1
|
Leistikow KR, May DS, Suh WS, Vargas Asensio G, Schaenzer AJ, Currie CR, Hristova KR. Bacillus subtilis-derived peptides disrupt quorum sensing and biofilm assembly in multidrug-resistant Staphylococcus aureus. mSystems 2024; 9:e0071224. [PMID: 38990088 PMCID: PMC11334493 DOI: 10.1128/msystems.00712-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 06/15/2024] [Indexed: 07/12/2024] Open
Abstract
Multidrug-resistant Staphylococcus aureus is one of the most clinically important pathogens in the world, with infections leading to high rates of morbidity and mortality in both humans and animals. The ability of S. aureus to form biofilms protects cells from antibiotics and promotes the transfer of antibiotic resistance genes; therefore, new strategies aimed at inhibiting biofilm growth are urgently needed. Probiotic species, including Bacillus subtilis, are gaining interest as potential therapies against S. aureus for their ability to reduce S. aureus colonization and virulence. Here, we search for strains and microbially derived compounds with strong antibiofilm activity against multidrug-resistant S. aureus by isolating and screening Bacillus strains from a variety of agricultural environments. From a total of 1,123 environmental isolates, we identify a single strain B. subtilis 6D1, with a potent ability to inhibit biofilm growth, disassemble mature biofilm, and improve antibiotic sensitivity of S. aureus biofilms through an Agr quorum sensing interference mechanism. Biochemical and molecular networking analysis of an active organic fraction revealed multiple surfactin isoforms, and an uncharacterized peptide was driving this antibiofilm activity. Compared with commercial high-performance liquid chromatography grade surfactin obtained from B. subtilis, we show these B. subtilis 6D1 peptides are significantly better at inhibiting biofilm formation in all four S. aureus Agr backgrounds and preventing S. aureus-induced cytotoxicity when applied to HT29 human intestinal cells. Our study illustrates the potential of exploring microbial strain diversity to discover novel antibiofilm agents that may help combat multidrug-resistant S. aureus infections and enhance antibiotic efficacy in clinical and veterinary settings. IMPORTANCE The formation of biofilms by multidrug-resistant bacterial pathogens, such as Staphylococcus aureus, increases these microorganisms' virulence and decreases the efficacy of common antibiotic regimens. Probiotics possess a variety of strain-specific strategies to reduce biofilm formation in competing organisms; however, the mechanisms and compounds responsible for these phenomena often go uncharacterized. In this study, we identified a mixture of small probiotic-derived peptides capable of Agr quorum sensing interference as one of the mechanisms driving antibiofilm activity against S. aureus. This collection of peptides also improved antibiotic killing and protected human gut epithelial cells from S. aureus-induced toxicity by stimulating an adaptive cytokine response. We conclude that purposeful strain screening and selection efforts can be used to identify unique probiotic strains that possess specially desired mechanisms of action. This information can be used to further improve our understanding of the ways in which probiotic and probiotic-derived compounds can be applied to prevent bacterial infections or improve bacterial sensitivity to antibiotics in clinical and agricultural settings.
Collapse
Affiliation(s)
- Kyle R. Leistikow
- Department of Biological Sciences, Marquette University, Milwaukee, Wisconsin, USA
| | - Daniel S. May
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Chemistry, Washington College, Chestertown, Maryland, USA
| | - Won Se Suh
- Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | | - Adam J. Schaenzer
- Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Cameron R. Currie
- Department of Bacteriology, University of Wisconsin-Madison, Madison, Wisconsin, USA
- Department of Biochemistry & Biomedical Sciences, McMaster University, Hamilton, Ontario, Canada
| | | |
Collapse
|
2
|
Ren Y, Huang P, Zhang L, Tang YF, Luo SL, She Z, Peng H, Chen YQ, Luo JW, Duan WX, Liu LJ, Liu LQ. Dual Regulation Mechanism of Obesity: DNA Methylation and Intestinal Flora. Biomedicines 2024; 12:1633. [PMID: 39200098 PMCID: PMC11351752 DOI: 10.3390/biomedicines12081633] [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: 06/12/2024] [Revised: 07/15/2024] [Accepted: 07/18/2024] [Indexed: 09/01/2024] Open
Abstract
Obesity is a multifactorial chronic inflammatory metabolic disorder, with pathogenesis influenced by genetic and non-genetic factors such as environment and diet. Intestinal microbes and their metabolites play significant roles in the occurrence and development of obesity by regulating energy metabolism, inducing chronic inflammation, and impacting intestinal hormone secretion. Epigenetics, which involves the regulation of host gene expression without changing the nucleotide sequence, provides an exact direction for us to understand how the environment, lifestyle factors, and other risk factors contribute to obesity. DNA methylation, as the most common epigenetic modification, is involved in the pathogenesis of various metabolic diseases. The epigenetic modification of the host is induced or regulated by the intestinal microbiota and their metabolites, linking the dynamic interaction between the microbiota and the host genome. In this review, we examined recent advancements in research, focusing on the involvement of intestinal microbiota and DNA methylation in the etiology and progression of obesity, as well as potential interactions between the two factors, providing novel perspectives and avenues for further elucidating the pathogenesis, prevention, and treatment of obesity.
Collapse
Affiliation(s)
- Yi Ren
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
- Department of Pediatrics, Haikou Hospital of the Maternal and Child Health, Haikou 570100, China
- Department of Children’s Healthcare, Hainan Modern Women and Children’s Medical, Haikou 570100, China
| | - Peng Huang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Lu Zhang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yu-Fen Tang
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Sen-Lin Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Zhou She
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Hong Peng
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Yu-Qiong Chen
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Jin-Wen Luo
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Wang-Xin Duan
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Ling-Juan Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| | - Li-Qun Liu
- Department of Pediatrics, The Second Xiangya Hospital of Central South University, Changsha 410011, China; (Y.R.); (P.H.); (L.Z.); (Y.-F.T.); (S.-L.L.); (Z.S.); (H.P.); (Y.-Q.C.); (J.-W.L.); (W.-X.D.); (L.-J.L.)
- Children’s Brain Development and Brain Injury Research Office, The Second Xiangya Hospital of Central South University, Changsha 410011, China
| |
Collapse
|
3
|
Yu Q, Liu Y, Liu S, Li S, Zhai Y, Zhang Q, Zheng L, Zheng H, Zhai Y, Wang X. Lactobacillus melliventris promotes hive productivity and immune functionality in Bombus terrestris performance in the greenhouse. INSECT SCIENCE 2024; 31:911-926. [PMID: 37830269 DOI: 10.1111/1744-7917.13281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2023] [Revised: 08/18/2023] [Accepted: 09/15/2023] [Indexed: 10/14/2023]
Abstract
Bumblebees are important pollinators in agricultural ecosystems, but their abundance is declining globally. There is an urgent need to protect bumblebee health and their pollination services. Bumblebees possess specialized gut microbiota with potential to be used as probiotics to help defend at-risk bumblebee populations. However, evidence for probiotic benefits on bumblebees is lacking. Here, we evaluated how supplementation with Lactobacillus melliventris isolated from bumblebee gut affected the colony development of Bombus terrestris. This native strain colonized robustly and persisted long-term in bumblebees, leading to a significantly higher quality of offspring. Subsequently, the tyrosine pathway was upregulated in the brain and fat body, while the Wnt and mTOR pathways of the gut were downregulated. Notably, the field experiment in the greenhouse revealed the supplementation of L. melliventris led to a 2.5-fold increase in the bumblebee survival rate and a more than 10% increase in the number of flowers visited, indicating a better health condition and pollination ability in field conditions. Our study represents a first screening for the potential use of the native gut member, L. melliventris, as probiotic strains in hive supplement for bumblebee breeding, which may be a practical approach to improve immunity and hive health.
Collapse
Affiliation(s)
- Qianhui Yu
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan, China
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yan Liu
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Shanshan Liu
- BGI-Qingdao, BGI-Shenzhen, Qingdao, Shandong, China
| | - Shaogang Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yifan Zhai
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Qingchao Zhang
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan, China
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Li Zheng
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Hao Zheng
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan, China
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yifan Zhai
- Institute of Plant Protection, Shandong Academy of Agricultural Sciences, Jinan, China
- Key Laboratory of Natural Enemies Insects, Ministry of Agriculture and Rural Affairs, Jinan, China
| | - Xiaofei Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| |
Collapse
|
4
|
Fusco A, Savio V, Chiaromonte A, Alfano A, D’Ambrosio S, Cimini D, Donnarumma G. Evaluation of Different Activity of Lactobacillus spp. against Two Proteus mirabilis Isolated Clinical Strains in Different Anatomical Sites In Vitro: An Explorative Study to Improve the Therapeutic Approach. Microorganisms 2023; 11:2201. [PMID: 37764044 PMCID: PMC10534642 DOI: 10.3390/microorganisms11092201] [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: 07/30/2023] [Revised: 08/20/2023] [Accepted: 08/29/2023] [Indexed: 09/29/2023] Open
Abstract
Urinary tract infections (UTIs) and catheter-associated UTIs (CAUTIs) are the principal hospital-acquired infections. Between these, bacterial prostatitis is believed to be the leading cause of recurrent UTIs in men under 50 years of age and is often unresponsive to antibiotic treatment. Proteus mirabilis is more commonly associated with UTIs in these abnormalities, especially in patients undergoing catheterization. Lactobacillus spp. are an important component of the human microbiota and occur in large quantities in foods. Probiotics are proposed as an alternative to antibiotic therapy in the treatment of urinary tract infections. In addition to their ability to produce antimicrobial metabolites, they have immunomodulatory activity and do not cause side effects. For this reason, the combination of probiotic microorganisms and conventional drugs was considered. The aim of this work was to select the most active Lactobacillus strains against two clinical isolates of P. mirabilis on bladder and prostatic epithelium, potentially exploitable to improve the clinical management of UTIs.
Collapse
Affiliation(s)
- Alessandra Fusco
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.S.); (A.C.); (A.A.); (D.C.)
| | | | | | | | | | | | - Giovanna Donnarumma
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (V.S.); (A.C.); (A.A.); (D.C.)
| |
Collapse
|
5
|
Omotosho AO, Tajudeen YA, Oladipo HJ, Yusuff SI, AbdulKadir M, Muili AO, Egbewande OM, Yusuf RO, Faniran ZO, Afolabi AO, El‐Sherbini MS. Parkinson's disease: Are gut microbes involved? Brain Behav 2023; 13:e3130. [PMID: 37340511 PMCID: PMC10454343 DOI: 10.1002/brb3.3130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 06/05/2023] [Accepted: 06/05/2023] [Indexed: 06/22/2023] Open
Abstract
INTRODUCTION Parkinson's disease (PD) is a neurodegenerative disorder that affects more than 10 million individuals worldwide. It is characterized by motor and sensory deficits. Research studies have increasingly demonstrated a correlation between Parkinson's disease and alternations in the composition of the gut microbiota in affected patients. Also, the significant role of prebiotics and probiotics in gastrointestinal and neurological conditions is imperative to understand their relation to Parkinson's disease. METHOD To explore the scientific interaction of the gut-microbiota-brain axis and its association with Parkinson's disease, a comprehensive narrative review of the relevant literature was conducted. Articles were retrieved systematically from reputable sources, including PubMed, Science Direct, World Health Organization (WHO), and Advanced Google Scholar. Key search terms included are "Parkinson's Disease", "Gut Microbiome", "Braak's Theory", "Neurological Disorders", and "Gut-brain axis". Articles included in our review are published in English and they provide detailed information on the relationship between Parkinson's disease and gut microbiota RESULTS: This review highlights the impact of gut microbiota composition and associated factors on the progression of Parkinson's disease. Evidence-based studies highlighting the existing evidence of the relationship between Parkinson's disease and alteration in gut microbiota are discussed. Consequently, the potential mechanisms by which the gut microbiota may affect the composition of the gut microbiota were revealed, with a particular emphasis on the role of the gut-brain axis in this interplay. CONCLUSION Understanding the complex interplay between gut microbiota and Parkinson's disease is a potential implication for the development of novel therapeutics against Parkinson's disease. Following the existing relationship demonstrated by different evidence-based studies on Parkinson's disease and gut microbiota, our review concludes by providing recommendations and suggestions for future research studies with a particular emphasis on the impact of the microbiota-brain axis on Parkinson's disease.
