1
|
Copeland HM, Maye S, MacLeod G, Brabazon D, Loscher C, Freeland B. Statistical optimisation and analysis of biomass and exopolysaccharide production by Lacticaseibacillus rhamnosus LRH30. World J Microbiol Biotechnol 2025; 41:58. [PMID: 39888560 PMCID: PMC11785610 DOI: 10.1007/s11274-025-04273-2] [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: 11/21/2024] [Accepted: 01/21/2025] [Indexed: 02/01/2025]
Abstract
Exopolysaccharides (EPS) produced by lactic acid bacteria with immunomodulatory potential are promising natural food additives. This study employs small-scale, 250 mL bioreactors combined with a central composite design to optimise two important bioprocess parameters, namely temperature and airflow, to achieve high yields of biomass and EPS from Lacticaseibacillus rhamnosus LRH30 (L. rhamnosus LRH30). A quadratic model was determined to be the best fit for the production of both products. The optimum critical process parameters for maximised biomass were identified to be 37.01 °C with an airflow of 0.12 vvm, while optimum criteria was 20.1 °C with an airflow of 0.18 vvm for maximum EPS production. Under these optimized conditions, small-scale batch experiments yielded a biomass concentration of 10.1 g/L and an EPS yield of 520.2 mg/L. In comparison, scale-up experiments in 2L reactors resulted in a biomass concentration of 8.54 g/L (a reduction of 18%) and an EPS yield of 654.6 mg/L (an increase of 26%). The produced EPS was purified and characterised through Fourier transform infrared spectroscopy and showed characteristic peaks associated with polysaccharides. The immunomodulatory potential of the L. rhamnosus LRH30 cells and EPS was evaluated through cytokine and chemokine secretion in a J774A.1 murine macrophage, resulting in a predominantly anti-inflammatory effect of L. rhamnosus LRH30 and EPS.
Collapse
Affiliation(s)
- Helena Mylise Copeland
- School of Biotechnology, Dublin City University, Dublin, D9, Ireland.
- I-Form, Advanced Manufacturing Research Centre, Dublin City University, Dublin, D9, Ireland.
- Dairygold Co-Operative Society Limited, Clonmel Road, Co. Cork, Mitchelstown, P67 DD36, Ireland.
| | - Susan Maye
- Dairygold Co-Operative Society Limited, Clonmel Road, Co. Cork, Mitchelstown, P67 DD36, Ireland
| | - George MacLeod
- School of Biotechnology, Dublin City University, Dublin, D9, Ireland
- I-Form, Advanced Manufacturing Research Centre, Dublin City University, Dublin, D9, Ireland
- Dairygold Co-Operative Society Limited, Clonmel Road, Co. Cork, Mitchelstown, P67 DD36, Ireland
| | - Dermot Brabazon
- I-Form, Advanced Manufacturing Research Centre, Dublin City University, Dublin, D9, Ireland
| | - Christine Loscher
- School of Biotechnology, Dublin City University, Dublin, D9, Ireland
| | - Brian Freeland
- School of Biotechnology, Dublin City University, Dublin, D9, Ireland
| |
Collapse
|
2
|
Todorov SD, Tagg J, Algburi A, Tiwari SK, Popov I, Weeks R, Mitrokhin OV, Kudryashov IA, Kraskevich DA, Chikindas ML. The Hygienic Significance of Microbiota and Probiotics for Human Wellbeing. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10419-9. [PMID: 39688648 DOI: 10.1007/s12602-024-10419-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/01/2024] [Indexed: 12/18/2024]
Abstract
The human body can be viewed as a combination of ecological niches inhabited by trillions of bacteria, viruses, fungi, and parasites, all united by the microbiota concept. Human health largely depends on the nature of these relationships and how they are built and maintained. However, personal hygiene practices have historically been focused on the wholesale elimination of pathogens and "hygiene-challenging microorganisms" without considering the collateral damage to beneficial and commensal species. The microbiota can vary significantly in terms of the qualitative and quantitative composition both between different people and within one person during life, and the influence of various environmental factors, including age, nutrition, bad habits, genetic factors, physical activity, medication, and hygienic practices, facilitates these changes. Disturbance of the microbiota is a predisposing factor for the development of diseases and also greatly influences the course and severity of potential complications. Therefore, studying the composition of the microbiota of the different body systems and its appropriate correction is an urgent problem in the modern world. The application of personal hygiene products or probiotics must not compromise health through disruption of the healthy microbiota. Where changes in the composition or metabolic functions of the microbiome may occur, they must be carefully evaluated to ensure that essential biological functions are unaffected. As such, the purpose of this review is to consider the microbiota of each of the "ecological niches" of the human body and highlight the importance of the microbiota in maintaining a healthy body as well as the possibility of its modulation through the use of probiotics for the prevention and treatment of certain human diseases.
Collapse
Affiliation(s)
- Svetoslav D Todorov
- ProBacLab, Laboratório de Microbiologia de Alimentos, Departamento de Alimentos E Nutrição Experimental, Food Research Center, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo, São Paulo, SP, 05508-000, Brazil.
- Instituto Politécnico de Viana Do Castelo, 4900-347, Viana Do Castelo, Portugal.
| | - John Tagg
- Blis Technologies, South Dunedin, 9012, New Zealand
| | - Ammar Algburi
- Department of Microbiology, College of Veterinary Medicine, University of Diyala, Baqubah, Iraq
| | - Santosh Kumar Tiwari
- Department of Genetics, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Igor Popov
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq., 1344002, Rostov-On-Don, Russia
- Division of Immunobiology and Biomedicine, Center of Genetics and Life Sciences, Sirius University of Science and Technology, Federal Territory Sirius, Olimpijskij Ave., 1, Federal Territory Sirius, Sirius, 354340, Russia
- Centre for Healthy Eating & Food Innovation (HEFI), Maastricht University-Campus Venlo, Villafloraweg, 1, 5928 SZ, Venlo, The Netherlands
| | - Richard Weeks
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA
| | - Oleg V Mitrokhin
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Ilya A Kudryashov
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Denis A Kraskevich
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia
| | - Michael L Chikindas
- Center for Agrobiotechnology, Don State Technical University, Gagarina Sq., 1344002, Rostov-On-Don, Russia.
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers, the State University of New Jersey, New Brunswick, NJ, 08901, USA.
- Department of General Hygiene, I.M. Sechenov First Moscow State Medical University, Moscow, 119435, Russia.
| |
Collapse
|
3
|
Crowther TW, Rappuoli R, Corinaldesi C, Danovaro R, Donohue TJ, Huisman J, Stein LY, Timmis JK, Timmis K, Anderson MZ, Bakken LR, Baylis M, Behrenfeld MJ, Boyd PW, Brettell I, Cavicchioli R, Delavaux CS, Foreman CM, Jansson JK, Koskella B, Milligan-McClellan K, North JA, Peterson D, Pizza M, Ramos JL, Reay D, Remais JV, Rich VI, Ripple WJ, Singh BK, Smith GR, Stewart FJ, Sullivan MB, van den Hoogen J, van Oppen MJH, Webster NS, Zohner CM, van Galen LG. Scientists' call to action: Microbes, planetary health, and the Sustainable Development Goals. Cell 2024; 187:5195-5216. [PMID: 39303686 DOI: 10.1016/j.cell.2024.07.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 07/05/2024] [Accepted: 07/27/2024] [Indexed: 09/22/2024]
Abstract
Microorganisms, including bacteria, archaea, viruses, fungi, and protists, are essential to life on Earth and the functioning of the biosphere. Here, we discuss the key roles of microorganisms in achieving the United Nations Sustainable Development Goals (SDGs), highlighting recent and emerging advances in microbial research and technology that can facilitate our transition toward a sustainable future. Given the central role of microorganisms in the biochemical processing of elements, synthesizing new materials, supporting human health, and facilitating life in managed and natural landscapes, microbial research and technologies are directly or indirectly relevant for achieving each of the SDGs. More importantly, the ubiquitous and global role of microbes means that they present new opportunities for synergistically accelerating progress toward multiple sustainability goals. By effectively managing microbial health, we can achieve solutions that address multiple sustainability targets ranging from climate and human health to food and energy production. Emerging international policy frameworks should reflect the vital importance of microorganisms in achieving a sustainable future.
