1
|
Angulo M, Angulo C. Analysis of the potential long-lasting effects of probiotic Debaryomyces hansenii CBS 8339 on trained immunity in newborn goats. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2025; 162:105292. [PMID: 39577680 DOI: 10.1016/j.dci.2024.105292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 11/19/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Trained immunity has been described as the memory capacity of the innate immune system. Several microbial components have been shown to induce trained immunity. Research on the potential of probiotics to trigger these effects has been limited to a few in vitro studies but remains completely unknown in vivo. Components from the probiotic Debaryomyces hansenii CBS 8339 (Dh) have been shown to induce innate immune memory in goat kids and calves. In the present study, stimulating innate immune cells from newborn goats with probiotic Dh increased respiratory burst activity and nitric oxide production, while cell phagocytosis was unaffected. Glucose uptake was enhanced in goat's cells stimulated with Dh, but lactate production was decreased. In newborn goats, after the training scheme (via oral probiotic administration), cell phagocytosis, nitric oxide production and glycolysis - through the upregulation of AKT and HIF1A gene expression, glucose consumption and lactate production- were enhanced. The expression of IL1B gene was similar between the D. hansenii and control groups. Moreover, the potential long-lasting effects were assessed 30 days after initiation of the training scheme. Cell phagocytosis, respiratory burst and myeloperoxidase activity were enhanced, while glycolytic parameters remained unaffected. Altogether, the results of the present study suggest that the immune training scheme may induce trained immunity by the probiotic D. hansenii in newborn goats. However, our findings were not conclusive regarding the long-lasting (one-month) effects of trained immunity by probiotics.
Collapse
Affiliation(s)
- Miriam Angulo
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S, C.P. 23096, Mexico; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S, C.P. 23096, Mexico
| | - Carlos Angulo
- Immunology & Vaccinology Group. Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S, C.P. 23096, Mexico; Laboratorio Nacional CONAHCYT de Generación de Vacunas Veterinarias y Servicios de Diagnóstico (LNC-GVD). Centro de Investigaciones Biológicas del Noroeste, S.C., Instituto Politécnico Nacional 195, Playa Palo de Santa Rita Sur, La Paz, B.C. S, C.P. 23096, Mexico.
| |
Collapse
|
2
|
Neelawala RN, Edison LK, Kariyawasam S. Pre-Harvest Non-Typhoidal Salmonella Control Strategies in Commercial Layer Chickens. Animals (Basel) 2024; 14:3578. [PMID: 39765482 PMCID: PMC11672659 DOI: 10.3390/ani14243578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 12/03/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Non-typhoidal Salmonella (NTS) infections in poultry, particularly in commercial-layer chickens, pose a critical risk to food safety and public health worldwide. NTS bacteria can remain undetected in poultry flocks, contaminating products and potentially leading to gastroenteritis in humans. This review examines pre-harvest control strategies for NTS in layer chickens, including biosecurity protocols, vaccinations, feed additives, genetic selection, and environmental management. These strategies have substantially reduced Salmonella colonization and product contamination rates in the commercial layer industry. By evaluating these strategies, this review highlights the importance of integrated control measures to limit NTS colonization, reduce antimicrobial resistance, and improve poultry health. This review aims to provide producers, researchers, and policymakers with insights into effective practices to minimize Salmonella contamination and enhance both animal and human health outcomes.
Collapse
Affiliation(s)
| | | | - Subhashinie Kariyawasam
- Department of Comparative, Diagnostic, and Population Medicine, College of Veterinary Medicine, University of Florida, Gainesville, FL 32608, USA; (R.N.N.); (L.K.E.)
| |
Collapse
|
3
|
Taufer CR, da Silva J, Rampelotto PH. In Silico Analysis of Probiotic Bacteria Changes Across COVID-19 Severity Stages. Microorganisms 2024; 12:2353. [PMID: 39597740 PMCID: PMC11596909 DOI: 10.3390/microorganisms12112353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 11/29/2024] Open
Abstract
The gut microbiota plays a crucial role in modulating the immune response during COVID-19, with several studies reporting significant alterations in specific bacterial genera, including Akkermansia, Bacteroides, Bifidobacterium, Faecalibacterium, Lactobacillus, Oscillospira, and Ruminococcus. These genera are symbionts of the gut microbiota and contribute to host health. However, comparing results across studies is challenging due to differences in analysis methods and reference databases. We screened 16S rRNA raw datasets available in public databases on COVID-19, focusing on the V3-V4 region of the bacterial genome. In total, seven studies were included. All samples underwent the same bioinformatics pipeline, evaluating the differential abundance of these seven bacterial genera at each level of severity. The reanalysis identified significant changes in differential abundance. Bifidobacterium emerged as a potential biomarker of disease severity and a therapeutic target. Bacteroides presented a complex pattern, possibly related to disease-associated inflammation or opportunistic pathogen growth. Lactobacillus showed significant changes in abundance across the COVID-19 stages. On the other hand, Akkermansia and Faecalibacterium did not show significant differences, while Oscillospira and Ruminococcus produced statistically significant results but with limited relevance to COVID-19 severity. Our findings reveal new insights into the differential abundance of key bacterial genera in COVID-19, particularly Bifidobacterium and Bacteroides.
Collapse
Affiliation(s)
- Clarissa Reginato Taufer
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil; (C.R.T.); (J.d.S.)
| | - Juliana da Silva
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil; (C.R.T.); (J.d.S.)
- Graduate Program in Health and Human Development, Universidade La Salle, Canoas 92010-000, Brazil
| | - Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| |
Collapse
|
4
|
Zhang S, Ma J, Ma Y, Yi J, Wang B, Wang H, Yang Q, Zhang K, Yan X, Sun D, You J. Engineering Probiotics for Diabetes Management: Advances, Challenges, and Future Directions in Translational Microbiology. Int J Nanomedicine 2024; 19:10917-10940. [PMID: 39493275 PMCID: PMC11530765 DOI: 10.2147/ijn.s492651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 10/08/2024] [Indexed: 11/05/2024] Open
Abstract
Background Diabetes Mellitus (DM) is a substantial health concern worldwide, and its incidence is progressively escalating. Conventional pharmacological interventions frequently entail undesirable side effects, and while probiotics offer benefits, they are hindered by constraints such as diminished stability and effectiveness within the gastrointestinal milieu. Given these complications, the advent of bioengineered probiotics is a promising alternative for DM management. Aim of Review The objective of this review is to provide an exhaustive synthesis of the most recent studies on the use of engineered probiotics in the management of DM. This study aimed to clarify the mechanisms through which these probiotics function, evaluate their clinical effectiveness, and enhance public awareness of their prospective advantages in the treatment of DM. Key Scientific Concepts of Review Scholarly critiques have explored diverse methodologies of probiotic engineering, including physical alteration, bioenrichment, and genetic manipulation. These techniques augment the therapeutic potency of probiotics by ameliorating gut microbiota, fortifying the intestinal barrier, modulating metabolic pathways, and regulating immune responses. Such advancements have established engineered probiotics as a credible therapeutic strategy for DM, potentially providing enhanced results compared to conventional treatments.
Collapse
Affiliation(s)
- Shenghao Zhang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Yilei Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Jia Yi
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
| | - Beier Wang
- Department of Hepatobiliary-Pancreatic Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, People’s Republic of China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, Hong Kong SAR, 999077, People’s Republic of China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, People’s Republic of China
| | - Kun Zhang
- Chongqing Municipality Clinical Research Center for Endocrinology and Metabolic Diseases, Chongqing University Three Gorges Hospital, Chongqing, 404000, People’s Republic of China
| | - Xiaoqing Yan
- The Chinese-American Research Institute for Diabetic Complications, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, 325035, People’s Republic of China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, People’s Republic of China
- Department of Endocrinology, Yiwu Central Hospital, The Affiliated Yiwu hospital of Wenzhou Medical University, Yiwu, 322000, People’s Republic of China
| | - Jinfeng You
- Department of Obstetrics, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou, 324000, People’s Republic of China
| |
Collapse
|
5
|
Spencer KD, Bline H, Chen HJ, Verosky BG, Hilt ME, Jaggers RM, Gur TL, Mathé EA, Bailey MT. Modulation of anxiety-like behavior in galactooligosaccharide-fed mice: A potential role for bacterial tryptophan metabolites and reduced microglial reactivity. Brain Behav Immun 2024; 121:229-243. [PMID: 39067620 DOI: 10.1016/j.bbi.2024.07.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 07/02/2024] [Accepted: 07/20/2024] [Indexed: 07/30/2024] Open
Abstract
Prebiotic galactooligosaccharides (GOS) reduce anxiety-like behaviors in mice and humans. However, the biological pathways behind these behavioral changes are not well understood. To begin to study these pathways, we utilized C57BL/6 mice that were fed a standard diet with or without GOS supplementation for 3 weeks prior to testing on the open field. After behavioral testing, colonic contents and serum were collected for bacteriome (16S rRNA gene sequencing, colonic contents only) and metabolome (UPLC-MS, colonic contents and serum data) analyses. As expected, GOS significantly reduced anxiety-like behavior (i.e., increased time in the center) and decreased cytokine gene expression (Tnfa and Ccl2) in the prefrontal cortex. Notably, time in the center of the open field was significantly correlated with serum methyl-indole-3-acetic acid (methyl-IAA). This metabolite is a methylated form of indole-3-acetic acid (IAA) that is derived from bacterial metabolism of tryptophan. Sequencing analyses showed that GOS significantly increased Lachnospiraceae UCG006 and Akkermansia; these taxa are known to metabolize both GOS and tryptophan. To determine the extent to which methyl-IAA can affect anxiety-like behavior, mice were intraperitoneally injected with methyl-IAA. Mice given methyl-IAA had a reduction in anxiety-like behavior in the open field, along with lower Tnfa in the prefrontal cortex. Methyl-IAA was also found to reduce TNF-α (as well as CCL2) production by LPS-stimulated BV2 microglia. Together, these data support a novel pathway through which GOS reduces anxiety-like behaviors in mice and suggests that the bacterial metabolite methyl-IAA reduces microglial cytokine and chemokine production, which in turn reduces anxiety-like behavior.
Collapse
Affiliation(s)
- Kyle D Spencer
- Graduate Partnership Program, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA; Department of Biomedical Informatics, The Ohio State University, Columbus, OH, USA; Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Heather Bline
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Helen J Chen
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Branden G Verosky
- Medical Scientist Training Program, The Ohio State University, Columbus, OH, USA; Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Miranda E Hilt
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Biomedical Sciences Graduate Program, The Ohio State University College of Medicine, Columbus, OH, USA
| | - Robert M Jaggers
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - Tamar L Gur
- Department of Neuroscience, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Department of Psychiatry & Behavioral Health, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Ewy A Mathé
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, NIH, Rockville, MD, USA
| | - Michael T Bailey
- Center for Microbial Pathogenesis, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Institute for Behavioral Medicine Research, The Ohio State University Wexner Medical Center, Columbus, OH, USA; Oral and GI Research Affinity Group, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH, USA.
| |
Collapse
|
6
|
de Oliveira LD, de Carvalho LS, Xavier ACC, de Oliveira FE, Leão MVP, Diamantino MGG, Khoury RD, Valera MC, Carvalho CAT, Abu Hasna A. In Vitro Evaluation of Sodium Hypochlorite, Chlorhexidine, Propolis, and Calcium Hydroxide Effect on Lipoteichoic-Acid-Induced Proinflammatory Cytokines Production. Dent J (Basel) 2024; 12:286. [PMID: 39329852 PMCID: PMC11431833 DOI: 10.3390/dj12090286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 08/27/2024] [Accepted: 08/30/2024] [Indexed: 09/28/2024] Open
Abstract
This study aimed to evaluate the effects of sodium hypochlorite (NaOCl), chlorhexidine (CHX), and the glycolic extract of propolis (GEP) as endodontic irrigants and of calcium hydroxide [Ca(OH)2], CHX, or Ca(OH)2 + CHX as intracanal medications on the capacity of the lipoteichoic acid (LTA) of Enterococcus faecalis in macrophages' proinflammatory cytokines production. Freshly extracted 108 human single-rooted teeth were used in this study. The LTA of E. faecalis was standardized in double-distilled pyrogen-free water (250 µg/mL) and inoculated into the specimens subdivided into nine subgroups (n = 12). Cultures of murine macrophages (RAW 264.7) were treated with 30 µL of each sample collected from root canals and incubated (37 °C, 5% CO2) for 24 h. Lastly, anti-TNF-α, anti-IL-6, anti-IP-10, anti-MIP-1α, anti-G-CSF, and anti-IL-1β DuoSet kits were used to perform an ELISA assay. Data were analyzed using one-way ANOVA and Tukey test (p ≥ 0.05). It was found that 1% NaOCl was the most effective irrigant in reducing the capacity of LTA in cytokines production, followed by 12% GEP and 2% CHX, respectively. Ca(OH)2 + CHX presented the best results when associated with NaOCl or GEP. Thus, NaOCl or GEP associated with Ca(OH)2 + CHX were effective in reducing the capacity of LTA in different macrophages pro-inflammatory cytokines production.
Collapse
Affiliation(s)
- Luciane Dias de Oliveira
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, Campus of São José dos Campos, São Paulo State University, São Paulo 12245-000, Brazil; (L.D.d.O.); (L.S.d.C.); (M.V.P.L.)
| | - Lara Steffany de Carvalho
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, Campus of São José dos Campos, São Paulo State University, São Paulo 12245-000, Brazil; (L.D.d.O.); (L.S.d.C.); (M.V.P.L.)
| | - Ana Claudia Carvalho Xavier
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, Campus of São José dos Campos, São Paulo State University, São Paulo 12245-000, Brazil; (L.D.d.O.); (L.S.d.C.); (M.V.P.L.)
| | - Felipe Eduardo de Oliveira
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, Campus of São José dos Campos, São Paulo State University, São Paulo 12245-000, Brazil; (L.D.d.O.); (L.S.d.C.); (M.V.P.L.)
| | - Mariella Vieira Pereira Leão
- Department of Bioscience and Oral Diagnosis, Institute of Science and Technology, Campus of São José dos Campos, São Paulo State University, São Paulo 12245-000, Brazil; (L.D.d.O.); (L.S.d.C.); (M.V.P.L.)
| | - Mariana Gadelho Gimenez Diamantino
- Department of Restorative Dentistry, Endodontics Division, Institute of Science and Technology, Campus of São José dos Campos, São Paulo State University, São Paulo 12245-000, Brazil; (M.G.G.D.); (R.D.K.); (M.C.V.); (C.A.T.C.)
| | - Rayana Duarte Khoury
- Department of Restorative Dentistry, Endodontics Division, Institute of Science and Technology, Campus of São José dos Campos, São Paulo State University, São Paulo 12245-000, Brazil; (M.G.G.D.); (R.D.K.); (M.C.V.); (C.A.T.C.)
| | - Marcia Carneiro Valera
- Department of Restorative Dentistry, Endodontics Division, Institute of Science and Technology, Campus of São José dos Campos, São Paulo State University, São Paulo 12245-000, Brazil; (M.G.G.D.); (R.D.K.); (M.C.V.); (C.A.T.C.)
| | - Cláudio Antonio Talge Carvalho
- Department of Restorative Dentistry, Endodontics Division, Institute of Science and Technology, Campus of São José dos Campos, São Paulo State University, São Paulo 12245-000, Brazil; (M.G.G.D.); (R.D.K.); (M.C.V.); (C.A.T.C.)
| | - Amjad Abu Hasna
- Department of Restorative Dentistry, Endodontics Division, Institute of Science and Technology, Campus of São José dos Campos, São Paulo State University, São Paulo 12245-000, Brazil; (M.G.G.D.); (R.D.K.); (M.C.V.); (C.A.T.C.)