Collapse
Affiliation(s)
- Abass Olawale Omotosho
- Department of Microbiology, Faculty of Pure and Applied SciencesKwara State University, Malete‐IlorinIlorinNigeria
| | - Yusuf Amuda Tajudeen
- Department of Microbiology, Faculty of Life SciencesUniversity of IlorinIlorinNigeria
- Faculty of Pharmaceutical SciencesUniversity of IlorinIlorinNigeria
| | - Habeebullah Jayeola Oladipo
- Department of Microbiology, Faculty of Life SciencesUniversity of IlorinIlorinNigeria
- Department of Epidemiology and Medical Statistics, Faculty of Public Health, College of MedicineUniversity of IbadanIbadanNigeria
| | - Sodiq Inaolaji Yusuff
- Department of Medicine, Faculty of Clinical SciencesObafemi Awolowo UniversityIfeNigeria
| | - Muritala AbdulKadir
- Department of Epidemiology and Medical Statistics, Faculty of Public Health, College of MedicineUniversity of IbadanIbadanNigeria
| | | | - Oluwaseyi Muyiwa Egbewande
- Department of Epidemiology and Medical Statistics, Faculty of Public Health, College of MedicineUniversity of IbadanIbadanNigeria
| | - Rashidat Onyinoyi Yusuf
- Department of Epidemiology and Medical Statistics, Faculty of Public Health, College of MedicineUniversity of IbadanIbadanNigeria
| | | | - Abdullateef Opeyemi Afolabi
- Faculty of Biomedical Sciences, Department of Microbiology and ImmunologyKampala International UniversityBushenyiUganda
| | - Mona Said El‐Sherbini
- Narrative Medicine and Planetary Health, Integrated Program of Kasr Al-Ainy (IPKA), Faculty of MedicineCairo UniversityCairoEgypt
- Invited Facultythe Nova Institute for HealthBaltimoreMDUSA
- Department of Medical Parasitology, Faculty of MedicineCairo UniversityCairoEgypt
| |
Collapse
|
6
|
Elgamily HM, El-Sayed SM, El-Sayed HS, Youssef AM. Laboratory evaluation of anti-plaque and remineralization efficacy of sugarless probiotic jelly candy supplemented with natural nano prebiotic additive. Sci Rep 2023; 13:10977. [PMID: 37414826 PMCID: PMC10326239 DOI: 10.1038/s41598-023-37645-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Accepted: 06/25/2023] [Indexed: 07/08/2023] Open
Abstract
We evaluated the anti-cariogenic effect of an experimental synbiotic compound containing probiotic Lacticaseibacillus rhamnosus (NRRL B-442)-based jelly candy supplemented with natural prebiotic grape seed extract (GSE) in a nanoemulsion formula on the colonization and establishment of Streptococcus mutans (ATCC 25175) and Actinomyces viscosus (ATTCC 19246) biofilms through counting colony forming units, scanning electron microscopy (SEM), and transmission electron microscopy (TEM). We were then analysing the remineralizing effect of synbiotic jelly candy on human enamel surface lesions using Vickers microhardness testers, atomic force microscopy (AFM), SEM, energy-dispersive X-ray spectroscopy (EDAX), and confocal laser scanning microscopy (CLSM) at three stages (sound, after demineralization, and after pH cycling). We found after 21 days of treatment of the pH-cycled enamel discs with jelly candy for 10 min twice daily, a 68% decrease in S. mutans colony formation, reducing biofilm development, trapping S. mutans visualized in jelly candy under SEM examination, and significantly altering the morphological structure of these bacteria under TEM analysis. For remineralization measurements, statistically significant differences in microhardness integrated mineral loss, and lesion depth through CLSM between demineralization and treatment stages. These findings provide an effective anti-cariogenic synbiotic compound of grape seed extract and probiotic jelly candy with potential remineralizing activity.
Collapse
Affiliation(s)
- Hanaa M Elgamily
- Restorative and Dental Materials Department, Oral and Dental Research Institutes, National Research Centre, 33 El Bohouth St., Dokki, Giza, 12622, Egypt.
| | - Samah M El-Sayed
- Dairy Department, Food Industries and Nutrition Research Institute, National Research Centre, 33 El Bohouth St., Dokki, Giza, 12622, Egypt
| | - Hoda S El-Sayed
- Dairy Department, Food Industries and Nutrition Research Institute, National Research Centre, 33 El Bohouth St., Dokki, Giza, 12622, Egypt
| | - Ahmed M Youssef
- Packaging Materials Department, National Research Centre, 33 El Bohouth St., Dokki, Giza, 12622, Egypt
| |
Collapse
|
7
|
Porras-García E, Fernández-Espada Calderón I, Gavala-González J, Fernández-García JC. Potential neuroprotective effects of fermented foods and beverages in old age: a systematic review. Front Nutr 2023; 10:1170841. [PMID: 37396132 PMCID: PMC10313410 DOI: 10.3389/fnut.2023.1170841] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 05/30/2023] [Indexed: 07/04/2023] Open
Abstract
Purpose Numerous articles have recently studied the involvement of the gut microbiota in neurological diseases. Aging is associated with changes in the microbiome, which implies a reduction in microbial biodiversity among other changes. Considering that the consumption of a fermented-food diet improves intestinal permeability and barrier function, it seems of interest to study its participation in the prevention of neurodegenerative diseases. This article reviews existing studies to establish whether the consumption of fermented foods and fermented beverages prevents or ameliorates neurodegenerative decline in old age. Methods The protocol used was performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines. Details of the protocol for this systematic review are registered on PROSPERO (CRD42021250921). Results Out of 465 articles identified in the Pubmed, Scopus, and Cochrane Library databases, a total of 29 that examined the relationship of the consumption of fermented products with cognitive impairment in old people were selected (22 cohort, 4 case-control, and 3 cross-sectional studies). The results suggest that low-to-moderate alcohol consumption and daily intake of coffee, soy products, and fermented-food diets in general are associated with a lower risk of dementia and Alzheimer's disease. Conclusion Daily consumption of fermented foods and beverages, either alone or as part of a diet, has neuroprotective effects and slows cognitive decline in old people. Systematic review registration https://www.crd.york.ac.uk/prospero/display_record.php?RecordID=250921, identifier: CRD42021250921.
Collapse
Affiliation(s)
- Elena Porras-García
- Department of Physiology, Anatomy and Cellular Biology, University of Pablo de Olavide, Seville, Spain
| | | | - Juan Gavala-González
- Department of Physical Education and Sports, University of Seville, Seville, Spain
| | - José Carlos Fernández-García
- Department of Didactics of Languages, Arts and Sport, University of Malaga, Andalucía-Tech, Instituto de Investigación Biomédica de Málaga (IBIMA), Malaga, Spain
| |
Collapse
|
8
|
Feng L, Gu J, Guo L, Mu G, Tuo Y. Safety evaluation and application of lactic acid bacteria and yeast strains isolated from Sichuan broad bean paste. Food Sci Nutr 2023; 11:940-952. [PMID: 36789042 PMCID: PMC9922144 DOI: 10.1002/fsn3.3129] [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: 04/13/2022] [Revised: 10/03/2022] [Accepted: 10/25/2022] [Indexed: 12/23/2022] Open
Abstract
Broad bean paste is one of the most popular characteristic traditional fermented bean products in China, which is prepared by mixed fermentation of a variety of microorganisms, among which lactic acid bacteria and yeast played an important role in the improvement of the fermented broad bean paste quality. However, the traditional open-air fermentation of broad bean paste brought some risks of harmful microorganisms. In this study, the safety and fermentation ability of lactic acid bacteria and yeast strains isolated from traditional broad bean paste was evaluated. The results showed that the protease activity of the strain Lactobacillus plantarum DPUL-J5 (366.73 ± 9.00 U/L) and Pichia kudriavzevii DPUY-J5 (237.18 ± 10.93 U/L) were the highest. Both strains produced little biogenic amines, and did not exhibit α-hemolytic activity or antibiotic resistance for some of the antibiotics most used in human medicine. Furthermore, the broad bean paste fermentation involving DPUL-J5 and DPUY-J5 was beneficial for accumulating higher total acid (1.69 ± 0.01 g/100 g), amino-acid nitrogen (0.85 ± 0.03 g/100 g), and more volatile flavor compounds, meanwhile, reducing the levels of biogenic amines and aflatoxin B1. Therefore, this study provided a new strategy to improve the safety and quality of traditional broad bean paste.
Collapse
Affiliation(s)
- Lu Feng
- School of Food Science and TechnologyDalian Polytechnic UniversityDalianChina
| | - Jinhong Gu
- School of Food Science and TechnologyDalian Polytechnic UniversityDalianChina
| | - Linjie Guo
- School of Food Science and TechnologyDalian Polytechnic UniversityDalianChina
| | - Guangqing Mu
- School of Food Science and TechnologyDalian Polytechnic UniversityDalianChina
| | - Yanfeng Tuo
- School of Food Science and TechnologyDalian Polytechnic UniversityDalianChina
| |
Collapse
|
9
|
Bahaddad SA, Almalki MHK, Alghamdi OA, Sohrab SS, Yasir M, Azhar EI, Chouayekh H. Bacillus Species as Direct-Fed Microbial Antibiotic Alternatives for Monogastric Production. Probiotics Antimicrob Proteins 2023; 15:1-16. [PMID: 35092567 PMCID: PMC8799964 DOI: 10.1007/s12602-022-09909-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/09/2022] [Indexed: 01/18/2023]
Abstract
Antibiotic growth promoters have been utilized for long time at subtherapeutic levels as feed supplements in monogastric animal rations. Because of their side-effects such as antibiotic resistance, reduction of beneficial bacteria in the gut, and dysbiosis, it is necessary to look for non-therapeutic alternatives. Probiotics play an important role as the key substitutes to antibacterial agents due to their many beneficial effects on the monogastric animal host. For instance, enhancement of the gut microbiota balance can contribute to improvement of feed utilization efficiency, nutrients absorption, growth rate, and economic profitability of livestock. Probiotics are defined as "live microorganisms that, when administered in adequate amounts, confer a health benefit on the host." They are available in diverse forms for use as feed supplements. Their utilization as feed additives assists in good digestion of feed ingredients and hence, making the nutrients available for promoting growth. Immunity can also be enhanced by supplementing probiotics to monogastrics diets. Moreover, probiotics can help in improving major meat quality traits and countering a variety of monogastric animals infectious diseases. A proper selection of the probiotic strains is required in order to confer optimal beneficial effects. The present review focuses on the general functional, safety, and technological screening criteria for selection of ideal Bacillus probiotics as feed supplements as well as their mechanism of action and beneficial effects on monogastric animals for improving production performance and health status.
Collapse
Affiliation(s)
- Shifa A Bahaddad
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Kingdom of Saudi Arabia
| | - Meshal H K Almalki
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Kingdom of Saudi Arabia
| | - Othman A Alghamdi
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Kingdom of Saudi Arabia
| | - Sayed S Sohrab
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Muhammad Yasir
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Esam I Azhar
- Special Infectious Agents Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Kingdom of Saudi Arabia
| | - Hichem Chouayekh
- Department of Biological Sciences, College of Science, University of Jeddah, Jeddah, Kingdom of Saudi Arabia.
| |
Collapse
|
10
|
Beneficial Effects of Lactic Acid Bacteria on Animal Reproduction Function. Vet Med Int 2022; 2022:4570320. [PMID: 36505731 PMCID: PMC9729032 DOI: 10.1155/2022/4570320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 10/22/2022] [Accepted: 11/23/2022] [Indexed: 12/02/2022] Open
Abstract
Considering the importance of a healthy uterus to the success of breeding, the beneficial effects of lactic acid bacteria on animal reproduction function are of particular interest. In recent decades, infertility has become a widespread issue, with microbiological variables playing a significant role. According to reports, dysbiosis of the vaginal microbiota is connected with infertility; however, the effect of the normal vaginal microbiota on infertility is unknown. In addition, lactic acid bacteria dominate the reproductive system. According to evidence, vaginal lactic acid bacteria play a crucial role in limiting the invasion of pathogenic bacteria by triggering anti-inflammatory chemicals through IL-8, IL-1, and IL-6; immunological responses through inhibition of the adherence of other microorganisms, production of inhibiting substances, and stimulation of mucus production; and also reproductive hormones by increased testosterone hormone release, enhanced the levels of luteinizing hormone, follicle stimulating hormone, the amount of prostaglandin E (2), and prostaglandin F2 alpha. The objective of this study was to compare the advantages of lactic acid bacteria in animal reproduction based on the most recent literature. The administration of a single strain or numerous strains of lactic acid bacteria has a favourable impact on steroidogenesis, gametogenesis, and animal fertility.
Collapse
|
11
|
Yang J, Qin S, Zhang H. Precise strategies for selecting probiotic bacteria in treatment of intestinal bacterial dysfunctional diseases. Front Immunol 2022; 13:1034727. [PMID: 36341458 PMCID: PMC9632739 DOI: 10.3389/fimmu.2022.1034727] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/07/2022] [Indexed: 11/20/2022] Open
Abstract
Abundant microbiota resides in the organs of the body, which utilize the nutrition and form a reciprocal relationship with the host. The composition of these microbiota changes under different pathological conditions, particularly in response to stress and digestive diseases, making the microbial composition and health of the hosts body interdependent. Probiotics are living microorganisms that have demonstrated beneficial effects on physical health and as such are used as supplements to ameliorate symptoms of various digestive diseases by optimizing microbial composition of the gut and restore digestive balance. However, the supplementary effect does not achieve the expected result. Therefore, a targeted screening strategy on probiotic bacteria is crucial, owing to the presence of several bacterial strains. Core bacteria work effectively in maintaining microbiological homeostasis and stabilization in the gastrointestinal tract. Some of the core bacteria can be inherited and acquired from maternal pregnancy and delivery; others can be acquired from contact with the mother, feces, and the environment. Knowing the genera and functions of the core bacteria could be vital in the isolation and selection of probiotic bacteria for supplementation. In addition, other supporting strains of probiotic bacteria are also needed. A comprehensive strategy for mining both core and supporting bacteria before its clinical use is needed. Using metagenomics or other methods of estimation to discern the typically differentiated strains of bacteria is another important strategy to treat dysbiosis. Hence, these two factors are significant to carry out targeted isolation and selection of the functional strains to compose the resulting probiotic preparation for application in both research and clinical use. In conclusion, precise probiotic supplementation, by screening abundant strains of bacteria and isolating specific probiotic strains, could rapidly establish the core microbiota needed to confer resilience, particularly in bacterial dysfunctional diseases. This approach can help identify distinct bacteria which can be used to improve supplementation therapies.
