1
|
Chen M, Xia L, Wu C, Wang Z, Ding L, Xie Y, Feng W, Chen Y. Microbe-material hybrids for therapeutic applications. Chem Soc Rev 2024; 53:8306-8378. [PMID: 39005165 DOI: 10.1039/d3cs00655g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
As natural living substances, microorganisms have emerged as useful resources in medicine for creating microbe-material hybrids ranging from nano to macro dimensions. The engineering of microbe-involved nanomedicine capitalizes on the distinctive physiological attributes of microbes, particularly their intrinsic "living" properties such as hypoxia tendency and oxygen production capabilities. Exploiting these remarkable characteristics in combination with other functional materials or molecules enables synergistic enhancements that hold tremendous promise for improved drug delivery, site-specific therapy, and enhanced monitoring of treatment outcomes, presenting substantial opportunities for amplifying the efficacy of disease treatments. This comprehensive review outlines the microorganisms and microbial derivatives used in biomedicine and their specific advantages for therapeutic application. In addition, we delineate the fundamental strategies and mechanisms employed for constructing microbe-material hybrids. The diverse biomedical applications of the constructed microbe-material hybrids, encompassing bioimaging, anti-tumor, anti-bacteria, anti-inflammation and other diseases therapy are exhaustively illustrated. We also discuss the current challenges and prospects associated with the clinical translation of microbe-material hybrid platforms. Therefore, the unique versatility and potential exhibited by microbe-material hybrids position them as promising candidates for the development of next-generation nanomedicine and biomaterials with unique theranostic properties and functionalities.
Collapse
Affiliation(s)
- Meng Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
- School of Medicine, Shanghai University, Shanghai 200444, P. R. China.
| | - Lili Xia
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Chenyao Wu
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Zeyu Wang
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Li Ding
- Department of Medical Ultrasound, National Clinical Research Center of Interventional Medicine, Shanghai Tenth People's Hospital, Tongji University Cancer Center, Tongji University School of Medicine, Tongji University, Shanghai, 200072, P. R. China.
| | - Yujie Xie
- School of Medicine, Shanghai University, Shanghai 200444, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai 200444, P. R. China.
- Shanghai Institute of Materdicine, Shanghai 200051, P. R. China
| |
Collapse
|
2
|
Kalam N, Balasubramaniam VRMT. Crosstalk between COVID-19 and the gut-brain axis: a gut feeling. Postgrad Med J 2024; 100:539-554. [PMID: 38493312 DOI: 10.1093/postmj/qgae030] [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: 12/10/2023] [Accepted: 02/15/2024] [Indexed: 03/18/2024]
Abstract
The microbes in the gut are crucial for maintaining the body's immune system and overall gut health. However, it is not fully understood how an unstable gut environment can lead to more severe cases of SARS-CoV-2 infection. The gut microbiota also plays a role in the gut-brain axis and interacts with the central nervous system through metabolic and neuroendocrine pathways. The interaction between the microbiota and the host's body involves hormonal, immune, and neural pathways, and any disruption in the balance of gut bacteria can lead to dysbiosis, which contributes to pathogen growth. In this context, we discuss how dysbiosis could contribute to comorbidities that increase susceptibility to SARS-CoV-2. Probiotics and fecal microbiota transplantation have successfully treated infectious and non-infectious inflammatory-related diseases, the most common comorbidities. These treatments could be adjuvant therapies for COVID-19 infection by restoring gut homeostasis and balancing the gut microbiota.
Collapse
Affiliation(s)
- Nida Kalam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Malaysia
| | - Vinod R M T Balasubramaniam
- Infection and Immunity Research Strength, Jeffrey Cheah School of Medicine & Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500 Bandar Sunway, Malaysia
| |
Collapse
|
3
|
D'Accolti M, Soffritti I, Bini F, Mazziga E, Caselli E. Tackling transmission of infectious diseases: A probiotic-based system as a remedy for the spread of pathogenic and resistant microbes. Microb Biotechnol 2024; 17:e14529. [PMID: 39045894 PMCID: PMC11267305 DOI: 10.1111/1751-7915.14529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 07/04/2024] [Indexed: 07/25/2024] Open
Abstract
Built environments (BEs) currently represent the areas in which human beings spend most of their life. Consistently, microbes populating BEs mostly derive from human occupants and can be easily transferred from BE to occupants. The hospital microbiome is a paradigmatic example, representing a reservoir for harmful pathogens that can be transmitted to susceptible patients, causing the healthcare-associated infections (HAIs). Environmental cleaning is a crucial pillar in controlling BE pathogens and preventing related infections, and chemical disinfectants have been largely used so far towards this aim. However, despite their immediate effect, chemical-based disinfection is unable to prevent recontamination, has a high environmental impact, and can select/increase antimicrobial resistance (AMR) in treated microbes. To overcome these limitations, probiotic-based sanitation (PBS) strategies were recently proposed, built on the use of detergents added with selected probiotics able to displace surrounding pathogens by competitive exclusion. PBS was reported as an effective and low-impact alternative to chemical disinfection, providing stable rebalance of the BE microbiome and significantly reducing pathogens and HAIs compared to disinfectants, without exacerbating AMR and pollution concerns. This minireview summarizes the most significant results obtained by applying PBS in sanitary and non-sanitary settings, which overall suggest that PBS may effectively tackle the infectious risk meanwhile preventing the further spread of pathogenic and resistant microbes.
Collapse
Affiliation(s)
- Maria D'Accolti
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences, and LTTAUniversity of FerraraFerraraItaly
- CIAS Research CenterUniversity of FerraraFerraraItaly
| | - Irene Soffritti
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences, and LTTAUniversity of FerraraFerraraItaly
- CIAS Research CenterUniversity of FerraraFerraraItaly
| | - Francesca Bini
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences, and LTTAUniversity of FerraraFerraraItaly
- CIAS Research CenterUniversity of FerraraFerraraItaly
| | - Eleonora Mazziga
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences, and LTTAUniversity of FerraraFerraraItaly
- CIAS Research CenterUniversity of FerraraFerraraItaly
| | - Elisabetta Caselli
- Section of Microbiology, Department of Chemical, Pharmaceutical and Agricultural Sciences, and LTTAUniversity of FerraraFerraraItaly
- CIAS Research CenterUniversity of FerraraFerraraItaly
| |
Collapse
|
4
|
Petrariu OA, Barbu IC, Niculescu AG, Constantin M, Grigore GA, Cristian RE, Mihaescu G, Vrancianu CO. Role of probiotics in managing various human diseases, from oral pathology to cancer and gastrointestinal diseases. Front Microbiol 2024; 14:1296447. [PMID: 38249451 PMCID: PMC10797027 DOI: 10.3389/fmicb.2023.1296447] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/18/2023] [Indexed: 01/23/2024] Open
Abstract
The imbalance of microbial composition and diversity in favor of pathogenic microorganisms combined with a loss of beneficial gut microbiota taxa results from factors such as age, diet, antimicrobial administration for different infections, other underlying medical conditions, etc. Probiotics are known for their capacity to improve health by stimulating the indigenous gut microbiota, enhancing host immunity resistance to infection, helping digestion, and carrying out various other functions. Concurrently, the metabolites produced by these microorganisms, termed postbiotics, which include compounds like bacteriocins, lactic acid, and hydrogen peroxide, contribute to inhibiting a wide range of pathogenic bacteria. This review presents an update on using probiotics in managing and treating various human diseases, including complications that may emerge during or after a COVID-19 infection.
Collapse
Affiliation(s)
- Oana-Alina Petrariu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
| | - Ilda Czobor Barbu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
| | - Adelina-Gabriela Niculescu
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, Bucharest, Romania
| | - Marian Constantin
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Institute of Biology of Romanian Academy, Bucharest, Romania
| | - Georgiana Alexandra Grigore
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- Academy of Romanian Scientists, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| | - Roxana-Elena Cristian
- The Research Institute of the University of Bucharest, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Grigore Mihaescu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
| | - Corneliu Ovidiu Vrancianu
- Microbiology-Immunology Department, Faculty of Biology, University of Bucharest, Bucharest, Romania
- The Research Institute of the University of Bucharest, Bucharest, Romania
- National Institute of Research and Development for Biological Sciences, Bucharest, Romania
| |
Collapse
|
5
|
Lv C, Yang J, Zhao L, Zou Z, Kang C, Zhang Q, Wu C, Yang L, Cheng C, Zhao Y, Liao Q, Hu X, Li C, Sun X, Jin M. Bacillus subtilis partially inhibits African swine fever virus infection in vivo and in vitro based on its metabolites arctiin and genistein interfering with the function of viral topoisomerase II. J Virol 2023; 97:e0071923. [PMID: 37929962 PMCID: PMC10688316 DOI: 10.1128/jvi.00719-23] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/21/2023] [Indexed: 11/07/2023] Open
Abstract
IMPORTANCE African swine fever virus (ASFV) is a highly fatal swine disease that severely affects the pig industry. Although ASFV has been prevalent for more than 100 years, effective vaccines or antiviral strategies are still lacking. In this study, we identified four Bacillus subtilis strains that inhibited ASFV proliferation in vitro. Pigs fed with liquid biologics or powders derived from four B. subtilis strains mixed with pellet feed showed reduced morbidity and mortality when challenged with ASFV. Further analysis showed that the antiviral activity of B. subtilis was based on its metabolites arctiin and genistein interfering with the function of viral topoisomerase II. Our findings offer a promising new strategy for the prevention and control of ASFV that may significantly alleviate the economic losses in the pig industry.