- School of Dentistry, Universidad Espíritu Santo, Samborondón 092301, Ecuador
| |
Collapse
|
7
|
Xu C, Aqib AI, Fatima M, Muneer S, Zaheer T, Peng S, Ibrahim EH, Li K. Deciphering the Potential of Probiotics in Vaccines. Vaccines (Basel) 2024; 12:711. [PMID: 39066349 PMCID: PMC11281421 DOI: 10.3390/vaccines12070711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 06/13/2024] [Accepted: 06/18/2024] [Indexed: 07/28/2024] Open
Abstract
The demand for vaccines, particularly those prepared from non-conventional sources, is rising due to the emergence of drug resistance around the globe. Probiotic-based vaccines are a wise example of such vaccines which represent new horizons in the field of vaccinology in providing an enhanced and diversified immune response. The justification for incorporating probiotics into vaccines lies in the fact that that they hold the capacity to regulate immune function directly or indirectly by influencing the gastrointestinal microbiota and related pathways. Several animal-model-based studies have also highlighted the efficacy of these vaccines. The aim of this review is to collect and summarize the trends in the recent scientific literature regarding the role of probiotics in vaccines and vaccinology, along with their impact on target populations.
Collapse
Affiliation(s)
- Chang Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| | - Amjad Islam Aqib
- Department of Medicine, Cholistan University of Veterinary and Animal Sciences, Bahawalpur 63100, Pakistan
| | - Mahreen Fatima
- Faculty of Biosciences, Cholistan University of Veterinary and Animal Sciences, Bahawalpur 63100, Pakistan;
| | - Sadia Muneer
- Institute of Microbiology, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan
| | - Tean Zaheer
- Department of Parasitology, University of Agriculture Faisalabad, Faisalabad 38000, Pakistan;
| | - Song Peng
- Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China;
| | - Essam H. Ibrahim
- Biology Department, Faculty of Science, King Khalid University, P.O. Box 9004, Abha 61413, Saudi Arabia
| | - Kun Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, China
| |
Collapse
|
8
|
Shi Z, Nan Y, Zhou X, Zhang W, Zhang Z, Zhang C, Duan H, Ge J, Zhao L. Molecular Mechanisms of Intestinal Protection by Levilactobacillus brevis 23017 against Salmonella typhimurium C7731-Induced Damage: Role of Nrf2. Microorganisms 2024; 12:1135. [PMID: 38930517 PMCID: PMC11205325 DOI: 10.3390/microorganisms12061135] [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: 04/11/2024] [Revised: 05/29/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
The treatment and prevention of pathogenic diseases by lactic acid bacteria (LAB) has attracted more and more attention. As a special LAB, Levilactobacillus brevis (L. brevis) has relatively less research on its antibacterial infection in vivo, and its protective effect and mechanism still need to be fully studied. In this study, we selected L. brevis 23017, which can regulate the intestinal immunity of the host animal and resist pathogen infection, to evaluate its protective role and potential molecular mechanisms in the mouse model of S. typhimurium C7731 infection. As expected, we confirmed that L. brevis 23017 reduced the diarrhea rate and increased the daily weight gain and survival rate of the mouse model, and inhibited S. typhimurium colonization in the jejunum and liver. It also reduced the level of oxidative damage and protected the integrity of intestinal tissue by increasing the activity of intestinal antioxidant enzymes (SOD, GSH-Px and T-AOC). From the perspective of intestinal mucosal barrier injury and repair, it was confirmed that L. brevis 23017 could increase the expression levels of intestinal tight junction proteins (ZO-1 and OCLN). Our research results also show that L. brevis 23017 inhibits the inflammatory response and promotes the occurrence of cellular immunity in the body by promoting the increase in IL-10 and inhibiting IL-13 in serum and intestinal tissue. Notably, L. brevis 23017 increased total secretory immunoglobulin A (SIgA) levels in the intestine, which were closely associated with elevated levels of IL-5, IL-13, pIgR, j-chain, and IgAα-chain. In addition, L. brevis 23017 increased the expression of antioxidant proteins Nrf2, NQO1, and HO-1 associated with Nrf2 signaling to inhibit intestinal oxidative damage. This mechanism may be responsible for its protective effect against S. typhimurium-infected intestine. Our study provides new evidence and theoretical support for the analysis of the anti-bacterial infection effect and mechanism of L. brevis, which will contribute to the development of L. brevis and the treatment of pathogenic bacteria intestinal infection.
Collapse
Affiliation(s)
- Ziqi Shi
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China;
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Yongchao Nan
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Xinyao Zhou
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Wenzhi Zhang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Zheng Zhang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Chuankun Zhang
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Haoyuan Duan
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Junwei Ge
- Heilongjiang Provincial Key Laboratory of Zoonosis, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China; (Y.N.); (X.Z.); (W.Z.); (Z.Z.); (C.Z.); (H.D.)
| | - Lili Zhao
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research of the Ministry of Education, Institute of Zoonosis, and College of Veterinary Medicine, Jilin University, Changchun 130062, China;
| |
Collapse
|
9
|
Taufer CR, da Silva J, Rampelotto PH. The Influence of Probiotic Lactobacilli on COVID-19 and the Microbiota. Nutrients 2024; 16:1350. [PMID: 38732597 PMCID: PMC11085918 DOI: 10.3390/nu16091350] [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: 03/07/2024] [Revised: 04/01/2024] [Accepted: 04/03/2024] [Indexed: 05/13/2024] Open
Abstract
This comprehensive review explores the potential of using lactobacilli as a probiotic in the management of COVID-19. Our findings suggest that lactobacilli show promise in reducing the risk of death, gastrointestinal and overall symptoms, and respiratory failure, as well as in lowering cytokines and inflammatory markers associated with the disease. The molecular mechanisms by which lactobacilli protect against COVID-19 and other viral infections may be related to the reduction in inflammation, modulation of the immune response, and direct interaction with viruses to produce antiviral substances. However, the selected studies demonstrate the presence of mixed findings for various clinical, biochemical, hematological, and immunological parameters, which may be attributed to methodological differences among studies. We highlight the importance of clearly describing randomization processes to minimize bias and caution against small sample sizes and inappropriate statistical tests that could lead to errors. This review offers valuable insights into the therapeutic potential of lactobacilli in the context of COVID-19 and identifies avenues for further research and applications. These findings hold promise for the development of novel approaches to managing COVID-19 and warrant further investigation into the potential benefits of lactobacilli in combating the disease.
Collapse
Affiliation(s)
- Clarissa Reginato Taufer
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | - Juliana da Silva
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
- Graduate Program in Health and Human Development, Universidade La Salle, Canoas 92010-000, Brazil
| | - Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| |
Collapse
|
10
|
Junaid M, Lu H, Din AU, Yu B, Liu Y, Li Y, Liu K, Yan J, Qi Z. Deciphering Microbiome, Transcriptome, and Metabolic Interactions in the Presence of Probiotic Lactobacillus acidophilus against Salmonella Typhimurium in a Murine Model. Antibiotics (Basel) 2024; 13:352. [PMID: 38667028 PMCID: PMC11047355 DOI: 10.3390/antibiotics13040352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 04/06/2024] [Accepted: 04/08/2024] [Indexed: 04/29/2024] Open
Abstract
Salmonella enterica serovar Typhimurium (S. Typhimurium), a foodborne pathogen that poses significant public health risks to humans and animals, presents a formidable challenge due to its antibiotic resistance. This study explores the potential of Lactobacillus acidophilus (L. acidophilus 1.3251) probiotics as an alternative strategy to combat antibiotic resistance associated with S. Typhimurium infection. In this investigation, twenty-four BALB/c mice were assigned to four groups: a non-infected, non-treated group (CNG); an infected, non-treated group (CPG); a group fed with L. acidophilus but not infected (LAG); and a group fed with L. acidophilus and challenged with Salmonella (LAST). The results revealed a reduction in Salmonella levels in the feces of mice, along with restored weight and improved overall health in the LAST compared to the CPG. The feeding of L. acidophilus was found to downregulate pro-inflammatory cytokine mRNA induced by Salmonella while upregulating anti-inflammatory cytokines. Additionally, it influenced the expression of mRNA transcript, encoding tight junction protein, oxidative stress-induced enzymes, and apoptosis-related mRNA expression. Furthermore, the LEfSe analysis demonstrated a significant shift in the abundance of critical commensal genera in the LAST, essential for maintaining gut homeostasis, metabolic reactions, anti-inflammatory responses, and butyrate production. Transcriptomic analysis revealed 2173 upregulated and 506 downregulated differentially expressed genes (DEGs) in the LAST vs. the CPG. Functional analysis of these DEGs highlighted their involvement in immunity, metabolism, and cellular development. Kyoto Encyclopedia of Genes and Genome (KEGG) pathway analysis indicated their role in tumor necrosis factor (TNF), mitogen-activated protein kinase (MAPK), chemokine, Forkhead box O (FOXO), and transforming growth factor (TGF-β) signaling pathway. Moreover, the fecal metabolomic analysis identified 929 differential metabolites, with enrichment observed in valine, leucine, isoleucine, taurine, glycine, and other metabolites. These findings suggest that supplementation with L. acidophilus promotes the growth of beneficial commensal genera while mitigating Salmonella-induced intestinal disruption by modulating immunity, gut homeostasis, gut barrier integrity, and metabolism.
Collapse
Affiliation(s)
| | - Hongyu Lu
- Medical College, Guangxi University, Nanning 530004, China
| | - Ahmad Ud Din
- Plants for Human Health Institute, North Carolina State University, 600 Laureate Way, Kannapolis, NC 28081, USA
| | - Bin Yu
- Medical College, Guangxi University, Nanning 530004, China
| | - Yu Liu
- Medical College, Guangxi University, Nanning 530004, China
| | - Yixiang Li
- Medical College, Guangxi University, Nanning 530004, China
| | - Kefei Liu
- Tianjin Shengji Group., Co., Ltd., No. 2, Hai Tai Development 2nd Road, Huayuan Industrial Zone, Tianjin 300384, China
| | - Jianhua Yan
- Medical College, Guangxi University, Nanning 530004, China
| | - Zhongquan Qi
- Medical College, Guangxi University, Nanning 530004, China
| |
Collapse
|
11
|
Taufer CR, Rampelotto PH. Lactobacilli in COVID-19: A Systematic Review Based on Next-Generation Sequencing Studies. Microorganisms 2024; 12:284. [PMID: 38399688 PMCID: PMC10891515 DOI: 10.3390/microorganisms12020284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 01/25/2024] [Accepted: 01/26/2024] [Indexed: 02/25/2024] Open
Abstract
The global pandemic was caused by the SARS-CoV-2 virus, known as COVID-19, which primarily affects the respiratory and intestinal systems and impacts the microbial communities of patients. This systematic review involved a comprehensive search across the major literature databases to explore the relationship between lactobacilli and COVID-19. Our emphasis was on investigations employing NGS technologies to explore this connection. Our analysis of nine selected studies revealed that lactobacilli have a reduced abundance in the disease and an association with disease severity. The protective mechanisms of lactobacilli in COVID-19 and other viral infections are likely to be multifaceted, involving complex interactions between the microbiota, the host immune system, and the virus itself. Moreover, upon closely examining the NGS methodologies and associated statistical analyses in each research study, we have noted concerns regarding the approach used to delineate the varying abundance of lactobacilli, which involves potential biases and the exclusion of pertinent data elements. These findings provide new insight into the relationship between COVID-19 and lactobacilli, highlighting the potential for microbiota modulation in COVID-19 treatment.
Collapse
Affiliation(s)
- Clarissa Reginato Taufer
- Graduate Program in Genetics and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| | - Pabulo Henrique Rampelotto
- Bioinformatics and Biostatistics Core Facility, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre 91501-970, Brazil
| |
Collapse
|
12
|
Zaib S, Hayat A, Khan I. Probiotics and their Beneficial Health Effects. Mini Rev Med Chem 2024; 24:110-125. [PMID: 37291788 DOI: 10.2174/1389557523666230608163823] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/10/2023]
Abstract
Probiotics are living microorganisms that are present in cultured milk and fermented food. Fermented foods are a rich source for the isolation of probiotics. They are known as good bacteria. They have various beneficial effects on human health including antihypertensive effects, antihypercholesterolemic effects, prevention of bowel disease, and improving the immune system. Microorganisms including bacteria, yeast, and mold are used as probiotics but the major microorganisms that are used as probiotics are bacteria from the genus Lactobacillus, Lactococcus, Streptococcus, and Bifidobacterium. Probiotics are beneficial in the prevention of harmful effects. Recently, the use of probiotics for the treatment of various oral and skin diseases has also gained significant attention. Clinical studies indicate that the usage of probiotics can alter gut microbiota composition and provoke immune modulation in a host. Due to their various health benefits, probiotics are attaining more interest as a substitute for antibiotics or anti-inflammatory drugs leading to the growth of the probiotic market.