Collapse
Affiliation(s)
- Jiajun Yang
- School of Animal Husbandry and Veterinary Medicine, Jiangsu Vocational College of Agriculture and Forestry, Jurong, China
| | - Shunyi Qin
- Key Laboratory of Agricultural Animal Breeding and Healthy Breeding of Tianjin, College of Animal Science and Veterinary Medicine, Tianjin Agricultural University, Tianjin, China
| | - Hao Zhang
- College of Animal Science and Technology, Chinese Agricultural University, Beijing, China
- *Correspondence: Hao Zhang,
| |
Collapse
|
12
|
Ahmed HM, Shehata HH, El-Saeed GSM, Gabal HHA, El-Daly SM. Ameliorative effect of Lactobacillus rhamnosus GG on acetaminophen-induced hepatotoxicity via PKC/Nrf2/PGC-1α pathway. J Genet Eng Biotechnol 2022; 20:142. [PMID: 36201094 PMCID: PMC9537380 DOI: 10.1186/s43141-022-00422-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 09/23/2022] [Indexed: 07/10/2024]
Abstract
Background Acetaminophen (APAP) overdose is a common cause of hepatotoxicity. Antioxidants like N-acetyl cysteine are recommended as a therapeutic option; nevertheless, it has limitations. The search for efficient alternatives is ongoing. Probiotics are live microorganisms that maintain a healthy gut microecology. Lactobacillus rhamnosus GG (LGG) is one of the widely used probiotics. Our study aimed to assess the protective and therapeutic effects of probiotic LGG on APAP-induced hepatotoxicity and evaluate the molecular pathways behind this effect. Methods Wistar Albino male rats were randomly distributed into the following experimental groups: group 1, non-treated rats (vehicle); group 2, rats received oral gavage of suspension of probiotic LGG (5 × 1010 CFU GG/0.5 ml in PBS) daily for 2 weeks (probiotic control); group 3, rats received APAP dose of 2 g/kg body weight (positive control); group 4, rats received oral gavage of suspension of probiotic LGG for 2 weeks followed by a single dose of APAP injection (prophylactic); and group 5, rats received a single dose of APAP and then 24 h later treated with oral gavage of probiotic LGG daily for 2 weeks (treatment). Results Our study revealed that administration of probiotic LGG (either as prophylactic or treatment) exhibited a remarkable reduction in APAP-induced liver injury as resembled by the decrease in liver enzymes (ALT and AST) and the histopathological features of liver sections. Moreover, the significant reduction in the oxidative marker malondialdehyde, along with the enhancement in glutathione reductase, and the significant reduction in inflammatory markers (nitric oxide and tumor necrosis factor-α) were all indicators of the efficiency of LGG in ameliorating the alterations accompanied with APAP-induced hepatotoxicity. Our findings also demonstrate that LGG administration boosted the expression of Nrf2 and PGC-1 while decreasing the expression of protein kinase C (PKC). As a result, the nuclear abundance of Nrf2 is increased, and the expression of various antioxidants is eventually upregulated. Conclusion Our study shows that probiotic LGG supplementation exerts a prophylactic and therapeutic effect against APAP-induced hepatotoxicity through modulating the expression of PKC and the Nrf2/PGC-1α signaling pathway and eventually suppressing oxidative damage from APAP overdose.
Collapse
Affiliation(s)
- Hend M Ahmed
- Medical Biochemistry Department, Medicine and Clinical Studies Research Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Hanan H Shehata
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Gamila S M El-Saeed
- Medical Biochemistry Department, Medicine and Clinical Studies Research Institute, National Research Centre, Dokki, Cairo, 12622, Egypt
| | - Hoda H Abou Gabal
- Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Sherien M El-Daly
- Medical Biochemistry Department, Medicine and Clinical Studies Research Institute, National Research Centre, Dokki, Cairo, 12622, Egypt. .,Cancer Biology and Genetics Laboratory, Centre of Excellence for Advanced Sciences, National Research Centre, Cairo, Egypt.
| |
Collapse
|
13
|
Okeke ES, Chukwudozie KI, Nyaruaba R, Ita RE, Oladipo A, Ejeromedoghene O, Atakpa EO, Agu CV, Okoye CO. Antibiotic resistance in aquaculture and aquatic organisms: a review of current nanotechnology applications for sustainable management. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:69241-69274. [PMID: 35969340 PMCID: PMC9376131 DOI: 10.1007/s11356-022-22319-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 07/27/2022] [Indexed: 05/13/2023]
Abstract
Aquaculture has emerged as one of the world's fastest-growing food industries in recent years, helping food security and boosting global economic status. The indiscriminate disposal of untreated or improperly managed waste and effluents from different sources including production plants, food processing sectors, and healthcare sectors release various contaminants such as bioactive compounds and unmetabolized antibiotics, and antibiotic-resistant organisms into the environment. These emerging contaminants (ECs), especially antibiotics, have the potential to pollute the environment, particularly the aquatic ecosystem due to their widespread use in aquaculture, leading to various toxicological effects on aquatic organisms as well as long-term persistence in the environment. However, various forms of nanotechnology-based technologies are now being explored to assist other remediation technologies to boost productivity, efficiency, and sustainability. In this review, we critically highlighted several ecofriendly nanotechnological methods including nanodrug and vaccine delivery, nanoformulations, and nanosensor for their antimicrobial effects in aquaculture and aquatic organisms, potential public health risks associated with nanoparticles, and their mitigation measures for sustainable management.
Collapse
Affiliation(s)
- Emmanuel Sunday Okeke
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Nsukka, 41000, Enugu State, Nigeria
- Natural Science Unit, School of General Studies, University of Nigeria, Nsukka, 41000, Enugu State, Nigeria
- Institute of Environmental Health and Ecological Security, School of Environment & Safety Engineering, Jiangsu University, Zhenjiang, 212013, China
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
| | - Kingsley Ikechukwu Chukwudozie
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Nigeria
- Department of Clinical Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Raphael Nyaruaba
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- Center for Biosafety Megascience, Wuhan Institute of Virology, CAS, Wuhan, China
| | - Richard Ekeng Ita
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- Ritman University, Ikot Ekpene, Akwa Ibom State, Nigeria
| | - Abiodun Oladipo
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- Co-Innovation Center for Sustainable Forestry in Southern China, College of Forestry, Nanjing Forestry University, Nanjing, 210037, Jiangsu, People's Republic of China
| | - Onome Ejeromedoghene
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- School of Chemistry and Chemical Engineering, Southeast University, Jiangning District, Nanjing, Jiangsu Province, 211189, People's Republic of China
| | - Edidiong Okokon Atakpa
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya
- Institute of Marine Biology & Pharmacology, Ocean College, Zhejiang University, Zhoushan, 316021, Zhejiang, China
- Department of Animal & Environmental Biology, University of Uyo, Uyo, 1017, Akwa Ibom State, Nigeria
| | | | - Charles Obinwanne Okoye
- Organisation of African Academic Doctors (OAAD), Off Kamiti Road, Nairobi, Kenya.
- Department of Zoology & Environmental Biology, Faculty of Biological Sciences, University of Nigeria, Nsukka, 410001, Nigeria.
- School of Environment & Safety Engineering, Biofuels Institute, Jiangsu University, Zhenjiang, 212013, China.
- Key Laboratory of Intelligent Agricultural Machinery Equipment, Jiangsu University, Zhenjiang, 212013, China.
| |
Collapse
|
14
|
Li C, Xie J, Wang J, Cao Y, Pu M, Gong Q, Lu Q. Therapeutic effects and mechanisms of plant-derived natural compounds against intestinal mucositis. Front Pharmacol 2022; 13:969550. [PMID: 36210837 PMCID: PMC9533105 DOI: 10.3389/fphar.2022.969550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Accepted: 09/05/2022] [Indexed: 01/26/2023] Open
Abstract
Intestinal mucositis is a clinically related adverse reaction of antitumor treatment. Majority of patients receiving high-dose chemical therapy, radiotherapy, and bone-marrow transplant suffer from intestinal mucositis. Clinical manifestations of intestinal mucositis mainly include pain, body-weight reduction, inflammatory symptom, diarrhea, hemoproctia, and infection, which all affect regular nutritional input and enteric function. Intestinal mucositis often influences adherence to antitumor treatment because it frequently restricts the sufferer’s capacity to tolerate treatment, thus resulting in schedule delay, interruption, or premature suspension. In certain circumstances, partial and general secondary infections are found, increasing the expenditures on medical care and hospitalization. Current methods of treating intestinal mucositis are provided, which do not always counteract this disorder. Against this background, novel therapeutical measures are extremely required to prevent and treat intestinal mucositis. Plant-derived natural compounds have lately become potential candidates against enteric injury ascribed to the capacity to facilitate mucosal healing and anti-inflammatory effects. These roles are associated with the improvement of intestinal mucosal barrier, suppression of inflammatory response and oxidant stress, and modulation of gut microflora and immune system. The present article aims at systematically discussing the recent progress of plant-derived natural compounds as promising treatments for intestinal mucositis.
Collapse
Affiliation(s)
- Cailan Li
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Jianhui Xie
- The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jiahao Wang
- Department of Pharmacology, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
| | - Ying Cao
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
| | - Min Pu
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
| | - Qihai Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China
- Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, China
- *Correspondence: Qihai Gong, ; Qiang Lu,
| | - Qiang Lu
- Department of Pharmaceutical Sciences, Zunyi Medical University, Zhuhai Campus, Zhuhai, China
- *Correspondence: Qihai Gong, ; Qiang Lu,
| |
Collapse
|
15
|
Emergence and spread of antibiotic-resistant foodborne pathogens from farm to table. Food Sci Biotechnol 2022; 31:1481-1499. [PMID: 36065433 PMCID: PMC9435411 DOI: 10.1007/s10068-022-01157-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/26/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022] Open
Abstract
Antibiotics have been overused and misused for preventive and therapeutic purposes. Specifically, antibiotics are frequently used as growth promoters for improving productivity and performance of food-producing animals such as pigs, cattle, and poultry. The increasing use of antibiotics has been of great concern worldwide due to the emergence of antibiotic resistant bacteria. Food-producing animals are considered reservoirs for antibiotic resistance genes (ARGs) and residual antibiotics that transfer from the farm through the table. The accumulation of residual antibiotics can lead to additional antibiotic resistance in bacteria. Therefore, this review evaluates the risk of carriage and spread of antibiotic resistance through food chain and the potential impact of antibiotic use in food-producing animals on food safety. This review also includes in-depth discussion of promising antibiotic alternatives such as vaccines, immune modulators, phytochemicals, antimicrobial peptides, probiotics, and bacteriophages.
Collapse
|
16
|
Yadav M, Kumar T, Kanakan A, Maurya R, Pandey R, Chauhan NS. Isolation and Characterization of Human Intestinal Bacteria Cytobacillus oceanisediminis NB2 for Probiotic Potential. Front Microbiol 2022; 13:932795. [PMID: 35910631 PMCID: PMC9326467 DOI: 10.3389/fmicb.2022.932795] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Accepted: 06/17/2022] [Indexed: 11/27/2022] Open
Abstract
Systemic characterization of the human gut microbiota highlighted its vast therapeutic potential. Despite having enormous potential, the non-availability of their culture representatives created a bottleneck to understand the concept of microbiome-based therapeutics. The present study is aimed to isolate and evaluate the probiotic potential of a human gut isolate. Physiochemical, morphological, and phylogenetic characterization of a human gut isolate identifies it as a rod-shaped gram-negative microbe taxonomically affiliated with the Cytobacillus genus, having an optimal growth at 37°C in a partially alkaline environment (pH 8.0). This human gut isolate showed continuous growth in the presence of salts (up to 7% NaCl and 10% KCl), antibiotics, metals and metalloids [silver nitrate (up to 2 mM); lead acetate (up to 2 mM); sodium arsenate (up to 10 mM); potassium dichromate (up to 2 mM)], gastric and intestinal conditions, diverse temperature (25–50°C), and pH (5–9) conditions making it fit to survive in the highly variable gut environment. Genomic characterization identified the presence of gene clusters for diverse bio-catalytic activity, stress response, and antimicrobial activity, as well as it indicated the absence of pathogenic gene islands. A combination of functional features like anti-amylase, anti-lipase, glutenase, prolyl endopeptidase, lactase, bile salt hydrolase, cholesterol oxidase, and anti-pathogenic activity is indicative of its probiotic potential in various disorders. This was further substantiated by the CaCo-2 cell line assay confirming its cellular adherence and biosafety. Conclusively, human gut isolate possessed significant probiotic potential that can be used to promote animal and human health.
