, Yang G, Dai Z, Zeng X, Qiao S. Biosynthetic Microcin J25 Exerts Strong Antibacterial, Anti-Inflammatory Activities, Low Cytotoxicity Without Increasing Drug-Resistance to Bacteria Target. Front Immunol 2022; 13:811378. [PMID: 35250983 PMCID: PMC8894198 DOI: 10.3389/fimmu.2022.811378] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/27/2022] [Indexed: 12/15/2022] Open
Abstract
Multidrug resistant (MDR) bacterial infection has emerged, raising concerns about untreatable infections, and posing the highest health risks. Antimicrobial peptides (AMPs) are thought to be the best remedy for this problem. Here, we showed biosynthetic microcin J25 (MccJ25) exhibited excellent bactericidal activity against standard and clinically relevant veterinary MDR strains with high stability, no cytotoxicity, and no increase in drug resistance. Analysis of antimicrobial mechanism possessed by sensitive enterotoxigenic Escherichia coli (ETEC) based on electron microscopy and Sytox Green methods was carried out. Results showed excellent activity against ETEC was due to permeabilizing bacterial membranes and strong affinity. MccJ25 exhibited high endotoxin-neutralizing activity in both in vivo and in vitro environments, and mice exposed to lipopolysaccharide (LPS) showed decreased plasma LPS levels and improved survival after administration of MccJ25. In an LPS-treated mouse septicemia model, MccJ25 treatment significantly alleviated inflammatory responses by inhibiting proinflammatory factor secretion and expression. In a mouse E. coli infection model, administration of MccJ25 effectively improved host defense against clinically source cocktail of multidrug-resistant E. coli strains induced intestinal inflammation and bacteria dissemination. Results of studies on anti-inflammatory mechanisms showed that MccJ25 downregulated nuclear factor kappa B kinase and mitogen-activated protein kinase, thereby reducing the production of toll-like receptor 4, myeloid differentiation factor 88 and decreasing the key proinflammatory cytokines. These findings clarify MccJ25 may be an ideal antibacterial/antiendotoxic drug candidate that has the potential to further guide the development of anti-inflammatory and/or antimicrobial agents in the war against MDR bacterial infection.
Collapse
Affiliation(s)
- Haitao Yu
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China
- Department of Immunology, Beijing Key Laboratory of Tumor Systems Biology, Institute of Systems Biomedicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Lijun Shang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Biofeed Additives, China Agricultural University, Beijing, China
| | - Guangxin Yang
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Biofeed Additives, China Agricultural University, Beijing, China
| | - Ziqi Dai
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Biofeed Additives, China Agricultural University, Beijing, China
| | - Xiangfang Zeng
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Biofeed Additives, China Agricultural University, Beijing, China
| | - Shiyan Qiao
- State Key Laboratory of Animal Nutrition, Ministry of Agriculture and Rural Affairs Feed Industry Center, China Agricultural University, Beijing, China
- Beijing Key Laboratory of Biofeed Additives, China Agricultural University, Beijing, China
- *Correspondence: Shiyan Qiao,
| |
Collapse
|
33
|
Bacteria and bacterial derivatives as delivery carriers for immunotherapy. Adv Drug Deliv Rev 2022; 181:114085. [PMID: 34933064 DOI: 10.1016/j.addr.2021.114085] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 11/16/2021] [Accepted: 12/14/2021] [Indexed: 02/08/2023]
Abstract
There is growing interest in the role of microorganisms in human health and disease, with evidence showing that new types of biotherapy using engineered bacterial therapeutics, including bacterial derivatives, can address specific mechanisms of disease. The complex interactions between microorganisms and metabolic/immunologic pathways underlie many diseases with unmet medical needs, suggesting that targeting these interactions may improve patient treatment. Using tools from synthetic biology and chemical engineering, non-pathogenic bacteria or bacterial products can be programmed and designed to sense and respond to environmental signals to deliver therapeutic effectors. This review describes current progress in biotherapy using live bacteria and their derivatives to achieve therapeutic benefits against various diseases.