Collapse
Affiliation(s)
- Sumera Zaib
- Department of Basic and Applied Chemistry, Faculty of Sciences and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Aqsa Hayat
- Department of Basic and Applied Chemistry, Faculty of Sciences and Technology, University of Central Punjab, Lahore, 54590, Pakistan
| | - Imtiaz Khan
- Department of Chemistry, Manchester Institute of Biotechnology, The University of Manchester, 131, Princess Street, Manchester M1 7DN, United Kingdom
| |
Collapse
|
13
|
Angriman I, Scarpa M, Savarino E, Patuzzi I, Rigo A, Kotsafti A, Stepanyan A, Sciuto E, Celotto F, Negro S, Caruso A, Ruffolo C, Bardini R, Pucciarelli S, Barberio B, Spolverato G, Zingone F, D'Incà R, Castagliuolo I, Scarpa M. Oral administration of Lactobacillus casei DG® after ileostomy closure in restorative proctocolectomy: a randomized placebo-controlled trial (microbiota and immune microenvironment in pouchitis -MEP1). Gut Microbes 2024; 16:2423037. [PMID: 39485259 PMCID: PMC11540070 DOI: 10.1080/19490976.2024.2423037] [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: 05/09/2024] [Revised: 07/29/2024] [Accepted: 10/24/2024] [Indexed: 11/03/2024] Open
Abstract
Pouchitis is an idiopathic inflammatory disease that may occur in ileal pouches, and it can lead to ileal pouch failure. This was a single-center, randomized, double-blinded, placebo-controlled trial that assessed the effect of Lactobacillus casei (L. casei) DG®, a probiotic strain, on the ileal pouch mucosa to determine the crosstalk between microbiota and mucosal immune system. Fifty-two patients undergoing restorative proctocolectomy were recruited and randomly assigned to receive a daily oral supplementation of L. casei DG® (n = 26) or placebo (n = 26) for 8 weeks from the ileostomy closure (T0) to a pouch endoscopy after 8 weeks (T1) and 1 year (T2). Ileal pouch mucosa samples were collected at T0, T1, and T2. At T1, the L. casei DG®-supplemented group showed a significant reduction of inflammatory cytokines levels compared to T0 baseline levels in the pouch mucosa, whereas in the placebo group cytokines levels resulted stable. In conclusion, probiotic manipulation of mucosal microbiota by L. casei DG®-supplementation after stoma closure in patients who underwent restorative proctocolectomy has a beneficial impact on the ileal pouch microenvironment. Registration number: NCT03136419 (http://www.clinicaltrials.gov).
Collapse
Affiliation(s)
- Imerio Angriman
- General Surgery 3 Unit, Department of Surgical, Oncological and Gastroenterological Sciences, DiSCOG, University of Padova, Padova, Italy
| | - Melania Scarpa
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Edoardo Savarino
- Gastroenterology Unit, Department of Surgical Oncological and Gastroenterological Sciences DiSCOG, University of Padova, Padova, Italy
| | | | - Alessandra Rigo
- Gastroenterology Unit, Department of Surgical Oncological and Gastroenterological Sciences DiSCOG, University of Padova, Padova, Italy
| | - Andromachi Kotsafti
- Immunology and Molecular Oncology Diagnostics, Veneto Institute of Oncology IOV-IRCCS, Padova, Italy
| | - Astghik Stepanyan
- General Surgery 3 Unit, Department of Surgical, Oncological and Gastroenterological Sciences, DiSCOG, University of Padova, Padova, Italy
| | - Elisa Sciuto
- General Surgery 3 Unit, Department of Surgical, Oncological and Gastroenterological Sciences, DiSCOG, University of Padova, Padova, Italy
| | - Francesco Celotto
- General Surgery 3 Unit, Department of Surgical, Oncological and Gastroenterological Sciences, DiSCOG, University of Padova, Padova, Italy
| | - Silvia Negro
- General Surgery 3 Unit, Department of Surgical, Oncological and Gastroenterological Sciences, DiSCOG, University of Padova, Padova, Italy
| | - Antonino Caruso
- Gastroenterology Unit, Department of Surgical Oncological and Gastroenterological Sciences DiSCOG, University of Padova, Padova, Italy
| | - Cesare Ruffolo
- General Surgery 3 Unit, Department of Surgical, Oncological and Gastroenterological Sciences, DiSCOG, University of Padova, Padova, Italy
| | - Romeo Bardini
- General Surgery 3 Unit, Department of Surgical, Oncological and Gastroenterological Sciences, DiSCOG, University of Padova, Padova, Italy
| | - Salvatore Pucciarelli
- General Surgery 3 Unit, Department of Surgical, Oncological and Gastroenterological Sciences, DiSCOG, University of Padova, Padova, Italy
| | - Brigida Barberio
- Gastroenterology Unit, Department of Surgical Oncological and Gastroenterological Sciences DiSCOG, University of Padova, Padova, Italy
| | - Gaya Spolverato
- General Surgery 3 Unit, Department of Surgical, Oncological and Gastroenterological Sciences, DiSCOG, University of Padova, Padova, Italy
| | - Fabiana Zingone
- Gastroenterology Unit, Department of Surgical Oncological and Gastroenterological Sciences DiSCOG, University of Padova, Padova, Italy
| | - Renata D'Incà
- Gastroenterology Unit, Department of Surgical Oncological and Gastroenterological Sciences DiSCOG, University of Padova, Padova, Italy
| | | | - Marco Scarpa
- General Surgery 3 Unit, Department of Surgical, Oncological and Gastroenterological Sciences, DiSCOG, University of Padova, Padova, Italy
| |
Collapse
|
14
|
Gupta U, Dey P. Rise of the guardians: Gut microbial maneuvers in bacterial infections. Life Sci 2023; 330:121993. [PMID: 37536616 DOI: 10.1016/j.lfs.2023.121993] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 07/23/2023] [Accepted: 07/29/2023] [Indexed: 08/05/2023]
Abstract
AIMS Bacterial infections are one of the major causes of mortality globally. The gut microbiota, primarily comprised of the commensals, performs an important role in maintaining intestinal immunometabolic homeostasis. The current review aims to provide a comprehensive understanding of how modulation of the gut microbiota influences opportunistic bacterial infections. MATERIALS AND METHODS Primarily centered around mechanisms related to colonization resistance, nutrient, and metabolite-associated factors, mucosal immune response, and commensal-pathogen reciprocal interactions, we discuss how gut microbiota can promote or prevent bacterial infections. KEY FINDINGS Opportunistic infections can occur directly due to obligate pathogens or indirectly due to the overgrowth of opportunistic pathobionts. Gut microbiota-centered mechanisms of altered intestinal immunometabolic and metabolomic homeostasis play a significant role in infection promotion and prevention. Depletion in the population of commensals, increased abundance of pathobionts, and overall decrease in gut microbial diversity and richness caused due to prolonged antibiotic use are risk factors of opportunistic bacterial infections, including infections from multidrug-resistant spp. Gut commensals can limit opportunistic infections by mechanisms including the production of antimicrobials, short-chain fatty acids, bile acid metabolism, promoting mucin formation, and maintaining immunological balance at the mucosa. Gut microbiota-centered strategies, including the administration of probiotics and fecal microbiota transplantation, could help attenuate opportunistic bacterial infections. SIGNIFICANCE The current review discussed the gut microbial population and function-specific aspects contributing to bacterial infection susceptibility and prophylaxis. Collectively, this review provides a comprehensive understanding of the mechanisms related to the dual role of gut microbiota in bacterial infections.
Collapse
Affiliation(s)
- Upasana Gupta
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, Punjab, India
| | - Priyankar Dey
- Department of Biotechnology, Thapar Institute of Engineering and Technology, Patiala 147004, Punjab, India.
| |
Collapse
|
15
|
LI C, YANG Y, FENG C, LI H, QU Y, WANG Y, WANG D, WANG Q, GUO J, SHI T, SUN X, WANG X, HOU Y, SUN Z, YANG T. Integrated 'omics analysis for the gut microbiota response to moxibustion in a rat model of chronic fatigue syndrome. J TRADIT CHIN MED 2023; 43:1176-1189. [PMID: 37946480 PMCID: PMC10623263 DOI: 10.19852/j.cnki.jtcm.20231018.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/08/2023] [Indexed: 11/12/2023]
Abstract
OBJECTIVE To observe the efficacy of moxibustion in the treatment of chronic fatigue syndrome (CFS) and explore the effects on gut microbiota and metabolic profiles. METHODS Forty-eight male Sprague-Dawley rats were randomly assigned to control group (Con), CFS model group (Mod, established by multiple chronic stress for 35 d), MoxA group (CFS model with moxibustion Shenque (CV8) and Guanyuan (CV4), 10 min/d, 28 d) and MoxB group (CFS model with moxibustion Zusanli (ST36), 10 min/d, 28 d). Open-field test (OFT) and Morris-water-maze test (MWMT) were determined for assessment the CFS model and the therapeutic effects of moxibustion.16S rRNA gene sequencing analysis based gut microbiota integrated untargeted liquid chromatograph-mass spectrometer (LC-MS) based fecal metabolomics were executed, as well as Spearman correlation analysis, was utilized to uncover the functional relevance between the potential metabolites and gut microbiota. RESULTS The results of our behavioral tests showed that moxibustion improved the performance of CFS rats in the OFT and the MWMT. Microbiome profiling analysis revealed that the gut microbiomes of CFS rats were less diverse with altered composition, including increases in pro-inflammatory species (such as Proteobacteria) and decreases in anti-inflammatory species (such as Bacteroides, Lactobacillus, Ruminococcus, and Prevotella). Moxibustion partially normalized these changes in the gut microbiota. Furthermore, CFS was associated with metabolic disorders, which were effectively ameliorated by moxibustion. This was demonstrated by the normalization of 33 microbiota-related metabolites, including mannose (P = 0.001), aspartic acid (P = 0.009), alanine (P = 0.007), serine (P = 0.000), threonine (P = 0.027), methionine (P = 0.023), 5-hydroxytryptamine (P = 0.008), alpha-linolenic acid (P = 0.003), eicosapentaenoic acid (P = 0.006), hypoxanthine (P = 0.000), vitamin B6 (P = 0.000), cholic acid (P = 0.013), and taurocholate (P = 0.002). Correlation analysis showed a significant association between the perturbed fecal microbiota and metabolite levels, with a notable negative relationship between LCA and Bacteroides. CONCLUSIONS In this study, we demonstrated that moxibustion has an antifatigue-like effect. The results from the 16S rRNA gene sequencing and metabolomics analysis suggest that the therapeutic effects of moxibustion on CFS are related to the regulation of gut microorganisms and their metabolites. The increase in Bacteroides and decrease in LCA may be key targets for the moxibustion treatment of CFS.
Collapse
Affiliation(s)
- Chaoran LI
- 1 Department of Acupuncture, Zhejiang Chinese Medical University, Hangzhou 310000, China
| | - Yan YANG
- 2 Department of Chinese Medical Literature, College of Basic Medicine, Heilongjiang University of Chinese medicine, Harbin 150040, China
| | - Chuwen FENG
- 3 Department of Rehabilitation, the First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Heng LI
- 7 Shanghai Applied Protein Technology Co., Ltd., Shanghai 200233, China
| | - Yuanyuan QU
- 5 Graduate School, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Yulin WANG
- 6 Department of Acupuncture, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Delong WANG
- 6 Department of Acupuncture, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Qingyong WANG
- 5 Graduate School, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Jing GUO
- 5 Graduate School, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Tianyu SHI
- 5 Graduate School, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Xiaowei SUN
- 4 Department of Acupuncture, the First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Xue WANG
- 8 Department of Acupuncture, Chongqing Changshou District People's Hospital, Chongqing 401220, China
| | - Yunlong HOU
- 9 College of integrated Chinese and Western Medicine, Hebei University of Chinese Medicine, and National Key Laboratory of Collateral Disease Research and Innovative Chinese Medicine, Hebei 050000, China
| | - Zhongren SUN
- 6 Department of Acupuncture, the Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin 150040, China
| | - Tiansong YANG
- 10 Department of Rehabilitation, the First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, and Traditional Chinese Medicine Informatics Key Laboratory of Heilongjiang Province, Harbin 150040, China
| |
Collapse
|
16
|
Summer M, Ali S, Fiaz U, Tahir HM, Ijaz M, Mumtaz S, Mushtaq R, Khan R, Shahzad H, Fiaz H. Therapeutic and immunomodulatory role of probiotics in breast cancer: A mechanistic review. Arch Microbiol 2023; 205:296. [PMID: 37486419 DOI: 10.1007/s00203-023-03632-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 07/03/2023] [Accepted: 07/09/2023] [Indexed: 07/25/2023]
Abstract
Breast cancer has become the most prevalent and noxious type of malignancy around the globe (Giaquinto et al., 2022). Multiple clinical strategies including chemotherapy, radiotherapy, and immunotherapy have been in practice to manage breast cancer. Besides the protective roles of conventional remedial approaches, and non-reversible and deteriorative impacts like healthy cell damage, organ failure, etc., the world scientific community is in a continuous struggle to find some alternative biocompatible and comparatively safe solutions. Among novel breast cancer management/treatment options, the role of probiotics has become immensely important. The current review encompasses the prevalence statistics of breast cancer across the globe concerning developed and undeveloped counties, intestinal microbiota linkage with breast cancer, and association of breast microbiome with breast carcinoma. Furthermore, this review also narrates the role of probiotics against breast cancer and their mode of action. In Vivo and In Vitro studies under breast cancer research regarding probiotics are mechanistically explained. The current review systematically explains the immunomodulatory role of probiotics to prevent breast cancer. Last, but not the least, current review concludes the use of probiotics in the treatment of breast cancer through various mechanisms and future recommendations for molecular basis studies.
Collapse
Affiliation(s)
- Muhammad Summer
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Shaukat Ali
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan.
| | - Umaima Fiaz
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Hafiz Muhammad Tahir
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Muhammad Ijaz
- Department of Veterinary Medicine, University of Veterinary and Animal Sciences Lahore, Lahore, Pakistan
| | - Shumaila Mumtaz
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Rabia Mushtaq
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Rida Khan
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Hafsa Shahzad
- Department of Zoology, Government College University Lahore, Lahore, 54000, Pakistan
| | - Hashim Fiaz
- Department of Medicine and Surgery, Ammer-ul-din Medical College Lahore, Lahore, Pakistan
| |
Collapse
|
17
|
Li Q, Zheng T, Ding H, Chen J, Li B, Zhang Q, Yang S, Zhang S, Guan W. Exploring the Benefits of Probiotics in Gut Inflammation and Diarrhea-From an Antioxidant Perspective. Antioxidants (Basel) 2023; 12:1342. [PMID: 37507882 PMCID: PMC10376667 DOI: 10.3390/antiox12071342] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/17/2023] [Accepted: 06/19/2023] [Indexed: 07/30/2023] Open
Abstract
Inflammatory bowel disease (IBD), characterized by an abnormal immune response, includes two distinct types: Crohn's disease (CD) and ulcerative colitis (UC). Extensive research has revealed that the pathogeny of IBD encompasses genetic factors, environmental factors, immune dysfunction, dysbiosis, and lifestyle choices. Furthermore, patients with IBD exhibit both local and systemic oxidative damage caused by the excessive presence of reactive oxygen species. This oxidative damage exacerbates immune response imbalances, intestinal mucosal damage, and dysbiosis in IBD patients. Meanwhile, the weaning period represents a crucial phase for pigs, during which they experience pronounced intestinal immune and inflammatory responses, leading to severe diarrhea and increased mortality rates. Pigs are highly similar to humans in terms of physiology and anatomy, making them a potential choice for simulating human IBD. Although the exact mechanism behind IBD and post-weaning diarrhea remains unclear, the oxidative damage, in its progression and pathogenesis, is well acknowledged. Besides conventional anti-inflammatory drugs, certain probiotics, particularly Lactobacillus and Bifidobacteria strains, have been found to possess antioxidant properties. These include the scavenging of reactive oxygen species, chelating metal ions to inhibit the Fenton reaction, and the regulation of host antioxidant enzymes. Consequently, numerous studies in the last two decades have committed to exploring the role of probiotics in alleviating IBD. Here, we sequentially discuss the oxidative damage in IBD and post-weaning diarrhea pathogenesis, the negative consequences of oxidative stress on IBD, the effectiveness of probiotics in IBD treatment, the application of probiotics in weaned piglets, and the potential antioxidant mechanisms of probiotics.