Collapse
Affiliation(s)
- Monika Yadav
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| | - Tarun Kumar
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
| | - Akshay Kanakan
- Integrative GENomics of Host-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
| | - Ranjeet Maurya
- Integrative GENomics of Host-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Rajesh Pandey
- Integrative GENomics of Host-PathogEn (INGEN-HOPE) Laboratory, Council of Scientific and Industrial Research-Institute of Genomics and Integrative Biology (CSIR-IGIB), New Delhi, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, India
| | - Nar Singh Chauhan
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, India
- *Correspondence: Nar Singh Chauhan
| |
Collapse
|
17
|
Li D, Li Y, Yang S, Lu J, Jin X, Wu M. Diet-gut microbiota-epigenetics in metabolic diseases: From mechanisms to therapeutics. Biomed Pharmacother 2022; 153:113290. [PMID: 35724509 DOI: 10.1016/j.biopha.2022.113290] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/26/2022] [Accepted: 06/09/2022] [Indexed: 11/02/2022] Open
Abstract
The prevalence of metabolic diseases, including obesity, dyslipidemia, type 2 diabetes mellitus (T2DM), and non-alcoholic fatty liver disease (NAFLD), is a severe burden in human society owing to the ensuing high morbidity and mortality. Various factors linked to metabolic disorders, particularly environmental factors (such as diet and gut microbiota) and epigenetic modifications, contribute to the progression of metabolic diseases. Dietary components and habits regulate alterations in gut microbiota; in turn, microbiota-derived metabolites, such as short-chain fatty acids (SCFAs), are influenced by diet. Interestingly, diet-derived microbial metabolites appear to produce substrates and enzymatic regulators for epigenetic modifications (such as DNA methylation, histone modifications, and non-coding RNA expression). Epigenetic changes mediated by microbial metabolites participate in metabolic disorders via alterations in intestinal permeability, immune responses, inflammatory reactions, and insulin resistance. In addition, microbial metabolites can trigger inflammatory immune responses and microbiota dysbiosis by directly binding to G-protein-coupled receptors (GPCRs). Hence, diet-gut microbiota-epigenetics may play a role in metabolic diseases. However, their complex relationships with metabolic diseases remain largely unknown and require further investigation. This review aimed to elaborate on the interactions among diet, gut microbiota, and epigenetics to uncover the mechanisms and therapeutics of metabolic diseases.
Collapse
Affiliation(s)
- Dan Li
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Yujuan Li
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Shengjie Yang
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Jing Lu
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Xiao Jin
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| | - Min Wu
- Guang'an men Hospital, China Academy of Chinese Medical Sciences, Beijing 100053, China.
| |
Collapse
|
18
|
Suryadi U, Hertamawati RT, Imam S. Hydrolyzation of snail (Achatina fulica) meat with rice water as novel probiotic supplements for animal feed. Vet World 2022; 15:937-942. [PMID: 35698513 PMCID: PMC9178601 DOI: 10.14202/vetworld.2022.937-942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 03/02/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: Snail meat and digestive tract hydrolyzate fermented with a consortium of preserved rice water microorganisms could serve as new sources of probiotics. Microorganisms from the examined feed supplement were isolated, identified, and characterized for resistance at low pH and with bile salts. The study aimed to determine the potential hydrolysate of the snail meat and digestive tract as a novel probiotic supplement for animal feed at various pH values and Oxgall.
Materials and Methods: The submerged fermentation method was conducted for 21 days to examine the novel probiotic that originated from snail microorganisms in the hydrolyzed liquid fermented by finely ground snail meat and the digestive tract. The microorganisms in the hydrolyzate were isolated by a spread plate method, while the potential of the probiotic hydrolyzate was tested for resistance to pH values of 2, 2.5, 3, and 4, as well as resistance to bile salts at Oxgall concentrations of 0.2%, 0.3%, 0.5%, 1%, and 2%.
Results: The hydrolyzate profile of snail meat and digestive tract contained five isolates of lactic acid bacteria that could serve as potential probiotics.
Conclusion: The application of fermentation technology using a consortium of preserved rice water microorganisms can convert snail meat and the digestive tract into novel probiotic products that could be utilized in feed supplements.
Collapse
Affiliation(s)
- Ujang Suryadi
- Department of Animal Science, Politeknik Negeri Jember, Mastrip Street PO. BOX 164, Jember, East Java, Indonesia
| | - Rosa Tri Hertamawati
- Department of Animal Science, Politeknik Negeri Jember, Mastrip Street PO. BOX 164, Jember, East Java, Indonesia
| | - Shokhirul Imam
- Department of Animal Science, Politeknik Negeri Jember, Mastrip Street PO. BOX 164, Jember, East Java, Indonesia
| |
Collapse
|
19
|
Lactobacillus paracasei CNCM I 1572: A Promising Candidate for Management of Colonic Diverticular Disease. J Clin Med 2022; 11:jcm11071916. [PMID: 35407527 PMCID: PMC8999804 DOI: 10.3390/jcm11071916] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 03/15/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023] Open
Abstract
Diverticular disease (DD) is a common gastrointestinal condition. Patients with DD experience a huge variety of chronic nonspecific symptoms, including abdominal pain, bloating, and altered bowel habits. They are also at risk of complications such as acute diverticulitis, abscess formation, hemorrhage, and perforation. Intestinal dysbiosis and chronic inflammation have recently been recognized as potential key factors contributing to disease progression. Probiotics, due to their ability to modify colonic microbiota balance and to their immunomodulatory effects, could present a promising treatment option for patients with DD. Lactobacillus paracasei CNCM I 1572 (LCDG) is a probiotic strain with the capacity to rebalance gut microbiota and to decrease intestinal inflammation. This review summarizes the available clinical data on the use of LCDG in subjects with colonic DD.
Collapse
|
20
|
Evaluation of inhibitory and probiotic properties of lactic acid bacteria isolated from vaginal microflora. Folia Microbiol (Praha) 2022; 67:427-445. [PMID: 35083726 DOI: 10.1007/s12223-021-00942-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2021] [Accepted: 12/10/2021] [Indexed: 01/02/2023]
Abstract
Lactic acid bacteria (LABs) are known to secrete species-specific secondary metabolites that could be utilized as novel therapeutics against multi-drug resistant pathogens. This study aimed to investigate the antagonistic and probiotic properties of LABs isolated from the vaginal ecosystem of healthy women and to assess the stability of their antagonistic metabolites. Among 43 strains isolated from healthy women, eight LAB strains exhibited detectable BLISs (bacteriocin-like substances) producing ability against E. faecalis (JH-86), S. aureus (JH-68), Streptococcus sp. (JH-80), and E. coli (JH-101), with zone of inhibition (ZI) ranging from 9.00 to 20.33 mm and minimum inhibitory concentrations (MICs) from 62.5 to 500 μL/mL, respectively. The partially purified compounds extracted from cell free supernatant (CFS) displayed an increase in antagonistic activity based on ZI, 9.67-30.17 mm and MICs, 3.91-15.63 mg/mL, respectively. In a time-kill study, both crude and partially purified compounds of Limosilactobacillus reuteri (MT180537), Pediococcus pentosaceus (MT176555), Limosilactobacillus pontis (MW362838), and Levilactobacillus brevis (MW362790) exhibited significant bactericidal action against E. faecalis (MW051601), the most frequent etiological agent of aerobic vaginitis (AV). The active secondary metabolites from L. reuteri (MT180537), P. pentosaceus (MT176555), and L. pontis (MW362838) were protein in nature and remained stable under different physicochemical conditions. Regarding probiotic properties, the strains presented probiotic characteristics, i.e., good acid, bile salt tolerance, aggregation properties, and biofilm formation. The strains were susceptible to most of the commonly used antibiotics and had no hemolytic activity. In conclusion, antagonistic compounds or BLIS produced by L. reuteri (MT180537) could be investigated further for preparation of ointments to treat AV.
Collapse
|
21
|
Are We What We Eat? Impact of Diet on the Gut-Brain Axis in Parkinson's Disease. Nutrients 2022; 14:nu14020380. [PMID: 35057561 PMCID: PMC8780419 DOI: 10.3390/nu14020380] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/14/2022] [Accepted: 01/14/2022] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease is characterized by motor and non-motor symptoms, such as defects in the gut function, which may occur before the motor symptoms. To date, there are therapies that can improve these symptoms, but there is no cure to avoid the development or exacerbation of this disorder. Dysbiosis of gut microbiota could have a crucial role in the gut–brain axis, which is a bidirectional communication between the central nervous system and the enteric nervous system. Diet can affect the microbiota composition, impacting gut–brain axis functionality. Gut microbiome restoration through probiotics, prebiotics, synbiotics or other dietary means could have the potential to slow PD progression. In this review, we will discuss the influence of diet on the bidirectional communication between gut and brain, thus supporting the hypothesis that this disorder could begin in the gut. We also focus on how food-based therapies might then have an influence on PD and could ameliorate non-motor as well as motor symptoms.
Collapse
|
22
|
Lee S, You H, Lee Y, Baik H, Paik J, Lee H, Park S, Shim J, Lee J, Hyun S. Intake of MPRO3 over 4 Weeks Reduces Glucose Levels and Improves Gastrointestinal Health and Metabolism. Microorganisms 2021; 10:microorganisms10010088. [PMID: 35056536 PMCID: PMC8780283 DOI: 10.3390/microorganisms10010088] [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: 12/09/2021] [Revised: 12/24/2021] [Accepted: 12/30/2021] [Indexed: 01/01/2023] Open
Abstract
Human gut microbiota are involved in different metabolic processes, such as digestion and nutrient synthesis, among others. For the elderly, supplements are a major means of maintaining health and improving intestinal homeostasis. In this study, 51 elderly women were administered MPRO3 (n = 17), a placebo (n = 16), or both (MPRO3: 1 week, placebo: 3 weeks; n = 18) for 4 weeks. The fecal microbiota were analyzed by sequencing the 16S rRNA gene V3–V4 super-variable region. The dietary fiber intake increased, and glucose levels decreased with 4-week MPRO3 intake. Reflux, indigestion, and diarrhea syndromes gradually improved with MPRO3 intake, whereas constipation was maintained. The stool shape also improved. Bifidobacterium animalis, B. pseudolongum, Lactobacillus plantarum, and L. paracasei were relatively more abundant after 4 weeks of MPRO3 intake than in those subjects after a 1-week intake. Bifidobacterium and B. longum abundances increased after 1 week of MPRO3 intake but decreased when the intake was discontinued. Among different modules and pathways, all 10 modules analyzed showed a relatively high association with 4-week MPRO3 intake. The mineral absorption pathway and cortisol biosynthesis and secretion pathways correlated with the B. animalis and B. pseudolongum abundances at 4 weeks. Therefore, 4-week MPRO3 intake decreased the fasting blood glucose level and improved intestinal health and metabolism.
Collapse
Affiliation(s)
- Songhee Lee
- Department of Biomedical Laboratory Science, Graduate School, Eulji University, 712, Dongil-ro, Uijeongbu-si 11759, Korea; (S.L.); (Y.L.)
| | - Heesang You
- Department of Senior Healthcare, Graduate School, Eulji University, 712, Dongil-ro, Uijeongbu-si 11759, Korea;
| | - Yeongju Lee
- Department of Biomedical Laboratory Science, Graduate School, Eulji University, 712, Dongil-ro, Uijeongbu-si 11759, Korea; (S.L.); (Y.L.)
| | - Haingwoon Baik
- Department of Biochemistry and Molecular Biology, Graduate School, Eulji University School of Medicine, Daejeon 34824, Korea;
| | - Jeankyung Paik
- Department of Food and Nutrition, Graduate School, Eulji University, Seongnam 13135, Korea;
| | - Hayera Lee
- R&BD Center, hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Korea; (H.L.); (S.P.); (J.S.); (J.L.)
| | - Soodong Park
- R&BD Center, hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Korea; (H.L.); (S.P.); (J.S.); (J.L.)
| | - Jaejung Shim
- R&BD Center, hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Korea; (H.L.); (S.P.); (J.S.); (J.L.)
| | - Junglyoul Lee
- R&BD Center, hy Co., Ltd., 22, Giheungdanji-ro 24beon-gil, Giheung-gu, Yongin-si 17086, Korea; (H.L.); (S.P.); (J.S.); (J.L.)
| | - Sunghee Hyun
- Department of Biomedical Laboratory Science, Graduate School, Eulji University, 712, Dongil-ro, Uijeongbu-si 11759, Korea; (S.L.); (Y.L.)