Collapse
Affiliation(s)
- Changjie Lv
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Jingyu Yang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, China
| | - Li Zhao
- State Key Laboratory of Biocatalysis and Enzyme Engineering, College of Life Sciences, Hubei University, Wuhan, China
| | - Zhong Zou
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Chao Kang
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Qiang Zhang
- College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Chao Wu
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Li Yang
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Chuxing Cheng
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Ya Zhao
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Qi Liao
- Research Institute of Wuhan Keqian Biology Co., Ltd, Wuhan, China
| | - Xiaotong Hu
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Chengfei Li
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Xiaomei Sun
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| | - Meilin Jin
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
6
|
Ren J, Yu D, Li N, Liu S, Xu H, Li J, He F, Zou L, Cao Z, Wen J. Biological Characterization and Whole-Genome Analysis of Bacillus subtilis MG-1 Isolated from Mink Fecal Samples. Microorganisms 2023; 11:2821. [PMID: 38137965 PMCID: PMC10745379 DOI: 10.3390/microorganisms11122821] [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: 10/17/2023] [Revised: 11/12/2023] [Accepted: 11/15/2023] [Indexed: 12/24/2023] Open
Abstract
Bacillus subtilis is an important part of the gut microbiota and a commonly used probiotic. In the present study, to assess the biological characteristics and probiotic properties of B. subtilis derived from mink, we isolated B. subtilis MG-1 isolate from mink fecal samples, characterized its biological characteristics, optimized the hydrolysis of casein by its crude extract, and comprehensively analyzed its potential as a probiotic in combination with whole-genome sequencing. Biological characteristics indicate that, under low-pH conditions (pH 2), B. subtilis MG-1 can still maintain a survival rate of 64.75%; under the conditions of intestinal fluid, gastric acid, and a temperature of 70 °C, the survival rate was increased by 3, 1.15 and 1.17 times compared with the control group, respectively. This shows that it can tolerate severe environments. The results of hydrolyzed casein in vitro showed that the crude bacterial extract of isolate MG-1 exhibited casein hydrolyzing activity (21.56 U/mL); the enzyme activity increased to 32.04 U/mL under optimized reaction conditions. The complete genome sequencing of B. subtilis MG-1 was performed using the PacBio third-generation sequencing platform. Gene annotation analysis results revealed that B. subtilis MG-1 was enriched in several Kyoto Encyclopedia of Genes and Genomes (KEGG) metabolic pathways, and most genes were related to Brite hierarchy pathways (1485-35.31%) and metabolism pathways (1395-33.17%). The egg-NOG annotation revealed that most genes were related to energy production and conversion (185-4.10%), amino acid transport and metabolism (288-6.38%), carbohydrate transport and metabolism (269-5.96%), transcription (294-6.52%), and cell wall/membrane/envelope biogenesis (231-5.12%). Gene Ontology (GO) annotation elucidated that most genes were related to biological processes (8230-45.62%), cellular processes (3582-19.86%), and molecular processes (6228-34.52%). Moreover, the genome of B. subtilis MG-1 was predicted to possess 77 transporter-related genes. This study demonstrates that B. subtilis MG-1 has potential for use as a probiotic, and further studies should be performed to develop it as a probiotic additive in animal feed to promote animal health.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Jianxin Wen
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, China; (J.R.); (D.Y.); (N.L.); (S.L.); (H.X.); (J.L.); (F.H.); (L.Z.); (Z.C.)
| |
Collapse
|
7
|
Wang X, Hao G, Zhou M, Chen M, Ling H, Shang Y. Secondary metabolites of Bacillus subtilis L2 show antiviral activity against pseudorabies virus. Front Microbiol 2023; 14:1277782. [PMID: 37965547 PMCID: PMC10642297 DOI: 10.3389/fmicb.2023.1277782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 10/13/2023] [Indexed: 11/16/2023] Open
Abstract
Bacillus subtilis (B. subtilis) is a commercially important probiotic known to produce secondary metabolites with antibacterial, antifungal and anti-inflammatory activities. However, the potential ability of B. subtilis to combat viruses, especially DNA viruses, has not been extensively investigated. In this study, we identified two distinct B. subtilis strains and examined the efficiency of their secondary metabolites against pseudorabies virus (PRV), a swine herpesvirus resulting in economic losses worldwide. We found that treatment with the secondary metabolites of B. subtilis L2, but not the metabolites of B. subtilis V11, significantly inhibited PRV replication in multiple cells. Notably, the antiviral activity of the metabolites of B. subtilis L2 was thermal stable, resistant to protease digestion. Moreover, these metabolites effectively impeded PRV binding, entry and replication. Importantly, oral administration of the metabolites of B. subtilis L2 protected mice from lethal PRV infection, rescuing weight loss and reducing the viral load in vivo. In summary, our results reveal that the metabolites of B. subtilis L2 exhibit anti-PRV activity both in vitro and in vivo, providing a potential candidate for novel antiviral drugs.
Collapse
Affiliation(s)
- Xiaoli Wang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | - Guijuan Hao
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | - Meng Zhou
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | - Meng Chen
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China
| | | | - Yingli Shang
- Department of Preventive Veterinary Medicine, College of Veterinary Medicine, Shandong Agricultural University, Taian, China
- Shandong Provincial Key Laboratory of Animal Biotechnology and Disease Control and Prevention, Shandong Agricultural University, Taian, China
- Institute of Immunology, Shandong Agricultural University, Taian, China
| |
Collapse
|
8
|
Chathuranga K, Shin Y, Uddin MB, Paek J, Chathuranga WAG, Seong Y, Bai L, Kim H, Shin JH, Chang YH, Lee JS. The novel immunobiotic Clostridium butyricum S-45-5 displays broad-spectrum antiviral activity in vitro and in vivo by inducing immune modulation. Front Immunol 2023; 14:1242183. [PMID: 37881429 PMCID: PMC10595006 DOI: 10.3389/fimmu.2023.1242183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2023] [Accepted: 09/20/2023] [Indexed: 10/27/2023] Open
Abstract
Clostridium butyricum is known as a probiotic butyric acid bacterium that can improve the intestinal environment. In this study, we isolated a new strain of C. butyricum from infant feces and evaluated its physiological characteristics and antiviral efficacy by modulating the innate immune responses in vitro and in vivo. The isolated C. butyricum S-45-5 showed typical characteristics of C. butyricum including bile acid resistance, antibacterial ability, and growth promotion of various lactic acid bacteria. As an antiviral effect, C. butyricum S-45-5 markedly reduced the replication of influenza A virus (PR8), Newcastle Disease Virus (NDV), and Herpes Simplex Virus (HSV) in RAW264.7 cells in vitro. This suppression can be explained by the induction of antiviral state in cells by the induction of antiviral, IFN-related genes and secretion of IFNs and pro-inflammatory cytokines. In vivo, oral administration of C. butyricum S-45-5 exhibited prophylactic effects on BALB/c mice against fatal doses of highly pathogenic mouse-adapted influenza A subtypes (H1N1, H3N2, and H9N2). Before challenge with influenza virus, C. butyricum S-45-5-treated BALB/c mice showed increased levels of IFN-β, IFN-γ, IL-6, and IL-12 in serum, the small intestine, and bronchoalveolar lavage fluid (BALF), which correlated with observed prophylactic effects. Interestingly, after challenge with influenza virus, C. butyricum S-45-5-treated BALB/c mice showed reduced levels of pro-inflammatory cytokines and relatively higher levels of anti-inflammatory cytokines at day 7 post-infection. Taken together, these findings suggest that C. butyricum S-45-5 plays an antiviral role in vitro and in vivo by inducing an antiviral state and affects immune modulation to alleviate local and systemic inflammatory responses caused by influenza virus infection. Our study provides the beneficial effects of the new C. butyricum S-45-5 with antiviral effects as a probiotic.
Collapse
Affiliation(s)
- Kiramage Chathuranga
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Yeseul Shin
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Md Bashir Uddin
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
- Department of Medicine, Sylhet Agricultural University, Sylhet, Bangladesh
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, United States
| | - Jayoung Paek
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | | | - Yebin Seong
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| | - Lu Bai
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Hongik Kim
- Research and Development Division, Vitabio, Inc., Daejeon, Republic of Korea
| | - Jeong Hwan Shin
- Department of Laboratory Medicine, Inje University College of Medicine, Busan, Republic of Korea
| | - Young-Hyo Chang
- Access to Genetic Resources and Benefit-Sharing (ABS) Research Support Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Jong-Soo Lee
- College of Veterinary Medicine, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
9
|
Chioma Mgbodile F, Nwagu TNT. Probiotic therapy, African fermented foods and food-derived bioactive peptides in the management of SARS-CoV-2 cases and other viral infections. BIOTECHNOLOGY REPORTS 2023; 38:e00795. [PMID: 37041970 PMCID: PMC10066861 DOI: 10.1016/j.btre.2023.e00795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 03/20/2023] [Accepted: 03/27/2023] [Indexed: 04/04/2023]
Abstract
The current paper focuses on the impact of probiotics, African fermented foods and bioactive peptides on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection severity and related viral infections. Using probiotics or bioactive peptides as therapeutic adjuncts appears superior to standard care alone. Probiotics play critical roles in innate and adaptive immune modulation by balancing the gut microbiota to combat viral infections, secondary bacterial infections and microbial dysbiosis. African fermented foods contain abundant potential probiotic microorganisms such as the lactic acid bacteria (LAB), Saccharomyces, and Bacillus. More so, fermented food-derived bioactive peptides play vital roles in preventing cardiovascular diseases, hypertension, lung injury, diabetes, and other COVID-19 comorbidities. Regularly incorporating potential probiotics and bioactive peptides into diets should enable a build-up of the benefits in the body system that may result in a better prognosis, especially in COVID-19 patients with underlying complexities. Despite the reported therapeutic potentials of probiotics and fermented foods, numerous setbacks exist regarding their application in disease management. These shortfalls underscore an evident need for more studies to evaluate the specific potentials of probiotics and traditional fermented foods in ameliorating SARS-CoV-2 and other viral infections.
Collapse
|
10
|
Lactic Acid Bacteria as Mucosal Immunity Enhancers and Antivirals through Oral Delivery. Appl Microbiol 2022. [DOI: 10.3390/applmicrobiol2040064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Mucosal vaccination offer an advantage over systemic inoculation from the immunological viewpoint. The development of an efficient vaccine is now a priority for emerging diseases such as COVID-19, that was declared a pandemic in 2020 and caused millions of deaths globally. Lactic acid bacteria (LAB) especially Lactobacillus are the vital microbiota of the gut, which is observed as having valuable effects on animals’ and human health. LAB produce lactic acid as the major by-product of carbohydrate degradation and play a significant role in innate immunity enhancement. LAB have significant characteristics to mimic pathogen infections and intrinsically possess adjuvant properties to enhance mucosal immunity. Increasing demand and deliberations are being substantially focused on probiotic organisms that can enhance mucosal immunity against viral diseases. LAB can also strengthen their host’s antiviral defense system by producing antiviral peptides, and releasing metabolites that prevent viral infections and adhesion to mucosal surfaces. From the perspectives of “one health” and the use of probiotics, conventional belief has opened up a new horizon on the use of LAB as antivirals. The major interest of this review is to depict the beneficial use of LAB as antivirals and mucosal immunity enhancers against viral diseases.