Collapse
Affiliation(s)
- Qihui Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Tenghui Zheng
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Hanting Ding
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Jiaming Chen
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Baofeng Li
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Qianzi Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Siwang Yang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
| | - Shihai Zhang
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| | - Wutai Guan
- Guangdong Province Key Laboratory of Animal Nutrition Control, College of Animal Science, South China Agricultural University, Guangzhou 510642, China
- College of Animal Science and National Engineering Research Center for Breeding Swine Industry, South China Agricultural University, Guangzhou 510642, China
- Guangdong Laboratory for Lingnan Modern Agriculture, South China Agricultural University, Guangzhou 510642, China
| |
Collapse
|
18
|
Rashid M, Narang A, Thakur S, Jain SK, Kaur S. Therapeutic and prophylactic effects of oral administration of probiotic Enterococcus faecium Smr18 in Salmonella enterica-infected mice. Gut Pathog 2023; 15:23. [PMID: 37208771 DOI: 10.1186/s13099-023-00548-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 04/25/2023] [Indexed: 05/21/2023] Open
Abstract
Salmonella enterica serotype Typhi causes chronic enteric fever known as typhoid. Prolonged treatment regimen used for the treatment of typhoid and indiscriminate use of antibiotics has led to the emergence of resistant strains of S. enterica that has further increased the severity of the disease. Therefore, alternative therapeutic agents are urgently required. In this study, probiotic and enterocin-producing bacteria Enterococcus faecium Smr18 was compared for both its prophylactic and therapeutic efficacy in S. enterica infection mouse model. E. faecium Smr18 possessed high tolerance to bile salts and simulated gastric juice, as treatment for 3 and 2 h resulted in 0.5 and 0.23 log10 reduction in the colony forming units, respectively. It exhibited 70% auto aggregation after 24 h of incubation and formed strong biofilms at both pH 5 and 7. Oral administration of E. faecium in BALB/c mice infected with S. enterica significantly (p < 0.05) reduced the mortality of the infected mice and prevented the weight loss in mice. Administration of E. faecium prior to infection inhibited the translocation of S. enterica to liver and spleen, whereas, its administration post-infection completely cleared the pathogen from the organs within 8 days. Further, in both pre- and post-E. faecium-treated infected groups, sera levels of liver enzymes were restored back to normal; whereas the levels of creatinine, urea and antioxidant enzymes were significantly (p < 0.05) reduced compared to the untreated-infected group. E. faecium Smr18 administration significantly increased the sera levels of nitrate by 1.63-fold and 3.22-fold in pre- and post-administration group, respectively. Sera levels of interferon-γ was highest (tenfold) in the untreated-infected group, whereas the levels of interleukin-10 was highest in the post-infection E. faecium-treated group thereby indicating the resolution of infection in the probiotic-treated group, plausibly due to the increased production of reactive nitrogen intermediates.
Collapse
Affiliation(s)
- Muzamil Rashid
- Department of Microbiology, Guru Nanak Dev University, Amritsar, India
| | - Anmol Narang
- Department of Microbiology, Guru Nanak Dev University, Amritsar, India
| | - Shubham Thakur
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Subheet Kumar Jain
- Department of Pharmaceutical Sciences, Guru Nanak Dev University, Amritsar, India
| | - Sukhraj Kaur
- Department of Microbiology, Guru Nanak Dev University, Amritsar, India.
| |
Collapse
|
19
|
Xu X, Qiao Y, Peng Q, Shi B. Probiotic Properties of Loigolactobacillus coryniformis NA-3 and In Vitro Comparative Evaluation of Live and Heat-Killed Cells for Antioxidant, Anticancer and Immunoregulatory Activities. Foods 2023; 12:foods12051118. [PMID: 36900635 PMCID: PMC10001366 DOI: 10.3390/foods12051118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 03/03/2023] [Accepted: 03/04/2023] [Indexed: 03/09/2023] Open
Abstract
Some Latiactobacilli are often used as probiotics due to their functional activities, including antioxidant, anticancer and immunoregulation effect. Loigolactobacillus coryniformis NA-3 obtained from our laboratory is a promising probiotic according to the previous study. Coculture, the Oxford cup test and disk-diffusion methods were used to evaluate the probiotic properties and antibiotic resistance of L. coryniformis NA-3. The antioxidant activities of live and heat-killed L. coryniformis NA-3 were assessed via radicals' scavenging ability. The potential anticancer and immunoregulatory capacity was determined in vitro using cell lines. The results indicate that L. coryniformis NA-3 has antibacterial activity and cholesterol removal ability and is sensitive to most antibiotics. Dead L. coryniformis NA-3 can scavenge free radicals as well as live strains. Live L. coryniformis NA-3 can significantly inhibit the proliferation of colon cancer cells; however, dead cells cannot. After RAW 264.7 macrophages were treated with live and heat-killed L. coryniformis NA-3, the production of NO, IL-6, TNF-α and reactive oxygen species (ROS) was induced. The increased expression of inducible nitric oxide synthase (iNOS) in treated macrophages mediates the production of NO. In conclusion, L. coryniformis NA-3 showed potential probiotic properties, and the heat-killed strain also exhibited activities similar to those of live bacteria, suggesting the possible value of its further application in the food processing and pharmaceutical industries.
Collapse
|
20
|
Particulate Cell Wall Materials of Lactobacillus acidophilus as Vaccine Adjuvant. Vet Sci 2022; 9:vetsci9120698. [PMID: 36548859 PMCID: PMC9783621 DOI: 10.3390/vetsci9120698] [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: 10/22/2022] [Revised: 12/08/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
We evaluated Lactobacillus acidophilus (LA) for adjuvant application in animal vaccines. LA particles (LAPs) are made by treating LA with purification processes and high-pressure homogenization (HPH). We found that LAPs treated with HPH with trehalose and emulsifiers had an average particle size of 179 nm, considerably smaller than LAPs without additives. First, we evaluated the adjuvanticity of LAPs using a murine model with ovalbumin antigens, revealing that LAPs, especially in a five-fold concentration, could induce a considerable antibody response compared with other current adjuvants. In poultry vaccination tests using inactivated Newcastle disease virus, LAPs alone could induce a similar antibody response compared to commercial water-in-oil (W/O) adjuvant ISA70, a commercial adjuvant, at weeks 4 and 6; however, they declined faster than ISA70 at weeks 8 and 10. LAPs added to conventional adjuvant materials, such as mineral oil-based O/W emulsions, showed similar adjuvanticity to ISA70. LA-H5-C, composed of carbomer, emulsifiers and trehalose showed no significant body weight change in acute toxicity compared to other adjuvants including ISA70, making formulated LAPs a potential candidate for use as a veterinary vaccine adjuvant.
Collapse
|
21
|
Shojadoost B, Alizadeh M, Boodhoo N, Astill J, Karimi SH, Shoja Doost J, Taha-Abdelaziz K, Kulkarni R, Sharif S. Effects of Treatment with Lactobacilli on Necrotic Enteritis in Broiler Chickens. Probiotics Antimicrob Proteins 2022; 14:1110-1129. [PMID: 35044636 DOI: 10.1007/s12602-021-09901-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/25/2021] [Indexed: 12/25/2022]
Abstract
Growth promoter antibiotics have been commonly used for the control of necrotic enteritis (NE) in broilers for decades. However, due to a ban on the use of these antibiotics, alternatives such as probiotics have been tested widely for NE control. The present study tested the efficacy of four different species of lactobacilli (two isolates of Lactobacillus johnsonii and one of Ligilactobacillus (L.) salivarius, Limosilactobacillus (L.) reuteri, and L. crispatus) against NE. Day-old male broiler chickens were divided into six groups and orally inoculated with 1 × 107 or 1 × 108 colony-forming units (CFU) of lactobacilli on 1, 7, 14, and 20 days of age. While negative and positive control groups did not receive lactobacilli, the latter was challenged with Clostridium perfringens (CP). Chickens, at 21 days old, were challenged for 3 days with 3 × 108 CFU of a virulent strain of CP. Tissues were collected for immune system gene expression, immunophenotyping, intestinal histomorphometry, and microbiota analysis. Lactobacilli inoculation conferred partial protection in chickens against NE, marked by lowered lesion scores and improved villus:crypt ratio. Immunomodulatory effects were demonstrated by the significant alteration of interferon (IFN)-γ, interleukin (IL)-1β, IL-2, IL-12p35, IL-17, and transforming growth factor beta (TGF-β) gene transcription in the duodenum and jejunum as well as subtle changes in the frequency of CD8 + T cells and B cells in the cecal tonsil of the treated chickens. Microbiota analysis showed increased levels of some bacterial phyla including Actinobacteria, Lactobacillaceae, and Firmicutes. In conclusion, these findings suggest that the use of certain lactobacilli can reduce NE severity and modulate immune responses and intestinal microbiota composition in chickens.
Collapse
Affiliation(s)
- Bahram Shojadoost
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Mohammadali Alizadeh
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Nitish Boodhoo
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Jake Astill
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Seyed Hossein Karimi
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Janan Shoja Doost
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada
| | - Khaled Taha-Abdelaziz
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.,Pathology Department, Faculty of Veterinary Medicine, Beni-Suef University, Al Shamlah, Beni-Suef, 62511, Egypt.,Animal and Veterinary Sciences Department, Clemson University, Clemson, SC, 29634, USA
| | - Raveendra Kulkarni
- Dept of Population Health and Pathobiology, College of Vet Medicine, North Carolina State University, 1060 William Moore Dr, Raleigh, NC -27607, USA
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, ON, N1G 2W1, Canada.
| |
Collapse
|
22
|
Kim JY, Kim JY, Kim H, Moon EC, Heo K, Shim JJ, Lee JL. Immunostimulatory effects of dairy probiotic strains Bifidobacterium animalis ssp. lactis HY8002 and Lactobacillus plantarum HY7717. JOURNAL OF ANIMAL SCIENCE AND TECHNOLOGY 2022; 64:1117-1131. [PMID: 36812033 PMCID: PMC9890336 DOI: 10.5187/jast.2022.e84] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 10/05/2022] [Accepted: 10/13/2022] [Indexed: 12/14/2022]
Abstract
Previous studies reported that Bifidobacterium animalis ssp. lactis HY8002 (HY8002) improved intestinal integrity and had immunomodulatory effects. Lactobacillus plantarum HY7717 (HY7717) was screened in vitro from among 21 other lactic acid bacteria (LAB) and demonstrated nitric oxide (NO) production. The aims of this study were to investigate the individual and combined ex vivo and in vivo effects of LAB strains HY8002 and HY7717 at immunostimulating mice that have been challenged with an immunosuppressant drug. The combination of HY8002 and HY7717 increased the secretion of cytokines such as interferon (IFN)-γ, interleukin (IL)-12, and tumor necrosis factor (TNF)-α in splenocytes. In a cyclophosphamide (CTX)-induced immunosuppression model, administration of the foregoing LAB combination improved the splenic and hematological indices, activated natural killer (NK) cells, and up-regulated plasma immunoglobulins and cytokines. Moreover, this combination treatment increased Toll-like receptor 2 (TLR2) expression. The ability of the combination treatment to upregulate IFN-γ and TNF-α in the splenocytes was inhibited by anti-TLR2 antibody. Hence, the immune responses stimulated by the combination of HY8002 and HY7717 are associated with TLR2 activation. The preceding findings suggest that the combination of the HY8002 and HY7717 LAB strains could prove to be a beneficial and efficacious immunostimulant probiotic supplement. The combination of the two probiotic strains will be applied on the dairy foods including yogurt and cheese.
Collapse
Affiliation(s)
- Ju-Yeon Kim
- R&BD Center, hy Co.,
Ltd., Yongin 17086, Korea
| | - Joo Yun Kim
- R&BD Center, hy Co.,
Ltd., Yongin 17086, Korea
| | - Hyeonji Kim
- R&BD Center, hy Co.,
Ltd., Yongin 17086, Korea
| | | | - Keon Heo
- R&BD Center, hy Co.,
Ltd., Yongin 17086, Korea,Corresponding author: Keon Heo,
R&BD Center, hy Co., Ltd., Yongin 17086, Korea. Tel: +82-70-7835-6040,
E-mail:
| | | | | |
Collapse
|
23
|
Lacticaseibacillus casei Strain Shirota Modulates Macrophage-Intestinal Epithelial Cell Co-Culture Barrier Integrity, Bacterial Sensing and Inflammatory Cytokines. Microorganisms 2022; 10:microorganisms10102087. [PMID: 36296363 PMCID: PMC9607601 DOI: 10.3390/microorganisms10102087] [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: 08/23/2022] [Revised: 10/03/2022] [Accepted: 10/06/2022] [Indexed: 11/06/2022] Open
Abstract
Probiotic bacteria modulate macrophage immune inflammatory responses, with functional cytokine responses determined by macrophage subset polarisation, stimulation and probiotic strain. Mucosal macrophages exhibit subset functional heterogeneity but are organised in a 3-dimensional tissue, over-laid by barrier epithelial cells. This study aimed to investigate the effects of the probiotic Lacticaseibacillus casei strain Shirota (LcS) on macrophage-epithelial cell cytokine responses, pattern recognition receptor (PRR) expression and LPS responses and the impacts on barrier integrity. THP-1-derived M1 and M2 subset macrophages were co-cultured in a transwell system with differentiated Caco-2 epithelial cells in the presence or absence of enteropathogenic LPS. Both Caco-2 cells in monoculture and macrophage co-culture were assayed for cytokines, PRR expression and barrier integrity (TEER and ZO-1) by RT-PCR, ELISA, IHC and electrical resistance. Caco-2 monocultures expressed distinct cytokine profiles (IL-6, IL-8, TNFα, endogenous IL-10), PRRs and barrier integrity, determined by inflammatory context (TNFα or IL-1β). In co-culture, LcS rescued ZO-1 and TEER in M2/Caco-2, but not M1/Caco-2. LcS suppressed TLR2, TLR4, MD2 expression in both co-cultures and differentially regulated NOD2, TLR9, Tollip and cytokine secretion. In conclusion, LcS selectively modulates epithelial barrier integrity, pathogen sensing and inflammatory cytokine profile; determined by macrophage subset and activation status.