- Department of Senior Healthcare, Graduate School, Eulji University, 712, Dongil-ro, Uijeongbu-si 11759, Korea;
- Correspondence: ; Tel.: +82-10-9412-8853
| |
Collapse
|
23
|
Diallo D, Somboro AM, Diabate S, Baya B, Kone A, Sarro YS, Kone B, Diarra B, Diallo S, Diakite M, Doumbia S, Toloba Y, Murphy RL, Maiga M. Antituberculosis Therapy and Gut Microbiota: Review of Potential Host Microbiota Directed-Therapies. Front Cell Infect Microbiol 2021; 11:673100. [PMID: 34950603 PMCID: PMC8688706 DOI: 10.3389/fcimb.2021.673100] [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: 02/26/2021] [Accepted: 11/12/2021] [Indexed: 12/29/2022] Open
Abstract
Tuberculosis (TB) remains a major public health concern with millions of deaths every year. The overlap with HIV infections, long treatment duration, and the emergence of drug resistance are significant obstacles to the control of the disease. Indeed, the standard first-line regimen TB treatment takes at least six months and even longer for the second-line therapy, resulting in relapses, drug resistance and re-infections. Many recent reports have also shown prolonged and significant damage of the gut microbial community (dysbiosis) from anti-TB drugs that can detrimentally persist several months after the cessation of treatment and could lead to the impairment of the immune response, and thus re-infections and drug resistance. A proposed strategy for shortening the treatment duration is thus to apply corrective measures to the dysbiosis for a faster bacterial clearance and a better treatment outcome. In this review, we will study the role of the gut microbiota in both TB infection and treatment, and its potential link with treatment duration. We will also discuss, the new concept of "Host Microbiota Directed-Therapies (HMDT)" as a potential adjunctive strategy to improve the treatment effectiveness, reduce its duration and or prevent relapses. These strategies include the use of probiotics, prebiotics, gut microbiota transfer, and other strategies. Application of this innovative solution could lead to HMDT as an adjunctive tool to shorten TB treatment, which will have enormous public health impacts for the End TB Strategy worldwide.
Collapse
Affiliation(s)
- Dramane Diallo
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Anou M Somboro
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali.,School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban, South Africa
| | - Seydou Diabate
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Bacar Baya
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Amadou Kone
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Yeya S Sarro
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Bourahima Kone
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Bassirou Diarra
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Souleymane Diallo
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Mahamadou Diakite
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Seydou Doumbia
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Yacouba Toloba
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali
| | - Robert L Murphy
- Institute for Global Health, Northwestern University, Chicago, IL, United States
| | - Mamoudou Maiga
- University Clinical Research Center (UCRC) of the University of Sciences, Techniques and Technologies of Bamako (USTTB), Bamako, Mali.,Institute for Global Health, Northwestern University, Chicago, IL, United States
| |
Collapse
|
24
|
Comberiati P, Di Cicco M, Paravati F, Pelosi U, Di Gangi A, Arasi S, Barni S, Caimmi D, Mastrorilli C, Licari A, Chiera F. The Role of Gut and Lung Microbiota in Susceptibility to Tuberculosis. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph182212220. [PMID: 34831976 PMCID: PMC8623605 DOI: 10.3390/ijerph182212220] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/16/2021] [Accepted: 11/18/2021] [Indexed: 12/12/2022]
Abstract
Tuberculosis is one of the most common infectious diseases and infectious causes of death worldwide. Over the last decades, significant research effort has been directed towards defining the understanding of the pathogenesis of tuberculosis to improve diagnosis and therapeutic options. Emerging scientific evidence indicates a possible role of the human microbiota in the pathophysiology of tuberculosis, response to therapy, clinical outcomes, and post-treatment outcomes. Although human studies on the role of the microbiota in tuberculosis are limited, published data in recent years, both from experimental and clinical studies, suggest that a better understanding of the gut-lung microbiome axis and microbiome-immune crosstalk could shed light on the specific pathogenetic mechanisms of Mycobacterium tuberculosis infection and identify new therapeutic targets. In this review, we address the current knowledge of the host immune responses against Mycobacterium tuberculosis infection, the emerging evidence on how gut and lung microbiota can modulate susceptibility to tuberculosis, the available studies on the possible use of probiotic-antibiotic combination therapy for the treatment of tuberculosis, and the knowledge gaps and future research priorities in this field.
Collapse
Affiliation(s)
- Pasquale Comberiati
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.D.C.); (A.D.G.)
- Allergology and Pulmonology Section, Pediatrics Unit, Pisa University Hospital, 56126 Pisa, Italy
- Department of Clinical Immunology and Allergology, I.M. Sechenov First Moscow State Medical University, Moscow 119991, Russia
- Correspondence:
| | - Maria Di Cicco
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.D.C.); (A.D.G.)
- Allergology and Pulmonology Section, Pediatrics Unit, Pisa University Hospital, 56126 Pisa, Italy
| | - Francesco Paravati
- Department of Pediatrics, San Giovanni di Dio Hospital, 88900 Crotone, Italy; (F.P.); (F.C.)
| | - Umberto Pelosi
- Pediatric Unit, Santa Barbara Hospital, 09016 Iglesias, Italy;
| | - Alessandro Di Gangi
- Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy; (M.D.C.); (A.D.G.)
- Allergology and Pulmonology Section, Pediatrics Unit, Pisa University Hospital, 56126 Pisa, Italy
| | - Stefania Arasi
- Area of Translational Research in Pediatric Specialities, Allergy Unit, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy;
| | - Simona Barni
- Allergic Unit, Department of Pediatric, Meyer Children’s Hospital, 50139 Florence, Italy;
| | - Davide Caimmi
- Allergy Unit, CHU de Montpellier, Université de Montpellier, 34295 Montpellier, France;
- IDESP, UMR A11, Université de Montpellier, 34093 Montpellier, France
| | - Carla Mastrorilli
- Department of Pediatrics, University Hospital Consortium Corporation Polyclinic of Bari, Pediatric Hospital Giovanni XXIII, 70124 Bari, Italy;
| | - Amelia Licari
- Pediatric Clinic, Pediatrics Department, Policlinico San Matteo, University of Pavia, 27100 Pavia, Italy;
| | - Fernanda Chiera
- Department of Pediatrics, San Giovanni di Dio Hospital, 88900 Crotone, Italy; (F.P.); (F.C.)
| |
Collapse
|
25
|
Yadav M, Chauhan NS. Microbiome therapeutics: exploring the present scenario and challenges. Gastroenterol Rep (Oxf) 2021; 10:goab046. [PMID: 35382166 PMCID: PMC8972995 DOI: 10.1093/gastro/goab046] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 09/24/2021] [Accepted: 09/25/2021] [Indexed: 12/11/2022] Open
Abstract
Human gut-microbiome explorations have enriched our understanding of microbial colonization, maturation, and dysbiosis in health-and-disease subsets. The enormous metabolic potential of gut microbes and their role in the maintenance of human health is emerging, with new avenues to use them as therapeutic agents to overcome human disorders. Microbiome therapeutics are aimed at engineering the gut microbiome using additive, subtractive, or modulatory therapy with an application of native or engineered microbes, antibiotics, bacteriophages, and bacteriocins. This approach could overcome the limitation of conventional therapeutics by providing personalized, harmonized, reliable, and sustainable treatment. Its huge economic potential has been shown in the global therapeutics market. Despite the therapeutic and economical potential, microbiome therapeutics is still in the developing stage and is facing various technical and administrative issues that require research attention. This review aims to address the current knowledge and landscape of microbiome therapeutics, provides an overview of existing health-and-disease applications, and discusses the potential future directions of microbiome modulations.
Collapse
Affiliation(s)
- Monika Yadav
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, India
| | - Nar Singh Chauhan
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, Haryana, India
| |
Collapse
|
26
|
Qiao N, Du G, Zhong X, Sun X. Recombinant lactic acid bacteria as promising vectors for mucosal vaccination. EXPLORATION (BEIJING, CHINA) 2021; 1:20210026. [PMID: 37323212 PMCID: PMC10191043 DOI: 10.1002/exp.20210026] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 09/13/2021] [Indexed: 06/15/2023]
Abstract
Lactic acid bacteria (LAB), a diverse family of gram-positive bacteria, has been proven effective in delivering varieties of therapeutic and prophylactic molecules such as antigens and cytokines. Featuring the properties of acid-resistant, high uptake into Peyer's patches, and superior capacity for inducing secretory IgA antibodies, LAB have good potential to be used as vaccine vectors for mucosal vaccination. Mucosal immunization enables both mucosal and systemic immune responses, which are critical for resisting pathogens that invade the host through the mucosal surfaces. With the development of genetic engineering, LAB strains, primarily Lactococcus and Lactobacillus have been exploited to express a range of heterologous antigens. Numerous studies have demonstrated that LAB mucosal vaccines can stimulate all arms of the immune system to provide adequate protection against pathogen infections. Additionally, several LAB-based human papillomavirus vaccines have entered the clinical trial studies, which suggest the great promise of LAB vaccines for new interventions in mucosal transport diseases. Herein, we will discuss the factors that influence the immunogenicity of LAB vaccines, including LAB strains, the location of antigens, and administration routes, and focus on the current strategies that have been reported for optimizing LAB vaccines.
Collapse
Affiliation(s)
- Nan Qiao
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan ProvinceSichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of Pharmacy, Sichuan UniversityChengduChina
| | - Guangsheng Du
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan ProvinceSichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of Pharmacy, Sichuan UniversityChengduChina
| | - Xiaofang Zhong
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan ProvinceSichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of Pharmacy, Sichuan UniversityChengduChina
| | - Xun Sun
- Key Laboratory of Drug‐Targeting and Drug Delivery System of the Education Ministry and Sichuan ProvinceSichuan Engineering Laboratory for Plant‐Sourced Drug and Sichuan Research Center for Drug Precision Industrial TechnologyWest China School of Pharmacy, Sichuan UniversityChengduChina
| |
Collapse
|
27
|
Lei Q, Wu T, Wu J, Hu X, Guan Y, Wang Y, Yan J, Shi G. Roles of α‑synuclein in gastrointestinal microbiome dysbiosis‑related Parkinson's disease progression (Review). Mol Med Rep 2021; 24:734. [PMID: 34414447 PMCID: PMC8404091 DOI: 10.3892/mmr.2021.12374] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/16/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disease amongst the middle-aged and elderly populations. Several studies have confirmed that the microbiota-gut-brain axis (MGBA) serves a key role in the pathogenesis of PD. Changes to the gastrointestinal microbiome (GM) cause misfolding and abnormal aggregation of α-synuclein (α-syn) in the intestine. Abnormal α-syn is not eliminated via physiological mechanisms and is transported into the central nervous system (CNS) via the vagus nerve. The abnormal levels of α-syn aggregate in the substantia nigra pars compacta, not only leading to the formation of eosinophilic Lewis Bodies in the cytoplasm and mitochondrial dysfunction in dopaminergic (DA) neurons, but also leading to the stimulation of an inflammatory response in the microglia. These pathological changes result in an increase in oxidative stress (OS), which triggers nerve cell apoptosis, a characteristic of PD. This increase in OS further oxidizes and intensifies abnormal aggregation of α-syn, eventually forming a positive feedback loop. The present review discusses the abnormal accumulation of α-syn in the intestine caused by the GM changes and the increased levels of α-syn transport to the CNS via the MGBA, resulting in the loss of DA neurons and an increase in the inflammatory response of microglial cells in the brain of patients with PD. In addition, relevant clinical therapeutic strategies for improving the GM and reducing α-syn accumulation to relieve the symptoms and progression of PD are described.
Collapse
Affiliation(s)
- Qingchun Lei
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Tingting Wu
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Jin Wu
- Department of Neurosurgery, Puer People's Hospital, Pu'er, Yunnan 665000, P.R. China
| | - Xiaogang Hu
- Department of Neurosurgery, Puer People's Hospital, Pu'er, Yunnan 665000, P.R. China
| | - Yingxia Guan
- Department of Vasculocardiology, The Affiliated Hospital of Yunnan University, Kunming, Yunnan 650021, P.R. China
| | - Ying Wang
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Jinyuan Yan
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| | - Guolin Shi
- Department of Neurosurgery, The Second Affiliated Hospital of Kunming Medical University, Kunming, Yunnan 650101, P.R. China
| |
Collapse
|
28
|
Zheng SY, Li HX, Xu RC, Miao WT, Dai MY, Ding ST, Liu HD. Potential roles of gut microbiota and microbial metabolites in Parkinson's disease. Ageing Res Rev 2021; 69:101347. [PMID: 33905953 DOI: 10.1016/j.arr.2021.101347] [Citation(s) in RCA: 48] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 04/06/2021] [Accepted: 04/22/2021] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a complicated neurodegenerative disease attributed to multifactorial changes. However, its pathological mechanism remains undetermined. Accumulating evidence has revealed the emerging functions of gut microbiota and microbial metabolites, which can affect both the enteric nervous system and the central nervous system via the microbiota-gut-brain axis. Accordingly, intestinal dysbiosis might be closely associated with PD. This review explores alterations to gut microbiota, correlations with clinical manifestations of PD, and briefly probes the underlying mechanisms. Next, the highly controversial roles of microbial metabolites including short-chain fatty acids (SCFAs), H2 and H2S are discussed. Finally, the pros and cons of the current treatments for PD, including those targeting microbiota, are assessed. Advancements in research techniques, further studies on levels of specific strains and longitudinal prospective clinical trials are urgently needed for the identification of early diagnostic markers and the development of novel therapeutic approaches for PD.