Collapse
Affiliation(s)
- Thomas W Crowther
- Department of Environmental Systems Science, Institute of Integrative Biology, ETH Zürich (Swiss Federal Institute of Technology), Zürich 8092, Switzerland; Restor Eco AG, Zürich 8001, Switzerland.
| | - Rino Rappuoli
- Fondazione Biotecnopolo di Siena, Siena 53100, Italy.
| | - Cinzia Corinaldesi
- Department of Materials, Environmental Sciences and Urban Planning, Polytechnic University of Marche, Ancona 60131, Italy; National Biodiversity Future Center, Palermo 90133, Italy
| | - Roberto Danovaro
- National Biodiversity Future Center, Palermo 90133, Italy; Department of Life and Environmental Sciences, Polytechnic University of Marche, Ancona 60131, Italy
| | - Timothy J Donohue
- Wisconsin Energy Institute, Department of Bacteriology, University of Wisconsin-Madison, Madison, WI 53726, USA
| | - Jef Huisman
- Department of Freshwater and Marine Ecology, Institute for Biodiversity and Ecosystem Dynamics, University of Amsterdam, Amsterdam 94240, the Netherlands
| | - Lisa Y Stein
- Department of Biological Sciences, University of Alberta, Edmonton, AB T6G 2E9, Canada
| | - James Kenneth Timmis
- Institute of Political Science, University of Freiburg, Freiburg 79085, Germany; Athena Institute for Research on Innovation and Communication in Health and Life Sciences, Vrije Universiteit Amsterdam, Amsterdam 1081, the Netherlands
| | - Kenneth Timmis
- Institute of Microbiology, Technical University of Braunschweig, Braunschweig 38106, Germany
| | - Matthew Z Anderson
- Center for Genomic Science Innovation, University of Wisconsin-Madison, Madison, WI 53706, USA; Laboratory of Genetics, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Lars R Bakken
- Faculty of Chemistry, Biotechnology and Food Science, Norwegian University of Life Sciences, Aas 1433, Norway
| | - Matthew Baylis
- Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Leahurst Campus, Cheshire, Neston CH64 7TE, UK
| | - Michael J Behrenfeld
- Department of Botany and Plant Pathology, Oregon State University, Corvallis, OR 97331, USA
| | - Philip W Boyd
- Institute for Marine and Antarctic Studies, University of Tasmania, Hobart, TAS 7004, Australia
| | - Ian Brettell
- Department of Environmental Systems Science, Institute of Integrative Biology, ETH Zürich (Swiss Federal Institute of Technology), Zürich 8092, Switzerland
| | - Ricardo Cavicchioli
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - Camille S Delavaux
- Department of Environmental Systems Science, Institute of Integrative Biology, ETH Zürich (Swiss Federal Institute of Technology), Zürich 8092, Switzerland
| | - Christine M Foreman
- Department of Chemical and Biological Engineering and Center for Biofilm Engineering, Montana State University, Bozeman, MT 59718, USA
| | - Janet K Jansson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA 99352, USA
| | - Britt Koskella
- Department of Integrative Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Kat Milligan-McClellan
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT 06269-3125, USA
| | - Justin A North
- Department of Microbiology, The Ohio State University, Columbus, OH 43210, USA
| | - Devin Peterson
- Department of Food Science and Technology, The Ohio State University, Columbus, OH 43210, USA
| | - Mariagrazia Pizza
- Department of Life Sciences, CBRB Center, Imperial College, London SW7 2AZ, UK
| | - Juan L Ramos
- Consejo Superior de Investigaciones Científicas, Estación Experimental del Zaidín, Granada 18008, Spain
| | - David Reay
- School of GeoSciences, The University of Edinburgh, Edinburgh EH8 9XP, UK
| | - Justin V Remais
- Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Virginia I Rich
- Center of Microbiome Science, Byrd Polar and Climate Research, and Microbiology Department, The Ohio State University, Columbus, OH 43214, USA
| | - William J Ripple
- Department of Forest Ecosystems and Society, Oregon State University, Corvallis, OR 97331-5704, USA
| | - Brajesh K Singh
- Hawkesbury Institute for the Environment, Western Sydney University, Penrith, NSW 2751, Australia
| | - Gabriel Reuben Smith
- Department of Environmental Systems Science, Institute of Integrative Biology, ETH Zürich (Swiss Federal Institute of Technology), Zürich 8092, Switzerland
| | - Frank J Stewart
- Department of Microbiology and Cell Biology, Montana State University, Bozeman, MT 59717, USA
| | - Matthew B Sullivan
- Departments of Microbiology and Civil, Environmental, and Geodetic Engineering, Center of Microbiome Science, and EMERGE Biology Integration Institute, Ohio State University, Columbus, OH 43210, USA
| | - Johan van den Hoogen
- Department of Environmental Systems Science, Institute of Integrative Biology, ETH Zürich (Swiss Federal Institute of Technology), Zürich 8092, Switzerland
| | - Madeleine J H van Oppen
- Australian Institute of Marine Science, Townsville, QLD 4810, Australia; School of Biosciences, The University of Melbourne, Parkville, VIC 3010, Australia
| | - Nicole S Webster
- Institute for Marine and Antarctic Studies, University of Tasmania, Hobart, TAS 7004, Australia; Australian Institute of Marine Science, Townsville, QLD 4810, Australia; Australian Centre for Ecogenomics, University of Queensland, Brisbane, QLD 4072, Australia
| | - Constantin M Zohner
- Department of Environmental Systems Science, Institute of Integrative Biology, ETH Zürich (Swiss Federal Institute of Technology), Zürich 8092, Switzerland
| | - Laura G van Galen
- Department of Environmental Systems Science, Institute of Integrative Biology, ETH Zürich (Swiss Federal Institute of Technology), Zürich 8092, Switzerland; Society for the Protection of Underground Networks (SPUN), Dover, DE 19901, USA.
| |
Collapse
|
4
|
Karmazyn M, Gan XT. Probiotics as potential treatments to reduce myocardial remodelling and heart failure via the gut-heart axis: State-of-the-art review. Mol Cell Biochem 2023; 478:2539-2551. [PMID: 36892791 DOI: 10.1007/s11010-023-04683-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Accepted: 02/17/2023] [Indexed: 03/10/2023]
Abstract
Probiotics are considered to represent important modulators of gastrointestinal health through increased colonization of beneficial bacteria thus altering the gut microflora. Although these beneficial effects of probiotics are now widely recognized, emerging evidence suggests that alterations in the gut microflora also affect numerous other organ systems including the heart through a process generally referred to as the gut-heart axis. Moreover, cardiac dysfunction such as that seen in heart failure can produce an imbalance in the gut flora, known as dysbiosis, thereby further contributing to cardiac remodelling and dysfunction. The latter occurs by the production of gut-derived pro-inflammatory and pro-remodelling factors which exacerbate cardiac pathology. One of the key contributors to gut-dependent cardiac pathology is trimethylamine N-oxide (TMAO), a choline and carnitine metabolic by-product first synthesized as trimethylamine which is then converted into TMAO by a hepatic flavin-containing monooxygenase. The production of TMAO is particularly evident with regular western diets containing high amounts of both choline and carnitine. Dietary probiotics have been shown to reduce myocardial remodelling and heart failure in animal models although the precise mechanisms for these effects are not completely understood. A large number of probiotics have been shown to possess a reduced capacity to synthesize gut-derived trimethylamine and therefore TMAO thereby suggesting that inhibition of TMAO is a factor mediating the beneficial cardiac effects of probiotics. However, other potential mechanisms may also be important contributing factors. Here, we discuss the potential benefit of probiotics as effective therapeutic tools for attenuating myocardial remodelling and heart failure.