Collapse
|
24
|
Lactobacillus supports Clostridiales to restrict gut colonization by multidrug-resistant Enterobacteriaceae. Nat Commun 2022; 13:5617. [PMID: 36153315 PMCID: PMC9509339 DOI: 10.1038/s41467-022-33313-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 09/09/2022] [Indexed: 11/10/2022] Open
Abstract
Infections by multidrug-resistant Enterobacteriaceae (MRE) are life-threatening to patients. The intestinal microbiome protects against MRE colonization, but antibiotics cause collateral damage to commensals and open the way to colonization and subsequent infection. Despite the significance of this problem, the specific commensals and mechanisms that restrict MRE colonization remain largely unknown. Here, by performing a multi-omic prospective study of hospitalized patients combined with mice experiments, we find that Lactobacillus is key, though not sufficient, to restrict MRE gut colonization. Lactobacillus rhamnosus and murinus increase the levels of Clostridiales bacteria, which induces a hostile environment for MRE growth through increased butyrate levels and reduced nutrient sources. This mechanism of colonization resistance, an interaction between Lactobacillus spp. and Clostridiales involving cooperation between microbiota members, is conserved in mice and patients. These results stress the importance of exploiting microbiome interactions for developing effective probiotics that prevent infections in hospitalized patients. Multidrug-resistant Enterobacteriaceae (MRE) represent a major threat for patients’ health. Here, the authors describe how cooperation between gut commensal bacteria (Lactobacillus spp. And Clostridiales) restrict MRE colonization in mice and patients
Collapse
|
25
|
Ali MS, Lee EB, Quah Y, Birhanu BT, Suk K, Lim SK, Park SC. Heat-killed Limosilactobacillus reuteri PSC102 Ameliorates Impaired Immunity in Cyclophosphamide-induced Immunosuppressed Mice. Front Microbiol 2022; 13:820838. [PMID: 36033865 PMCID: PMC9413535 DOI: 10.3389/fmicb.2022.820838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
The immune functions of heat-killed Limosilactobacillus reuteri PSC102 (hLR) were investigated in cyclophosphamide (CP)-treated immunosuppressed mice. BALB/c mice were randomly divided into five groups: normal control group, CP group, CP treated with levamisole (positive control group), and CP treated with low- and high-dose hLR. After receiving the samples for 21 days, mice were sacrificed, and different parameters, such as immune organ index, immune blood cells, splenocyte proliferation, lymphocyte subpopulations, cytokines, and immunoglobulins, were analyzed. Results showed that the immune organ (thymus and spleen) indices of hLR treatment groups were significantly increased compared to the CP group (p < 0.05). hLR administration prevented CP-induced reduction in the numbers of white blood cells, lymphocytes, midrange absolute, and granulocytes, providing supporting evidence for hematopoietic activities. Splenocyte proliferation and T-lymphocyte (CD4+ and CD8+) subpopulations were also significantly augmented in mice treated with hLR compared to the CP group (p < 0.05). Moreover, Th1-type [interferon-γ, interleukin (IL)-2, and tumor necrosis factor-α] and Th2-type (IL-4 and IL-10) immune factors and immunoglobulin (IgG) showed significant increasing trends (p < 0.05). Additionally, the other proinflammatory cytokines (IL-1β and IL-6) were also significantly elevated (p < 0.05). Taken together, this investigation suggested that orally administered hLR could recover immunosuppression caused by CP and be considered a potential immunostimulatory agent for the treatment of immunosuppressive disorders.
Collapse
Affiliation(s)
- Md. Sekendar Ali
- Department of Biomedical Science and Department of Pharmacology, School of Medicine, Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu, South Korea
- Department of Pharmacy, International Islamic University Chittagong, Kumira, Bangladesh
| | - Eon-Bee Lee
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu, South Korea
| | - Yixian Quah
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu, South Korea
| | - Biruk Tesfaye Birhanu
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu, South Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, South Korea
| | - Kyoungho Suk
- Department of Biomedical Science and Department of Pharmacology, School of Medicine, Brain Science and Engineering Institute, Kyungpook National University, Daegu, South Korea
| | - Suk-Kyung Lim
- Bacterial Disease Division, Animal and Plant Quarantine Agency, Gimcheon-si, South Korea
| | - Seung-Chun Park
- Laboratory of Veterinary Pharmacokinetics and Pharmacodynamics, College of Veterinary Medicine, Kyungpook National University, Daegu, South Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, South Korea
- *Correspondence: Seung-Chun Park,
| |
Collapse
|
26
|
Gou HZ, Zhang YL, Ren LF, Li ZJ, Zhang L. How do intestinal probiotics restore the intestinal barrier? Front Microbiol 2022; 13:929346. [PMID: 35910620 PMCID: PMC9330398 DOI: 10.3389/fmicb.2022.929346] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/27/2022] [Indexed: 12/14/2022] Open
Abstract
The intestinal barrier is a structure that prevents harmful substances, such as bacteria and endotoxins, from penetrating the intestinal wall and entering human tissues, organs, and microcirculation. It can separate colonizing microbes from systemic tissues and prevent the invasion of pathogenic bacteria. Pathological conditions such as shock, trauma, stress, and inflammation damage the intestinal barrier to varying degrees, aggravating the primary disease. Intestinal probiotics are a type of active microorganisms beneficial to the health of the host and an essential element of human health. Reportedly, intestinal probiotics can affect the renewal of intestinal epithelial cells, and also make cell connections closer, increase the production of tight junction proteins and mucins, promote the development of the immune system, regulate the release of intestinal antimicrobial peptides, compete with pathogenic bacteria for nutrients and living space, and interact with the host and intestinal commensal flora to restore the intestinal barrier. In this review, we provide a comprehensive overview of how intestinal probiotics restore the intestinal barrier to provide new ideas for treating intestinal injury-related diseases.
Collapse
Affiliation(s)
- Hong-Zhong Gou
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yu-Lin Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Long-Fei Ren
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Zhen-Jiao Li
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
| | - Lei Zhang
- The First Clinical Medical College, Lanzhou University, Lanzhou, China
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Biotherapy and Regenerative Medicine of Gansu Province, The First Hospital of Lanzhou University, Lanzhou, China
- *Correspondence: Lei Zhang,
| |
Collapse
|
27
|
Darbandi A, Mirkalantari S, Golmoradi Zadeh R, Esghaei M, Talebi M, Kakanj M. Safety evaluation of mutagenicity, genotoxicity, and cytotoxicity of Lactobacillus spp. isolates as probiotic candidates. J Clin Lab Anal 2022; 36:e24481. [PMID: 35582746 PMCID: PMC9279957 DOI: 10.1002/jcla.24481] [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: 09/30/2021] [Revised: 04/15/2022] [Accepted: 04/23/2022] [Indexed: 11/05/2022] Open
Abstract
BACKGROUND Probiotics promote a healthy balance of gut bacteria and have many beneficial effects on human digestive physiology. Although, few side effects of probiotics have been reported. This study aimed to assess the safety of five probiotic candidate Lactobacillus strains isolated from healthy individuals by examining mutagenicity, genotoxicity, and oral toxic effects. METHODS Five selected candidate probiotic (SCPs) strains were evaluated for genotoxicity (Ames test with Salmonella typhimurium), in vitro mammalian chromosome aberration test and an in vivo mouse micronucleus assay on peripheral blood of mice. To evaluate the oral dose toxicity, BALB/c mice models were treated repeatedly (2000, 1000, and 500 mg/kg body weight /day) for 28-days. RESULTS The Ames test performed for two S. typhimurium strains TA 98 and TA100 (both in the absence and in the presence of S-9 metabolic activation system) did not show an increase in reverse mutation because of exposure to the SCPs in any of the doses (5.0, 2.5, 1.25, 0.625, and 0.3125 mg/plate). There was no genotoxicity in the SCPs treatment in the vitro chromosome aberration assay with Chinese hamster ovary cells (CHO-K1). In addition, none of the tested strains increased the frequency of micronucleated reticulocytes in reticulocytes, the SCPs with the studied doses caused no substantial variation in the experimental groups compared to the negative control group (p > 0.05). SCPs were not acutely toxic when administered to male and female BALB/c mice by single gavage at (2000, 1000, and 500 mg/kg b.w/day) with no mortality or clinical signs, change in body weight or macroscopic abnormalities were observed in this dose range. CONCLUSION As a result, SCPs did not induce mutagenic potential in vitro with bacterial reverse mutation, clastogenicity, and in vivo tests in the ranges of concentrations evaluated in our study.
Collapse
Affiliation(s)
- Atieh Darbandi
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shiva Mirkalantari
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rezvan Golmoradi Zadeh
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Esghaei
- Department of Virology, Iran University of Medical Sciences, Tehran, Iran
| | - Malihe Talebi
- Microbial Biotechnology Research Centre, Iran University of Medical Sciences, Tehran, Iran.,Department of Microbiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maryam Kakanj
- Food and Drug Laboratory Research Center, Food and Drug Administration, MOH&ME, Tehran, Iran
| |
Collapse
|
28
|
Phillippi DT, Daniel S, Nguyen KN, Penaredondo BA, Lund AK. Probiotics Function as Immunomodulators in the Intestine in C57Bl/6 Male Mice Exposed to Inhaled Diesel Exhaust Particles on a High-Fat Diet. Cells 2022; 11:cells11091445. [PMID: 35563751 PMCID: PMC9101602 DOI: 10.3390/cells11091445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 04/10/2022] [Accepted: 04/20/2022] [Indexed: 12/04/2022] Open
Abstract
Epidemiological studies reveal a correlation between air pollution exposure and gastrointestinal (GI) diseases, yet few studies have investigated the role of inhaled particulate matter on intestinal integrity in conjunction with a high-fat (HF) diet. Additionally, there is currently limited information on probiotics in mitigating air-pollutant responses in the intestines. Thus, we investigated the hypothesis that exposure to inhaled diesel exhaust particles (DEP) and a HF diet can alter intestinal integrity and inflammation, which can be attenuated with probiotics. 4-6-w-old male C57Bl/6 mice on a HF diet (45% kcal fat) were randomly assigned to be exposed via oropharyngeal aspiration to 35 µg of DEP suspended in 35 µL of 0.9% sterile saline or sterile saline (CON) only twice a week for 4 w. A subset of mice was treated with 0.3 g/day of Winclove Ecologic® barrier probiotics (PRO) in drinking water throughout the duration of the study. Our results show that DEP exposure ± probiotics resulted in increased goblet cells and mucin (MUC)-2 expression, as determined by AB/PAS staining. Immunofluorescent quantification and/or RT-qPCR showed that DEP exposure increases claudin-3, occludin, zona occludens (ZO)-1, matrix metalloproteinase (MMP)-9, and toll-like receptor (TLR)-4, and decreases tumor necrosis factor (TNF)-α and interleukin (IL)-10 expression compared to CON. DEP exposure + probiotics increases expression of claudin-3, occludin, ZO-1, TNF-α, and IL-10 and decreases MMP-9 and TLR-4 compared to CON + PRO in the small intestine. Collectively, these results show that DEP exposure alters intestinal integrity and inflammation in conjunction with a HF diet. Probiotics proved fundamental in understanding the role of the microbiome in protecting and altering inflammatory responses in the intestines following exposure to inhaled DEP.
Collapse
Affiliation(s)
| | | | | | | | - Amie K. Lund
- Correspondence: ; Tel.: +1-(940)-369-8946; Fax: +1-(940)-565-4297
| |
Collapse
|
29
|
Williams LM, Stoodley IL, Berthon BS, Wood LG. The Effects of Prebiotics, Synbiotics, and Short-Chain Fatty Acids on Respiratory Tract Infections and Immune Function: A Systematic Review and Meta-Analysis. Adv Nutr 2022; 13:167-192. [PMID: 34543378 PMCID: PMC8803493 DOI: 10.1093/advances/nmab114] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/25/2021] [Accepted: 09/14/2021] [Indexed: 11/15/2022] Open
Abstract
Prebiotics, synbiotics, and SCFAs have been shown to decrease systemic inflammation and play a protective role in chronic respiratory conditions. However, their effects on infection and immune function are unclear. The objective of this systematic review was to summarize the current evidence for prebiotic, synbiotic, and SCFA supplementation on respiratory tract infections (RTIs) and immune function. The protocol for this systematic review was registered with PROSPERO (National Institute for Health Research, University of York, UK), accessed online at https://www.crd.york.ac.uk/prospero (CRD42019118786). Relevant English-language articles up to May 2021 were identified via online databases: MEDLINE, EMBASE, CINAHL, and Cochrane Library. Included studies (n = 58) examined the effect of prebiotics, synbiotics, or SCFA, delivered orally, on the incidence, severity, or duration of RTIs and/or markers of immune function (e.g., peripheral blood immunophenotyping, NK cell activity). The majority of studies were randomized controlled trials reporting on RTIs in infants and children. The meta-analysis indicated that the numbers of subjects with ≥1 RTI were reduced with prebiotic (OR, 0.73; 95% CI: 0.62-0.86; P = 0.0002; n = 17) and synbiotic (OR, 0.75; 95% CI: 0.65-0.87; P = 0.0001; n = 9) supplementation compared to placebo. Further, NK cell activity was increased with synbiotic (standardized mean difference, 0.74; 95% CI: 0.42-1.06; P < 0.0001, n = 3) supplementation. This review provides evidence that prebiotic, specifically oligosaccharide, supplementation may play a protective role in RTIs in infants and children. There is less evidence for this effect in adults. Supplementation with prebiotic and synbiotic treatment may alter immune function by increasing NK cell activity, though effects on immunophenotype were less clear.
Collapse
Affiliation(s)
- Lily M Williams
- School of Biomedical Sciences & Pharmacy, University of Newcastle, Callaghan, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Isobel L Stoodley
- School of Biomedical Sciences & Pharmacy, University of Newcastle, Callaghan, Australia
| | - Bronwyn S Berthon
- School of Biomedical Sciences & Pharmacy, University of Newcastle, Callaghan, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, Australia
| | - Lisa G Wood
- School of Biomedical Sciences & Pharmacy, University of Newcastle, Callaghan, Australia
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute, New Lambton Heights, Australia
| |
Collapse
|
30
|
Balcells F, Martínez Monteros MJ, Gómez AL, Cazorla SI, Perdigón G, Maldonado-Galdeano C. Probiotic Consumption Boosts Thymus in Obesity and Senescence Mouse Models. Nutrients 2022; 14:nu14030616. [PMID: 35276973 PMCID: PMC8838891 DOI: 10.3390/nu14030616] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 01/21/2022] [Accepted: 01/23/2022] [Indexed: 02/01/2023] Open
Abstract
The ability of the immune system to respond to different pathogens throughout life requires the constant production and selection of T cells in the thymus. This immune organ is very sensitive to age, infectious processes and nutrition disorders (obesity and malnutrition). Several studies have shown that the incorporation of some probiotic bacteria or probiotic fermented milk in the diet has beneficial effects, not only at the intestinal level but also on distant mucosal tissues, improving the architecture of the thymus in a malnutrition model. The aim of the present study was to determine whether supplementation with the probiotic strain Lactobacillus casei CRL 431 and/or its cell wall could improve body weight, intestinal microbiota and thymus structure and function in both obese and aging mice. We evaluated probiotic administration to BALB/c mice in 2 experimental mouse models: obesity and senescence, including mice of different ages (21, 28, 45, 90 and 180 days). Changes in thymus size and histology were recorded. T-lymphocyte population and cytokine production were also determined. The consumption of probiotics improved the cortical/medullary ratio, the production and regulation of cytokines and the recovery of mature T-lymphocyte populations of the thymus in obese and old mice. Probiotic incorporation into the diet could not only modulate the immune system but also lead to thymus function recovery, thus improving quality of life.