Collapse
|
34
|
Pandey M, Bhati A, Priya K, Sharma KK, Singhal B. Precision Postbiotics and Mental Health: the Management of Post-COVID-19 Complications. Probiotics Antimicrob Proteins 2021; 14:426-448. [PMID: 34806151 PMCID: PMC8606251 DOI: 10.1007/s12602-021-09875-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/11/2021] [Indexed: 01/14/2023]
Abstract
The health catastrophe originated by COVID-19 pandemic construed profound impact on a global scale. However, a plethora of research studies corroborated convincing evidence conferring severity of infection of SARS-CoV-2 with the aberrant gut microbiome that strongly speculated its importance for development of novel therapeutic modalities. The intense exploration of probiotics has been envisaged to promote the healthy growth of the host, and restore intestinal microecological balance through various metabolic and physiological processes. The demystifying effect of probiotics cannot be defied, but there exists a strong skepticism related to their safety and efficacy. Therefore, molecular signature of probiotics termed as "postbiotics" are of paramount importance and there is continuous surge of utilizing postbiotics for enhancing health benefits, but little is explicit about their antiviral effects. Therefore, it is worth considering their prospective role in post-COVID regime that pave the way for exploring the pastoral vistas of postbiotics. Based on previous research investigations, the present article advocates prospective role of postbiotics in alleviating the health burden of viral infections, especially SARS-CoV-2. The article also posits current challenges and proposes a futuristic model describing the concept of "precision postbiotics" for effective therapeutic and preventive interventions that can be used for management of this deadly disease.
Collapse
Affiliation(s)
- Muskan Pandey
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - Archana Bhati
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - Kumari Priya
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India
| | - K K Sharma
- Department of Microbiology, Maharshi Dayanand University, Rohtak, Haryana, 124001, India
| | - Barkha Singhal
- School of Biotechnology, Gautam Buddha University, Greater Noida, Uttar Pradesh, 201312, India.
| |
Collapse
|
35
|
Abstract
The steadfast advance of the synthetic biology field has enabled scientists to use genetically engineered cells, instead of small molecules or biologics, as the basis for the development of novel therapeutics. Cells endowed with synthetic gene circuits can control the localization, timing and dosage of therapeutic activities in response to specific disease biomarkers and thus represent a powerful new weapon in the fight against disease. Here, we conceptualize how synthetic biology approaches can be applied to programme living cells with therapeutic functions and discuss the advantages that they offer over conventional therapies in terms of flexibility, specificity and predictability, as well as challenges for their development. We present notable advances in the creation of engineered cells that harbour synthetic gene circuits capable of biological sensing and computation of signals derived from intracellular or extracellular biomarkers. We categorize and describe these developments based on the cell scaffold (human or microbial) and the site at which the engineered cell exerts its therapeutic function within its human host. The design of cell-based therapeutics with synthetic biology is a rapidly growing strategy in medicine that holds great promise for the development of effective treatments for a wide variety of human diseases.
Collapse
|
36
|
Design and in situ biosynthesis of precision therapies against gastrointestinal pathogens. CURRENT OPINION IN PHYSIOLOGY 2021. [DOI: 10.1016/j.cophys.2021.06.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
37
|
Śmiałek M, Kowalczyk J, Koncicki A. The Use of Probiotics in the Reduction of Campylobacter spp. Prevalence in Poultry. Animals (Basel) 2021; 11:1355. [PMID: 34068764 PMCID: PMC8150830 DOI: 10.3390/ani11051355] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 04/21/2021] [Accepted: 05/06/2021] [Indexed: 12/23/2022] Open
Abstract
Campylobacter spp. are widely distributed microorganisms, many of which are commensals of gastrointestinal tract in multiple animal species, including poultry. Most commonly detected are C. jejuni and C. coli. Although infections are usually asymptomatic in poultry, poultry meat and products represent main sources of infection with these bacteria to humans. According to recent EFSA report, campylobacteriosis is the most commonly reported zoonotic disease. In 2018, EFSA Panel on Biological Hazards indicated that use of feed and water additives is the second most likely strategy that can be successful in minimizing Campylobacter spp. colonization rate in broiler chickens. One of those feed and water additives are probiotics. From numerous research papers it can be concluded that probiotics exhibit plenty of mechanisms of anti-Campylobacter activity, which were evaluated under in vitro conditions. These results, to some extent, can explain the efficacy of probiotics in in vivo studies, although different outcome can be observed under these two laboratory conditions. Probiotics are capable of reducing Campylobacter spp. population count in poultry gastrointestinal tract and they can reduce carcass contamination. Potential modes of anti-Campylobacter activity of probiotics, results of in vivo studies and studies performed at a farm level are widely discussed in the paper.