Collapse
Affiliation(s)
- Morris Karmazyn
- Department of Pharmacology and Physiology, University of Western Ontario, London, ON, N6G 2X6, Canada.
| | - Xiaohong Tracey Gan
- Department of Pharmacology and Physiology, University of Western Ontario, London, ON, N6G 2X6, Canada
| |
Collapse
|
5
|
Łoniewski I, Szulińska M, Kaczmarczyk M, Podsiadło K, Styburski D, Skonieczna-Żydecka K, Bogdański P. Multispecies probiotic affects fecal short-chain fatty acids in postmenopausal women with obesity: A post hoc analysis of a randomized, double-blind, placebo-controlled study. Nutrition 2023; 114:112109. [PMID: 37441828 DOI: 10.1016/j.nut.2023.112109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 04/25/2023] [Accepted: 05/26/2023] [Indexed: 07/15/2023]
Abstract
OBJECTIVES Probiotics are known to regulate host metabolism. The aim of this study was to assess whether interventions with a multi-strain probiotic formula affect fecal short-chain fatty acids (SCFAs). METHODS The analysis was carried out in 56 obese, postmenopausal women randomized to three groups: probiotic dose 2.5 × 109 CFU/d (n = 18; lower probiotic dose [LPD]), 1 × 1010 CFU/d (n = 18; higher probiotic dose [HPD]), or placebo (n = 20). RESULTS An increase in three SCFA fecal concentrations in the HPD group was observed: acetic acid (C2; effect [E] = 1.72, SE = 0.73; 95% confidence interval [CI], 0.28-3.16; P = 0.019), butyric acid (C4; E = 0.98, SE = 0.46; 95% CI, 0.08-1.88; P = 0.033), and valeric acid (C5; E = 0.68, SE = 0.23; 95% CI, 0.23-1.12; P = 0.003). The mediation analysis showed that the decrease in uric acid under HPD may be transmitted through the elevation of C5 content. Multi-strain probiotic increases the SCFA content in the stool in a dose-dependent manner, which may diminish some cardiovascular risk factors because of a reduction in blood uric acid levels. CONCLUSION Assessing long-term health benefits requires further research, including assessment of blood SCFA concentrations and multiomic and mechanistic approaches.
Collapse
Affiliation(s)
- Igor Łoniewski
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Szczecin, Poland; Department of Human Nutrition and Metabolomics, Pomeranian Medical University, Szczecin, Poland
| | - Monika Szulińska
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, University of Medical Sciences in Poznań, Poznań, Poland
| | | | - Konrad Podsiadło
- Department of Clinical Biochemistry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | | | | | - Paweł Bogdański
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, University of Medical Sciences in Poznań, Poznań, Poland
| |
Collapse
|
6
|
Yeramilli V, Cheddadi R, Shah J, Brawner K, Martin C. A Review of the Impact of Maternal Prenatal Stress on Offspring Microbiota and Metabolites. Metabolites 2023; 13:metabo13040535. [PMID: 37110193 PMCID: PMC10142778 DOI: 10.3390/metabo13040535] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/06/2023] [Accepted: 04/07/2023] [Indexed: 04/29/2023] Open
Abstract
Maternal prenatal stress exposure affects the development of offspring. We searched for articles in the PubMed database and reviewed the evidence for how prenatal stress alters the composition of the microbiome, the production of microbial-derived metabolites, and regulates microbiome-induced behavioral changes in the offspring. The gut-brain signaling axis has gained considerable attention in recent years and provides insights into the microbial dysfunction in several metabolic disorders. Here, we reviewed evidence from human studies and animal models to discuss how maternal stress can modulate the offspring microbiome. We will discuss how probiotic supplementation has a profound effect on the stress response, the production of short chain fatty acids (SCFAs), and how psychobiotics are emerging as novel therapeutic targets. Finally, we highlight the potential molecular mechanisms by which the effects of stress are transmitted to the offspring and discuss how the mitigation of early-life stress as a risk factor can improve the birth outcomes.
Collapse
Affiliation(s)
- Venkata Yeramilli
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Riadh Cheddadi
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Juhi Shah
- Burnett School of Medicine, Texas Christian University, Fort Worth, TX 76129, USA
| | - Kyle Brawner
- Department of Biology, Lipscomb University, Nashville, TN 37204, USA
| | - Colin Martin
- Division of Pediatric Surgery, Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| |
Collapse
|
7
|
Dincer E, Kivanc M. Evaluation of metabolic activities and probiotic characteristics of two Latilactobacillus sakei strains isolated from pastırma. World J Microbiol Biotechnol 2022; 38:237. [DOI: 10.1007/s11274-022-03431-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 10/02/2022] [Indexed: 10/17/2022]
|
8
|
Liu G, Khan I, Li Y, Yang Y, Lu X, Wang Y, Li J, Zhang C. Overcoming Anxiety Disorder by Probiotic Lactiplantibacillus plantarum LZU-J-TSL6 through Regulating Intestinal Homeostasis. Foods 2022; 11:foods11223596. [PMID: 36429192 PMCID: PMC9689226 DOI: 10.3390/foods11223596] [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: 09/26/2022] [Revised: 10/14/2022] [Accepted: 11/04/2022] [Indexed: 11/16/2022] Open
Abstract
Lactiplantibacillus plantarum LZU-J-TSL6 with high γ-aminobutyric acid (GABA) production (3.838 g/L) was screened and isolated from the Chinese fermented food snack “Jiangshui”. The improvement effect on anxiety disorder was explored using mice as animal models. In vitro results revealed that LZU-J-TSL6 had the potential to colonize the intestine (p < 0.01) and the anxiety-like behavior of the mice after seven days’ gavage with LZU-J-TSL6 was significantly improved (p < 0.01) when compared to the model group. LZU-J-TSL6 was able to effectively increase the GABA content in the mice hippocampus (p < 0.0001) and restore some markers related to anxiety such as brain-derived neurotrophic factor (BDNF), glial fibrillary acidic protein (GFAP), and 5-hydroxytryptamine (5-HT). Simultaneously, it had a certain repair effect on Nissl bodies and colon tissue in mice hippocampus. In addition, LZU-J-TSL6 increased the relative abundance of beneficial bacteria Bacteroides and Muribaculum, thereby regulating the imbalance of intestinal microbiota caused by anxiety disorder. It also affects the nerve pathway and intestinal mucosal barrier by increasing the content of glutamine and γ-aminobutyric acid and other related metabolites, thereby improving anxiety. Therefore, the GABA-producing Lactobacillus plantus LZU-J-TSL6 can be used as a probiotic to exert an indirect or direct anti-anxiety effect by maintaining the balance of the intestinal environment, producing related metabolites that affect nerve pathways and repair the intestinal mucosal barrier. It can be used as an adjuvant treatment to improve anxiety disorders.
Collapse
Affiliation(s)
- Guanlan Liu
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Israr Khan
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Yuxi Li
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Yun Yang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Xuerui Lu
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Yafei Wang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Junxiang Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Chunjiang Zhang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China
- Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
- Gansu Key Laboratory of Functional Genomics and Molecular Diagnosis, Lanzhou University, Lanzhou 730000, China
- Correspondence:
| |
Collapse
|
9
|
Timmis JK, Roussilhon DF, van de Burgwal LHM. Innovations for microbiome targeting interventions - a patent landscape analysis indicating overall patenting activity decline and promising target disease areas. Benef Microbes 2022; 13:265-282. [PMID: 35979711 DOI: 10.3920/bm2021.0050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The human microbiota have been implicated in the aetiology and remedy of a host of disorders. However, due to the pervasive uncertainty inherent in the field of microbiota-targeting interventions and associated issues with establishing rigorous safety and efficacy profiles, regulatory oversight is suboptimal. This can dissuade innovators from further exploring novel and much needed health interventions. Modification of regulatory protocols and practices requires focussed efforts and funding to build the evidence base around future regulatory needs. Such modification can be critically informed by identification of changes and trends in technology fields to facilitate identification of regulatory gaps. To this purpose, this study rigorously collected and analysed patent data from Espacenet - covering the years 2013-2018 - and created a patent landscape analysis of microbiome targeting interventions with a focus on medicinal products. Pertinent patenting activity has declined overall. While, in absolute terms, patents most frequently claimed inventions targeting disorders of the gut and alimentary tract, relative year-on-year interest increases have been substantial for cancer, and disorders of the (neuro-)muscular and respiratory systems - driven by the private sector. Academic stakeholders showed top interest in disorders of the metabolism, anti-infectives, and skeletal and dermatological diseases. Although medicinal preparation claims dominated our dataset, a third of patents claimed food preparations, while only 1% claimed application as a diagnostic. Finally, China is, by an inordinate margin, a market of particular interest for both domestic and foreign innovators, indicating that microbiome targeting intervention innovation for EU and US markets might be frustrated. This study is the first to empirically demonstrate that live biotherapeutic product innovation is decelerating and potentially frustrated, supporting the urgent need for improved regulatory standards. Our results indicate which disease areas deserve particular attention for research funding to facilitate proper regulatory appraisal in the near- to mid-term future.