Collapse
|
29
|
Rahardja F, Prasetyo D, Shahib MN, Tjahjani S. The Influence of Lactobacillus Acidophilus on MUC1, GAL-3, IL-1β and IL-17 Gene Expression in BALB/c Mice Stomach. Open Microbiol J 2021. [DOI: 10.2174/1874285802115010067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Background and Objective:
Lactobacillus acidophilus has been widely used for the management of gastrointestinal carcinoma owing to its immunomodulation effect; however, the role of L. acidophilus and its specific mechanism of action in the stomach is not fully comprehended. The present study evaluated the expression profile of MUC-1, GAL-3, IL -1β, and IL-17 in the L. acidophilus treated mice stomach.
Methods:
The study was conducted utilizing three groups of mice, 6 mice for each group, administered with different doses of L. acidophilus and a control group treated with normal saline. The results were analyzed with the Mann-Whitney Test.
Results:
The results demonstrated that L. acidophilus elevated IL-1β insignificantly and inhibited the expression of IL-17. The MUC-1 expression is influenced by L. acidophilus and inversely proportional to GAL-3 expression.
Conclusion:
Lactobacillus acidophilus plays a prominent role against inflammatory responses and has a potential in the treatment of gastrointestinal cancer.
Collapse
|
30
|
Barreto MO, Soust M, Moore RJ, Olchowy TWJ, Alawneh JI. Systematic review and meta-analysis of probiotic use on inflammatory biomarkers and disease prevention in cattle. Prev Vet Med 2021; 194:105433. [PMID: 34298303 DOI: 10.1016/j.prevetmed.2021.105433] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Revised: 07/08/2021] [Accepted: 07/11/2021] [Indexed: 10/20/2022]
Abstract
The aim of this study was to appraise the available evidence on the effectiveness of probiotic treatment on mature cattle immunity, inflammation, and disease prevention. A systematic review with meta-analysis was conducted to analyse studies that were eligible to answer the following research question: "in cattle of at least 6-months of age, is the use of probiotics associated with immunomodulatory and inflammatory responses, and clinical disease outcomes?" Our literature search yielded 25 studies that fit the inclusion criteria. From these studies, only 19 were suitable for inclusion in the meta-analysis due to data limitations and differences in study population characteristics. Included studies were assessed for bias using a risk assessment tool adapted from the Cochrane Collaboration's tool for assessing risk of bias in randomised trials. GRADE guidelines were used to assess the quality of the body of evidence at the outcome level. The meta-analysis was performed using Review Manager and R. The overall quality of evidence at the outcome level was assessed as being very low. On average, the treatment effect on immunoglobulin G (IgG), serum amyloid A (SAA), haptoglobin (Hp) and β-hydroxybutyrate (BoHB) for cows receiving probiotics did not differ from control cows. Exposure to probiotics was not associated with reduced risk of reproductive disorders (pooled RR = 1.02 95 % CI = 0.81-1.27, P = 0.88). There is insufficient evidence to support any significant positive effects of probiotics on cattle immunity and disease prevention. This lack of consistent evidence could be due to dissimilarities in the design of the included studies such as differences in dosage, dose schedule, diet composition and/or physiological state of the host at the time of treatment.
Collapse
Affiliation(s)
- Michelle O Barreto
- The University of Queensland, School of Veterinary Science, Gatton, Queensland, 4343, Australia; The University of Queensland, Good Clinical Practice Research Group (GCPRG), Gatton, Queensland, 4343, Australia
| | - Martin Soust
- Terragen Biotech Pty Ltd., Coolum Beach, Queensland, 4573, Australia
| | - Robert J Moore
- School of Science, RMIT University, Bundoora, Melbourne, Victoria, 3083, Australia
| | - Timothy W J Olchowy
- The University of Queensland, Good Clinical Practice Research Group (GCPRG), Gatton, Queensland, 4343, Australia; Faculty of Veterinary Medicine, University of Calgary, Calgary, Alberta, T3R 1J3, Canada
| | - John I Alawneh
- The University of Queensland, School of Veterinary Science, Gatton, Queensland, 4343, Australia; The University of Queensland, Good Clinical Practice Research Group (GCPRG), Gatton, Queensland, 4343, Australia; Murdoch University, School of Veterinary Medicine, Perth, Western Australia, 6150, Australia.
| |
Collapse
|
31
|
Lee CS, Kim SH. Anti-inflammatory and Anti-osteoporotic Potential of Lactobacillus plantarum A41 and L. fermentum SRK414 as Probiotics. Probiotics Antimicrob Proteins 2021; 12:623-634. [PMID: 31372901 DOI: 10.1007/s12602-019-09577-y] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
This study involves an investigation on the probiotic properties of lactic acid bacteria and their potential applications in an in vitro model of lipopolysaccharide (LPS)-stimulated inflammation and dexamethasone-induced osteoporosis. Nine strains were pre-screened from 485 lactic acid bacteria based on their survival at a low pH and in a solution containing bile salts. All candidates were capable of surviving in an environment with low pH and with bile salts and could successfully colonize the intestine. Furthermore, their functional properties, such as anti-oxidation and anti-inflammation, were evaluated. Of the nine probiotic candidates, Lactobacillus plantarum A41 and L. fermentum SRK414 exhibited the highest anti-oxidative capacity. Moreover, only L. plantarum A41 and L. fermentum SRK414 could increase gut barrier function by upregulating the mRNA expression of tight junction proteins and inhibit the expression of inflammatory mediators induced by LPS-stimulated inflammation. Interestingly, these two strains were also capable of regulating several bone metabolism-related markers playing a role in bone homeostasis and osteoblast differentiation. In brief, L. plantarum A41 and L. fermentum SRK414 exhibited high probiotic potential and potentially impact immune-related bone health by modulating pro-inflammatory cytokines and bone metabolism-related markers.
Collapse
Affiliation(s)
- Chul Sang Lee
- College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea.,Institute of Life Science and Natural Resources, Korea University, Seoul, 02841, Republic of Korea
| | - Sae Hun Kim
- College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea. .,Institute of Life Science and Natural Resources, Korea University, Seoul, 02841, Republic of Korea.
| |
Collapse
|
32
|
Ng E, Tay JRH, Ong MMA, Bostanci N, Belibasakis GN, Seneviratne CJ. Probiotic therapy for periodontal and peri-implant health - silver bullet or sham? Benef Microbes 2021; 12:215-230. [PMID: 34057054 DOI: 10.3920/bm2020.0182] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Probiotics are thought to be beneficial microbes that influence health-related outcomes through host immunomodulation and modulation of the bacteriome. Its reported success in the treatment of gastrointestinal disorders has led to further research on its potential applicability within the dental field due to similarities such as a polymicrobial aetiology and disease associated microbial-shifts. Although the literature is replete with studies demonstrating its efficacy, the use of probiotics in dentistry continues to polarise opinion. Here, we explore the evidence for probiotics and its effect on periodontal and peri-implant health. MEDLINE, EMBASE, and CENTRAL were systemically searched from June 2010 to June 2020 based on a formulated search strategy. Of 1,956 potentially relevant articles, we selected 27 double-blinded randomised clinical trials in the areas of gingivitis, periodontitis, residual pockets during supportive periodontal therapy, and peri-implant diseases, and reviewed their efficacy in these clinical situations. We observed substantial variation in treatment results and protocols between studies. Overall, the evidence for probiotic therapy for periodontal and peri-implant health appears unconvincing. The scarcity of trials with adequate power and follow-up precludes any meaningful clinical recommendations. Thus, the routine use of probiotics for these purposes are currently unsubstantiated. Further multi-centre trials encompassing a standardised investigation on the most promising strains and administration methods, with longer observation times are required to confirm the benefits of probiotic therapy for these applications.
Collapse
Affiliation(s)
- E Ng
- Department of Restorative Dentistry, National Dental Centre Singapore, 5 Second Hospital Ave, 168938, Singapore
| | - J R H Tay
- Department of Restorative Dentistry, National Dental Centre Singapore, 5 Second Hospital Ave, 168938, Singapore
| | - M M A Ong
- Department of Restorative Dentistry, National Dental Centre Singapore, 5 Second Hospital Ave, 168938, Singapore.,Oral Health Academic Clinical Programme, Duke-NUS Medical School, 8 College Road, 169857, Singapore
| | - N Bostanci
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, P.O. Box 4064, 14104 Huddinge, Sweden
| | - G N Belibasakis
- Division of Oral Diseases, Department of Dental Medicine, Karolinska Institutet, P.O. Box 4064, 14104 Huddinge, Sweden
| | - C J Seneviratne
- Oral Health Academic Clinical Programme, Duke-NUS Medical School, 8 College Road, 169857, Singapore.,Singapore Oral Microbiomics Initiative, National Dental Research Institute Singapore, National Dental Centre Singapore, Second Hospital Ave, 168938, Singapore
| |
Collapse
|
33
|
Vega L, Herrera G, Muñoz M, Patarroyo MA, Maloney JG, Santín M, Ramírez JD. Gut microbiota profiles in diarrheic patients with co-occurrence of Clostridioides difficile and Blastocystis. PLoS One 2021; 16:e0248185. [PMID: 33725006 PMCID: PMC7963057 DOI: 10.1371/journal.pone.0248185] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 02/19/2021] [Indexed: 02/07/2023] Open
Abstract
Blastocystis and Clostridioides difficile co-occurrence is considered a rare event since the colonization by Blastocystis is prevented under a decrease in beneficial bacteria in the microbiota when there is C. difficile infection (CDI). This scenario has been reported once, but no information on the gut microbiota profiling is available. The present study is motivated by knowing which members of the microbiota can be found in this rare scenario and how this co-occurrence may impact the abundance of other bacteria, eukaryotes or archaea present in the gut microbiota. This study aimed to describe the bacterial and eukaryotic communities using amplicon-based sequencing of the 16S- and 18S-rRNA regions of three patient groups: (1) Blastocystis and C. difficile infection (B+/C+, n = 31), (2) C. difficile infection only (B˗/C+, n = 44), and (3) without Blastocystis or C. difficile (B˗/C˗, n = 40). Blastocystis was subtyped using amplicon-based sequencing of the 18S-rRNA gene, revealing circulation of subtypes ST1 (43.4%), ST3 (35.85%) and ST5 (20.75%) among the study population. We found that B+/C+ patients had a higher abundance of some beneficial bacteria (such as butyrate producers or bacteria with anti-inflammatory properties) compared with non-Blastocystis-colonized patients, which may suggest a shift towards an increase in beneficial bacteria when Blastocystis colonizes patients with CDI. Regarding eukaryotic communities, statistical differences in the abundance of some eukaryotic genera between the study groups were not observed. Thus, this study provides preliminary descriptive information of a potential microbiota profiling of differential presence by Blastocystis and C. difficile.
Collapse
Affiliation(s)
- Laura Vega
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Giovanny Herrera
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Marina Muñoz
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
| | - Manuel A. Patarroyo
- Molecular Biology and Immunology Department, Fundación Instituto de Inmunología de Colombia (FIDIC), Bogotá, Colombia
- Microbiology Department, Faculty of Medicine, Universidad Nacional de Colombia, Bogotá, Colombia
| | - Jenny G. Maloney
- USDA ARS, Environmental Microbial and Food Safety Laboratory, BARC, Beltsville, MD, United States of America
| | - Monica Santín
- USDA ARS, Environmental Microbial and Food Safety Laboratory, BARC, Beltsville, MD, United States of America
| | - Juan David Ramírez
- Centro de Investigaciones en Microbiología y Biotecnología-UR (CIMBIUR), Facultad de Ciencias Naturales, Universidad del Rosario, Bogotá, Colombia
- * E-mail:
| |
Collapse
|
34
|
Probiotic microorganisms and herbs in ruminant nutrition as natural modulators of health and production efficiency – a review. ANNALS OF ANIMAL SCIENCE 2021. [DOI: 10.2478/aoas-2020-0081] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Probiotics, prebiotics, synbiotics, direct-fed microbials, and herbs may improve the production efficiency in ruminants. The beneficial effect of selected specific microbes on animal health is reflected in protection against pathogens, stimulation of immunological response, increased production capacity, and mitigation of stress effects. Phytobiotic plants used in the nutrition of ruminant animals increase feed palatability. This in turn has a positive effect on feed intake and, consequently, increases production performance. Pectins, terpenes, phenols, saponins, and antibioticlike substances contained in phytobiotics prevent irritation, diarrhea, and increase the activity of digestive enzymes. Thanks to the abundance of biologically active substances such as flavonoids, glycosides, coumarins, carotenoids, polyphenols, etc., phytobiotics exhibit immunostimulatory and antioxidant properties as well. Given such a wide range of effects on health status and production parameters in animals, an attempt was made in this review to compile the current knowledge on the possible application of these natural growth stimulants in ruminant nutrition and to demonstrate their potential benefits and/or risks for breeding these animals.