Collapse
|
11
|
Nasal-spraying Bacillus spores as an effective symptomatic treatment for children with acute respiratory syncytial virus infection. Sci Rep 2022; 12:12402. [PMID: 35858943 PMCID: PMC9297280 DOI: 10.1038/s41598-022-16136-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 07/05/2022] [Indexed: 01/30/2023] Open
Abstract
Respiratory syncytial virus (RSV) is a leading cause of Acute Respiratory Tract Infections (ARTIs) in young children. However, there is currently no vaccine or treatment available for children. Here, we demonstrated that nasal-spraying probiotics containing 5 billion of Bacillus spores (LiveSpo Navax) is an effective symptomatic treatment in a 6-day randomized controlled clinical study for RSV-infected children (n = 40–46/group). Navax treatment resulted in 1-day faster recovery-time and 10–50% better efficacy in relieving ARTI symptoms. At day 3, RSV load and level of pro-inflammatory cytokines in nasopharyngeal samples was reduced by 630 folds and 2.7–12.7 folds respectively. This showed 53-fold and 1.8–3.6-fold more effective than those in the control-standard of care-group. In summary, nasal-spraying Bacillus spores can rapidly and effectively relieve symptoms of RSV-induced ARTIs while exhibit strong impacts in reducing viral load and inflammation. Our nasal-spraying probiotics may provide a basis for simple-to-use, low-cost, and effective treatment against viral infection in general.
Collapse
|
12
|
Sasi M, Kumar S, Hasan M, S R A, Garcia-Gutierrez E, Kumari S, Prakash O, Nain L, Sachdev A, Dahuja A. Current trends in the development of soy-based foods containing probiotics and paving the path for soy-synbiotics. Crit Rev Food Sci Nutr 2022; 63:9995-10013. [PMID: 35611888 DOI: 10.1080/10408398.2022.2078272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the world of highly processed foods, special attention is drawn to the nutrient composition and safety of consumed food products. Foods fortified with probiotic bacteria confer beneficial effects on human health and are categorized as functional foods. The salubrious activities of probiotics include the synthesis of vital bioactives, prevention of inflammatory diseases, anticancerous, hypocholesterolemic, and antidiarrheal effects. Soy foods are exemplary delivery vehicles for probiotics and prebiotics and there are diverse strategies to enhance their functionality like employing mixed culture fermentation, engineering probiotics, and incorporating prebiotics in fermented soy foods. High potential is ascribed to the concurrent use of probiotics and prebiotics in one product, termed as "synbiotics," which implicates synergy, in which a prebiotic ingredient particularly favors the growth and activity of a probiotic micro-organism. The insights on emended bioactive profile, metabolic role, and potential health benefits of advanced soy-based probiotic and synbiotic hold a promise which can be profitably implemented to meet consumer needs. This article reviews the available knowledge about strategies to enhance the nutraceutical potential, mechanisms, and health-promoting effects of advanced soy-based probiotics. Traditional fermentation merged with diverse strategies to improve the efficiency and health benefits of probiotics considered vital, are also discussed.
Collapse
Affiliation(s)
- Minnu Sasi
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Sandeep Kumar
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
- Quality and Productivity Improvement Division, ICAR-Indian Institute of Natural Resins and Gums, Ranchi, India
| | - Muzaffar Hasan
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
- Agro Produce Processing Division, ICAR-Central Institute of Agricultural Engineering, Bhopal, India
| | - Arpitha S R
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | | | - Sweta Kumari
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Om Prakash
- National Centre for Microbial Resource (NCMR), National Centre for Cell Science, Pune, India
| | - Lata Nain
- Division of Microbiology, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Archana Sachdev
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| | - Anil Dahuja
- Division of Biochemistry, ICAR-Indian Agricultural Research Institute, New Delhi, India
| |
Collapse
|
13
|
Abstract
Fermented foods (FFs) hold global attention because of their huge advantages. Their health benefits, palatability, preserved, tasteful, and aromatic properties impart potential importance in the comprehensive evaluation of FFs. The bioactive components, such as minerals, vitamins, fatty acids, amino acids, and other phytochemicals synthesized during fermentation, provide consumers with several health benefits. Fermentation of food is an ancient process that has met with many remarkable changes owing to the development of scientific technologies over the years. Initially, fermentation relied on back-slapping. Nowadays, starter cultures strains are specifically chosen for the type of fermentation process. Modern biotechnological methods are being implemented in the fermentation process to achieve the desired product in high quality. Respiratory and gastrointestinal tract infections are the most severe health issues affecting human beings of all age groups, especially children and older adults, during this COVID-19 pandemic period. Studies suggest that the consumption of probiotic Lactobacillus strains containing fermented foods protects the subjects from common infectious diseases (CIDs, which is classified as upper respiratory tract infections, lower respiratory tract infections and gastrointestinal infections) by improving the host’s immune system. Further studies are obligatory to develop probiotic-based functional FFs that are effective against CIDs. Presently, we are urged to find alternative, safe, and cost-effective prevention measures against CIDs. The current manuscript briefs the production of FFs, functional properties of FFs, and their beneficial effects against respiratory tract infections. It summarizes the outcomes of clinical trials using human subjects on the effects of supplementation of FFs.
Collapse
|
14
|
Song L, Huang Y, Liu G, Li X, Xiao Y, Liu C, Zhang Y, Li J, Xu J, Lu S, Ren Z. A Novel Immunobiotics Bacteroides dorei Ameliorates Influenza Virus Infection in Mice. Front Immunol 2022; 12:828887. [PMID: 35154087 PMCID: PMC8826429 DOI: 10.3389/fimmu.2021.828887] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Accepted: 12/29/2021] [Indexed: 12/24/2022] Open
Abstract
Objective Probiotics can modulate immune responses to resist influenza infection. This study aims to evaluate the anti-viral efficacy of B. dorei. Methods C57BL/6J mice were infected with influenza virus together with treatment of PBS vehicle, B. dorei, or oseltamivir respectively. Anti-influenza potency of B. dorei and the underlying mechanism were determined by measuring survival rate, lung viral load and pathology, gene expression and production of cytokines and chemokines, and analysis of gut microbiota. Results Administration of B. dorei increased (by 30%) the survival of influenza-infected mice, and improved their weight loss, lung pathology, lung index, and colon length compared to the vehicle control group. B. dorei treatment reduced (by 61%) the viral load of lung tissue and increased expression of type 1 interferon more rapidly at day 3 postinfection. At day 7 postinfection, B. dorei-treated mice showed lower local (lung) and systemic (serum) levels of interferon and several proinflammatory cytokines or chemokines (IL-1β, IL-6, TNF-α, IL-10, MCP-1 and IP-10) with a efficacy comparable to oseltamivi treatment. B. dorei treatment also altered gut microbiota as indicated by increased levels of Bacteroides, Prevotella, and Lactobacillus and decreased levels of Escherichia, Shigella, and Parabacteroides. Conclusion B. dorei has anti-influenza effect. Its working mechanisms involve promoting earlier interferon expression and down-regulating both local and systemic inflammatory response. B. dorei changes the composition of gut microbiota, which may also contribute to its beneficial effects.
Collapse
Affiliation(s)
- Liqiong Song
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Yuanming Huang
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Guoxing Liu
- Respiratory Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China.,Traditional Chinese Medicine Department, Linwei Liu Zunji Clinic of Traditional Chinese Medicine, Weinan, China
| | - Xianping Li
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Yuchun Xiao
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Chang Liu
- Respiratory Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Yue Zhang
- Respiratory Department, Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, China
| | - Jintong Li
- Respiratory Department, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Jianguo Xu
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Shan Lu
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| | - Zhihong Ren
- State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Research Units of Discovery of Unknown Bacteria and Function (2018 RU010), Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
15
|
Akhmedov VA. Correction of intestinal microbial composition disturbances as a potential link in complex therapy of patients with COVID-19. TERAPEVT ARKH 2022; 94:277-282. [DOI: 10.26442/00403660.2022.02.201388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 03/29/2022] [Indexed: 12/23/2022]
Abstract
The article reflects the potential for correcting intestinal microbiota disorders in the complex therapy of patients with COVID-19. It has been noted that the inclusion of dietary fiber in the diet contributes to protection against disruption of the integrity of the intestinal barrier and may limit bacterial translocation into the systemic circulation. The possibility of using psyllium (Mucofalk) is reflected, the action of which is realized both through its sorption, cytoprotective and anti-inflammatory properties in viral lesions of the gastrointestinal tract, and through stimulation of the own beneficial intestinal microbiota. The paper presents studies of the prospects for the use of probiotics, synbiotics in the complex therapy of patients with COVID-19. Detailed data are provided on the mechanisms of the positive effect of short-chain fatty acid preparations on reducing the severity of the disease in patients with COVID-19. It was noted that taking the drug Zacofalk leads to a significant increase in its own butyrate-producing microbiota (Faecalibacterium prausnitzii) and suppression of the growth of opportunistic flora with pro-inflammatory activity. The results of a recent study are presented showing that in patients with a mild course of COVID infection with respiratory and intestinal symptoms, the administration of Zakofalk for 30 days (3 tablets per day) led to significantly faster stool normalization (by day 7), persistent normalization of the frequency and consistency of stools by the 21st day and a significantly more pronounced regression of bloating and abdominal pain, as well as a decrease in the risk of developing post-infectious irritable bowel syndrome.
Collapse
|
16
|
Khodavirdipour A, Chamanrokh P, Alikhani MY, Alikhani MS. Potential of Bacillus subtilis Against SARS-CoV-2 - A Sustainable Drug Development Perspective. Front Microbiol 2022; 13:718786. [PMID: 35222320 PMCID: PMC8874248 DOI: 10.3389/fmicb.2022.718786] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 01/17/2022] [Indexed: 12/11/2022] Open
Abstract
The COVID-19 pandemic had anomalous yet inevitable impacts on the world's economies, healthcare systems, and all other aspects of life. Researchers began to uncover hidden routes to find a new horizon of hope using underrated resources. Biosurfactants are sustainable biomolecules with an active surface, unique characteristics, and extensive uses. Bacillus species showed the highest amount of biosurfactant activities and Bacillus subtilis is one of them. The antiviral, antimicrobial, and anti-inflammatory activity of B. subtilis was proven recently. The great advantage is its non-toxic nature. Pro-inflammatory cytokines including IL-1 β, 6, 8, 12, 18, and TNF-(α are secreted in higher amounts when neutrophils and monocytes are triggered by biosurfactant bacteria. This point of view furnishes the potential application of B. subtilis and its biomolecules against COVID-19, either in the form of a vaccine/therapeutic agent, for a greener environment, healthier life, and environmental sustainability. Further in vivo and clinical trials are needed to validate this hypothesis.