Collapse
Affiliation(s)
- Florencia Balcells
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (F.B.); (M.J.M.M.); (S.I.C.); (G.P.)
| | - María José Martínez Monteros
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (F.B.); (M.J.M.M.); (S.I.C.); (G.P.)
| | - Alba Lorena Gómez
- PatLab Laboratorio de Anatomía Patológica Citopatología e Inmunohistoquímica, San Miguel de Tucumán 4000, Argentina;
| | - Silvia Inés Cazorla
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (F.B.); (M.J.M.M.); (S.I.C.); (G.P.)
- Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán 4000, Argentina
| | - Gabriela Perdigón
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (F.B.); (M.J.M.M.); (S.I.C.); (G.P.)
| | - Carolina Maldonado-Galdeano
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán 4000, Argentina; (F.B.); (M.J.M.M.); (S.I.C.); (G.P.)
- Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán 4000, Argentina
- Correspondence:
| |
Collapse
|
31
|
Kaur H, Ali SA. Probiotics and gut microbiota: mechanistic insights into gut immune homeostasis through TLR pathway regulation. Food Funct 2022; 13:7423-7447. [DOI: 10.1039/d2fo00911k] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Consumption of probiotics as a useful functional food improves the host's wellbeing, and, when paired with prebiotics (indigestible dietary fibre/carbohydrate), often benefits the host through anaerobic fermentation.
Collapse
Affiliation(s)
- Harpreet Kaur
- Animal Biochemistry Division, ICAR-NDRI, 132001, India
| | - Syed Azmal Ali
- Cell Biology and Proteomics Lab, Animal Biotechnology Center, ICAR-NDRI, 132001, India
- Division of Proteomics of Stem Cells and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
32
|
Li Q, Tang X, Xu J, Ren X, Wang R, Jiang S. Study on alleviation effect of stachyose on food allergy through TLR2/NF-κB signal pathway in a mouse model. Life Sci 2021; 286:120038. [PMID: 34653427 DOI: 10.1016/j.lfs.2021.120038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 08/27/2021] [Accepted: 09/05/2021] [Indexed: 10/20/2022]
Abstract
AIMS To explore the effect on food allergy of stachyose. MAIN METHODS The egg allergen ovalbumin (OVA) was used to induce a food allergy model of BALB/c mice, and different doses of stachyose were given in process. Using enzyme-linked immunosorbent assay (ELISA) methods to detect the levels of IgE, IgG1, histamine and cytokines. And flow cytometry was used to analyze TH1/TH2 balance further. Besides, Hematoxylin-eosin (HE) staining was used to observe changes of intestinal morphology. Lastly, Reverse Transcription-Polymerase Chain Reaction (RT-qPCR) and western Blot was conducted to explore the possible mechanism. KEY FINDINGS Compared with OVA group, serum IgE and IgG1 levels in the low-dose (1mg/mouse) group and high-dose (5mg/mouse) group of stachyose were significantly reduced (P < 0.05); the level of plasma histamine was also decreased significantly (P < 0.05) and the body temperature were decreased. In all, allergic symptoms were alleviated after stachyose treatment. Furthermore, TH1/TH2 balance was improved after stachyose treatment. Lastly, the expression of TLR2 and NF-κB were increased significantly (P < 0.05) in both mRNA and protein levels after stachyose treatment. SIGNIFICANCE Food allergy was alleviated through improving TH1/TH2 balance by activating TLR2/NF-κB signal by stachyose.
Collapse
Affiliation(s)
- Qian Li
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Xinlei Tang
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Jianghao Xu
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Xingyuan Ren
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Rui Wang
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, Jiangsu 225127, China
| | - Songsong Jiang
- College of Tourism and Culinary Science, Yangzhou University, Yangzhou, Jiangsu 225127, China.
| |
Collapse
|
33
|
Probiotics and Trained Immunity. Biomolecules 2021; 11:biom11101402. [PMID: 34680035 PMCID: PMC8533468 DOI: 10.3390/biom11101402] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Revised: 09/09/2021] [Accepted: 09/15/2021] [Indexed: 12/17/2022] Open
Abstract
The characteristics of innate immunity have recently been investigated in depth in several research articles, and original findings suggest that innate immunity also has a memory capacity, which has been named “trained immunity”. This notion has revolutionized our knowledge of the innate immune response. Thus, stimulation of trained immunity represents a therapeutic alternative that is worth exploring. In this context, probiotics, live microorganisms which when administered in adequate amounts confer a health benefit on the host, represent attractive candidates for the stimulation of trained immunity; however, although numerous studies have documented the beneficial proprieties of these microorganisms, their mechanisms of action are not yet fully understood. In this review, we propose to explore the putative connection between probiotics and stimulation of trained immunity.
Collapse
|
34
|
Buddhasiri S, Sukjoi C, Kaewsakhorn T, Nambunmee K, Nakphaichit M, Nitisinprasert S, Thiennimitr P. Anti-inflammatory Effect of Probiotic Limosilactobacillus reuteri KUB-AC5 Against Salmonella Infection in a Mouse Colitis Model. Front Microbiol 2021; 12:716761. [PMID: 34497597 PMCID: PMC8419263 DOI: 10.3389/fmicb.2021.716761] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Accepted: 08/04/2021] [Indexed: 01/31/2023] Open
Abstract
Acute non-typhoidal salmonellosis (NTS) caused by Salmonella enterica Typhimurium (STM) is among the most prevalent of foodborne diseases. A global rising of antibiotic resistance strains of STM raises an urgent need for alternative methods to control this important pathogen. Major human food animals which harbor STM in their gut are cattle, swine, and poultry. Previous studies showed that the probiotic Limosilactobacillus (Lactobacillus) reuteri KUB-AC5 (AC5) exhibited anti-Salmonella activities in chicken by modulating gut microbiota and the immune response. However, the immunobiotic effect of AC5 in a mammalian host is still not known. Here, we investigated the anti-Salmonella and anti-inflammatory effects of AC5 on STM infection using a mouse colitis model. Three groups of C57BL/6 mice (prophylactic, therapeutic, and combined) were fed with 109 colony-forming units (cfu) AC5 daily for 7, 4, and 11 days, respectively. Then, the mice were challenged with STM compared to the untreated group. By using a specific primer pair, we found that AC5 can transiently colonize mouse gut (colon, cecum, and ileum). Interestingly, AC5 reduced STM gut proliferation and invasion together with attenuated gut inflammation and systemic dissemination in mice. The decreased STM numbers in mouse gut lumen, gut tissues, and spleen possibly came from longer AC5 feeding duration and/or the combinatorial (direct and indirect inhibitory) effect of AC5 on STM. However, AC5 attenuated inflammation (both in the gut and in the spleen) with no difference between these three approaches. This study demonstrated that AC5 confers both direct and indirect inhibitory effects on STM in the inflamed gut.
Collapse
Affiliation(s)
- Songphon Buddhasiri
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Chutikarn Sukjoi
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Thattawan Kaewsakhorn
- Department of Veterinary Biosciences and Veterinary Public Health, Faculty of Veterinary Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Kowit Nambunmee
- Major of Occupational Health and Safety, School of Health Science, Mae Fah Luang University, Chiang Rai, Thailand.,Urban Safety Innovation Research Group, Mae Fah Luang University, Chiang Rai, Thailand
| | - Massalin Nakphaichit
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
| | - Sunee Nitisinprasert
- Department of Biotechnology, Faculty of Agro-Industry, Kasetsart University, Bangkok, Thailand
| | - Parameth Thiennimitr
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.,Research Center of Microbial Diversity and Sustainable Utilization, Chiang Mai University, Chiang Mai, Thailand.,Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine, Chiang Mai University, Chiang Mai, Thailand
| |
Collapse
|
35
|
Lai YH, Tsai BY, Hsu CY, Chen YH, Chou PH, Chen YL, Liu HC, Ko WC, Tsai PJ, Hung YP. The Role of Toll-Like Receptor-2 in Clostridioides difficile Infection: Evidence From a Mouse Model and Clinical Patients. Front Immunol 2021; 12:691039. [PMID: 34322122 PMCID: PMC8313301 DOI: 10.3389/fimmu.2021.691039] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2021] [Accepted: 06/24/2021] [Indexed: 01/12/2023] Open
Abstract
Background Clostridioides difficile is the leading cause of nosocomial infectious diarrhea. Toll-like receptors (TLRs) are the major components of innate immunity that sense pathogens. The relationship between TLRs and C. difficile infection (CDI) was analyzed in clinical patients and a mouse model. Materials and Methods A prospective investigation was conducted in medical wards of Tainan Hospital, Ministry of Health and Welfare, Tainan, Taiwan, from January 2011 to January 2013. Adult patients were followed up for the development of CDI. Single nucleotide polymorphisms (SNPs) of TLR2 and TLR4 were analyzed to assess the relationship between genetic polymorphisms and the development of CDI. A mouse model of CDI was used to investigate the pathogenic role of TLRs in CDI, TLR2 and TLR4 knockout (Tlr2-/- and Tlr4-/-) mice. Results In the prospective study, 556 patients were enrolled, and 6.5% (36) of patients, accounting for 3.59 episodes per 1000 patient-days, developed CDI. Of 539 patients with available blood samples, the TLR2 rs3804099 polymorphism was more often noted in those with CDI than in those without CDI (64.5% vs. 46.1%; P = 0.046) but was not significant in multivariate analysis. Because the TLR2 rs3804099 polymorphism was moderately associated with CDI, the role of TLR2 and TLR4 was further evaluated in a mouse model. Both Tlr2-/- and Tlr4-/- mice showed more severe CDI disease than wild-type mice in terms of body weight change and fecal content five days after oral challenge with C. difficile. Furthermore, Tlr2-/- mice suffered from more severe disease than Tlr4-/- mice, as evidenced by stool consistency, cecum weight, and survival rate. Conclusion The TLR2 rs3804099 polymorphism is marginally associated with the development of CDI, and the pathogenic role of TLR2 is further supported by a mouse model.
Collapse
Affiliation(s)
- Yi-Hsin Lai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Bo-Yang Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Chih-Yu Hsu
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Yi-Hsuan Chen
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Po-Han Chou
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Yueh-Lin Chen
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
| | - Hsiao-Chieh Liu
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- Departments of Internal Medicine, Tainan Hospital, Ministry of Health & Welfare, Tainan, Taiwan
| | - Wen-Chien Ko
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| | - Pei-Jane Tsai
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan
- Department of Medical Laboratory Science and Biotechnology, National Cheng Kung University, Tainan, Taiwan
- Center of Infectious Disease and Signaling Research, National Cheng Kung University, Tainan City, Taiwan
- Department of Pathology, National Cheng Kung University Hospital, Tainan, Taiwan
| | - Yuan-Pin Hung
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, Taiwan
- Departments of Internal Medicine, Tainan Hospital, Ministry of Health & Welfare, Tainan, Taiwan
- Department of Medicine, College of Medicine, National Cheng Kung University, Tainan City, Taiwan
| |
Collapse
|
36
|
Tagliari E, Campos LF, Casagrande TAC, Fuchs T, de Noronha L, Campos ACL. Effects of oral probiotics administration on the expression of transforming growth factor β and the proinflammatory cytokines interleukin 6, interleukin 17, and tumor necrosis factor α in skin wounds in rats. JPEN J Parenter Enteral Nutr 2021; 46:721-729. [PMID: 34173254 DOI: 10.1002/jpen.2216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Cytokines and growth factors play key roles during the tissue repair process. We aim to evaluate the effect of perioperative oral of probiotics, on the healing process in skin wound in rats, by histological aspects, and by the expression of TGF-β, and the pro-inflammatory cytokines IL6, IL7, and TNF-α. METHODS 72 adult male Wistar rats were split into two groups control (n = 36) and probiotic group (n = 36). Each group was subdivided into three subgroups with 12 animals each according to euthanasia day: 3rd, 7th, and 10th postoperative(PO) day. RESULTS Wound contraction was faster with the use of probiotics (p = .013). Also fibrosis was significantly higher in the Probiotic group in the 7th PO day (p = .028). In the probiotic group, there was a reduction of TNF-α at 3th PO day (p = .023); and a reduction of IL6 in 7th PO day (p = .030). There was also a reduction of the expression of IL-17 in 3rd PO day (p = .039) and 7rd PO day (P = .024). In contrast, TGF-β was lower in the 10th PO day (p = .031) in the probiotic group as compared to controls, indicating that the increase of the fibrosis caused negative feedback with the TGF-β. CONCLUSION Probiotics are associated with a shorter inflammatory phase by attenuating the expression of cytokines IL-6 and TNF-α and accelerating the reduction of IL-17 and TGF-β, leading to faster and improved cutaneous healing in rats.
Collapse
Affiliation(s)
- Eliane Tagliari
- Graduate Program in Surgery, Division of Health Sciences, Federal University of Paraná, Curitiba, Brasil
| | - Letícia Fuganti Campos
- Graduate Program in Surgery, Division of Health Sciences, Federal University of Paraná, Curitiba, Brasil
| | | | - Taise Fuchs
- Professional Masters Program in Industrial Biotechnology, Positivo University
| | - Lúcia de Noronha
- Experimental Pathology Laboratory, Pontifical Catholic University of Paraná, Curitiba, Brasil
| | | |
Collapse
|
37
|
Villafuerte KRV, Martinez CJH, Nobre AVV, Maia LP, Tirapelli C. What are microbiological effects of the adjunctive use of probiotics in the treatment of periodontal diseases? A systematic review. Benef Microbes 2021; 12:1-13. [PMID: 34169804 DOI: 10.3920/bm2020.0143] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Probiotics have aroused great interest as an adjunctive treatment to periodontal therapy, due to the frequent colonisation by periodontopathogens after therapy. The aim of this systematic review was to analyse in the scientific literature, evidence of the microbiological effects of probiotics as an adjunct to periodontal therapy in the treatment of periodontal diseases (PD). Only randomised controlled trials (RCT), evaluating the microbiological effect of probiotics as an adjunct to periodontal therapy. The authors conducted a search in PubMed/MEDLINE, LILACS, ScienceDirect, Web of Science and Cochrane Library to identify articles published in English until February 2020. The quality of the studies was assessed using the JADAD scale and the risk of bias was assessed according to the Cochrane Collaboration assessment tool. Of the 265 articles potentially relevant to this review, 10 studies were included. The most frequently used probiotic bacteria were those of the genus Lactobacillus spp. and the time of administration of the probiotics was between 14 days to 3 months. Most studies have shown that the adjuvant use of probiotics reduces the total mean counts of gram-negative anaerobic species (Porphyromonas gingivalis, Tannerella forsythia, Treponema denticola and Prevotella intermedia) and gram-negative coccobacillus (Aggregatibacter actinomycetemcomitans) of subgingival plaque samples. Probiotics adjuvant to periodontal therapy reduces periodontopathogenic species in a greater proportion, compared only to periodontal therapy. Especially the Lactobacillus reuteri strain, without combination with other strains, offered a greater reduction in pathogenic bacteria associated with greater destruction of periodontal tissues and deep periodontal pockets. Researchers should perform high-quality RCT, evaluating single strains without combinations, in order to observe the microbiological benefits as adjunctive treatment of PD.