Collapse
Affiliation(s)
- J K Timmis
- Athena Institute for Research on Innovation and Communication in Health and Life Sciences, Vrije Universiteit Amsterdam, Boelelaan, 1081 HV Amsterdam, the Netherlands
| | - D Flaherty Roussilhon
- Athena Institute for Research on Innovation and Communication in Health and Life Sciences, Vrije Universiteit Amsterdam, Boelelaan, 1081 HV Amsterdam, the Netherlands
| | - L H M van de Burgwal
- Athena Institute for Research on Innovation and Communication in Health and Life Sciences, Vrije Universiteit Amsterdam, Boelelaan, 1081 HV Amsterdam, the Netherlands
| |
Collapse
|
10
|
Beyoğlu D, Idle JR. The gut microbiota - a vehicle for the prevention and treatment of hepatocellular carcinoma. Biochem Pharmacol 2022; 204:115225. [PMID: 35998677 DOI: 10.1016/j.bcp.2022.115225] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2022] [Revised: 08/15/2022] [Accepted: 08/16/2022] [Indexed: 12/24/2022]
Abstract
Hepatocellular carcinoma (HCC) arises principally against a background of cirrhosis and these two diseases are responsible globally for over 2 million deaths a year. There are few treatment options for liver cirrhosis and HCC, so it is vital to arrest these pathologies early in their development. To do so, we propose dietary and therapeutic solutions that involve the gut microbiota and its consequences. Integrated dietary, environmental and intrinsic signals result in a bidirectional connection between the liver and the gut with its microbiota, known as the gut-liver axis. Numerous lifestyle factors can result in dysbiosis with a change in the functional composition and metabolic activity of the microbiota. A panoply of metabolites can be produced by the microbiota, including ethanol, secondary bile acids, trimethylamine, indole, quinolone, phenazine and their derivatives and the quorum sensor acyl homoserine lactones that may contribute to HCC but have yet to be fully investigated. Gram-negative bacteria can activate the pattern recognition receptor toll-like receptor 4 (TLR4) in the liver leading to nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling, which can contribute to HCC initiation and progression. The goal in preventing HCC should be to ensure a healthy gut microbiota using probiotic supplements containing beneficial bacteria and prebiotic plant fibers such as oligosaccharides that stimulate their growth. The clinical development of TLR4 antagonists is urgently needed to counteract the pathological effects of dysbiosis on the liver and other organs. Further nutrigenomic studies are required to understand better how the diet influences the gut microbiota and its adverse effects on the liver.
Collapse
Affiliation(s)
- Diren Beyoğlu
- Arthur G. Zupko Institute for Systems Pharmacology and Pharmacogenomics, Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, New York 11201, USA
| | - Jeffrey R Idle
- Arthur G. Zupko Institute for Systems Pharmacology and Pharmacogenomics, Arnold and Marie Schwartz College of Pharmacy and Health Sciences, Long Island University, Brooklyn, New York 11201, USA.
| |
Collapse
|
11
|
Cruz N, Abernathy GA, Dichosa AEK, Kumar A. The Age of Next-Generation Therapeutic-Microbe Discovery: Exploiting Microbe-Microbe and Host-Microbe Interactions for Disease Prevention. Infect Immun 2022; 90:e0058921. [PMID: 35384688 PMCID: PMC9119102 DOI: 10.1128/iai.00589-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Humans are considered "superorganisms," harboring a diverse microbial collective that outnumbers human cells 10 to 1. Complex and gravely understudied host- and microbe-microbe interactions-the product of millions of years of host-microbe coevolution-govern the superorganism in almost every aspect of life functions and overall well-being. Abruptly disrupting these interactions via extrinsic factors has undesirable consequences for the host. On the other hand, supplementing commensal or beneficial microbes may mitigate perturbed interactions or enhance the interactive relationships that ultimately benefit all parties. Hence, immense efforts have focused on dissecting the innumerable host- and microbe-microbe relationships to characterize if a "positive" or "negative" interaction is at play and to exploit such behavior for broader implications. For example, microbiome research has worked to identify and isolate naturally antipathogenic microbes that may offer therapeutic potential either in a direct, one-on-one application or by leveraging its unique metabolic properties. However, the discovery and isolation of such desired therapeutic microbes from complex microbiota have proven challenging. Currently, there is no conventional technique to universally and functionally screen for these microbes. With this said, we first describe in this review the historical (probiotics) and current (fecal microbiota or defined consortia) perspectives on therapeutic microbes, present the discoveries of therapeutic microbes through exploiting microbe-microbe and host-microbe interactions, and detail our team's efforts in discovering therapeutic microbes via our novel microbiome screening platform. We conclude this minireview by briefly discussing challenges and possible solutions with therapeutic microbes' applications and paths ahead for discovery.
Collapse
Affiliation(s)
- Nathan Cruz
- B-10: Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - George A. Abernathy
- B-10: Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Armand E. K. Dichosa
- B-10: Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| | - Anand Kumar
- B-10: Biosecurity and Public Health Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, USA
| |
Collapse
|
12
|
Ivashkin VT, Maev IV, Alekseeva OP, Alekseenko SA, Korochanskaya NV, Poluektova EA, Simanenkov VI, Trukhmanov AS, Khlynov IB, Tsukanov VV, Shifrin OS, Lapina TL, Maslennikov RV, Ulyanin AI. Determination of Probiotics Prescription Indications in Patients with Irritable Bowel Syndrome (Materials of the Expert Council and Literature Review). RUSSIAN JOURNAL OF GASTROENTEROLOGY, HEPATOLOGY, COLOPROCTOLOGY 2022; 32:9-18. [DOI: 10.22416/1382-4376-2022-32-2-9-18] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/26/2024]
Abstract
Aim. To review the main indications for probiotics prescription in patients with irritable bowel syndrome and to present the materials of an Expert Council, which was held on 18 March 2022 in Moscow.Key points. Gut microbiota disturbance is an integral part of irritable bowel syndrome (IBS) pathogenesis. Changes of colonic microbiota composition are associated with its functional potential modification, which leads to an increasing of the pro-inflammatory immune response, as well as to an exacerbation of the disease symptoms and quality of life decreasing in patients with IBS. The novel coronavirus infection (COVID-19) is an independent risk factor for both exacerbation and onset of IBS, which predispose to increase IBS incidence. Correction of gut microbiota composition with probiotics seems to be a promising therapeutic target for IBS treatment optimizing. The optimal probiotic should be effective, safe, strain-specific, and its dose and duration of administration should be confirmed by the results of clinical studies. Some of the probiotics with proven efficacy in IBS are Alflorex® and Enterol®.Conclusion. Prescription of certain probiotics in IBS is advisable to normalize the frequency and consistency of stools, relieve abdominal pain and bloating, as well as improve patients’ quality of life.