Collapse
Affiliation(s)
| | - Parastoo Chamanrokh
- Dr. Rokh International Institute of Education and Health, Los Angeles, CA, United States
| | | | | |
Collapse
|
17
|
Yadav MK, Kumari I, Singh B, Sharma KK, Tiwari SK. Probiotics, prebiotics and synbiotics: Safe options for next-generation therapeutics. Appl Microbiol Biotechnol 2022; 106:505-521. [PMID: 35015145 PMCID: PMC8749913 DOI: 10.1007/s00253-021-11646-8] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Revised: 10/11/2021] [Accepted: 10/14/2021] [Indexed: 12/16/2022]
Abstract
Probiotics have been considered as an economical and safe alternative for the treatment of a large number of chronic diseases and improvement of human health. They are known to modulate the host immunity and protect from several infectious and non-infectious diseases. The colonization, killing of pathogens and induction of host cells are few of the important probiotic attributes which affect several functions of the host. In addition, prebiotics and non-digestible food substances selectively promote the growth of probiotics and human health through nutrient enrichment, and modulation of gut microbiota and immune system. This review highlights the role of probiotics and prebiotics alone and in combination (synbiotics) in the modulation of immune system, treatment of infections, management of inflammatory bowel disease and cancer therapy. KEY POINTS: • Probiotics and their derivatives against several human diseases. • Prebiotics feed probiotics and induce several functions in the host. • Discovery of novel and biosafe products needs attention for human health.
Collapse
Affiliation(s)
- Manoj Kumar Yadav
- Department of Genetics, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Indu Kumari
- Department of Genetics, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Bijender Singh
- Department of Microbiology, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
- Department of Biotechnology, Central University of Haryana, Jant-Pali 123031, Mahendragarh, Haryana, India
| | - Krishna Kant Sharma
- Department of Microbiology, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Santosh Kumar Tiwari
- Department of Genetics, Maharshi Dayanand University, Rohtak, 124001, Haryana, India.
| |
Collapse
|
18
|
Mironenko A, Holubka O, Radchenko L, Zakharchuk I, Teteriuk N. VIRULICIDAL EFFECT OF THE PROBIOTIC DRUG "SVITECO-MULTI" ON POLIOVIRUS TYPE 1 AND INFLUENZA VIRUS. BULLETIN OF TARAS SHEVCHENKO NATIONAL UNIVERSITY OF KYIV. SERIES: BIOLOGY 2022. [DOI: 10.17721/1728.2748.2022.91.24-27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The experimental work shows the antiviral activity of the probiotic drug "Sviteco-Multi" which contains bacteria of the genus Bacillusin model system in cell culturesMDCKandHEp-2, against influenza A (H1N1)pdm2009 virus and vaccine poliovirus type 1, which allows to recommend it for use, in particular, as an alternative to traditional antiviral disinfectants.
Collapse
|
19
|
Gouda AS, Adbelruhman FG, Sabbah Alenezi H, Mégarbane B. Theoretical benefits of yogurt-derived bioactive peptides and probiotics in COVID-19 patients - A narrative review and hypotheses. Saudi J Biol Sci 2021; 28:5897-5905. [PMID: 34177317 PMCID: PMC8213517 DOI: 10.1016/j.sjbs.2021.06.046] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/02/2021] [Accepted: 06/14/2021] [Indexed: 01/09/2023] Open
Abstract
The world is currently facing a frightening coronavirus disease-2019 (COVID-19) epidemic. Severity of COVID-19 presentation is highly variable among infected individuals with increasingly recognized risk factors. Although observational studies suggested lower COVID-19 severity in populations consuming fermented foods, no controlled study investigated the role of diet. Yogurt, a fermented dairy product, exhibits interesting properties related to the presence of bioactive peptides and probiotics that may play a beneficial role in COVID-19 presentation and outcome. Peptides contained in yogurt are responsible for angiotensin-converting enzyme-inhibitory, bradykinin potentiating, antiviral, anti-inflammatory, antithrombotic, and antioxidant effects. The types and activity of these peptides vary widely depending on their amino acid sequence, on the probiotics used in yogurt production and on intestinal digestion. Additionally, probiotics used in yogurt exhibit direct angiotensin-converting enzyme-inhibitory, antiviral and immune boosting activities. Since COVID-19 pathogenesis involves angiotensin II accumulation and bradykinin deficiency, yogurt bioactive peptides appear as potentially beneficial. Therefore, epidemiological investigations and randomized controlled clinical trials to evaluate the exact role of yogurt consumption on COVID-19 manifestations and outcome should be encouraged.
Collapse
Affiliation(s)
- Ahmed S. Gouda
- National Egyptian Center for Toxicological Researches, Faculty of Medicine, Cairo University, Cairo, Egypt,Poison Control and Forensic Chemistry Center, Northern Borders, Ministry of Health, Saudi Arabia
| | - Fatima G. Adbelruhman
- Department of Clinical Pathology, Faculty of Medicine, Al-Azhar University, Cairo, Egypt
| | - Hamedah Sabbah Alenezi
- Poison Control and Forensic Chemistry Center, Northern Borders, Ministry of Health, Saudi Arabia
| | - Bruno Mégarbane
- Department of Medical and Toxicological Critical Care, Lariboisière Hospital, University of Paris, INSERM UMRS-1144, Paris, France,Corresponding author at: Department of Medical and Toxicological Critical Care, Lariboisière Hospital, Paris University, Paris, France.
| |
Collapse
|
20
|
Cruz CS, Ricci MF, Vieira AT. Gut Microbiota Modulation as a Potential Target for the Treatment of Lung Infections. Front Pharmacol 2021; 12:724033. [PMID: 34557097 PMCID: PMC8453009 DOI: 10.3389/fphar.2021.724033] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
The gastrointestinal and respiratory systems are colonized by a complex ecosystem of microorganisms called the microbiota. These microorganisms co-evolved over millions of years with the host, creating a symbiotic relationship that is fundamental for promoting host homeostasis by producing bioactive metabolites and antimicrobial molecules, and regulating the immune and inflammatory responses. Imbalance in the abundance, diversity, and function of the gut microbiota (known as dysbiosis) have been shown to increase host susceptibility to infections in the lungs, suggesting crosstalk between these organs. This crosstalk is now referred to as the gut-lung axis. Hence, the use of probiotics, prebiotics, and synbiotics for modulation of gut microbiota has been studied based on their effectiveness in reducing the duration and severity of respiratory tract infections, mainly owing to their effects on preventing pathogen colonization and modulating the immune system. This review discusses the role and responses of probiotics, prebiotics, and synbiotics in the gut-lung axis in the face of lung infections.
Collapse
Affiliation(s)
- Clênio Silva Cruz
- Laboratory of Microbiota and Immunomodulation (LMI), Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Mayra Fernanda Ricci
- Laboratory of Microbiota and Immunomodulation (LMI), Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| | - Angélica Thomaz Vieira
- Laboratory of Microbiota and Immunomodulation (LMI), Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, Brazil
| |
Collapse
|
21
|
Colom J, Freitas D, Simon A, Brodkorb A, Buckley M, Deaton J, Winger AM. Presence and Germination of the Probiotic Bacillus subtilis DE111 ® in the Human Small Intestinal Tract: A Randomized, Crossover, Double-Blind, and Placebo-Controlled Study. Front Microbiol 2021; 12:715863. [PMID: 34408741 PMCID: PMC8366289 DOI: 10.3389/fmicb.2021.715863] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 07/12/2021] [Indexed: 12/04/2022] Open
Abstract
Spore-based probiotics offer important advantages over other probiotics as they can survive the harsh gastric conditions of the stomach and bile salts in the small intestine, ultimately germinating in the digestive tract. A novel clinical trial in 11 ileostomy participants was conducted to directly investigate the presence and germination of the probiotic strain Bacillus subtilis DE111® in the small intestine. Three hours following ingestion of DE111®, B. subtilis spores (6.4 × 104 ± 1.3 × 105 CFU/g effluent dry weight) and vegetative cells (4.7 × 104 ± 1.1 × 105 CFU/g effluent dry weight) began to appear in the ileum effluent. Six hours after ingestion, spore concentration increased to 9.7 × 107 ± 8.1 × 107 CFU/g and remained constant to the final time point of 8 h. Vegetative cells reached a concentration of 7.3 × 107 ± 1.4 × 108 CFU/g at 7 h following ingestion. These results reveal orally ingested B. subtilis DE111® spores are able to remain viable during transit through the stomach and germinate in the small intestine of humans within 3 h of ingestion.
Collapse
Affiliation(s)
- Joan Colom
- Deerland Probiotics and Enzymes, Food Science Building, University College Cork, Cork, Ireland
| | | | - Annie Simon
- Deerland Probiotics and Enzymes, Food Science Building, University College Cork, Cork, Ireland
| | - Andre Brodkorb
- Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | | | - John Deaton
- Deerland Probiotics and Enzymes, Kennesaw, GA, United States
| | - Alison M Winger
- Deerland Probiotics and Enzymes, Food Science Building, University College Cork, Cork, Ireland
| |
Collapse
|
22
|
The Interaction Between Viruses and Intestinal Microbiota: A Review. Curr Microbiol 2021; 78:3597-3608. [PMID: 34350485 PMCID: PMC8336530 DOI: 10.1007/s00284-021-02623-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 07/28/2021] [Indexed: 02/07/2023]
Abstract
As the main pathogen threatening human and animal health, viruses can affect the immunity and metabolism of bodies. There are innate microbial barriers in the digestive tract of the body to preserve the homeostasis of the animal body, which directly or indirectly influences the host defence against viral infection. Understanding the interaction between viruses and intestinal microbiota or probiotics is helpful to study the pathogenesis of diseases. Here, we review recent studies on the interaction mechanism between intestinal microbiota and viruses. The interaction can be divided into two aspects: inhibition of viral infection by microbiota and promotion of viral infection by microbiota. The treatment of viral infection by probiotics is summarized.