Collapse
Affiliation(s)
- Marcin Śmiałek
- Department of Poultry Diseases, Faculty of Veterinary Medicine, University of Warmia and Mazury, Oczapowskiego 13, 10-719 Olsztyn, Poland; (J.K.); (A.K.)
| | | | | |
Collapse
|
38
|
Nair D, Vazhakkattu Thomas J, Dewi G, Brannon J, Noll S, Johnson T, Cox R, Kollanoor Johny A. Propionibacterium freudenreichii freudenreichii B3523 reduces cecal colonization and internal organ dissemination of multidrug-resistant Salmonella Heidelberg in finishing turkeys. J APPL POULTRY RES 2021. [DOI: 10.1016/j.japr.2020.10.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
39
|
Kaur J, Singh BP, Chaudhary V, Elshaghabee FMF, Singh J, Singh A, Rokana N, Panwar H. Probiotics as Live Bio-therapeutics: Prospects and Perspectives. MICROORGANISMS FOR SUSTAINABILITY 2021:83-120. [DOI: 10.1007/978-981-15-6795-7_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
40
|
Telhig S, Ben Said L, Zirah S, Fliss I, Rebuffat S. Bacteriocins to Thwart Bacterial Resistance in Gram Negative Bacteria. Front Microbiol 2020; 11:586433. [PMID: 33240239 PMCID: PMC7680869 DOI: 10.3389/fmicb.2020.586433] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/16/2020] [Indexed: 12/16/2022] Open
Abstract
An overuse of antibiotics both in human and animal health and as growth promoters in farming practices has increased the prevalence of antibiotic resistance in bacteria. Antibiotic resistant and multi-resistant bacteria are now considered a major and increasing threat by national health agencies, making the need for novel strategies to fight bugs and super bugs a first priority. In particular, Gram-negative bacteria are responsible for a high proportion of nosocomial infections attributable for a large part to Enterobacteriaceae, such as pathogenic Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa. To cope with their highly competitive environments, bacteria have evolved various adaptive strategies, among which the production of narrow spectrum antimicrobial peptides called bacteriocins and specifically microcins in Gram-negative bacteria. They are produced as precursor peptides that further undergo proteolytic cleavage and in many cases more or less complex posttranslational modifications, which contribute to improve their stability and efficiency. Many have a high stability in the gastrointestinal tract where they can target a single pathogen whilst only slightly perturbing the gut microbiota. Several microcins and antibiotics can bind to similar bacterial receptors and use similar pathways to cross the double-membrane of Gram-negative bacteria and reach their intracellular targets, which they also can share. Consequently, bacteria may use common mechanisms of resistance against microcins and antibiotics. This review describes both unmodified and modified microcins [lasso peptides, siderophore peptides, nucleotide peptides, linear azole(in)e-containing peptides], highlighting their potential as weapons to thwart bacterial resistance in Gram-negative pathogens and discusses the possibility of cross-resistance and co-resistance occurrence between antibiotics and microcins in Gram-negative bacteria.
Collapse
Affiliation(s)
- Soufiane Telhig
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
- Laboratory Molecules of Communication and Adaptation of Microorganisms, Muséum National d’Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| | - Laila Ben Said
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| | - Séverine Zirah
- Laboratory Molecules of Communication and Adaptation of Microorganisms, Muséum National d’Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| | - Ismail Fliss
- Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| | - Sylvie Rebuffat
- Laboratory Molecules of Communication and Adaptation of Microorganisms, Muséum National d’Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| |
Collapse
|
41
|
Oral delivery of bacteria: Basic principles and biomedical applications. J Control Release 2020; 327:801-833. [PMID: 32926886 DOI: 10.1016/j.jconrel.2020.09.011] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Accepted: 09/05/2020] [Indexed: 12/18/2022]
|
42
|
Mining and Statistical Modeling of Natural and Variant Class IIa Bacteriocins Elucidate Activity and Selectivity Profiles across Species. Appl Environ Microbiol 2020; 86:AEM.01646-20. [PMID: 32917749 DOI: 10.1128/aem.01646-20] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Accepted: 09/08/2020] [Indexed: 12/24/2022] Open
Abstract