Collapse
Affiliation(s)
- V. T. Ivashkin
- Sechenov First Moscow State Medical University (Sechenov University)
| | - I. V. Maev
- A.I. Yevdokimov Moscow State University of Medicine and Dentistry
| | | | | | | | - E. A. Poluektova
- Sechenov First Moscow State Medical University (Sechenov
University)
| | | | - A. S. Trukhmanov
- Sechenov First Moscow State Medical University (Sechenov University)
| | | | - V. V. Tsukanov
- Research Institute for Medical Problems in the North — Division of Krasnoyarsk Scientific Centre of the Siberian Branch of the RAS
| | - O. S. Shifrin
- Sechenov First Moscow State Medical University (Sechenov University)
| | - T. L. Lapina
- Sechenov First Moscow State Medical University (Sechenov University)
| | - R. V. Maslennikov
- Sechenov First Moscow State Medical University (Sechenov University)
| | - A. I. Ulyanin
- Sechenov First Moscow State Medical University (Sechenov University)
| |
Collapse
|
13
|
Kaczmarczyk M, Szulińska M, Łoniewski I, Kręgielska-Narożna M, Skonieczna-Żydecka K, Kosciolek T, Bezshapkin V, Bogdański P. Treatment With Multi-Species Probiotics Changes the Functions, Not the Composition of Gut Microbiota in Postmenopausal Women With Obesity: A Randomized, Double-Blind, Placebo-Controlled Study. Front Cell Infect Microbiol 2022; 12:815798. [PMID: 35360106 PMCID: PMC8963764 DOI: 10.3389/fcimb.2022.815798] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Accepted: 02/11/2022] [Indexed: 12/12/2022] Open
Abstract
Probiotics are known to regulate host metabolism. In randomized controlled trial we aimed to assess whether interventions with probiotic containing following strains: Bifidobacterium bifidum W23, Bifidobacterium lactis W51, Bifidobacterium lactis W52, Lactobacillus acidophilus W37, Levilactobacillus brevis W63, Lacticaseibacillus casei W56, Ligilactobacillus salivarius W24, Lactococcus lactis W19, and Lactococcus lactis W58 affect gut microbiota to promote metabolic effects. By 16S rRNA sequencing we analyzed the fecal microbiota of 56 obese, postmenopausal women randomized into three groups: (1) probiotic dose 2.5 × 109 CFU/day (n = 18), (2) 1 × 1010 CFU/day (n = 18), or (3) placebo (n = 20). In the set of linear mixed-effects models, the interaction between pre- or post-treatment bacterial abundance and time on cardiometabolic parameters was significantly (FDR-adjusted) modified by type of intervention (26 and 19 three-way interactions for the pre-treatment and post-treatment abundance, respectively), indicating the modification of the bio-physiological role of microbiota by probiotics. For example, the unfavorable effects of Erysipelotrichi, Erysipelotrichales, and Erysipelotrichaceae on BMI might be reversed, but the beneficial effect of Betaproteobacteria on BMI was diminished by probiotic treatment. Proinflammatory effect of Bacteroidaceae was alleviated by probiotic administration. However, probiotics did not affect the microbiota composition, and none of the baseline microbiota-related features could predict therapeutic response as defined by cluster analysis. Conclusions: Probiotic intervention alters the influence of microbiota on biochemical, physiological and immunological parameters, but it does not affect diversity and taxonomic composition. Baseline microbiota is not a predictor of therapeutic response to a multispecies probiotic. Further multi-omic and mechanistic studies performed on the bigger cohort of patients are needed to elucidate the cardiometabolic effect of investigated probiotics in postmenopausal obesity.
Collapse
Affiliation(s)
- Mariusz Kaczmarczyk
- Department of Clinical and Molecular Biochemistry, Pomeranian Medical University in Szczecin, Szczecin, Poland
| | - Monika Szulińska
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, University of Medical Sciences in Poznań, Poznań, Poland
| | - Igor Łoniewski
- Department of Biochemical Sciences, Pomeranian Medical University in Szczecin, Szczecin, Poland
- Department of Human Nutrition and Metabolomics, Pomeranian Medical University in Szczecin, Szczecin, Poland
- *Correspondence: Igor Łoniewski,
| | - Matylda Kręgielska-Narożna
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, University of Medical Sciences in Poznań, Poznań, Poland
| | | | - Tomasz Kosciolek
- Małopolska Centre of Biotechnology, Jagiellonian University, Kraków, Poland
| | | | - Paweł Bogdański
- Department of Treatment of Obesity, Metabolic Disorders and Clinical Dietetics, University of Medical Sciences in Poznań, Poznań, Poland
| |
Collapse
|
14
|
Happel AU, Kullin BR, Gamieldien H, Jaspan HB, Varsani A, Martin D, Passmore JAS, Froissart R. In Silico Characterisation of Putative Prophages in Lactobacillaceae Used in Probiotics for Vaginal Health. Microorganisms 2022; 10:214. [PMID: 35208669 PMCID: PMC8879116 DOI: 10.3390/microorganisms10020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2021] [Revised: 12/15/2021] [Accepted: 12/18/2021] [Indexed: 01/27/2023] Open
Abstract
While live biotherapeutics offer a promising approach to optimizing vaginal microbiota, the presence of functional prophages within introduced Lactobacillaceae strains could impact their safety and efficacy. We evaluated the presence of prophages in 895 publicly available Lactobacillaceae genomes using Phaster, Phigaro, Phispy, Prophet and Virsorter. Prophages were identified according to stringent (detected by ≥4 methods) or lenient criteria (detected by ≥2 methods), both with >80% reciprocal sequence overlap. The stringent approach identified 448 prophages within 359 genomes, with 40.1% genomes harbouring at least one prophage, while the lenient approach identified 1671 prophages within 83.7% of the genomes. To confirm our in silico estimates in vitro, we tested for inducible prophages in 57 vaginally-derived and commercial Lactobacillaceae isolates and found inducible prophages in 61.4% of the isolates. We characterised the in silico predicted prophages based on weighted gene repertoire relatedness and found that most belonged to the Siphoviridae or Myoviridae families. ResFam and eggNOG identified four potential antimicrobial resistance genes within the predicted prophages. Our results suggest that while Lactobacillaceae prophages seldomly carry clinically concerning genes and thus unlikely a pose a direct risk to human vaginal microbiomes, their high prevalence warrants the characterisation of Lactobacillaceae prophages in live biotherapeutics.
Collapse
Affiliation(s)
- Anna-Ursula Happel
- Department of Pathology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Anzio Road, Cape Town 7925, South Africa; (A.-U.H.); (B.R.K.); (H.G.); (H.B.J.); (J.-A.S.P.)
| | - Brian R. Kullin
- Department of Pathology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Anzio Road, Cape Town 7925, South Africa; (A.-U.H.); (B.R.K.); (H.G.); (H.B.J.); (J.-A.S.P.)
| | - Hoyam Gamieldien
- Department of Pathology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Anzio Road, Cape Town 7925, South Africa; (A.-U.H.); (B.R.K.); (H.G.); (H.B.J.); (J.-A.S.P.)
| | - Heather B. Jaspan
- Department of Pathology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Anzio Road, Cape Town 7925, South Africa; (A.-U.H.); (B.R.K.); (H.G.); (H.B.J.); (J.-A.S.P.)
- Seattle Children’s Research Institute, 307 Westlake Ave. N, Seattle, WA 98109, USA
- Department of Pediatrics and Global Health, University of Washington, 1410 NE Campus Parkway NE, Seattle, WA 98195, USA
| | - Arvind Varsani
- The Biodesign Center of Fundamental and Applied Microbiomics, Center for Evolution and Medicine, School of Life Sciences, Arizona State University, 1001 S. McAllister Ave., Tempe, AZ 85281, USA;
- Structural Biology Research Unit, Department of Integrative Biomedical Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Anzio Road, Cape Town 7925, South Africa
| | - Darren Martin
- Division of Computational Biology, Department of Integrative Biomedical Sciences, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Anzio Road, Cape Town 7925, South Africa;
| | - Jo-Ann S. Passmore
- Department of Pathology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Anzio Road, Cape Town 7925, South Africa; (A.-U.H.); (B.R.K.); (H.G.); (H.B.J.); (J.-A.S.P.)