Collapse
|
23
|
Hung YP, Lee CC, Lee JC, Tsai PJ, Ko WC. Gut Dysbiosis during COVID-19 and Potential Effect of Probiotics. Microorganisms 2021; 9:1605. [PMID: 34442684 PMCID: PMC8402052 DOI: 10.3390/microorganisms9081605] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), an RNA virus of the family Coronaviridae, causes coronavirus disease 2019 (COVID-19), an influenza-like disease that chiefly infects the lungs through respiratory transmission. The spike protein of SARS-CoV-2, a transmembrane protein in its outer portion, targets angiotensin-converting enzyme 2 (ACE2) as the binding receptor for the cell entry. As ACE2 is highly expressed in the gut and pulmonary tissues, SARS-CoV-2 infections frequently result in gastrointestinal inflammation, with presentations ordinarily ranging from intestinal cramps to complications with intestinal perforations. However, the evidence detailing successful therapy for gastrointestinal involvement in COVID-19 patients is currently limited. A significant change in fecal microbiomes, namely dysbiosis, was characterized by the enrichment of opportunistic pathogens and the depletion of beneficial commensals and their crucial association to COVID-19 severity has been evidenced. Oral probiotics had been evidenced to improve gut health in achieving homeostasis by exhibiting their antiviral effects via the gut-lung axis. Although numerous commercial probiotics have been effective against coronavirus, their efficacies in treating COVID-19 patients remain debated. In ClinicalTrials.gov, 19 clinical trials regarding the dietary supplement of probiotics, in terms of Lactobacillus and mixtures of Bifidobacteria and Lactobacillus, for treating COVID-19 cases are ongoing. Accordingly, the preventive or therapeutic role of probiotics for COVID-19 patients can be elucidated in the near future.
Collapse
Affiliation(s)
- Yuan-Pin Hung
- Department of Internal Medicine, Tainan Hospital, Ministry of Health and Welfare, Tainan 700, Taiwan;
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
| | - Ching-Chi Lee
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
- Clinical Medicine Research Center, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 705, Taiwan
- Graduate Institute of Medical Sciences, College of Health Sciences, Chang Jung Christian University, Tainan 711, Taiwan
| | - Jen-Chieh Lee
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
| | - Pei-Jane Tsai
- Department of Medical Laboratory Science and Biotechnology, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan;
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan
- Department of Pathology, National Cheng Kung University Hospital, Tainan 705, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, College of Medicine, National Cheng Kung University Hospital, National Cheng Kung University, Tainan 704, Taiwan; (C.-C.L.); (J.-C.L.)
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan 705, Taiwan
| |
Collapse
|
24
|
Heidari Z, Tajbakhsh A, Gheibi-Hayat SM, Moattari A, Razban V, Berenjian A, Savardashtaki A, Negahdaripour M. Probiotics/ prebiotics in viral respiratory infections: implication for emerging pathogens. Recent Pat Biotechnol 2021; 15:112-136. [PMID: 33874878 DOI: 10.2174/1872208315666210419103742] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/01/2021] [Accepted: 03/10/2021] [Indexed: 01/08/2023]
Abstract
BACKGROUND Viral respiratory infections could result in perturbation of the gut microbiota due to a probable cross-talk between lungs and gut microbiota. This can affect the pulmonary health and the gastrointestinal system. OBJECTIVE This review aimed to discuss the impact of probiotics/ prebiotics and supplements on the prevention and treatment of respiratory infections, especially emerging pathogens. METHODS The data were searched were searched in PubMed, Scopus, Google Scholar, Google Patents, and The Lens-Patent using keywords of probiotics and viral respiratory infections in the title, abstract, and keywords. RESULT Probiotics consumption could decrease the susceptibility to viral respiratory infections, such as COVID-19 and simultaneously enhance vaccine efficiency in infectious disease prevention through the immune system enhancement. Probiotics improve the gut microbiota and the immune system via regulating the innate system response and production of anti-inflammatory cytokines. Moreover, treatment with probiotics contributes to the intestinal homeostasis restitution under antibiotic pressure and decreasing the risk of secondary infections due to viral respiratory infections. Probiotics present varied performances in different conditions; thus, promoting their efficacy through combining with supplements (prebiotics, postbiotics, nutraceuticals, berberine, curcumin, lactoferrin, minerals, and vitamins) is important. Several supplements reported to enhance the probiotics' efficacy and their mechanisms as well as probiotics related patents are summarized in this review. Using nanotechnology and microencapsulation techniques can also improve probiotics efficiency. CONCLUSION Given the global challenge of COVID-19, probiotic/prebiotic and following nutritional guidelines should be regarded seriously. Additionally, their role as an adjuvant in vaccination for immune response augmentation needs attention.
Collapse
Affiliation(s)
- Zahra Heidari
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz. Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz. Iran
| | - Seyed Mohammad Gheibi-Hayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd. Iran
| | - Afagh Moattari
- Department of Parasitology and Mycology, Shiraz University of Medical Sciences, Shiraz. Iran
| | - Vahid Razban
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz. Iran
| | - Aydin Berenjian
- School of Engineering, Faculty of Science and Engineering, The University of Waikato, Hamilton. New Zealand
| | - Amir Savardashtaki
- Epilepsy Research Center, Shiraz University of Medical Sciences, Shiraz. Iran
| | - Manica Negahdaripour
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz. Iran
| |
Collapse
|
25
|
Brink LR, Chichlowski M, Pastor N, Thimmasandra Narayanappa A, Shah N. In the Age of Viral Pandemic, Can Ingredients Inspired by Human Milk and Infant Nutrition Be Repurposed to Support the Immune System? Nutrients 2021; 13:870. [PMID: 33800961 PMCID: PMC7999376 DOI: 10.3390/nu13030870] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 12/14/2022] Open
Abstract
In 2020, with the advent of a pandemic touching all aspects of global life, there is a renewed interest in nutrition solutions to support the immune system. Infants are vulnerable to infection and breastfeeding has been demonstrated to provide protection. As such, human milk is a great model for sources of functional nutrition ingredients, which may play direct roles in protection against viral diseases. This review aims to summarize the literature around human milk (lactoferrin, milk fat globule membrane, osteopontin, glycerol monolaurate and human milk oligosaccharides) and infant nutrition (polyunsaturated fatty acids, probiotics and postbiotics) inspired ingredients for support against viral infections and the immune system more broadly. We believe that the application of these ingredients can span across all life stages and thus apply to both pediatric and adult nutrition. We highlight the opportunities for further research in this field to help provide tangible nutrition solutions to support one's immune system and fight against infections.
Collapse
Affiliation(s)
- Lauren R. Brink
- Medical and Scientific Affairs, Nutrition, Reckitt Benckiser, Evansville, IN 47721, USA; (M.C.); (N.P.)
| | - Maciej Chichlowski
- Medical and Scientific Affairs, Nutrition, Reckitt Benckiser, Evansville, IN 47721, USA; (M.C.); (N.P.)
| | - Nitida Pastor
- Medical and Scientific Affairs, Nutrition, Reckitt Benckiser, Evansville, IN 47721, USA; (M.C.); (N.P.)
| | | | - Neil Shah
- Medical and Scientific Affairs, Nutrition, Reckitt Benckiser, Slough SL1 3UH, UK;
- University College London, Great Ormond Street, London WC1N 3JH, UK
| |
Collapse
|
26
|
Singh K, Rao A. Probiotics: A potential immunomodulator in COVID-19 infection management. Nutr Res 2021; 87:1-12. [PMID: 33592454 PMCID: PMC7881295 DOI: 10.1016/j.nutres.2020.12.014] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 12/02/2020] [Accepted: 12/15/2020] [Indexed: 02/07/2023]
Abstract
COVID-19 caused by SARS-CoV-2 is an ongoing global pandemic. SARS-CoV-2 affects the human respiratory tract's epithelial cells, leading to a proinflammatory cytokine storm and chronic lung inflammation. With numerous patients dying daily, a vaccine and specific antiviral drug regimens are being explored. Probiotics are live microorganisms with proven beneficial effects on human health. While probiotics as nutritional supplements are long practiced in different cuisines across various countries, the emerging scientific evidence supports the antiviral and general immune-strengthening health effects of the probiotics. Here, we present an overview of the experimental studies published in the last 10 years that provide a scientific basis for unexplored probiotics as a preventive approach to respiratory viral infections. Based on collated insights from these experimental data, we identify promising microbial strains that may serve as lead prophylactic and immune-boosting probiotics in COVID-19 management.
Collapse
Affiliation(s)
- Kuljit Singh
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh 160036 India
| | - Alka Rao
- CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh 160036 India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad, Uttar Pradesh 201002 India.
| |
Collapse
|
27
|
Santhi Sudha S, Aranganathan V. Experimental elucidation of an antimycobacterial bacteriocin produced by ethnomedicinal plant-derived Bacillus subtilis (MK733983). Arch Microbiol 2021; 203:1995-2006. [PMID: 33544157 PMCID: PMC7863612 DOI: 10.1007/s00203-020-02173-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 11/24/2020] [Accepted: 12/27/2020] [Indexed: 11/30/2022]
Abstract
A bacteriocin from Bacillus subtilis (MK733983) originated from ethnomedicinal plant was purified using Preparative RP-HPLC. The HPLC fraction eluted with 65% acetonitrile showed the highest antimicrobial activity with Mycobacterium smegmatis as an indicator. Its specific activity and purification fold increased by 70.5% and 44%, respectively, compared to the crude bacteriocin. The bacteriocin showed stability over a wide range of pH (3.0-8.0) and preservation (- 20 °C and 4 °C), also thermal stability up to 80 °C for 20 min. Its proteinaceous nature was confirmed with complete loss of activity on its treatment with Trypsin, Proteinase K, and α-Chymotrypsin. Nevertheless, the bacteriocin retained up to 45% activity with Papainase treatment and was unaffected by salivary Amylase. It maintained ~ 95% activity on UV exposure up to 3 h and its activity was augmented by ethyl alcohol and metal ions like Fe2+ and Mn2+. Most of the common organic solvents, general surfactants, preservatives, and detergents like Sulfobetaine-14, Deoxy-cholic-acid did not affect the bacteriocin's action. Its molecular weight was estimated to be 3.4KDa by LC-ESI-MS/MS analysis. The bacteriocin is non-hemolytic and exhibited a broad inhibition spectrum with standard strains of Staphylococcus aureus, Pseudomonas aeruginosa, Klebsiella pneumoniae, Escherichia coli and Chromobacterium violaceum with MICs ranging 0.225 ± 0.02-0.55 ± 0.05 mg/mL. Scanning Electron Microscopy showed cell annihilation with pores in cell membranes of S. aureus and P. aeruginosa treated with the bacteriocin, implicating bactericidal mode of action. These promising results suggest that the bacteriocin is significant and has wide-ranging application prospects.