Collapse
Affiliation(s)
- K R V Villafuerte
- Craniofacial Anomalies Rehabilitation Hospital - HRAC at University of São Paulo, Bauru, SP, 17012-900, Brazil.,Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry, University of São Paulo, Avenida do Café, S/N, Ribeirão Preto, SP, 14040-904, Brazil
| | - C J H Martinez
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry, University of São Paulo, Avenida do Café, S/N, Ribeirão Preto, SP, 14040-904, Brazil
| | - A V V Nobre
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry, University of São Paulo, Avenida do Café, S/N, Ribeirão Preto, SP, 14040-904, Brazil
| | - L P Maia
- Graduate Program in Dentistry, University of Western São Paulo - UNOESTE, Presidente Prudente, SP, 19050-680, Brazil
| | - C Tirapelli
- Integrated Dental Clinic, Department of Dental Materials and Prosthodontics, School of Dentistry, University of São Paulo, Avenida do Café, S/N, Ribeirão Preto, SP, 14040-904, Brazil
| |
Collapse
|
38
|
Simon E, Călinoiu LF, Mitrea L, Vodnar DC. Probiotics, Prebiotics, and Synbiotics: Implications and Beneficial Effects against Irritable Bowel Syndrome. Nutrients 2021; 13:nu13062112. [PMID: 34203002 PMCID: PMC8233736 DOI: 10.3390/nu13062112] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 06/05/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022] Open
Abstract
Irritable bowel syndrome (IBS) is still a common functional gastrointestinal disease that presents chronic abdominal symptoms but with a pathophysiology that is not yet fully elucidated. Moreover, the use of the synergistic combination of prebiotics and probiotics, known as synbiotics, for IBS therapy is still in the early stages. Advancements in technology led to determining the important role played by probiotics in IBS, whereas the present paper focuses on the detailed review of the various pathophysiologic mechanisms of action of probiotics, prebiotics, and synbiotics via multidisciplinary domains involving the gastroenterology (microbiota modulation, alteration of gut barrier function, visceral hypersensitivity, and gastrointestinal dysmotility) immunology (intestinal immunological modulation), and neurology (microbiota–gut–brain axis communication and co-morbidities) in mitigating the symptoms of IBS. In addition, this review synthesizes literature about the mechanisms involved in the beneficial effects of prebiotics and synbiotics for patients with IBS, discussing clinical studies testing the efficiency and outcomes of synbiotics used as therapy for IBS.
Collapse
Affiliation(s)
- Elemer Simon
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania; (E.S.); (L.F.C.)
| | - Lavinia Florina Călinoiu
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania; (E.S.); (L.F.C.)
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania;
| | - Laura Mitrea
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania;
| | - Dan Cristian Vodnar
- Faculty of Food Science and Technology, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania; (E.S.); (L.F.C.)
- Institute of Life Sciences, University of Agricultural Sciences and Veterinary Medicine, Calea Mănăştur 3–5, 400372 Cluj-Napoca, Romania;
- Correspondence: ; Tel.: +40-747-341-881
| |
Collapse
|
39
|
Milk Fermented by Lactobacillus paracasei NCC 2461 (ST11) Modulates the Immune Response and Microbiota to Exert its Protective Effects Against Salmonella typhimurium Infection in Mice. Probiotics Antimicrob Proteins 2021; 12:1398-1408. [PMID: 31970649 DOI: 10.1007/s12602-020-09634-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Probiotics form a promising strategy to maintain intestinal health. Milks fermented with probiotic strains, such as the Lactobacillus paracasei ST11, are largely commercialized in Brazil and form a low-cost alternative to probiotic pharmaceutical formulations. In this study, we assessed the probiotic effects of milk fermented by L. paracasei ST11 (administered through fermented milk) in a Salmonella typhimurium infection model in BALB/c mice. We observed in this murine model that the applied probiotic conferred protective effects against S. typhimurium infection, since its administration reduced mortality, weight loss, translocation to target organs (liver and spleen) and ileum injury. Moreover, a reduction in the mRNA expression of pro-inflammatory cytokines such as IFN-γ, IL-6, TNF-α and IL-17 in animals that received the probiotic before challenge was observed. Additionally, the ileum microbiota was better preserved in these animals. The present study highlights a multifactorial protective aspect of this commercial probiotic strain against a common gastrointestinal pathogen.
Collapse
|
40
|
Müller L, Kuhn T, Koch M, Fuhrmann G. Stimulation of Probiotic Bacteria Induces Release of Membrane Vesicles with Augmented Anti-inflammatory Activity. ACS APPLIED BIO MATERIALS 2021; 4:3739-3748. [PMID: 35006804 DOI: 10.1021/acsabm.0c01136] [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] [Indexed: 12/20/2022]
Abstract
During infection, inflammation is an important contributor to tissue regeneration and healing, but it may also negatively affect these processes should chronic overstimulation take place. Similar issues arise in chronic inflammatory gastrointestinal diseases such as inflammatory bowel diseases or celiac disease, which show increasing incidences worldwide. For these dispositions, probiotic microorganisms, including lactobacilli, are studied as an adjuvant therapy to counterbalance gut dysbiosis. However, not all who are affected can benefit from the probiotic treatment, as immunosuppressed or hospitalized patients can suffer from bacteremia or sepsis when living microorganisms are administered. A promising alternative is the treatment with bacteria-derived membrane vesicles that confer similar beneficial effects as the progenitor strains themselves. Membrane vesicles from lactobacilli have shown anti-inflammatory therapeutic effects, but it remains unclear whether the stimulation of probiotics induces vesicles that are more efficient. Here, the influence of culture conditions on the anti-inflammatory characteristics of Lactobacillus membrane vesicles was investigated. We reveal that the culture conditions of two Lactobacillus strains, namely, L. casei and L. plantarum, can be optimized to increase the anti-inflammatory effect of their vesicles. Five different cultivation conditions were tested, including pH manipulation, agitation rate, and oxygen supply, and the produced membrane vesicles were characterized physico-chemically regarding size, yield, and zeta potential. We furthermore analyzed the anti-inflammatory effect of the purified vesicles in macrophage inflammation models. Compared to standard cultivation conditions, vesicles obtained from L. casei cultured at pH 6.5 and agitation induced the strongest interleukin-10 release and tumor necrosis factor-α reduction. For L. plantarum, medium adjusted to pH 5 had the most pronounced effect on the anti-inflammatory activity of their vesicles. Our results reveal that the anti-inflammatory effect of probiotic vesicles may be potentiated by expanding different cultivation conditions for lactobacilli. This study creates an important base for the utilization of probiotic membrane vesicles to treat inflammation.
Collapse
Affiliation(s)
- Lisann Müller
- Helmholtz Institute for Pharmaceutical Research Saarland, Department of Pharmacy, Saarland University, Campus E8 1, 66123 Saarbrücken, Germany
| | - Thomas Kuhn
- Helmholtz Institute for Pharmaceutical Research Saarland, Department of Pharmacy, Saarland University, Campus E8 1, 66123 Saarbrücken, Germany
| | - Marcus Koch
- INM - Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany
| | - Gregor Fuhrmann
- Helmholtz Institute for Pharmaceutical Research Saarland, Department of Pharmacy, Saarland University, Campus E8 1, 66123 Saarbrücken, Germany
| |
Collapse
|
41
|
Lemme-Dumit JM, Cazorla SI, Perdigón GDV, Maldonado-Galdeano C. Probiotic Bacteria and Their Cell Walls Induce Th1-Type Immunity Against Salmonella Typhimurium Challenge. Front Immunol 2021; 12:660854. [PMID: 34054825 PMCID: PMC8149796 DOI: 10.3389/fimmu.2021.660854] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Accepted: 04/27/2021] [Indexed: 01/13/2023] Open
Abstract
Probiotics have been associated with a variety of health benefits. They can act as adjuvant to enhance specific immune response. Bacterial cell wall (CW) molecules are key structures that interact with host receptors promoting probiotic effects. The adjuvant capacity underlying this sub-cellular fraction purified from Lactobacillus casei CRL431 and L. paracasei CNCMI-1518 remains to be characterized. We interrogated the molecular and cellular events after oral feeding with probiotic-derived CW in addition to heat-inactivated Salmonella Typhimurium and their subsequent protective capacity against S. Typhimurium challenge. Intact probiotic bacteria were orally administered for comparison. We find that previous oral feeding with probiotics or their sub-cellular fraction reduce bacterial burden in spleen and liver after Salmonella challenge. Antibody responses after pathogen challenge were negligible, characterized by not major changes in the antibody-mediated phagocytic activity, and in the levels of total and Salmonella-specific intestinal sIgA and serum IgG, respectively. Conversely, the beneficial effect of probiotic-derived CW after S. Typhimurium challenge were ascribed to a Th1-type cell-mediated immunity which was characterized by augmentation of the delayed-type hypersensitivity response. The cell-mediated immunity associated with the oral feeding with probiotic-derived CW was accompanied with a Th1-cell polarizing cytokines, distinguished by increase IFN-γ/IL-4 ratio. Similar results were observed with the intact probiotics. Our study identified molecular events associated with the oral administration of sub-cellular structures derived from probiotics and their adjuvant capacity to exert immune modulatory function.
Collapse
Affiliation(s)
- José María Lemme-Dumit
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina.,Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán, Argentina
| | - Silvia Inés Cazorla
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina.,Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán, Argentina
| | - Gabriela Del Valle Perdigón
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina
| | - Carolina Maldonado-Galdeano
- Laboratorio de Inmunología, Centro de Referencia para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Argentina.,Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, San Miguel de Tucumán, Argentina
| |
Collapse
|
42
|
Methiwala HN, Vaidya B, Addanki VK, Bishnoi M, Sharma SS, Kondepudi KK. Gut microbiota in mental health and depression: role of pre/pro/synbiotics in their modulation. Food Funct 2021; 12:4284-4314. [PMID: 33955443 DOI: 10.1039/d0fo02855j] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The microbiome residing in the human gut performs a wide range of biological functions. Recently, it has been elucidated that a change in dietary habits is associated with alteration in the gut microflora which results in increased health risks and vulnerability towards various diseases. Falling in line with the same concept, depression has also been shown to increase its prevalence around the globe, especially in the western world. Various research studies have suggested that changes in the gut microbiome profile further result in decreased tolerance of stress. Although currently available medications help in relieving the symptoms of depressive disorders briefly, these drugs are not able to completely reverse the multifactorial pathology of depression. The discovery of the communication pathway between gut microbes and the brain, i.e. the Gut-Brain Axis, has led to new areas of research to find more effective and safer alternatives to current antidepressants. The use of probiotics and prebiotics has been suggested as being effective in various preclinical studies and clinical trials for depression. Therefore, in the present review, we address the new antidepressant mechanisms via gut microbe alterations and provide insight into how these can provide an alternative to antidepressant therapy without the side effects and risk of adverse drug reactions.
Collapse
Affiliation(s)
- Hasnain N Methiwala
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Punjab, India.
| | | | | | | | | | | |
Collapse
|
43
|
In vivo evidence: Repression of mucosal immune responses in mice with colon cancer following sustained administration of Streptococcus thermophiles. Saudi J Biol Sci 2021; 28:4751-4761. [PMID: 34354463 PMCID: PMC8324971 DOI: 10.1016/j.sjbs.2021.04.090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Revised: 04/27/2021] [Accepted: 04/28/2021] [Indexed: 12/20/2022] Open
Abstract
Probiotics have attracted considerable attention because of their ability to ameliorate disease and prevent cancer. In this study, we examined the immunomodulatory effects of a Streptococcus thermophilus probiotic on the intestinal mucosa azoxymethane-induced colon cancer. Sixty female mice were divided into four groups (n = 15 each). One group of untreated mice was used as a control (C group). Another mouse group was injected with azoxymethane once weekly for 8 weeks to induce colon cancer (CC group). Finally, two groups of mice were continuously treated twice per week from week 2 to 16 with either the Lactobacillus plantarum (Lac CC group) or S. thermophilus (Strep CC group) bacterial strain pre-and post-treatment as performed for the CC group. Remarkably, Tlr2, Ifng, Il4, Il13, Il10, and Tp53 transcription were significantly downregulated in the Strep CC intestinal mucosa group. Additionally, IL2 expression was decreased significantly in the Strep CC mouse serum, whereas TNFα was remarkably elevated compared to that in the CC, Lac CC, and untreated groups. This study suggested that Streptococcus thermophilus did not interrupt or hinder colon cancer development in mice when administered as a prophylactic.
Collapse
|
44
|
Raheem A, Liang L, Zhang G, Cui S. Modulatory Effects of Probiotics During Pathogenic Infections With Emphasis on Immune Regulation. Front Immunol 2021; 12:616713. [PMID: 33897683 PMCID: PMC8060567 DOI: 10.3389/fimmu.2021.616713] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Accepted: 02/12/2021] [Indexed: 12/11/2022] Open
Abstract
In order to inhibit pathogenic complications and to enhance animal and poultry growth, antibiotics have been extensively used for many years. Antibiotics applications not only affect target pathogens but also intestinal beneficially microbes, inducing long-lasting changes in intestinal microbiota associated with diseases. The application of antibiotics also has many other side effects like, intestinal barrier dysfunction, antibiotics residues in foodstuffs, nephropathy, allergy, bone marrow toxicity, mutagenicity, reproductive disorders, hepatotoxicity carcinogenicity, and antibiotic-resistant bacteria, which greatly compromise the efficacy of antibiotics. Thus, the development of new antibiotics is necessary, while the search for antibiotic alternatives continues. Probiotics are considered the ideal antibiotic substitute; in recent years, probiotic research concerning their application during pathogenic infections in humans, aquaculture, poultry, and livestock industry, with emphasis on modulating the immune system of the host, has been attracting considerable interest. Hence, the adverse effects of antibiotics and remedial effects of probiotics during infectious diseases have become central points of focus among researchers. Probiotics are live microorganisms, and when given in adequate quantities, confer good health effects to the host through different mechanisms. Among them, the regulation of host immune response during pathogenic infections is one of the most important mechanisms. A number of studies have investigated different aspects of probiotics. In this review, we mainly summarize recent discoveries and discuss two important aspects: (1) the application of probiotics during pathogenic infections; and (2) their modulatory effects on the immune response of the host during infectious and non-infectious diseases.