- NRF-DST CAPRISA Centre of Excellence in HIV Prevention, 719 Umbilo Road, Congella, Durban 4013, South Africa
- National Health Laboratory Service, Cape Town 7925, South Africa
| | - Rémy Froissart
- CNRS, IRD, Université Montpellier, UMR 5290, MIVEGEC, 34394 Montpellier, France
| |
Collapse
|
15
|
Ebrahimi V, Tarhriz V, Talebi M, Rasouli A, Farjami A, Razi Soofiyani S, Soleimanian A, Forouhandeh H. A new insight on feasibility of pre-, pro-, and synbiotics-based therapies in Alzheimer’s disease. JOURNAL OF REPORTS IN PHARMACEUTICAL SCIENCES 2022. [DOI: 10.4103/jrptps.jrptps_170_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
16
|
Zhou X, Guo Q, Guo M, Li B, Peng W, Wang D, Ming D, Zheng B. Nanoarmour-shielded single-cell factory for bacteriotherapy of Parkinson's disease. J Control Release 2021; 338:742-753. [PMID: 34517041 DOI: 10.1016/j.jconrel.2021.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 09/06/2021] [Accepted: 09/08/2021] [Indexed: 01/15/2023]
Abstract
Cell-based therapy for Parkinson's disease (PD) is a novel and promising approach in recent years. However, exogenous cells are easy to be captured and destroyed by the harsh environment in vivo, so their application prospects have been severely limited. Here, a facile yet versatile approach for decorating individual living cells with nano-armor coatings is reported. By simply self-assembly with liposome under a cyto-compatible condition, the lipid bimolecular coating on the surface of each cell acts as armor to effectively protect it from the attack and destruction of strong acids and digestive enzymes during the oral treatment of PD. Our results demonstrated that the liposome coated B. adolescentis (LCB) could significantly improve the colonization rate in the intestinal tract. LCB, as a living cell factory, can self-regulate to produce a constant concentration of γ-aminobutyric acid and maintain a longer half-life for the treatment of PD. Then, we also explored the specific mechanism of LCB to improve the behavior of murine models of PD, including abating inflammatory effects, reducing neuronal apoptosis, regulating the activity of dopaminergic neurons and microglia. The simple nano-armor shielded single-cell factory can produce neurotransmitters-like drugs on demand in vivo, introducing novel strategies of integration of producing and using to the research of drug delivery field.
Collapse
Affiliation(s)
- Xin Zhou
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Xincheng Hospital of Tianjin University, Tianjin University, Tianjin 300072, China; College of Medical Imaging, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Qinglu Guo
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Xincheng Hospital of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Mingming Guo
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Xincheng Hospital of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Bowen Li
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Xincheng Hospital of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Wenchang Peng
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Xincheng Hospital of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Deping Wang
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Xincheng Hospital of Tianjin University, Tianjin University, Tianjin 300072, China; College of Medical Imaging, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, China
| | - Dong Ming
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Xincheng Hospital of Tianjin University, Tianjin University, Tianjin 300072, China
| | - Bin Zheng
- Academy of Medical Engineering and Translational Medicine, Tianjin Key Laboratory of Brain Science and Neural Engineering, Xincheng Hospital of Tianjin University, Tianjin University, Tianjin 300072, China; Healthina Academy of Cellular Intelligence Manufacturing & Neurotrauma Repair, Beijing Tangyi Huikang Biomedical Technology Co., Ltd, Beijing 100010, China.
| |
Collapse
|
17
|
Isenring J, Geirnaert A, Lacroix C, Stevens MJA. Bistable auto-aggregation phenotype in Lactiplantibacillus plantarum emerges after cultivation in in vitro colonic microbiota. BMC Microbiol 2021; 21:268. [PMID: 34610822 PMCID: PMC8493755 DOI: 10.1186/s12866-021-02331-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 09/24/2021] [Indexed: 11/28/2022] Open
Abstract
Background Auto-aggregation is a desired property for probiotic strains because it is suggested to promote colonization of the human intestine, to prevent pathogen infections and to modulate the colonic mucosa. We recently reported the generation of adapted mutants of Lactiplantibacillus plantarum NZ3400, a derivative of the model strain WCFS1, for colonization under adult colonic conditions of PolyFermS continuous intestinal fermentation models. Here we describe and characterize the emerge of an auto-aggregating phenotype in L. plantarum NZ3400 derivatives recovered from the modelled gut microbiota. Results L. plantarum isolates were recovered from reactor effluent of four different adult microbiota and from spontaneously formed reactor biofilms. Auto-aggregation was observed in L. plantarum recovered from all microbiota and at higher percentage when recovered from biofilm than from effluent. Further, auto-aggregation percentage increased over time of cultivation in the microbiota. Starvation of the gut microbiota by interrupting the inflow of nutritive medium enhanced auto-aggregation, suggesting a link to nutrient availability. Auto-aggregation was lost under standard cultivation conditions for lactobacilli in MRS medium. However, it was reestablished during growth on sucrose and maltose and in a medium that simulates the abiotic gut environment. Remarkably, none of these conditions resulted in an auto-aggregation phenotype in the wild type strain NZ3400 nor other non-aggregating L. plantarum, indicating that auto-aggregation depends on the strain history. Whole genome sequencing analysis did not reveal any mutation responsible for the auto-aggregation phenotype. Transcriptome analysis showed highly significant upregulation of LP_RS05225 (msa) at 4.1–4.4 log2-fold-change and LP_RS05230 (marR) at 4.5–5.4 log2-fold-change in all auto-aggregating strains compared to non-aggregating. These co-expressed genes encode a mannose-specific adhesin protein and transcriptional regulator, respectively. Mapping of the RNA-sequence reads to the promoter region of the msa-marR operon reveled a DNA inversion in this region that is predominant in auto-aggregating but not in non-aggregating strains. This strongly suggests a role of this inversion in the auto-aggregation phenotype. Conclusions L. plantarum NZ3400 adapts to the in vitro colonic environment by developing an auto-aggregation phenotype. Similar aggregation phenotypes may promote gut colonization and efficacy of other probiotics and should be further investigated by using validated continuous models of gut fermentation such as PolyFermS. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-021-02331-x.
Collapse
Affiliation(s)
- Julia Isenring
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstrasse 7, 8092, Zürich, Switzerland
| | - Annelies Geirnaert
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstrasse 7, 8092, Zürich, Switzerland
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstrasse 7, 8092, Zürich, Switzerland.
| | - Marc J A Stevens
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, Schmelzbergstrasse 7, 8092, Zürich, Switzerland.,Institute for Food Hygiene and Safety, University of Zürich, Zurich, Switzerland
| |
Collapse
|
18
|
Peck J, Shepherd JP. Recurrent Urinary Tract Infections: Diagnosis, Treatment, and Prevention. Obstet Gynecol Clin North Am 2021; 48:501-513. [PMID: 34416934 DOI: 10.1016/j.ogc.2021.05.005] [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: 01/14/2023]
Abstract
Urinary tract infections are common infections seen by obstetrician/gynecologists and primary care providers. Recurrent urinary tract infections are difficult to treat and decrease quality of life. This article addresses the diagnosis, treatment, and prevention of recurrent urinary tract infections, including pharmacologic and nonpharmacologic interventions. Most notably, increased interest in the urinary microbiome is leading to improved understanding of virulent and commensal bacteria in the genitourinary tract. Understanding the causative factors of recurrent urinary tract infections will lead to improved detection, treatment, and prevention.