Collapse
Affiliation(s)
- S Santhi Sudha
- Department of Biochemistry, Jain (Deemed To-Be) University, 18/3, 3rd Block, 9th Main Rd, Jayanagar, Bangalore, Karnataka, 560011, India
| | - V Aranganathan
- Department of Biochemistry, Jain (Deemed To-Be) University, 18/3, 3rd Block, 9th Main Rd, Jayanagar, Bangalore, Karnataka, 560011, India.
| |
Collapse
|
28
|
Ran C, Li Y, Ma X, Xie Y, Xie M, Zhang Y, Zhou W, Yang Y, Zhang Z, Zhou L, Wei K, Zhou Z. Interactions between commensal bacteria and viral infection: insights for viral disease control in farmed animals. SCIENCE CHINA-LIFE SCIENCES 2021; 64:1437-1448. [PMID: 33420920 DOI: 10.1007/s11427-020-1721-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 09/30/2020] [Indexed: 12/11/2022]
Abstract
Viral diseases cause serious economic loss in farmed animals industry. However, the efficacy of remedies for viral infection in farmed animals is limited, and treatment strategies are generally lacking for aquatic animals. Interactions of commensal microbiota and viral infection have been studied in recent years, demonstrating a third player in the interaction between hosts and viruses. Here, we discuss recent developments in the research of interactions between commensal bacteria and viral infection, including both promotion and inhibition effect of commensal bacteria on viral pathogenesis, as well as the impact of viral infection on commensal microbiota. The antiviral effect of commensal bacteria is mostly achieved through priming or regulation of the host immune responses, involving differential microbial components and host signaling pathways, and gives rise to various antiviral probiotics. Moreover, we summarize studies related to the interaction between commensal bacteria and viral infection in farmed animals, including pigs, chickens, fish and invertebrate species. Further studies in this area will deepen our understanding of antiviral immunity of farmed animals in the context of commensal microbiota, and promote the development of novel strategies for treatment of viral diseases in farmed animals.
Collapse
Affiliation(s)
- Chao Ran
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Yu Li
- Sino-Norway Joint Lab on Fish Gut Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Xufa Ma
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China
| | - Yadong Xie
- Sino-Norway Joint Lab on Fish Gut Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Mingxu Xie
- Sino-Norway Joint Lab on Fish Gut Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Yuting Zhang
- Sino-Norway Joint Lab on Fish Gut Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Wei Zhou
- Sino-Norway Joint Lab on Fish Gut Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Yalin Yang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Zhen Zhang
- Key Laboratory for Feed Biotechnology of the Ministry of Agriculture and Rural Affairs, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China
| | - Li Zhou
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, The Innovation Academy of Seed Design, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Kaijian Wei
- College of Fisheries, Huazhong Agricultural University, Wuhan, 430070, China.
| | - Zhigang Zhou
- Sino-Norway Joint Lab on Fish Gut Microbiota, Feed Research Institute, Chinese Academy of Agricultural Sciences, Beijing, 100081, China.
| |
Collapse
|
29
|
Baindara P, Chakraborty R, Holliday Z, Mandal S, Schrum A. Oral probiotics in coronavirus disease 2019: connecting the gut-lung axis to viral pathogenesis, inflammation, secondary infection and clinical trials. New Microbes New Infect 2021; 40:100837. [PMID: 33425362 PMCID: PMC7785423 DOI: 10.1016/j.nmni.2021.100837] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 12/12/2020] [Accepted: 12/31/2020] [Indexed: 01/07/2023] Open
Abstract
Defined as helpful live bacteria that can provide medical advantages to the host when administered in tolerable amounts, oral probiotics might be worth considering as a possible preventive or therapeutic modality to mitigate coronavirus disease 2019 (COVID-19) symptom severity. This hypothesis stems from an emerging understanding of the gut-lung axis wherein probiotic microbial species in the digestive tract can influence systemic immunity, lung immunity, and possibly viral pathogenesis and secondary infection co-morbidities. We review the principles underlying the gut-lung axis, examples of probiotic-associated antiviral activities, and current clinical trials in COVID-19 based on oral probiotics.
Collapse
Affiliation(s)
- P. Baindara
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO, USA,Corresponding author: P. Baindara, Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO, USA.
| | - R. Chakraborty
- Department of Biotechnology, North Bengal University, Darjeeling, India
| | - Z.M. Holliday
- Pulmonary Disease, Critical Care Medicine, School of Medicine, University of Missouri, Columbia, MO, USA
| | - S.M. Mandal
- Central Research Facility, Indian Institute of Technology Kharagpur, Kharagpur, India,Corresponding author: S.M. Mandal, Central Research Facility, Indian Institute of Technology Kharagpur, Kharagpur, India.
| | - A.G. Schrum
- Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO, USA,Department of Surgery, School of Medicine, University of Missouri, Columbia, MO, USA,Department of Biomedical, Biological, & Chemical Engineering, College of Engineering, University of Missouri, Columbia, MO, USA,Corresponding author: A. Schrum, Department of Molecular Microbiology & Immunology, School of Medicine, University of Missouri, Columbia, MO, USA.
| |
Collapse
|
30
|
Hamida RS, Shami A, Ali MA, Almohawes ZN, Mohammed AE, Bin-Meferij MM. Kefir: A protective dietary supplementation against viral infection. Biomed Pharmacother 2021; 133:110974. [PMID: 33186795 PMCID: PMC7655491 DOI: 10.1016/j.biopha.2020.110974] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 10/26/2020] [Accepted: 11/01/2020] [Indexed: 12/14/2022] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an infectious disease caused by a recently discovered coronavirus termed 'severe acute respiratory syndrome coronavirus 2' (SARS-CoV-2). Several scholars have tested antiviral drugs and compounds to overcome COVID-19. 'Kefir' is a fermented milk drink similar to a thin yogurt that is made from kefir grains. Kefir and its probiotic contents can modulate the immune system to suppress infections from viruses (e.g., Zika, hepatitis C, influenza, rotaviruses). The antiviral mechanisms of kefir involve enhancement of macrophage production, increasing phagocytosis, boosting production of cluster of differentiation-positive (CD4+), CD8+, immunoglobulin (Ig)G+ and IgA+ B cells, T cells, neutrophils, as well as cytokines (e.g., interleukin (IL)-2, IL-12, interferon gamma-γ). Kefir can act as an anti-inflammatory agent by reducing expression of IL-6, IL-1, TNF-α, and interferon-γ. Hence, kefir might be a significant inhibitor of the 'cytokine storm' that contributes to COVID-19. Here, we review several studies with a particular emphasis on the effect of kefir consumption and their microbial composition against viral infection, as well as discussing the further development of kefir as a protective supplementary dietary against SARS-CoV-2 infection via modulating the immune response.
Collapse
Affiliation(s)
- Reham Samir Hamida
- Molecular Biology Unit, Department of Zoology, Faculty of Science, Alexandria University, Egypt.
| | - Ashwag Shami
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.
| | - Mohamed Abdelaal Ali
- Biotechnology Unit, Department of Plant Production, College of Food and Agriculture Science, King Saud University, Riyadh, Saudi Arabia.
| | - Zakiah Nasser Almohawes
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.
| | - Afrah E Mohammed
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, Saudi Arabia.
| | | |
Collapse
|
31
|
The Role of Mucosal Immunity and Recombinant Probiotics in SARS-CoV2 Vaccine Development. Probiotics Antimicrob Proteins 2021; 13:1239-1253. [PMID: 33770348 PMCID: PMC7996120 DOI: 10.1007/s12602-021-09773-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2021] [Indexed: 01/07/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV2), causing the 2019 novel coronavirus disease (COVID-19), was introduced by WHO (World Health Organization) as "pandemic" in March 2020. According to WHO, thus far (23 November 2020) 58,425,681 infected cases including 1,385,218 deaths have been reported worldwide. In order to reduce transmission and spread of this lethal virus, attempts are globally being made to develop an appropriate vaccine. Intending to neutralize pathogens at their initial entrance site, protective mucosal immunity is inevitably required. In SARS-CoV2 infection and transmission, respiratory mucosa plays a key role; hence, apparently mucosal vaccination could be a superior approach to elicit mucosal and systemic immune responses simultaneously. In this review, the advantages of mucosal vaccination to control COVID-19 infection, limitations, and outcomes of mucosal vaccines have been highlighted. Considering the gut microbiota dysregulation in COVID-19, we further provide evidences on utilization of recombinant probiotics, particularly lactic acid bacteria (LAB) as vaccine carrier. Their intrinsic immunomodulatory features, natural adjuvanticity, and feasible expression of relevant antigen in the mucosal surface make them more appealing as live cell factory. Among all available platforms, bioengineered probiotics are considered as the most affordable, most practical, and safest vaccination approach to halt this emerging virus.