Class IIa bacteriocin antimicrobial peptides (AMPs) are a compelling alternative to current antimicrobials because of potential specific activity toward antibiotic-resistant bacteria, including vancomycin-resistant enterococci. Engineering of these molecules would be enhanced by a better understanding of AMP sequence-activity relationships to improve efficacy in vivo and limit effects of off-target activity. Toward this goal, we experimentally evaluated 210 natural and variant class IIa bacteriocins for antimicrobial activity against six strains of enterococci. Inhibitory activity was ridge regressed to AMP sequence to predict performance, achieving an area under the curve of 0.70 and demonstrating the potential of statistical models for identifying and designing AMPs. Active AMPs were individually produced and evaluated against eight enterococcus strains and four Listeria strains to elucidate trends in susceptibility. It was determined that the mannose phosphotransferase system (manPTS) sequence is informative of susceptibility to class IIa bacteriocins, yet other factors, such as membrane composition, also contribute strongly to susceptibility. A broadly potent bacteriocin variant (lactocin DT1) from a Lactobacillus ruminis genome was identified as the only variant with inhibitory activity toward all tested strains, while a novel enterocin variant (DT2) from an Enterococcus faecium genome demonstrated specificity toward Listeria strains. Eight AMPs were evaluated for proteolytic stability to trypsin, chymotrypsin, and pepsin, and three C-terminal disulfide-containing variants, including divercin V41, were identified as compelling for future in vivo studies, given their high potency and proteolytic stability.IMPORTANCE Class IIa bacteriocin antimicrobial peptides (AMPs), an alternative to traditional small-molecule antibiotics, are capable of selective activity toward various Gram-positive bacteria, limiting negative side effects associated with broad-spectrum activity. This selective activity is achieved through targeting of the mannose phosphotransferase system (manPTS) of a subset of Gram-positive bacteria, although factors affecting this mechanism are not entirely understood. Peptides identified from genomic data, as well as variants of previously characterized AMPs, can offer insight into how peptide sequence affects activity and selectivity. The experimental methods presented here identify promising potent and selective bacteriocins for further evaluation, highlight the potential of simple computational modeling for prediction of AMP performance, and demonstrate that factors beyond manPTS sequence affect bacterial susceptibility to class IIa bacteriocins.
Collapse
|
43
|
Abd El-Hack ME, El-Saadony MT, Shafi ME, Qattan SYA, Batiha GE, Khafaga AF, Abdel-Moneim AME, Alagawany M. Probiotics in poultry feed: A comprehensive review. J Anim Physiol Anim Nutr (Berl) 2020; 104:1835-1850. [PMID: 32996177 DOI: 10.1111/jpn.13454] [Citation(s) in RCA: 176] [Impact Index Per Article: 35.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 01/06/2023]
Abstract
The use of antibiotics to maintain animal well-being, promote growth and improve efficiency has been practised for more than 50 years. However, as early as the 1950s, researchers identified concern on the development of resistant bacteria for the antibiotics streptomycin and tetracycline used in turkeys and broilers respectively. These findings laid the groundwork for agricultural officials to impose stricter regulatory parameters on the use of antibiotics in poultry feeds. Probiotics are live micro-organisms included in the diet of animals as feed additives or supplements. Commonly known as a direct-fed microbial, probiotics provide beneficial properties to the host, primarily through action in the gastrointestinal tract (GIT) of the animal. Supplementation of probiotics in the diet can improve animal health and performance, through contributions to gut health and nutrient use. For instance, supplementation of probiotics has been demonstrated to benefit farm animals in immune modulation, structural modulation and increased cytokine production, which positively affect the intestinal mucosal lining against pathogens. Bacillus subtilis has been a popular bacterium used within the industry and was shown to improve intestinal villus height. Increasing the villus height and structure of the crypts in the GIT allows for the improvement of nutrient digestion and absorption. Tight junctions maintain important defences against pathogenic bacteria and cellular homeostasis. Heat stress can be a major environmental challenge in the poultry industry. Heat stress causes the bird to fluctuate its internal core temperature beyond their comfort zone. To overcome such challenges, poultry will attempt to balance its heat production and dissipation through behavioural and physiological adaptation mechanisms.