Collapse
|
35
|
Abboud M, Rizk R, AlAnouti F, Papandreou D, Haidar S, Mahboub N. The Health Effects of Vitamin D and Probiotic Co-Supplementation: A Systematic Review of Randomized Controlled Trials. Nutrients 2020; 13:nu13010111. [PMID: 33396898 PMCID: PMC7824176 DOI: 10.3390/nu13010111] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 12/24/2020] [Accepted: 12/24/2020] [Indexed: 12/14/2022] Open
Abstract
Evidence of synergic health effects of co-supplementation with vitamin D and probiotics is emerging. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses PRISMA statement, scientific databases and the grey literature were searched, and a narrative review and risk of bias assessment were conducted. Seven randomized controlled trials were included, which had low risk of bias. Six studies were double-blind, and once single-blind, extended over 6–12 weeks, and included 50–105 participants. Conditions explored included schizophrenia, gestational diabetes, type 2 diabetes and coronary heart disease, polycystic ovarian syndrome, osteopenia, irritable bowel syndrome (IBS), and infantile colic. Supplementation frequency was daily or bi-monthly, with mainly vitamin D3, and Lactobacillus, Bifidobacterium, and Streptococcus. Comparators were placebo, vitamin D, lower vitamin D dose, and probiotics and lower vitamin D dose. The co-supplementation yielded greater health benefits than its comparators did in all studies except in one assessing IBS. Beneficial effects included decreased disease severity, improved mental health, metabolic parameters, mainly insulin sensitivity, dyslipidemia, inflammation, and antioxidative capacity, and lower use of healthcare. Co-supplementation of vitamin D and probiotics generated greater health benefits than its comparators did. More studies in other diseases and various populations are needed to confirm these findings and to elucidate the optimal form, composition, and frequency of this co-supplementation.
Collapse
Affiliation(s)
- Myriam Abboud
- Department of Health, College of Natural and Health Sciences, Zayed University, Dubai 19282, UAE; (F.A.); (D.P.)
- Correspondence:
| | - Rana Rizk
- Institut National de Santé Publique, d’Épidémiologie Clinique et de Toxicologie (INSPECT-Lb), Beirut, Lebanon;
| | - Fatme AlAnouti
- Department of Health, College of Natural and Health Sciences, Zayed University, Dubai 19282, UAE; (F.A.); (D.P.)
| | - Dimitrios Papandreou
- Department of Health, College of Natural and Health Sciences, Zayed University, Dubai 19282, UAE; (F.A.); (D.P.)
| | - Suzan Haidar
- Department of Nutrition and Food Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut 657314, Lebanon; (S.H.); (N.M.)
| | - Nadine Mahboub
- Department of Nutrition and Food Sciences, Faculty of Arts and Sciences, Lebanese International University, Beirut 657314, Lebanon; (S.H.); (N.M.)
- Department of Health Promotion, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6229 GT Maastricht, The Netherlands
| |
Collapse
|
36
|
van Zyl WF, Deane SM, Dicks LM. Molecular insights into probiotic mechanisms of action employed against intestinal pathogenic bacteria. Gut Microbes 2020; 12:1831339. [PMID: 33112695 PMCID: PMC7595611 DOI: 10.1080/19490976.2020.1831339] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Gastrointestinal (GI) diseases, and in particular those caused by bacterial infections, are a major cause of morbidity and mortality worldwide. Treatment is becoming increasingly difficult due to the increase in number of species that have developed resistance to antibiotics. Probiotic lactic acid bacteria (LAB) have considerable potential as alternatives to antibiotics, both in prophylactic and therapeutic applications. Several studies have documented a reduction, or prevention, of GI diseases by probiotic bacteria. Since the activities of probiotic bacteria are closely linked with conditions in the host's GI-tract (GIT) and changes in the population of enteric microorganisms, a deeper understanding of gut-microbial interactions is required in the selection of the most suitable probiotic. This necessitates a deeper understanding of the molecular capabilities of probiotic bacteria. In this review, we explore how probiotic microorganisms interact with enteric pathogens in the GIT. The significance of probiotic colonization and persistence in the GIT is also addressed.
Collapse
Affiliation(s)
- Winschau F. van Zyl
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Shelly M. Deane
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa
| | - Leon M.T. Dicks
- Department of Microbiology, Stellenbosch University, Stellenbosch, South Africa,CONTACT Leon M.T. Dicks; Department of Microbiology; Stellenbosch University, Stellenbosch7602, South Africa
| |
Collapse
|
37
|
Younis N, Mahasneh A. Probiotics and the envisaged role in treating human infertility. MIDDLE EAST FERTILITY SOCIETY JOURNAL 2020. [DOI: 10.1186/s43043-020-00039-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
Abstract
Background
Considerable attention is being directed nowadays towards using probiotics as an alternative therapy in treating several human diseases including gastrointestinal tract diseases especially colorectal cancers, cardiovascular diseases, hyperlipidemia, and blood pressure cases. However, infertility as affected by the microbiome and the probable role of probiotics in alleviating infertility problems did not receive the deserved attention, especially in IVF patients with male or female factors.
Main body
In this review, we tried to draw the attention of researchers in the medical settings to the importance of the forthcoming role of probiotics use in elucidating the role of the microbiome in infertile patients. The hope is to attain the best performance of both male and female reproductive systems and to shed some light on infertility problems.
Conclusion
More in vivo experiments are still needed to address many aspects of probiotics like proper administration, exact functional strains, required dose, application method, duration of treatment, and combination with antibiotics before considering probiotics as an alternative treatment.
Collapse
|
38
|
Brown EG, Goldman SM. Modulation of the Microbiome in Parkinson's Disease: Diet, Drug, Stool Transplant, and Beyond. Neurotherapeutics 2020; 17:1406-1417. [PMID: 33034846 PMCID: PMC7851230 DOI: 10.1007/s13311-020-00942-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/28/2020] [Indexed: 12/13/2022] Open
Abstract
The gastrointestinal microbiome is altered in Parkinson's disease and likely plays a key role in its pathophysiology, affecting symptoms and response to therapy and perhaps modifying progression or even disease initiation. Gut dysbiosis therefore has a significant potential as a therapeutic target in Parkinson's disease, a condition elusive to disease-modifying therapy thus far. The gastrointestinal environment hosts a complex ecology, and efforts to modulate the relative abundance or function of established microorganisms are still in their infancy. Still, these techniques are being rapidly developed and have important implications for our understanding of Parkinson's disease. Currently, modulation of the microbiome can be achieved through non-pharmacologic means such as diet, pharmacologically through probiotic, prebiotic, or antibiotic use and procedurally through fecal transplant. Novel techniques being explored include the use of small molecules or genetically engineered organisms, with vast potential. Here, we review how some of these approaches have been used to date, important areas of ongoing research, and how microbiome modulation may play a role in the clinical management of Parkinson's disease in the future.
Collapse
Affiliation(s)
- Ethan G Brown
- Division of Movement Disorders and Neuromodulation, Weill Institute of Neurology, University of California, San Francisco, CA, USA.
| | - Samuel M Goldman
- Division of Movement Disorders and Neuromodulation, Weill Institute of Neurology, University of California, San Francisco, CA, USA
- Division of Occupational and Environmental Medicine, University of California, San Francisco, CA, USA
| |
Collapse
|
39
|
KOWALSKA JUSTYNAD, NOWAK ADRIANA, ŚLIŻEWSKA KATARZYNA, STAŃCZYK MAŁGORZATA, ŁUKASIAK MAGDALENA, DASTYCH JAROSŁAW. Anti-Salmonella Potential of New Lactobacillus Strains with the Application in the Poultry Industry. Pol J Microbiol 2020; 69:5-18. [PMID: 32189480 PMCID: PMC7256722 DOI: 10.33073/pjm-2020-001] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/18/2019] [Accepted: 12/02/2019] [Indexed: 12/22/2022] Open
Abstract
Probiotics are considered an alternative to antibiotics in the prevention and treatment of Salmonella diseases in poultry. However, to use probiotics as proposed above, it is necessary to evaluate their properties in detail and to select the most effective bacterial strains in the application targeted. In this study, probiotic properties of new Lactobacillus sp. strains were investigated and their antimicrobial activity against 125 environmental strains of Salmonella sp. was determined using the agar slab method. Furthermore, their survival in the presence of bile salts and at low pH, antibiotics susceptibility, aggregation and coaggregation ability, adherence to polystyrene and Caco-2 cells, and cytotoxicity were investigated. Each strain tested showed antagonistic activity against at least 96% of the environmental Salmonella sp. strains and thus representing a highly epidemiologically differentiated collection of poultry isolates. In addition, the probiotic properties of new Lactobacillus strains are promising. Therefore, all strains examined showed a high potential for use in poultry against salmonellosis. Probiotics are considered an alternative to antibiotics in the prevention and treatment of Salmonella diseases in poultry. However, to use probiotics as proposed above, it is necessary to evaluate their properties in detail and to select the most effective bacterial strains in the application targeted. In this study, probiotic properties of new Lactobacillus sp. strains were investigated and their antimicrobial activity against 125 environmental strains of Salmonella sp. was determined using the agar slab method. Furthermore, their survival in the presence of bile salts and at low pH, antibiotics susceptibility, aggregation and coaggregation ability, adherence to polystyrene and Caco-2 cells, and cytotoxicity were investigated. Each strain tested showed antagonistic activity against at least 96% of the environmental Salmonella sp. strains and thus representing a highly epidemiologically differentiated collection of poultry isolates. In addition, the probiotic properties of new Lactobacillus strains are promising. Therefore, all strains examined showed a high potential for use in poultry against salmonellosis.
Collapse
Affiliation(s)
| | - ADRIANA NOWAK
- Department of Environmental Biotechnology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Lodz, Poland
| | - KATARZYNA ŚLIŻEWSKA
- Institute of Fermentation Technology and Microbiology, Faculty of Biotechnology and Food Sciences, Lodz University of Technology, Lodz, Poland
| | | | | | | |
Collapse
|
40
|
Isolation and safety characterisation of lactobacilli strains with antimicrobial properties as potential probiotics for human use. Lebensm Wiss Technol 2020. [DOI: 10.1016/j.lwt.2020.109796] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
41
|
Probiotics and Preterm Infants: A Position Paper by the European Society for Paediatric Gastroenterology Hepatology and Nutrition Committee on Nutrition and the European Society for Paediatric Gastroenterology Hepatology and Nutrition Working Group for Probiotics and Prebiotics. J Pediatr Gastroenterol Nutr 2020; 70:664-680. [PMID: 32332478 DOI: 10.1097/mpg.0000000000002655] [Citation(s) in RCA: 126] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
More than 10,000 preterm infants have participated in randomised controlled trials on probiotics worldwide, suggesting that probiotics in general could reduce rates of necrotising enterocolitis (NEC), sepsis, and mortality. Answers to relevant clinical questions as to which strain to use, at what dosage, and how long to supplement are, however, not available. On the other hand, an increasing number of commercial products containing probiotics are available from sometimes suboptimal quality. Also, a large number of units around the world are routinely offering probiotic supplementation as the standard of care despite lacking solid evidence. Our recent network meta-analysis identified probiotic strains with greatest efficacy regarding relevant clinical outcomes for preterm neonates. Efficacy in reducing mortality and morbidity was found for only a minority of the studied strains or combinations. In the present position paper, we aim to provide advice, which specific strains might potentially be used and which strains should not be used. In addition, we aim to address safety issues of probiotic supplementation to preterm infants, who have reduced immunological capacities and occasional indwelling catheters. For example, quality reassurance of the probiotic product is essential, probiotic strains should be devoid of transferable antibiotic resistance genes, and local microbiologists should be able to routinely detect probiotic sepsis. Provided all safety issues are met, there is currently a conditional recommendation (with low certainty of evidence) to provide either Lactobacillus rhamnosus GG ATCC53103 or the combination of Bifidobacterium infantis Bb-02, Bifidobacterium lactis Bb-12, and Streptococcus thermophilus TH-4 in order to reduce NEC rates.