Collapse
Affiliation(s)
- Julie Peck
- St Francis Hospital Trinity Health, 114 Woodland Street, OB Administration 43, Hartford, CT 06105, USA.
| | - Jonathan P Shepherd
- St Francis Hospital Trinity Health, 114 Woodland Street, OB Administration 43, Hartford, CT 06105, USA
| |
Collapse
|
19
|
Langlois L, Akhtar N, Tam KC, Dixon B, Reid G. Fishing for the right probiotic: Host-microbe interactions at the interface of effective aquaculture strategies. FEMS Microbiol Rev 2021; 45:6284803. [PMID: 34037775 DOI: 10.1093/femsre/fuab030] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 05/24/2021] [Indexed: 02/06/2023] Open
Abstract
Effective aquaculture management strategies are paramount to global food security. Growing demands stimulate the intensification of production and create the need for practices that are both economically viable and environmentally sustainable. Importantly, pathogenic microbes continue to be detrimental to fish growth and survival. In terms of host health, the intestinal mucosa and its associated consortium of microbes have a critical role in modulating fitness and present an attractive opportunity to promote health at this interface. In light of this, the administration of probiotic microorganisms is being considered as a means to restore and sustain health in fish. Current evidence suggests that certain probiotic strains might be able to augment immunity, enhance growth rate, and protect against infection in salmonids, the most economically important family of farmed finfish. This review affirms the relevance of host-microbe interactions in salmonids in light of emerging evidence, with an emphasis on intestinal health. In addition, the current understanding of the mode of action of probiotics in salmonid fish is discussed, along with delivery systems that can effectively carry the living microbes.
Collapse
Affiliation(s)
- Luana Langlois
- Canadian Centre for Human Microbiome and Probiotics Research, Lawson Health Research Institute, 268 Grosvenor St, N6A 4V2, London, Ontario, Canada.,Department of Microbiology and Immunology, The University of Western Ontario, 1151 Richmond St, N6A 5C1, London, Ontario, Canada
| | - Nadeem Akhtar
- Department of Chemical Engineering, University of Waterloo, 200 University Ave W, N2L 3G1, Waterloo, Ontario, Canada.,Department of Biology, University of Waterloo, 200 University Avenue W, N2L 3G1, Waterloo, Ontario, Canada
| | - Kam C Tam
- Department of Chemical Engineering, University of Waterloo, 200 University Ave W, N2L 3G1, Waterloo, Ontario, Canada
| | - Brian Dixon
- Department of Biology, University of Waterloo, 200 University Avenue W, N2L 3G1, Waterloo, Ontario, Canada
| | - Gregor Reid
- Canadian Centre for Human Microbiome and Probiotics Research, Lawson Health Research Institute, 268 Grosvenor St, N6A 4V2, London, Ontario, Canada.,Department of Microbiology and Immunology, The University of Western Ontario, 1151 Richmond St, N6A 5C1, London, Ontario, Canada.,Department of Surgery, The University of Western Ontario, St. Joseph's Health Care London, 268 Grosvenor St, N6A 4V2, London, Ontario, Canada
| |
Collapse
|
20
|
Martínez B, Rodríguez A, Kulakauskas S, Chapot-Chartier MP. Cell wall homeostasis in lactic acid bacteria: threats and defences. FEMS Microbiol Rev 2021; 44:538-564. [PMID: 32495833 PMCID: PMC7476776 DOI: 10.1093/femsre/fuaa021] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 06/03/2020] [Indexed: 12/16/2022] Open
Abstract
Lactic acid bacteria (LAB) encompasses industrially relevant bacteria involved in food fermentations as well as health-promoting members of our autochthonous microbiota. In the last years, we have witnessed major progresses in the knowledge of the biology of their cell wall, the outermost macrostructure of a Gram-positive cell, which is crucial for survival. Sophisticated biochemical analyses combined with mutation strategies have been applied to unravel biosynthetic routes that sustain the inter- and intra-species cell wall diversity within LAB. Interplay with global cell metabolism has been deciphered that improved our fundamental understanding of the plasticity of the cell wall during growth. The cell wall is also decisive for the antimicrobial activity of many bacteriocins, for bacteriophage infection and for the interactions with the external environment. Therefore, genetic circuits involved in monitoring cell wall damage have been described in LAB, together with a plethora of defence mechanisms that help them to cope with external threats and adapt to harsh conditions. Since the cell wall plays a pivotal role in several technological and health-promoting traits of LAB, we anticipate that this knowledge will pave the way for the future development and extended applications of LAB.
Collapse
Affiliation(s)
- Beatriz Martínez
- DairySafe research group. Department of Technology and Biotechnology of Dairy Products. Instituto de Productos Lácteos de Asturias, IPLA-CSIC. Paseo Río Linares s/n. 33300 Villaviciosa, Spain
| | - Ana Rodríguez
- DairySafe research group. Department of Technology and Biotechnology of Dairy Products. Instituto de Productos Lácteos de Asturias, IPLA-CSIC. Paseo Río Linares s/n. 33300 Villaviciosa, Spain
| | - Saulius Kulakauskas
- Université Paris-Saclay, INRAE, AgroParisTech, Micalis Institute, 78350, Jouy-en-Josas, France
| | | |
Collapse
|
21
|
Puebla-Barragan S, Reid G. Probiotics in Cosmetic and Personal Care Products: Trends and Challenges. Molecules 2021; 26:1249. [PMID: 33652548 PMCID: PMC7956298 DOI: 10.3390/molecules26051249] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/20/2021] [Accepted: 02/22/2021] [Indexed: 12/11/2022] Open
Abstract
Probiotics, defined as "live microorganisms that, when administered in adequate amounts, confer a health benefit on the host," are becoming increasingly popular and marketable. However, too many of the products currently labelled as probiotics fail to comply with the defining characteristics. In recent years, the cosmetic industry has increased the number of products classified as probiotics. While there are several potential applications for probiotics in personal care products, specifically for oral, skin, and intimate care, proper regulation of the labelling and marketing standards is still required to guarantee that consumers are indeed purchasing a probiotic product. This review explores the current market, regulatory aspects, and potential applications of probiotics in the personal care industry.
Collapse
Affiliation(s)
- Scarlett Puebla-Barragan
- Centre for Human Microbiome and Probiotics, Lawson Health Research Institute, London, ON N6C 2R5, Canada
- Departments of Microbiology & Immunology and Surgery, University of Western Ontario, London, ON N6A 3K7, Canada;
| | - Gregor Reid
- Centre for Human Microbiome and Probiotics, Lawson Health Research Institute, London, ON N6C 2R5, Canada
- Departments of Microbiology & Immunology and Surgery, University of Western Ontario, London, ON N6A 3K7, Canada;
| |
Collapse
|
22
|
Peluzio MDCG, Martinez JA, Milagro FI. Postbiotics: Metabolites and mechanisms involved in microbiota-host interactions. Trends Food Sci Technol 2021. [DOI: 10.1016/j.tifs.2020.12.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
23
|
Probiotic consumption relieved human stress and anxiety symptoms possibly via modulating the neuroactive potential of the gut microbiota. Neurobiol Stress 2021; 14:100294. [PMID: 33511258 PMCID: PMC7816019 DOI: 10.1016/j.ynstr.2021.100294] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 01/02/2021] [Accepted: 01/03/2021] [Indexed: 02/08/2023] Open
Abstract
Stress has been shown to disturb the balance of human intestinal microbiota and subsequently causes mental health problems like anxiety and depression. Our previous study showed that ingesting the probiotic strain, Lactobacillus (L.) plantarum P-8, for 12 weeks could alleviate stress and anxiety of stressed adults. The current study was a follow-up work aiming to investigate the functional role of the gut metagenomes in the observed beneficial effects. The fecal metagenomes of the probiotic (n = 43) and placebo (n = 36) receivers were analyzed in depth. The gut microbiomes of the placebo group at weeks 0 and 12 showed a significantly greater Aitchison distance (P < 0.001) compared with the probiotic group. Meanwhile, the Shannon diversity index of the placebo group (P < 0.05) but not the probiotic group decreased significantly at week 12. Additionally, significantly more species-level genome bins (SGBs) of Bifidobacterium adolescentis, Bifidobacterium longum, and Fecalibacterium prausnitzii (P < 0.01) were identified in the fecal metagenomes of the probiotic group, while the abundances of SGBs representing the species Roseburia faecis and Fusicatenibacter saccharivorans decreased significantly (P < 0.05). Furthermore, the 12-week probiotic supplementation enhanced the diversity of neurotransmitter-synthesizing/consuming SGBs and the levels of some predicted microbial neuroactive metabolites (e.g., short-chain fatty acids, gamma-aminobutyric acid, arachidonic acid, and sphingomyelin). Our results showed a potential link between probiotic-induced gut microbiota modulation and stress/anxiety alleviation in stressed adults, supporting that the gut-brain axis was involved in relieving stress-related symptoms. The beneficial effect relied not only on microbial diversity changes but more importantly gut metagenome modulations at the SGB and functional gene levels. Stress and anxiety are related to gut dysbiosis. Ingesting Lactobacillus plantarum P-8 (P-8) improved stress/anxiety symptoms. Symptoms were relieved by modulating gut microbiota and neuroactive potential.