Collapse
Affiliation(s)
| | - Mohamed T El-Saadony
- Agricultural Microbiology Department, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| | - Manal E Shafi
- Department of Biological Sciences, Zoology, Abdulaziz University, Jeddah, Saudi Arabia
| | - Shaza Y A Qattan
- Department of Biological Sciences, Microbiology, Faculty of Science, Abdulaziz University, Jeddah, Saudi Arabia
| | - Gaber E Batiha
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan.,Department of Pharmacology and Therapeutics, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Al-Beheira, Egypt
| | - Asmaa F Khafaga
- Department of Pathology, Faculty of Veterinary Medicine, Alexandria University, Edfina, Egypt
| | | | - Mahmoud Alagawany
- Department of Poultry, Faculty of Agriculture, Zagazig University, Zagazig, Egypt
| |
Collapse
|
44
|
Liu X, Jiang L, Li L, Yu H, Nie S, Xie M, Gong J. The Role of Neurotransmitters in the Protection of Caenorhabditis Elegans for Salmonella Infection by Lactobacillus. Front Cell Infect Microbiol 2020; 10:554052. [PMID: 33134188 PMCID: PMC7550654 DOI: 10.3389/fcimb.2020.554052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/20/2020] [Indexed: 12/20/2022] Open
Abstract
Salmonellosis is a common foodborne disease. We previously reported the protection of Caenorhabditis elegans from Salmonella Typhimurium DT104 infection by Lactobacillus zeae LB1. However, the mechanism is not fully understood. C. elegans exhibits behavior plasticity when presented with diverse pathogenic or commensal bacteria. Whether it can exert approach avoidance to S. Typhimurium through altering its neurological activity remains to be determined. In the current study, both the wild type and mutants defective in serotonin or dopamine production of C. elegans were used to investigate olfactory preference of the nematode to L. zeae LB1, DT104, and Escherichia coli OP50 by choice assays, and its resistance to DT104 infection and the protection offered by L. zeae LB1 using a life-span assay. The expression of target genes in C. elegans was also examined by real-time quantitative PCR. Results showed that pre-exposure to L. zeae LB1 did not elicit aversive olfactory behavior of the nematode toward DT104. Both mutants tph-1 and cat-2 succumbed faster than the wild type when infected with DT104. While pre-exposure to L. zeae LB1 significantly increased the survival of both the wild type and mutant tph-1, it provided no protection to mutant cat-2. Supplementation of dopamine resulted in both the resistance of mutant cat-2 to S. Typhimurium infection and the protection from L. zeae LB1 to the same mutant. Gene expression data also supported the observations in the life-span assay. These results suggest that both serotonin and dopamine play a positive role in the host defense of C. elegans to S. Typhimurium infection and that the L. zeae LB1 protection is not dependent on modifying olfactory preference of the nematode but mediated by dopamine that may have involved the regulation of p38-mitogen-activated protein kinase and insulin/insulin-like growth factor signaling pathways.
Collapse
Affiliation(s)
- Xiaozhen Liu
- Engineering Research Center of Health Food Design & Nutrition Regulation, School of Chemical Engineering and Energy Technology, Dongguan University of Technology, Dongguan, China.,Guelph Research and Development Center, Agriculture and Agri-Food Canada, Guelph, ON, Canada.,State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Leming Jiang
- Guelph Research and Development Center, Agriculture and Agri-Food Canada, Guelph, ON, Canada.,State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Linyan Li
- Guelph Research and Development Center, Agriculture and Agri-Food Canada, Guelph, ON, Canada.,State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Hai Yu
- Guelph Research and Development Center, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| | - Shaoping Nie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China
| | - Mingyong Xie
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, China.,National R&D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, China
| | - Joshua Gong
- Guelph Research and Development Center, Agriculture and Agri-Food Canada, Guelph, ON, Canada
| |
Collapse
|
45
|
Lasso Peptide Microcin J25 Effectively Enhances Gut Barrier Function and Modulates Inflammatory Response in an Enterotoxigenic Escherichia coli-Challenged Mouse Model. Int J Mol Sci 2020; 21:ijms21186500. [PMID: 32899529 PMCID: PMC7555725 DOI: 10.3390/ijms21186500] [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: 07/16/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 02/07/2023] Open
Abstract
Bacterial resistance leads to severe public health and safety issues worldwide. Alternatives to antibiotics are currently needed. A promising lasso peptide, microcin J25 (MccJ25), is considered to be the best potential substitute for antibiotics to treat pathogen infection, including enterotoxigenic Escherichia coli (ETEC). This study evaluated the efficacy of MccJ25 in the prevention of ETEC infection. Forty-five female BALB/c mice of clean grade (aged seven weeks, approximately 16.15 g) were randomly divided into three experimental groups as follows: (i) control group (uninfected); (ii) ETEC infection group; (iii) MccJ25 + ETEC group. Fifteen mice per group in five cages, three mice/cage. MccJ25 conferred effective protection against ETEC-induced body weight loss, decrease in rectal temperature and increase in diarrhea scores in mice. Moreover, in ETEC-challenged mice model, MccJ25 significantly improved intestinal morphology, decreased intestinal histopathological scores and attenuated intestinal inflammation by decreasing proinflammatory cytokines and intestinal permeability, including reducing serum diamine oxidase and D-lactate levels. MccJ25 enhanced epithelial barrier function by increasing occludin expression in the colon and claudin-1 expression in the jejunum, ultimately improving intestinal health of host. MccJ25 was further found to alleviate gut inflammatory responses by decreasing inflammatory cytokine production and expression via the activation of the mitogen-activated protein kinase and nuclear factor κB signaling pathways. Taken together, the results indicated that MccJ25 protects against ETEC-induced intestinal injury and intestinal inflammatory responses, suggesting the potential application of MccJ25 as an excellent antimicrobial or anti-inflammation agent against pathogen infections.