Collapse
|
42
|
Xu Y, Zhou T, Tang H, Li X, Chen Y, Zhang L, Zhang J. Probiotic potential and amylolytic properties of lactic acid bacteria isolated from Chinese fermented cereal foods. Food Control 2020. [DOI: 10.1016/j.foodcont.2019.107057] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
43
|
Gao R, Zhang X, Huang L, Shen R, Qin H. Gut Microbiota Alteration After Long-Term Consumption of Probiotics in the Elderly. Probiotics Antimicrob Proteins 2020. [PMID: 29520675 DOI: 10.1007/s12602-018-9403-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gut microbiota has been proven to be of crucial importance in maintaining human health. However, the microbiota profile changes with aging, while the loss of microbiota diversity and the alterations in the optimal composition and quantity of beneficial microbes are believed to increase the risk of many diseases. Although the short-term modulatory impact of probiotics on gut microbiota has been revealed in various studies, no studies focused on longer time consumption of probiotics have been demonstrated. In this study, we found that microbial diversity in the probiotic group was similar to that in the control. We identified a panel of microbiota changes, such as Blautia (10.24 vs. 3.76%, P = 0.006), Streptococcus (7.38 vs. 1.16%, P = 0.004), and Enterococcus (0.13 vs. 0.00%, P = 0.030) were more abundant in the probiotic group. Faecalibacterium, a genus containing anti-inflammatory property, also had a higher abundance in the probiotic group in the gut. The microbiota architecture in the different probiotic dose groups was also revealed. No statistical difference was observed in regard to the short-chain fatty acid concentration between the groups. High-dose intake of probiotics resulted in lower microbial richness. The profile of inflammatory factors indicated that only the level of IL-1β was higher in the probiotic population. Taken together, our study demonstrated that the long-time intake of probiotics caused significant changes in the gut microbiota structure, including an increase in the composition of beneficial microorganisms, which might contribute to the maintenance of host health and homeostasis of microenvironment. More prospective cohorts were needed to illustrate the influences of probiotics on the gut microbiota.
Collapse
Affiliation(s)
- Renyuan Gao
- Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Xiaohui Zhang
- Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China.,Medical College of Soochow University, Suzhou, China
| | - Linsheng Huang
- Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China.,Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China
| | - Rongrong Shen
- Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China
| | - Huanlong Qin
- Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China. .,Research Institute of Intestinal Diseases, Tongji University School of Medicine, Shanghai, China. .,Medical College of Soochow University, Suzhou, China.
| |
Collapse
|
44
|
Rana S, Bhawal S, Kumari A, Kapila S, Kapila R. pH-dependent inhibition of AHL-mediated quorum sensing by cell-free supernatant of lactic acid bacteria in Pseudomonas aeruginosa PAO1. Microb Pathog 2020; 142:104105. [PMID: 32114155 DOI: 10.1016/j.micpath.2020.104105] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Revised: 02/22/2020] [Accepted: 02/24/2020] [Indexed: 12/11/2022]
Abstract
Antibiotic mediated therapies target the growth-related processes of the pathogen hence imparting a strong selection pressure on the pathogen to develop antibiotic resistance. Recently anti-virulence strategies have gained lots of attention amongst the scientific community, wherein instead of inhibiting the normal growth of pathogens, it interferes with the regulation of virulence factors of the pathogens and impede their pathogenesis. In Pseudomonas aeruginosa, the virulence mechanism accountable for various types of infections in humans depends on N-acyl homoserine lactone (AHL) mediated quorum sensing. So quenching of these molecules, pose as a promising tool against P. aeruginosa pathogenesis. Lactic acid bacteria cell-free supernatant (acidic and neutralized) were evaluated in quorum quenching of P. aeruginosa PAO1 (MTCC 3541) after their initial screening for anti-biofilm potential against this pathogen.Though the reduction in biofilm formation with acidic and neutralized supernatants of lactic acid bacteria revealed strain specific response but acidic fractions showed much stronger (P ≤ 0.05) inhibition of biofilm irrespective of the type of challenge given to P. aeruginosa with lactic acid bacteria. The acidic fraction of supernatants (L. lactis, L. rhamnosus and L. fermentum) not only showed a significant reduction (P ≤ 0.05) in auto-inducer AHL levels but also diminished elastase activity which was among important virulence characters directly controlled by the quorum sensing signaling. Moreover, significant decrease (P ≤ 0.05) in mRNA expression of lasI and rhlI in presence of acidic fractions of lactic acid bacterial supernatants further confirmed the quorum quenching process in P. aeruginosa.
Collapse
Affiliation(s)
- Shikha Rana
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India
| | - Shalaka Bhawal
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India
| | - Ankita Kumari
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India
| | - Suman Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India
| | - Rajeev Kapila
- Animal Biochemistry Division, ICAR-National Dairy Research Institute, Karnal-132001, Haryana, India.
| |
Collapse
|
45
|
Goel A, Halami PM, Tamang JP. Genome Analysis of Lactobacillus plantarum Isolated From Some Indian Fermented Foods for Bacteriocin Production and Probiotic Marker Genes. Front Microbiol 2020; 11:40. [PMID: 32063893 PMCID: PMC7000354 DOI: 10.3389/fmicb.2020.00040] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Accepted: 01/09/2020] [Indexed: 01/09/2023] Open
Abstract
In this study, Lactobacillus plantarum strain DHCU70 isolated from dahi, a fermented milk product and L. plantarum strain DKP1 isolated from kinema, a fermented soybean food of India, respectively were evaluated for their bacteriocin production and probiotic properties. Both strains of L. plantarum (DHCU70 and DKP1) were found to have potent antimicrobial activity against Kocuria rhizophila ATCC 9341. Bacteriocin produced by L. plantarum strains DHCU70 and DKP1 did not exhibit inhibition of cell wall, DNA and fatty acids biosynthesis mechanisms as evaluated by whole cell reporter assays. We characterized the bacteriocin encoding genes in L. plantarum strains DHCU70 and DKP1 by whole genome sequence which consisted of a single and circular chromosome with genome size of 3.38 Mb (GC content of 44.3%) and 3.39 Mb, respectively and a GC content of 44.3%. L. plantarum DHCU70 has 3252 number of protein encoding genes comprising 89 number of RNA genes (69tRNA, 16rRNA, 4nc RNA) whereas L. plantarum DKP1 has total of 3277 number of protein encoding genes with 89 number. of RNA genes (69tRNA, 16S rRNA, 4nc RNA). Analysis revealed the presence of 20.5 kb long and 23 numbers of plantaricin encoding locus (pln locus) for production of antimicrobial compound. BAGEL analysis has shown that the pln locus of both the strains of L. plantarum showed maximum sequence similarity with plantaricin NC8 of L. plantarum NC8, originally isolated from grass silage. Annotated whole genome sequence of both strains DHCU70 and DKP1 was analyzed for the presence of probiotic marker genes. The probiotic properties of these strains of were also evaluated in vitro. Due to the presence of genes responsible for antimicrobial activity and probiotic properties, both strains of L. plantarum may be considered as a suitable probiotic candidate in food industry.
Collapse
Affiliation(s)
- Aditi Goel
- Department of Microbiology and Fermentation Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Prakash M. Halami
- Department of Microbiology and Fermentation Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Jyoti Prakash Tamang
- DBT-AIST International Centre for Translational and Environmental Research and Bioinformatics Centre, Department of Microbiology, School of Life Sciences, Sikkim University, Gangtok, India
| |
Collapse
|
46
|
Affiliation(s)
- Bastiaan W Haak
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers-Location AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - W Joost Wiersinga
- Center for Experimental and Molecular Medicine (CEMM), Amsterdam University Medical Centers-Location AMC, University of Amsterdam, Amsterdam, the Netherlands. .,Department of Internal Medicine, Division of Infectious Diseases, Amsterdam University Medical Centers-Location AMC, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
47
|
Ebrahimnezhad H, Barzegar L, Esmaeili D. Antibacterial Effects of Compound Bifilact on E.coli and Campylobacter jejuni. MEDICAL LABORATORY JOURNAL 2020. [DOI: 10.29252/mlj.14.1.15] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
|
48
|
Khatri I, Sharma G, Subramanian S. Composite genome sequence of Bacillus clausii, a probiotic commercially available as Enterogermina ®, and insights into its probiotic properties. BMC Microbiol 2019; 19:307. [PMID: 31888501 PMCID: PMC6937992 DOI: 10.1186/s12866-019-1680-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/11/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Some of the spore-forming strains of Bacillus probiotics are marketed commercially as they survive harsh gastrointestinal conditions and bestow health benefits to the host. RESULTS We report the composite genome of Bacillus clausii ENTPro from a commercially available probiotic Enterogermina® and compare it with the genomes of other Bacillus probiotics. We find that the members of B. clausii species harbor high heterogeneity at the species as well as genus level. The genes conferring resistance to chloramphenicol, streptomycin, rifampicin, and tetracycline in the B. clausii ENTPro strain could be identified. The genes coding for the bacteriocin gallidermin, which prevents biofilm formation in the pathogens Staphylococcus aureus and S. epidermidis, were also identified. KEGG Pathway analysis suggested that the folate biosynthesis pathway, which depicts one of the important roles of probiotics in the host, is conserved completely in B. subtilis and minimally in B. clausii and other probiotics. CONCLUSIONS We identified various antibiotic resistance, bacteriocins, stress-related, and adhesion-related domains, and industrially-relevant pathways, in the genomes of these probiotic bacteria that are likely to help them survive in the harsh gastrointestinal tract, facilitating adhesion to host epithelial cells, persistence during antibiotic treatment and combating bacterial infections.
Collapse
Affiliation(s)
- Indu Khatri
- CSIR-Institute of Microbial Technology, Sector-39A, Chandigarh, 160036, India.,Leiden University Medical Center, Leiden, the Netherlands
| | - Gaurav Sharma
- CSIR-Institute of Microbial Technology, Sector-39A, Chandigarh, 160036, India.,Institute of Bioinformatics and Applied Biotechnology, Bengaluru, Karnataka, India
| | | |
Collapse
|
49
|
Yang D, Zhao D, Ali Shah SZ, Wu W, Lai M, Zhang X, Li J, Guan Z, Zhao H, Li W, Gao H, Zhou X, Yang L. The Role of the Gut Microbiota in the Pathogenesis of Parkinson's Disease. Front Neurol 2019; 10:1155. [PMID: 31781020 PMCID: PMC6851172 DOI: 10.3389/fneur.2019.01155] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022] Open
Abstract
It is well-recognized that the gut microbiota (GM) is crucial for gut function, metabolism, and energy cycles. The GM also has effects on neurological outcomes via many mechanisms, such as metabolite production and the gut-brain axis. Emerging evidence has gradually indicated that GM dysbiosis plays a role in several neurological diseases, such as Parkinson's disease (PD), Alzheimer's disease, depression, and multiple sclerosis. Several studies have observed that PD patients generally suffer from gastrointestinal disorders and GM dysbiosis prior to displaying motor symptoms, but the specific link between the GM and PD is not clearly understood. In this review, we aim to summarize what is known regarding the correlation between the GM and PD pathologies, including direct, and indirect evidence.
Collapse
Affiliation(s)
- Dongming Yang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Deming Zhao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Syed Zahid Ali Shah
- Department of Pathology, Faculty of Veterinary Sciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur, Pakistan
| | - Wei Wu
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Mengyu Lai
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xixi Zhang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Jie Li
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Zhiling Guan
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Huafen Zhao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Wen Li
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Hongli Gao
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Xiangmei Zhou
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| | - Lifeng Yang
- Key Laboratory of Animal Epidemiology and Zoonosis, Ministry of Agriculture, National Animal Transmissible Spongiform Encephalopathy Laboratory, College of Veterinary Medicine, China Agricultural University, Beijing, China
| |
Collapse
|
50
|
Aziz K, Haseeb Zaidi A, Fatima HN, Tariq M. Lactobacillus fermentum strains of dairy-product origin adhere to mucin and survive digestive juices. J Med Microbiol 2019; 68:1771-1786. [PMID: 31613203 DOI: 10.1099/jmm.0.001090] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Introduction. There is an ever present need to isolate and characterize indigenous bacterial strains with potential probiotic health benefits for humans.Aim. Lactobacillus fermentum of dairy origin was focused because of its propensity to adhere to the intestinal glycoprotein, mucin.Methodology. The lactobacillus strains were screened for mucin adhesion, resistance to low pH and bile, autoaggregation, hydrophobicity, and survival in an in vitro digestion model. The cholesterol-lowering and oxalate-degrading effects of selected strains were also determined. Safety was assessed for haemolytic, mucinolytic and gelatinase activity, biogenic amine production, antibiotic resistance and phenol resistance. Expression of the 32-mmub adhesion-related gene was also measured following strain exposure to simulated gastrointestinal tract (GIT) digestion.Results. The selected mucin-adhesive strains were tolerant to acid (pH 3.0) and bile (0.25 %) and demonstrated >85 % survival following simulated human digestion in the presence of milk. The digestive treatment did not affect the adhesive potential of PL20, and PL27, regardless of the food matrix. The simulated digestion had less effect on their adhesion than on the type strain and it also did not correlate with the mmub gene expression level as determined by qPCR. The selected strains exhibited cholesterol removal (36-44 %) and degraded oxalate (66-55 %). Neither of these strains exhibited undesirable characteristics.Conclusion. These preliminary findings suggest a functionality in the two strains of L. fermentum with high colonization potential on GIT mucosal membranes and possible health-promoting effects. This prima facie evidence suggests the need for further studies to test these probiotic candidates as live biotherapeutic agents in vivo.
Collapse
Affiliation(s)
- Kanwal Aziz
- National Probiotic Lab-NIBGE, Jhang Road, Faisalabad 38000, Punjab, Pakistan
| | | | | | - Muhammad Tariq
- National Probiotic Lab-NIBGE, Jhang Road, Faisalabad 38000, Punjab, Pakistan
| |
Collapse
|