Collapse
|
24
|
Alfarrayeh I, Fekete C, Gazdag Z, Papp G. Propolis ethanolic extract has double-face in vitro effect on the planktonic growth and biofilm formation of some commercial probiotics. Saudi J Biol Sci 2021; 28:1033-1039. [PMID: 33424397 PMCID: PMC7785450 DOI: 10.1016/j.sjbs.2020.11.047] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 01/14/2023] Open
Abstract
This study investigated the in vitro effect of propolis ethanolic extract (PEE) on planktonic growth and biofilm forming abilities of five commercial probiotics (Enterol, Protexin, Normaflore, BioGaia and Linex). Broth microdilution method was used to investigate the susceptibility of the microbes of five commercial probiotics to PEE. Crystal violet assay was used for the quantitative assessment of biofilm formation and mature biofilm eradication tests. Effect of PEE on autoaggregation ability and swarming motility of Normaflore microbes was determined. Planktonic forms of probiotics showed varied susceptibilities with minimal inhibitory concentration values in the range of 100-800 µg/mL of PEE. However, low PEE concentrations significantly enhanced the planktonic growth of Linex and BioGaia microbes. Biofilm studies revealed that Enterol and Protexin were non-biofilm formers, while BioGaia, Linex and Normaflore showed weak biofilms, which were inhibited by 12.5, 25, and 800 µg/mL of PEE, respectively. PEE revealed double-face effect on the biofilms of Normaflore and Linex, which were enhanced at low concentrations of PEE and inhibited at higher concentrations. Interestingly, Normaflore biofilms were shifted from weak to strong biofilms at low PEE concentrations (12.5, 25, and 50 µg/mL). In conclusion, PEE has strain dependent controversial effects on the planktonic growth and biofilm forming ability of the tested probiotics, although high concentrations have inhibitory effect on all of them, low concentrations may have strain dependent prebiotic effect.
Collapse
Affiliation(s)
- Ibrahim Alfarrayeh
- Department of General and Environmental Microbiology, University of Pécs, Faculty of Sciences, Pécs, Hungary
| | - Csaba Fekete
- Department of General and Environmental Microbiology, University of Pécs, Faculty of Sciences, Pécs, Hungary
| | - Zoltán Gazdag
- Department of General and Environmental Microbiology, University of Pécs, Faculty of Sciences, Pécs, Hungary
| | - Gábor Papp
- Department of General and Environmental Microbiology, University of Pécs, Faculty of Sciences, Pécs, Hungary
| |
Collapse
|
25
|
García-Velasco JA, Budding D, Campe H, Malfertheiner SF, Hamamah S, Santjohanser C, Schuppe-Koistinen I, Nielsen HS, Vieira-Silva S, Laven J. The reproductive microbiome - clinical practice recommendations for fertility specialists. Reprod Biomed Online 2020; 41:443-453. [PMID: 32753361 DOI: 10.1016/j.rbmo.2020.06.014] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Revised: 06/12/2020] [Accepted: 06/19/2020] [Indexed: 02/08/2023]
Abstract
The interest in and understanding of the human microbiome has grown remarkably over recent years. Advances in molecular techniques have allowed researchers to identify and study the microbiota and also use this information to develop therapeutic solutions for a spectrum of conditions. Alongside the growing interest in the microbiome, societal changes have resulted in many couples looking to start families later in life, therefore increasing the demand for assisted reproductive technologies. Combining these trends, it makes sense that clinicians are eager to understand and exploit the microbiome of their patients, i.e. the reproductive microbiome, in order to help them achieve their goal of becoming parents. This paper aims to provide an overview of the current and future research into the reproductive microbiome in relation to fertility and also share clinical practice recommendations for physicians who are new to this field or unsure about how they can utilise what is known to help their patients.
Collapse
Affiliation(s)
- Juan A García-Velasco
- Department of Reproductive Endocrinology and Infertility, IVI Madrid, Rey Juan Carlos University, Madrid 28023, Spain.
| | | | | | | | - Samir Hamamah
- Centre Hospitalier Universitaire de Montpellier, Service Biologie de la Reproduction, 34295 Montpellier, Inserm U1203, France
| | | | - Ina Schuppe-Koistinen
- Karolinska Institutet, Department of Microbiology, Tumor and Cell Biology, Centre for Translational Microbiome Research (CTMR), Stockholm 171 77, Sweden
| | - Henriette Svarre Nielsen
- Department of Obstetrics and Gynecology, Hvidovre Hospital, Copenhagen University Hospital, Hvidovre 2650, Denmark
| | - Sara Vieira-Silva
- Laboratory of Molecular Bacteriology, Department of Microbiology and Immunology, Rega Institute, KU Leuven, Leuven 3000, Belgium; Center for Microbiology, VIB, Leuven, Belgium
| | - Joop Laven
- Erasmus MC, University Medical Center Rotterdam, Division of Reproductive Medicine, Rotterdam 3015 GD, the Netherlands
| |
Collapse
|
26
|
Dietary supplementation with spray-dried porcine plasma has prebiotic effects on gut microbiota in mice. Sci Rep 2020; 10:2926. [PMID: 32076042 PMCID: PMC7031359 DOI: 10.1038/s41598-020-59756-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 01/28/2020] [Indexed: 02/06/2023] Open
Abstract
In animal models of inflammation and in farm animals, dietary inclusion of spray-dried porcine plasma (SDP) reduces mucosal inflammation. Here, we study whether these effects could be mediated by changes in the intestinal microbiota and if these changes are similar to those induced by oral antibiotics. Weaned 21-day-old C57BL/6 mice were divided into 3 groups: the CTL group, fed the control diet; the COL group, administered low doses of neomycin and colistin; and the SDP group, supplemented with 8% SDP. After 14 days, analysis of the fecal microbiome showed that the microbiota profiles induced by SDP and the antibiotics were very different, thus, SDP has prebiotic rather than antibiotic effects. At the phylum level, SDP stimulated the presence of Firmicutes, considerably increasing the lactobacilli population. It also enhanced the growth of species involved in regulatory T-lymphocyte homeostasis and restoration of the mucosal barrier, as well as species negatively correlated with expression of pro-inflammatory cytokines. At the mucosal level, expression of toll-like receptors Tlr2, Tlr4 and Tlr9, and mucous-related genes Muc2 and Tff3 with regulatory and barrier stability functions, were increased. SDP also increased expression of Il-10 and Tgf-β, as well as markers of macrophages and dendritic cells eventually promoting an immune-tolerant environment.
Collapse
|
27
|
Abstract
Industrial biotechnology is a continuously expanding field focused on the application of microorganisms to produce chemicals using renewable sources as substrates. Currently, an increasing interest in new versatile processes, able to utilize a variety of substrates to obtain diverse products, can be observed. A robust microbial strain is critical in the creation of such processes. Lactic acid bacteria (LAB) are used to produce a wide variety of chemicals with high commercial interest. Lactic acid (LA) is the most predominant industrial product obtained from LAB fermentations, and its production is forecasted to rise as the result of the increasing demand of polylactic acid. Hence, the creation of new ways to revalorize LA production processes is of high interest and could further enhance its economic value. Therefore, this review explores some co-products of LA fermentations, derived from LAB, with special focus on bacteriocins, lipoteichoic acid, and probiotics. Finally, a multi-product process involving LA and the other compounds of interest is proposed.
Collapse
|