Collapse
|
46
|
Naimi S, Zirah S, Taher MB, Theolier J, Fernandez B, Rebuffat SF, Fliss I. Microcin J25 Exhibits Inhibitory Activity Against Salmonella Newport in Continuous Fermentation Model Mimicking Swine Colonic Conditions. Front Microbiol 2020; 11:988. [PMID: 32528437 PMCID: PMC7262971 DOI: 10.3389/fmicb.2020.00988] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 04/23/2020] [Indexed: 12/30/2022] Open
Abstract
Microcin J25 (MccJ25), a 21-amino acid bacteriocin produced by Escherichia coli (E. coli), is a potent inhibitor of Enterobacteriaceae, including pathogenic E. coli, Salmonella, and Shigella. Its lasso structure makes it highly stable and therefore of interest as a possible antimicrobial agent in foods or as an alternative to antibiotics in livestock production. In the present study, we aimed to evaluate in vitro the inhibitory activity of MccJ25 against Salmonella enterica subsp. enterica serovar Newport ATCC 6962 (Salmonella Newport) used as a model pathogen under conditions simulating those of the swine proximal colon. The growth inhibition activity of MccJ25 against Salmonella Newport was examined in lysogeny broth (LB) and in modified MacFarlane medium that allows miming the swine colonic conditions. The MccJ25 activity was further determined using the Polyfermentor intestinal model (PolyFermS), an in vitro continuous fermentation model that permits deciphering the activity of any antimicrobial molecule in real colon fermentation conditions using selected microbiota. It was set up here to simulate the porcine proximal colon fermentation. In these conditions, the inhibition activity of MccJ25 was compared to those of two antimicrobial agents, reuterin and rifampicin. The minimal inhibitory concentration (MIC) of MccJ25 was determined at 0.03 μM in LB medium, compared to 1,079 and 38 μM for reuterin and rifampicin, respectively, showing a significantly higher potency of MccJ25. Total inhibition of Salmonella Newport was observed in LB medium over 24 h of incubation at concentrations starting from the MIC. In the PolyFermS model, MccJ25 induced a significantly stronger inhibition of Salmonella Newport growth than reuterin or rifampicin. A specific and sensitive LC-MS method allowed to detect and quantify MccJ25 in the PolyFermS fermentation system, showing that MccJ25 remains stable and active against Salmonella in conditions mimicking those found in swine colon. This study paves the way for further exploring the potential of this bacteriocin as an alternative to antibiotics in livestock.
Collapse
Affiliation(s)
- Sabrine Naimi
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| | - Séverine Zirah
- Laboratoire Molécules de Communication et Adaptation des Microorganismes (MCAM), Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| | - Menel Ben Taher
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| | - Jérémie Theolier
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| | - Benoît Fernandez
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| | - Sylvie Françoise Rebuffat
- Laboratoire Molécules de Communication et Adaptation des Microorganismes (MCAM), Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Paris, France
| | - Ismail Fliss
- STELA Dairy Research Center, Institute of Nutrition and Functional Foods, Université Laval, Québec, QC, Canada
| |
Collapse
|
47
|
Nielsen B, Colle MJ, Ünlü G. Meat safety and quality: a biological approach. Int J Food Sci Technol 2020. [DOI: 10.1111/ijfs.14602] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Affiliation(s)
- Barbara Nielsen
- School of Food Science University of Idaho 875 Perimeter Drive Moscow ID 83844‐2312 USA
| | - Michael J. Colle
- Department of Animal and Veterinary Science University of Idaho 875 Perimeter Drive Moscow ID 83844‐2330 USA
| | - Gülhan Ünlü
- School of Food Science University of Idaho 875 Perimeter Drive Moscow ID 83844‐2312 USA
- School of Food Science Washington State University Pullman WA 99164‐6376 USA
- Department of Biological Engineering University of Idaho 875 Perimeter Drive Moscow ID 83844‐0904 USA
| |
Collapse
|
48
|
Ben Said L, Emond-Rheault JG, Soltani S, Telhig S, Zirah S, Rebuffat S, Diarra MS, Goodridge L, Levesque RC, Fliss I. Phenomic and genomic approaches to studying the inhibition of multiresistant Salmonella enterica by microcin J25. Environ Microbiol 2020; 22:2907-2920. [PMID: 32363677 DOI: 10.1111/1462-2920.15045] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/15/2020] [Accepted: 04/25/2020] [Indexed: 12/22/2022]
Abstract