Collapse
|
32
|
Salazar-Llorente E, Morales M, Sornoza I, Mariduena-Zavala MG, Gu G, Nou X, Ortiz J, Maldonado-Alvarado P, Cevallos-Cevallos JM. Microbiological Quality of High-Demand Food from Three Major Cities in Ecuador. J Food Prot 2021; 84:128-138. [PMID: 33411929 DOI: 10.4315/jfp-20-271] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Accepted: 09/07/2020] [Indexed: 11/11/2022]
Abstract
ABSTRACT Bacterial foodborne diseases are among the most important public health issues worldwide, but in Ecuador, reports on the microbiological quality of food are scarce. In this cross-sectional study, 450 samples of high-demand Ecuadorian food, including bolon, encebollado, sauces, ceviche, fruit, fruit juice, fruit salad, cheese, raw chicken, and ground beef, were collected from popular street markets in the cities of Guayaquil, Quito, and Cuenca. Populations of total aerobic mesophilic bacteria, total coliforms, fecal coliforms, Escherichia coli, Salmonella enterica, and Listeria monocytogenes were examined on composited samples by plate count following the local regulations (Norma Tecnica Ecuatoriana, Instituto Ecuatoriano de Normalización) for each kind of food. The individual and interaction effects of the city and food type on the levels of each bacterial group were assessed by two-way analysis of variance. Selected colonies from each culture were identified using Biolog OmniLog ID and sequencing of the V3 to V4 region on the 16S rRNA gene. Average total aerobic mesophilic bacteria, total coliform, fecal coliform, and E. coli levels were 5.10 ± 0.12, 2.50 ± 0.16, 1.09 ± 0.12, and 0.83 ± 0.12 log CFU/g or mL, respectively, with significant variations among the cities. The prevalence of Salmonella in chicken and sauces and L. monocytogenes in cheese and fruit salad was greater than 20%. Opportunistic pathogens including Klebsiella pneumoniae, Staphylococcus sciuri, and Enterococcus spp. were frequently identified in the samples from all three cities. High prevalence of spoilage microorganisms such as Bacillus amyloliquefaciens and biocontrol bacteria such as Lactococcus lactis was also observed. This is the first report on the microbiological quality of food from Ecuador. HIGHLIGHTS
Collapse
Affiliation(s)
- Enrique Salazar-Llorente
- Escuela Superior Politécnica del Litoral (ESPOL), Centro de Investigaciones Biotecnológicas del Ecuador
| | - Maria Morales
- ESPOL, Facultad de Ingeniería Mecánica y Ciencias de la Produccion, U.S. Department of Agriculture Agricultural Research Service, Beltsville, Maryland, USA
| | - Ivette Sornoza
- ESPOL, Facultad de Ingeniería Mecánica y Ciencias de la Produccion, U.S. Department of Agriculture Agricultural Research Service, Beltsville, Maryland, USA
| | | | - Ganyu Gu
- Environmental Microbiology and Food Safety Laboratory, U.S. Department of Agriculture Agricultural Research Service, Beltsville, Maryland, USA
| | - Xiangwu Nou
- Environmental Microbiology and Food Safety Laboratory, U.S. Department of Agriculture Agricultural Research Service, Beltsville, Maryland, USA
| | - Johana Ortiz
- Department of Biosciences, Food Nutrition and Health Research Unit, Faculty of Chemical Sciences, Cuenca University, Cuenca, Ecuador
| | - Pedro Maldonado-Alvarado
- Escuela Politécnica Nacional, Departamento de Alimentos y Biotecnología, P.O. Box 17-01-2759, Quito, Ecuador
| | - Juan Manuel Cevallos-Cevallos
- Escuela Superior Politécnica del Litoral (ESPOL), Centro de Investigaciones Biotecnológicas del Ecuador.,(ORCID: https://orcid.org/0000-0003-4609-7998 [J.M.C.C.]).,ESPOL, Facultad de Ciencias de la Vida, Campus Gustavo Galindo, Km. 30.5 Vía Perimetral, P.O. Box 09-01-5863, Guayaquil, Ecuador
| |
Collapse
|
33
|
Bacillus subtilis Inhibits Viral Hemorrhagic Septicemia Virus Infection in Olive Flounder ( Paralichthys olivaceus) Intestinal Epithelial Cells. Viruses 2020; 13:v13010028. [PMID: 33375689 PMCID: PMC7823535 DOI: 10.3390/v13010028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 12/15/2020] [Accepted: 12/22/2020] [Indexed: 12/13/2022] Open
Abstract
Viral hemorrhagic septicemia virus (VHSV) is a highly pathogenic virus that infects a wide range of host fish species causing high economic losses in aquaculture. Epithelial cells in mucosal organs are target sites for VHSV entry into fish. To protect fish against VHSV infection, there is a need to develop antiviral compounds able to prevent establishment of infection at portals of virus entry into fish. Bacillus subtilis is a probiotic with excellent antiviral properties, of which one of its secretions, surfactin, has been shown to inhibit viral infections in mammals. Herein, we demonstrate its ability to prevent VHSV infection in olive flounder (Paralichthys olivaceus) intestinal epithelial cells (IECs) and infection in internal organs. Our findings show inhibition of VHSV infection in IECs by B. subtilis and surfactin. In addition, our findings showed inhibition of VHSV in Epithelioma Papulosum Cyprini (EPC) cells inoculated with intestinal homogenates from the fish pretreated with B. subtilis by oral exposure, while the untreated fish had cytopathic effects (CPE) caused by VHSV infection in the intestines at 48 h after the VHSV challenge. At 96 h post-challenge, samples from the untreated fish had CPE from head kidney and spleen homogenates and no CPE were observed in the intestinal homogenates, while the B. subtilis-pretreated fish had no CPE in all organs. These findings demonstrate that inhibition of VHSV infection at portals of virus entry in the intestines culminated in prevention of infection in internal organs. In summary, our results show that B. subtilis has the potential to prevent VHSV infection in fish and that its use as a probiotic in aquaculture has the potential to serve as an antiviral therapeutic agent against different viral infections.
Collapse
|
34
|
Donati Zeppa S, Agostini D, Piccoli G, Stocchi V, Sestili P. Gut Microbiota Status in COVID-19: An Unrecognized Player? Front Cell Infect Microbiol 2020; 10:576551. [PMID: 33324572 PMCID: PMC7725702 DOI: 10.3389/fcimb.2020.576551] [Citation(s) in RCA: 71] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Accepted: 10/30/2020] [Indexed: 01/07/2023] Open
Abstract
Infection with the SARS-CoV-2 virus causes cardiopulmonary and vascular complications, ranging in severity. Understanding the pathogenic mechanisms of the novel SARS-CoV2 infection and progression can provide potential novel targets for its prevention and/or treatment. Virus microbiota reciprocal interactions have been studied in a variety of viral infections. For example, the integrity of Coronavirus particles can be disrupted by surfactin, a bacterial surface molecule that targets other viruses, including that of influenza A. In this light, intestinal microbiota likely influences COVID-19 virulence, while from its side SARS-CoV-2 may affect the intestinal microbiome promoting dysbiosis and other deleterious consequences. Hence, the microbiota pre-existing health status and its alterations in the course of SARS-CoV-2 infection, are likely to play an important, still underscored role in determining individual susceptibility and resilience to COVID-19. Indeed, the vast majority of COVID-19 worst clinical conditions and fatalities develop in subjects with specific risk factors such as aging and the presence of one or more comorbidities, which are intriguingly characterized also by unhealthy microbiome status. Moreover, these comorbidities require complex pharmacological regimens known as "polypharmacy" that may further affect microbiota integrity and worsen the resilience to viral infections. This complex situation may represent a further and underestimated risk with regard to COVID-19 clinical burden for the elderly and comorbid people. Here, we discuss the possible biological, physiopathological, and clinical implications of gut microbiota in COVID-19 and the strategies to improve/maintain its healthy status as a simple and adjunctive strategy to reduce COVID-19 virulence and socio-sanitary burden.
Collapse
Affiliation(s)
- Sabrina Donati Zeppa
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | | | | | | | | |
Collapse
|
35
|
The immunomodulatory effects of probiotics on respiratory viral infections: A hint for COVID-19 treatment? Microb Pathog 2020; 148:104452. [PMID: 32818576 PMCID: PMC7431320 DOI: 10.1016/j.micpath.2020.104452] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/07/2020] [Accepted: 08/11/2020] [Indexed: 02/07/2023]
Abstract
Respiratory virus infections are among the most prevalent diseases in humans and contribute to morbidity and mortality in all age groups. Moreover, since they can evolve fast and cross the species barrier, some of these viruses, such as influenza A and coronaviruses, have sometimes caused epidemics or pandemics and were associated with more serious clinical diseases and even mortality. The recently identified Coronavirus Disease 2019 (COVID-19) caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a Public Health Emergency of International concern and has been associated with rapidly progressive pneumonia. To ensure protection against emerging respiratory tract infections, the development of new strategies based on modulating the immune responses is essential. The use of probiotic components has substantially increased due to their effects on immune responses, in particular on those that occur in the upper/lower respiratory tract. Superinduction of inflammatory reaction, known as a cytokine storm, has been correlated directly with viral pneumonia and serious complications of respiratory infections. In this review, probiotics, as potential immunomodulatory agents, have been proposed to improve the host's response to respiratory viral infections. In addition, the effects of probiotics on different aspects of immune responses and their antiviral properties in both pre-clinical and clinical contexts have been described in detail.
Collapse
|
36
|
Akour A. Probiotics and COVID-19: is there any link? Lett Appl Microbiol 2020; 71:229-234. [PMID: 32495940 PMCID: PMC7300613 DOI: 10.1111/lam.13334] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 05/18/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022]
Abstract
Understanding mechanisms of the novel SARS-CoV2 infection and progression can provide potential novel targets for prevention and/or treatment. This could be achieved via the inhibition of viral entry and/or replication, or by suppression of the immunologic response that is provoked by the infection (known as the cytokine storm). Probiotics are defined as 'live microorganisms that, when administered in adequate amounts, confer a health benefit on the host'. There is scarcity of evidence about the relationship between COVID-19 and gut microbiota. So, whether or not these supplements can prevent or ameliorate COVID-19-associated symptoms is not fully understood. The aim of this study is to provide an indirect evidence about the utility of probiotics in combating COVID-19 or its associated symptoms, through the review of its antiviral and anti-inflammatory properties in vitro, animal models and human trials. SIGNIFICANCE AND IMPACT OF THE STUDY: The role of probiotics in alleviation of the novel COVID-19 has not been established. This review provides an insight about the anti-inflammatory, antiviral effects of probiotics in vitro, animal models and human. The latter can provide an indirect evidence and/or hypothesis-driven approach to investigate the use of probiotics as adjunctive therapy in the prophylaxis and/or alleviation of COVID-19 symptoms.
Collapse
Affiliation(s)
- A Akour
- Department of Biopharmaceutics and Clinical Pharmacy, The School of Pharmacy, The University of Jordan, Amman, Jordan.,Department of Pharmacy, Faculty of Pharmacy, Al-Zaytoonah University of Jordan, Amman, Jordan
| |
Collapse
|
37
|
Falasca F, Cavallari EN, Innocenti GP, Scagnolari C, Mezzaroma I, Santinelli L, Ceccarelli G, Vullo V, Turriziani O, d'Ettorre G. Antiviral Activity of Fecal Water Samples from HIV-1 Infected Subjects Treated with a Specific Probiotic Formulation. Curr HIV Res 2020; 17:183-189. [PMID: 31490760 DOI: 10.2174/1570162x17666190903230622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Revised: 08/11/2019] [Accepted: 08/21/2019] [Indexed: 12/27/2022]
Abstract
OBJECTIVES The aim of the study was to investigate if the supplementation with multistrain probiotics may be able to modulate T cell response in HIV-1 infected patients and to evaluate the anti-HIV activity of probiotic by studying fecal water (FW) samples. METHODS Three HIV-1-positive patients (Pt1, Pt2 and Pt3) on long-term suppressive combined antiretroviral therapy (cART) received a specific multi-strain probiotic supplementation (Vivomixx ®), for six months (T6). Levels of T cell subsets were evaluated by flow cytometry. Anti- HIV activity of FW samples was evaluated in vitro. RESULTS CD4+ T cells levels increased in all HIV-1 infected patients whereas activation markers (CD38 and HLA-DR) were decreased both on CD4+ and CD8+ T cells. FW samples presented an increased inhibitory activity against HIV-1 compared to T0 (FW-Pt1: T0 =40%, T6 = 65% of reduction; FW Pt2: T0 = 26%, T6 = 46% of reduction; FW Pt3: T0 = 47%, T6 = 94% of reduction). DISCUSSION Our data suggest that the administration of the specific probiotic formulation improves the antiviral status of people living with HIV-1 under cART, also modulating T cell response. CONCLUSION Anti-HIV activity of FW may have several public health and social implications for sexually transmitted diseases that need to be further explored.