In livestock production, antibiotics are used to promote animal growth, control infections and thereby increase profitability. This practice has led to the emergence of multiresistant bacteria such as Salmonella, of which some serovars are disseminated in the environment. The objective of this study is to evaluate microcin J25 as an inhibitor of Salmonella enterica serovars of various origins including human, livestock and food. Among the 116 isolates tested, 37 (31.8%) were found resistant to at least one antibiotic, and 28 were multiresistant with 19 expressing the penta-resistant phenotype ACSSuT. Microcin J25 inhibited all isolates, with minimal inhibitory concentration values ranging from 0.06 μg/ml (28.4 nM) to 400 μg/ml (189 μM). Interestingly, no cross-resistance was found between microcin J25 and antibiotics. Multiple sequence alignments of genes encoding for the different proteins involved in the recognition and transport of microcin J25 showed that only ferric-hydroxamate uptake is an essential determinant for susceptibility of S. enterica to microcin J25. Examination of Salmonella strains exposed to microcin J25 by transmission electronic microscopy showed for the first-time involvement of a pore formation mechanism. Microcin J25 was a strong inhibitor of several multiresistant isolates of Salmonella and may have a great potential as an alternative to antibiotics.
Collapse
Affiliation(s)
- Laila Ben Said
- Institute of Nutrition and Functional Foods, Université Laval, Québec, Quebec, G1V 0A6, Canada
| | | | - Samira Soltani
- Institute of Nutrition and Functional Foods, Université Laval, Québec, Quebec, G1V 0A6, Canada
| | - Sofiane Telhig
- Institute of Nutrition and Functional Foods, Université Laval, Québec, Quebec, G1V 0A6, Canada.,Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Laboratory of Communication Molecules and Adaptation of Micro-organisms, UMR 7245 CNRS-MNHN, Paris, CP 54, 57 rue Cuvier 75005, France
| | - Séverine Zirah
- Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Laboratory of Communication Molecules and Adaptation of Micro-organisms, UMR 7245 CNRS-MNHN, Paris, CP 54, 57 rue Cuvier 75005, France
| | - Sylvie Rebuffat
- Muséum National d'Histoire Naturelle, Centre National de la Recherche Scientifique, Laboratory of Communication Molecules and Adaptation of Micro-organisms, UMR 7245 CNRS-MNHN, Paris, CP 54, 57 rue Cuvier 75005, France
| | - Moussa Sory Diarra
- Guelph Research and Development Center, Agriculture and Agri-Food Canada, 93 Stone Road West, Guelph, Ontario, N1G 5C9, Canada
| | - Lawrence Goodridge
- Department of Food Science and Agriculture, McGill University, Ste Anne de Bellevue, Québec, Quebec, H9X3V9, Canada
| | - Roger C Levesque
- Institute of Integrative Biology and Systems, Université Laval, QC, Québec, G1V 0A6, Canada
| | - Ismail Fliss
- Institute of Nutrition and Functional Foods, Université Laval, Québec, Quebec, G1V 0A6, Canada
| |
Collapse
|
49
|
Tai HM, Huang HN, Tsai TY, You MF, Wu HY, Rajanbabu V, Chang HY, Pan CY, Chen JY. Dietary supplementation of recombinant antimicrobial peptide Epinephelus lanceolatus piscidin improves growth performance and immune response in Gallus gallus domesticus. PLoS One 2020; 15:e0230021. [PMID: 32160226 PMCID: PMC7065771 DOI: 10.1371/journal.pone.0230021] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Accepted: 02/18/2020] [Indexed: 12/24/2022] Open
Abstract
Supplementing chicken feed with antibiotics can improve survival and prevent disease outbreaks. However, overuse of antibiotics may promote the development of antibiotic-resistant bacteria. Recently, antimicrobial peptides have been proposed as alternatives to antibiotics in animal husbandry. Here, we evaluate the effects of antimicrobial peptide, Epinephelus lanceolatus piscidin (EP), in Gallus gallus domesticus. The gene encoding EP was isolated, sequenced, codon-optimized and cloned into a Pichia pastoris recombinant protein expression system. The expressed recombinant EP (rEP) was then used as a dietary supplement for G. g. domesticus; overall health, growth performance and immunity were assessed. Supernatant from rEP-expressing yeast showed in vitro antimicrobial activity against Gram-positive and Gram-negative bacteria, according to an inhibition-zone diameter (mm) assay. Moreover, the antimicrobial peptide function of rEP was temperature independent. The fermentation broth yielded a spray-dried powder formulation containing 262.9 μg EP/g powder, and LC-MS/MS (tandem MS) analysis confirmed that rEP had a molecular weight of 4279 Da, as expected for the 34-amino acid peptide; the DNA sequence of the expression vector was also validated. We then evaluated rEP as a feed additive for G. g. domesticus. Treatment groups included control, basal diet and rEP at different doses (0.75, 1.5, 3.0, 6.0 and 12%). Compared to control, rEP supplementation increased G. g. domesticus weight gain, feed efficiency, IL-10 and IFN-γ production. Our results suggest that crude rEP could provide an alternative to traditional antibiotic feed additives for G. g. domesticus, serving to enhance growth and health of the animals.
Collapse
Affiliation(s)
- Hsueh-Ming Tai
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Ilan, Taiwan
| | - Han-Ning Huang
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Ilan, Taiwan
| | - Tsung-Yu Tsai
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Ilan, Taiwan
| | - Ming-Feng You
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Ilan, Taiwan
| | - Hung-Yi Wu
- Department of Veterinary Medicine, College of Veterinary Medicine, National Pingtung University of Science and Technology, Neipu, Taiwan
| | - Venugopal Rajanbabu
- Anbil Dharmalingam Agricultural College and Research Institute, Tamil Nadu Agricultural university, Tiruchchirapalli, Tamil Nadu, India
| | - Hsiao-Yun Chang
- Biotechnology Department, Asia University, Wufeng, Taichung, Taiwan
| | - Chieh-Yu Pan
- Department and Graduate Institute of Aquaculture, National Kaohsiung University of Science and Technology, Kaohsiung, Taiwan
| | - Jyh-Yih Chen
- Marine Research Station, Institute of Cellular and Organismic Biology, Academia Sinica, Ilan, Taiwan
- The iEGG and Animal Biotechnology Center, National Chung Hsing University, Taichung, Taiwan
| |
Collapse
|
50
|
Zhou Z, Chen X, Sheng H, Shen X, Sun X, Yan Y, Wang J, Yuan Q. Engineering probiotics as living diagnostics and therapeutics for improving human health. Microb Cell Fact 2020; 19:56. [PMID: 32131831 PMCID: PMC7055047 DOI: 10.1186/s12934-020-01318-z] [Citation(s) in RCA: 79] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2019] [Accepted: 02/26/2020] [Indexed: 02/08/2023] Open
Abstract
The gut microbiota that inhabit our gastrointestinal tract are well known to play an important role in maintaining human health in many aspects, including facilitating the digestion and absorption of nutrients, protecting against pathogens and regulating immune system. Gut microbiota dysbiosis is associated with a lot of diseases, such as inflammatory bowel disease, allergy, obesity, cardiovascular and neurodegenerative diseases and cancers. With the increasing knowledge of the microbiome, utilization of probiotic bacteria in modulating gut microbiota to prevent and treat a large number of disorders and diseases has gained much interest. In recent years, aided by the continuous development of tools and techniques, engineering probiotic microbes with desired characteristics and functionalities to benefit human health has made significant progress. In this paper, we summarize the recent advances in design and construction of probiotics as living diagnostics and therapeutics for probing and treating a series of diseases including metabolic disorders, inflammation and pathogenic bacteria infections. We also discuss the current challenges and future perspectives in expanding the application of probiotics for disease treatment and detection. We intend to provide insights and ideas for engineering of probiotics to better serve disease therapy and human health.
Collapse
Affiliation(s)
- Zhao Zhou
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15# Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Xin Chen
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15# Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Huakang Sheng
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15# Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Xiaolin Shen
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15# Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Xinxiao Sun
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15# Beisanhuan East Road, Chaoyang District, Beijing, 100029, China
| | - Yajun Yan
- College of Engineering, The University of Georgia, Athens, GA, 30602, USA
| | - Jia Wang
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15# Beisanhuan East Road, Chaoyang District, Beijing, 100029, China.
| | - Qipeng Yuan
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, 15# Beisanhuan East Road, Chaoyang District, Beijing, 100029, China.
| |
Collapse
|