Collapse
Affiliation(s)
- Francesca Falasca
- Department of Molecular Medicine, Sapienza University of Rome, Rome, Italy
| | | | | | | | - Ivano Mezzaroma
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Letizia Santinelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy.,Azienda Policlinico Umberto I, Rome, Italy
| | - Vincenzo Vullo
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| | | | - Gabriella d'Ettorre
- Department of Public Health and Infectious Diseases, Sapienza University of Rome, Rome, Italy
| |
Collapse
|
38
|
van Praagh JB, Luo JN, Zaborina O, Alverdy JC. Involvement of the Commensal Organism Bacillus subtilis in the Pathogenesis of Anastomotic Leak. Surg Infect (Larchmt) 2020; 21:865-870. [PMID: 32310731 DOI: 10.1089/sur.2019.345] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: It is now well established that microbes play a key and causative role in the pathogenesis of anastomotic leak. Yet, in patients, determining whether a cultured pathogen retrieved from an anastomotic leak site is a cause or a consequence of the complication remains a challenge. The aim of this study was to test a methodology to invoke causality between a retrieved microbe from a leak site and its role in anastomotic leak. Methods: The commensal organism Bacillus subtilis was isolated from an esophagojejunostomy leak site in a 35-year-old patient with a CDH1 mutation after a prophylactic gastrectomy whose body mass index (BMI) was 35 kg/m2. The organism was screened for its ability to degrade collagen, shift human recombinant matrix metalloprotease-9 (MMP9) to its active form, and induce a clinical anastomotic leak when introduced to anastomotic tissues of mice fed their standard diet (SD) of chow or an obesogenic Western-type diet (WD). Results: The Bacillus subtilis strain retrieved from the anastomotic leak site displayed a high degree of collagenolytic activity and was able to activate human MMP9 consistent with other pathogens expressing this characteristic "leak phenotype." Exposure of the Bacillus subtilis to the anastomotic tissues of obese mice fed a WD led to dehiscence of the anastomosis, abscess formation with peritonitis, and mortality in 50% of mice (3/6). When anastomotic healing was evaluated by a validated anastomotic healing score (AHS), substantially worse healing was observed (i.e., higher AHS) in WD-fed mice exposed to Bacillus subtilis compared to SD-fed mice (analysis of variance [ANOVA], p = 0.0006). Conclusions: Microbial strains obtained from patients' anastomotic leak sites can be evaluated for their pathogenic in the leak process by assessing their ability to produce collagenase, activate MMP9 and cause clinical leaks in mice fed a WD. These studies may aid in identifying those bacterial strains that play a causal role in patients with an anastomotic leak.
Collapse
Affiliation(s)
- Jasper B van Praagh
- Department of Surgery, University of Chicago, Pritzker School of Medicine, Chicago, Illinois, USA.,Department of Surgery, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - James N Luo
- Department of Surgery, University of Chicago, Pritzker School of Medicine, Chicago, Illinois, USA
| | - Olga Zaborina
- Department of Surgery, University of Chicago, Pritzker School of Medicine, Chicago, Illinois, USA
| | - John C Alverdy
- Department of Surgery, University of Chicago, Pritzker School of Medicine, Chicago, Illinois, USA
| |
Collapse
|
39
|
Lee NK, Kim WS, Paik HD. Bacillus strains as human probiotics: characterization, safety, microbiome, and probiotic carrier. Food Sci Biotechnol 2019; 28:1297-1305. [PMID: 31695928 PMCID: PMC6811671 DOI: 10.1007/s10068-019-00691-9] [Citation(s) in RCA: 162] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/16/2019] [Accepted: 09/20/2019] [Indexed: 02/07/2023] Open
Abstract
Both spore and vegetative forms of Bacillus species have been used as probiotics, and they have high stability to the surrounding atmospheric conditions such as heat, gastric conditions, and moisture. The commercial Bacillus probiotic strains in use are B. cereus, B. clausii, B. coagulans, B. licheniformis, B. polyfermenticus, B. pumilus, and B. subtilis. These strains have antimicrobial, anticancer, antioxidant, and vitamin production properties. However, Bacillus probiotics can also produce toxins and biogenic amines and transfer antibiotic resistance genes; therefore, their safety is a concern. Studies on the microbiome using probiotic Bacillus strains are limited in humans. Most microbiome research has been conducted in chicken, mouse, and pig. Some Bacillus probiotics are used as fermentation starters in plant and soybean and dietary supplement of baking foods as a probiotic carrier. This review summarizes the characterization of Bacillus species as probiotics for human use and their safety, microbiome, and probiotic carrier.
Collapse
Affiliation(s)
- Na-Kyoung Lee
- Department of Food Science and Biotechnology of Animal Resource, Konkuk University, Seoul, 05029 Republic of Korea
| | - Won-Suck Kim
- College of Medical and Life Sciences, Silla University, Busan, 46958 Republic of Korea
| | - Hyun-Dong Paik
- Department of Food Science and Biotechnology of Animal Resource, Konkuk University, Seoul, 05029 Republic of Korea
| |
Collapse
|
40
|
Novik G, Savich V. Beneficial microbiota. Probiotics and pharmaceutical products in functional nutrition and medicine. Microbes Infect 2019; 22:8-18. [PMID: 31233819 DOI: 10.1016/j.micinf.2019.06.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 12/12/2022]
Abstract
The article is mainly devoted to such representatives of gut microbiota as lactic acid bacteria and bifidobacteria, with minor accent on less frequently used or new probiotic microorganisms. Positive effects in treatment and prevention of diseases by different microbial groups, their metabolites and mechanisms of action, management and market of probiotic products are considered.
Collapse
Affiliation(s)
- Galina Novik
- Belarusian Collection of Microorganisms, Institute of Microbiology, National Academy of Sciences of Belarus, 2 Academician V.F. Kuprevich Street, 220141 Minsk, the Republic of Belarus.
| | - Victoria Savich
- Belarusian Collection of Microorganisms, Institute of Microbiology, National Academy of Sciences of Belarus, 2 Academician V.F. Kuprevich Street, 220141 Minsk, the Republic of Belarus
| |
Collapse
|
41
|
Wang J, Wang Y, Zhang E, Zhou M, Lin J, Yang Q. Intranasal administration with recombinant Bacillus subtilis induces strong mucosal immune responses against pseudorabies. Microb Cell Fact 2019; 18:103. [PMID: 31170996 PMCID: PMC6555017 DOI: 10.1186/s12934-019-1151-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 05/28/2019] [Indexed: 12/22/2022] Open
Abstract
Background Pseudorabies caused by pseudorabies virus (PRV) mainly infects the swine and seriously threatens the biosafety of the other animals, including humans. Since 2011, the outbreaks of PRV mutants have caused enormous economic losses in the swine industry, and traditional vaccines cannot offer enough protection. PRV can transmit by direct contact, aerosol transmission and pollutants. PRV mainly transmit through the nasal mucosa. After infecting the nasal epithelial cells, PRV can quickly infect the olfactory nerve and establish a potential infection of sensory neurons. Therefore, nasal immunity can effectively prevent viral colonization infection. Recombinant Bacillus subtilis has been widely used to deliver antigen and achieve adequate protective immune responses. Results The present study successfully constructed recombinant Bacillus subtilis (B. subtilis) expressing the dominant antigen regions of PRV gC and gD proteins (named B. subtilis-gCa and B. subtilis-gDa). Furtherly, we evaluated the immunogenicity of the two recombinant B. subtilis in mice. The mice intranasal administration with B. subtilis-gCa and B. subtilis-gDa effectively stimulated IgA and IgG immune responses, further regulated specific T lymphocytes proliferative response by IFN-γ and IL-10, and ultimately produced high titers of neutralizing antibodies against PRV infection. In particular, B. subtilis-gDa possessed more excellent immune effect than B. subtilis-gCa in mice. Conclusions These results suggested that B. subtilis-gCa and B. subtilis-gDa could trigger high levels of mucosal and systemic immune responses and would be potential candidates for developing PRV vaccines. Electronic supplementary material The online version of this article (10.1186/s12934-019-1151-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Jialu Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Yongheng Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, 210095, Jiangsu, People's Republic of China
| | - En Zhang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Mengyun Zhou
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Jian Lin
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, 210095, Jiangsu, People's Republic of China
| | - Qian Yang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Weigang 1, Nanjing, 210095, Jiangsu, People's Republic of China.
| |
Collapse
|
42
|
Jing Y, Liu H, Xu W, Yang Q. Amelioration of the DSS-induced colitis in mice by pretreatment with 4,4'-diaponeurosporene-producing Bacillus subtilis. Exp Ther Med 2017; 14:6069-6073. [PMID: 29285159 PMCID: PMC5740520 DOI: 10.3892/etm.2017.5282] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 10/10/2017] [Indexed: 02/07/2023] Open
Abstract
Inflammatory bowel disease (IBD) is a chronically relapsing inflammatory disorder of the gastrointestinal tract. Current IBD treatments have poor tolerability and insufficient therapeutic efficacy, thus, alternative therapeutic approaches are required. Recently, a number of dietary supplements have emerged as promising interventions. In the present study oral administration of a carotenoid (4,4'-diaponeurosporene)-producing Bacillus subtilis markedly ameliorated dextran sulfate sodium salt-induced mouse colitis, as demonstrated by a reduction in weight loss and the severity of bleeding, which indicated that 4,4'-diaponeurosporene may have beneficial effects on treatments for colitis. This preliminary study indicated that 4,4'-diaponeurosporene may function synergistically with probiotics to provide a novel and effective strategy to prevent colitis.
Collapse
Affiliation(s)
- Yuchao Jing
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Haofei Liu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Wenwen Xu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| | - Qian Yang
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, Jiangsu 210095, P.R. China
| |
Collapse
|