Collapse
Affiliation(s)
- Abdul Raheem
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Lin Liang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Guangzhi Zhang
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| | - Shangjin Cui
- Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Scientific Observation and Experiment Station of Veterinary Drugs and Diagnostic Technology of Beijing, Ministry of Agriculture, Beijing, China
| |
Collapse
|
45
|
Chen F, Chen J, Chen Q, Yang L, Yin J, Li Y, Huang X. Lactobacillus delbrueckii Protected Intestinal Integrity, Alleviated Intestinal Oxidative Damage, and Activated Toll-Like Receptor-Bruton's Tyrosine Kinase-Nuclear Factor Erythroid 2-Related Factor 2 Pathway in Weaned Piglets Challenged with Lipopolysaccharide. Antioxidants (Basel) 2021; 10:antiox10030468. [PMID: 33809627 PMCID: PMC8002333 DOI: 10.3390/antiox10030468] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 03/10/2021] [Accepted: 03/13/2021] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress is increasingly being recognized as a player in the pathogenesis of intestinal pathologies, and probiotics are becoming an attractive means of addressing it. The present study investigated the effects of dietary supplementation with Lactobacillus delbrueckii (LAB) on intestinal integrity and oxidative damage in lipopolysaccharide (LPS)-challenged piglets. A total of 36 crossbred weaned piglets (Duroc × Landrace × Large Yorkshire) were randomly divided into three groups: (1) non-challenged controls (CON), (2) LPS-challenged controls (LPS), and (3) 0.2% LAB (2.01 × 1010 CFU/g) + LPS treatment (LAB + LPS). On the 29th day of the experiment, the LPS and CON groups were injected intraperitoneally with LPS and saline at 100 ug/kg body weight, respectively. The results show that the LPS-induced elevation of the serum diamine oxidase (DAO) level and small intestinal crypt depth (CD) were reversed by the dietary addition of LAB, which also markedly increased the ileal expression of tight junction proteins (occludin, ZO-1, and claudin-1) in the LPS-challenged piglets. Furthermore, LAB supplementation normalized other LPS-induced changes, such as by decreasing malondialdehyde (MDA) in both the serum and intestinal mucosa and 8-hydroxy-2-deoxyguanosine (8-OHdG) in the jejunal mucosa, increasing glutathione reductase (GR) and glutathione peroxidase (GSH-Px) in both the serum and intestinal mucosa, and increasing glutathione (GSH) and superoxide dismutase (SOD) in the jejunal mucosa. LAB also activated Toll-like receptor (TLR)–Bruton’s tyrosine kinase (Btk)–nuclear factor erythroid 2-related factor 2(Nrf2) signaling pathways in the intestine, suggesting that it plays a vital role in the ameliorative antioxidant capacity of weaned piglets. In summary, LAB increased intestinal integrity by improving the intestinal structure and tight junctions while enhancing antioxidant functions via the activation of the TLR–Btk–Nrf2 signaling pathway.
Collapse
Affiliation(s)
- Fengming Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (F.C.); (J.C.); (Q.C.); (L.Y.); (J.Y.)
- Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha 410128, China
- The Engineering Research Center of Feed Safety and Efficient Utilization, Education Ministry, Hunan Agriculture University, Changsha 410128, China
| | - Jiayi Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (F.C.); (J.C.); (Q.C.); (L.Y.); (J.Y.)
- Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha 410128, China
- The Engineering Research Center of Feed Safety and Efficient Utilization, Education Ministry, Hunan Agriculture University, Changsha 410128, China
| | - Qinghua Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (F.C.); (J.C.); (Q.C.); (L.Y.); (J.Y.)
- Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha 410128, China
- The Engineering Research Center of Feed Safety and Efficient Utilization, Education Ministry, Hunan Agriculture University, Changsha 410128, China
| | - Lingyuan Yang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (F.C.); (J.C.); (Q.C.); (L.Y.); (J.Y.)
- Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha 410128, China
- The Engineering Research Center of Feed Safety and Efficient Utilization, Education Ministry, Hunan Agriculture University, Changsha 410128, China
| | - Jie Yin
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (F.C.); (J.C.); (Q.C.); (L.Y.); (J.Y.)
- Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha 410128, China
- The Engineering Research Center of Feed Safety and Efficient Utilization, Education Ministry, Hunan Agriculture University, Changsha 410128, China
| | - Yinghui Li
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (F.C.); (J.C.); (Q.C.); (L.Y.); (J.Y.)
- Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha 410128, China
- The Engineering Research Center of Feed Safety and Efficient Utilization, Education Ministry, Hunan Agriculture University, Changsha 410128, China
- Correspondence: (Y.L.); (X.H.)
| | - Xingguo Huang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China; (F.C.); (J.C.); (Q.C.); (L.Y.); (J.Y.)
- Hunan Co-Innovation Center of Animal Production Safety, CICAPS, Changsha 410128, China
- The Engineering Research Center of Feed Safety and Efficient Utilization, Education Ministry, Hunan Agriculture University, Changsha 410128, China
- Correspondence: (Y.L.); (X.H.)
| |
Collapse
|
46
|
Monteros MJM, Galdeano CM, Balcells MF, Weill R, De Paula JA, Perdigón G, Cazorla SI. Probiotic lactobacilli as a promising strategy to ameliorate disorders associated with intestinal inflammation induced by a non-steroidal anti-inflammatory drug. Sci Rep 2021; 11:571. [PMID: 33436961 PMCID: PMC7803994 DOI: 10.1038/s41598-020-80482-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 12/09/2020] [Indexed: 02/07/2023] Open
Abstract
Damage to the small intestine caused by non-steroidal anti-inflammatory drugs (NSAIDs) occurs more frequently than in the upper gastrointestinal tract, is more difficult to diagnose and no effective treatments exist. Hence, we investigated whether probiotics can control the onset of this severe condition in a murine model of intestinal inflammation induced by the NSAID, indomethacin. Probiotic supplementation to mice reduce the body weight loss, anemia, shortening of the small intestine, cell infiltration into the intestinal tissue and the loss of Paneth and Goblet cells associated with intestinal inflammation. Furthermore, a high antimicrobial activity in the intestinal fluids of mice fed with probiotics compared to animals on a conventional diet was elicited against several pathogens. Interestingly, probiotics dampened the oxidative stress and several local and systemic markers of an inflammatory process, as well as increased the secretion of IL-10 by regulatory T cells. Even more importantly, probiotics induced important changes in the large intestine microbiota characterized by an increase in anaerobes and lactobacilli, and a significant decrease in total enterobacteria. We conclude that oral probiotic supplementation in NSAID-induced inflammation increases intestinal antimicrobial activity and reinforces the intestinal epithelial barrier in order to avoid pathogens and commensal invasion and maintain intestinal homeostasis.
Collapse
Affiliation(s)
- María José Martínez Monteros
- Laboratorio de Inmunología, Centro de Referencia Para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Chacabuco 145 - (T4000ILC), Tucumán, Argentina
| | - Carolina Maldonado Galdeano
- Laboratorio de Inmunología, Centro de Referencia Para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Chacabuco 145 - (T4000ILC), Tucumán, Argentina
- Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina
| | - María Florencia Balcells
- Laboratorio de Inmunología, Centro de Referencia Para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Chacabuco 145 - (T4000ILC), Tucumán, Argentina
| | | | | | - Gabriela Perdigón
- Laboratorio de Inmunología, Centro de Referencia Para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Chacabuco 145 - (T4000ILC), Tucumán, Argentina
| | - Silvia Inés Cazorla
- Laboratorio de Inmunología, Centro de Referencia Para Lactobacilos (CERELA-CONICET), San Miguel de Tucumán, Chacabuco 145 - (T4000ILC), Tucumán, Argentina.
- Cátedra de Inmunología, Facultad de Bioquímica, Química y Farmacia, Universidad Nacional de Tucumán, Tucumán, Argentina.
| |
Collapse
|
47
|
Alqazlan N, Astill J, Taha-Abdelaziz K, Nagy É, Bridle B, Sharif S. Probiotic Lactobacilli Enhance Immunogenicity of an Inactivated H9N2 Influenza Virus Vaccine in Chickens. Viral Immunol 2020; 34:86-95. [PMID: 33236974 DOI: 10.1089/vim.2020.0209] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Avian influenza viruses (AIVs) infect a wide range of hosts, including humans and many avian species. Efforts have been made to control this pathogen in chickens using vaccination programs, but that has been met with varying degrees of success. Therefore, identification of more efficacious vaccination strategies is warranted. This study was undertaken to investigate the potential effects of probiotics on the immunogenicity of a beta-propiolactone-whole inactivated virus (WIV) vaccine of H9N2 subtype adjuvanted with the Toll-like receptor-21 ligand, CpG oligodeoxynucleotides 2007 (CpG). Eighty-four 1-day-old White Leghorn layers were allocated into six groups. Two out of six groups received a mixture of probiotic Lactobacillus spp. (PROB) biweekly from days 1 35 of age. Chickens were intramuscularly vaccinated with WIV either alone or adjuvanted with AddaVax™ (WIV+Add) or CpG (WIV+CpG), and one group received saline (phosphate-buffered saline). Primary and secondary vaccinations occurred at days 14 and 28 of age, respectively. The results revealed that the group that received probiotics and was vaccinated with CpG-adjuvanted WIV H9N2 vaccine had higher hemagglutination inhibition titers than the other treatment groups at days 14 and 21 postprimary vaccination. Probiotics did not induce higher IgM or IgY titers in chickens receiving the WIV vaccine only. Concerning their effect on cell-mediated immune responses, probiotics enhanced interferon-gamma (IFN-γ) gene expression and significantly increased secretion of IFN-γ protein by splenocytes in chickens vaccinated with CpG-adjuvanted WIV H9N2. Together, these findings suggest the use of probiotics to enhance the immunogenicity of CpG-adjuvanted WIV H9N2 vaccines. Additional studies are required to better understand the specific interactions between probiotics and the gut microbiota and different types of cells of the gastrointestinal tract to decipher the underlying mechanisms of how probiotics modulate immune responses to vaccines.
Collapse
Affiliation(s)
- Nadiyah Alqazlan
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Jake Astill
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Khaled Taha-Abdelaziz
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada.,Pathology Department, Faculty of Veterinary Medicine, Beni-Suef University, Beni-Suef, Egypt
| | - Éva Nagy
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Byram Bridle
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| | - Shayan Sharif
- Department of Pathobiology, Ontario Veterinary College, University of Guelph, Guelph, Ontario, Canada
| |
Collapse
|
48
|
Invernici MM, Furlaneto FAC, Salvador SL, Ouwehand AC, Salminen S, Mantziari A, Vinderola G, Ervolino E, Santana SI, Silva PHF, Messora MR. Bifidobacterium animalis subsp lactis HN019 presents antimicrobial potential against periodontopathogens and modulates the immunological response of oral mucosa in periodontitis patients. PLoS One 2020; 15:e0238425. [PMID: 32960889 PMCID: PMC7508403 DOI: 10.1371/journal.pone.0238425] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2020] [Accepted: 08/14/2020] [Indexed: 02/07/2023] Open
Abstract
Objective To evaluate the effects of Bifidobacterium animalis subsp. lactis HN019 (HN019) on clinical periodontal parameters (plaque accumulation and gingival bleeding), on immunocompetence of gingival tissues [expression of beta-defensin (BD)-3, toll-like receptor 4 (TLR4), cluster of differentiation(CD)-57 and CD-4], and on immunological properties of saliva (IgA levels) in non-surgical periodontal therapy in generalized chronic periodontitis (GCP) patients. Adhesion to buccal epithelial cells (BEC) and the antimicrobial properties of HN019 were also investigated. Materials and methods Thirty patients were recruited and monitored clinically at baseline (before scaling and root planing—SRP) and after 30 and 90 days. Patients were randomly assigned to Test (SRP+Probiotic, n = 15) or Control (SRP+Placebo, n = 15) group. Probiotic lozenges were used for 30 days. Gingival tissues and saliva were immunologically analyzed. The adhesion of HN019 with or without Porphyromonas gingivalis in BEC and its antimicrobial properties were investigated in in vitro assays. Data were statistically analyzed (p<0.05). Results Test group presented lower plaque index (30 days) and lower marginal gingival bleeding (90 days) when compared with Control group. Higher BD-3, TLR4 and CD-4 expressions were observed in gingival tissues in Test group than in Control group. HN019 reduced the adhesion of P. gingivalis to BEC and showed antimicrobial potential against periodontopathogens. Conclusion Immunological and antimicrobial properties of B. lactis HN019 make it a potential probiotic to be used in non-surgical periodontal therapy of patients with GCP. Clinical relevance B. lactis HN019 may be a potential probiotic to improve the effects of non-surgical periodontal therapy. Name of the registry and registration number (ClinicalTrials.gov): “Effects of probiotic therapy in the treatment of periodontitis”—NCT03408548.
Collapse
Affiliation(s)
- Marcos M. Invernici
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo – USP, Ribeirão Preto, São Paulo, Brazil
| | - Flávia A. C. Furlaneto
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo – USP, Ribeirão Preto, São Paulo, Brazil
- * E-mail:
| | - Sérgio L. Salvador
- Department of Clinical Analyses, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo – USP, Ribeirão Preto, São Paulo, Brazil
| | | | - Seppo Salminen
- Functional Foods Forum, University of Turku, Turku, Finland
| | | | - Gabriel Vinderola
- Instituto de Lactología Industrial (UNL-CONICET), National University of the Litoral, Santa Fe, Argentina
| | - Edilson Ervolino
- Division of Histology, Department of Basic Sciences, Dental School of Araçatuba, São Paulo State University, São Paulo, Brazil
| | - Sandro Isaías Santana
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo – USP, Ribeirão Preto, São Paulo, Brazil
| | - Pedro Henrique Felix Silva
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo – USP, Ribeirão Preto, São Paulo, Brazil
| | - Michel R. Messora
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirão Preto, University of São Paulo – USP, Ribeirão Preto, São Paulo, Brazil
| |
Collapse
|
49
|
Lactobacillus johnsonii L531 Ameliorates Escherichia coli-Induced Cell Damage via Inhibiting NLRP3 Inflammasome Activity and Promoting ATG5/ATG16L1-Mediated Autophagy in Porcine Mammary Epithelial Cells. Vet Sci 2020; 7:vetsci7030112. [PMID: 32823867 PMCID: PMC7558184 DOI: 10.3390/vetsci7030112] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/06/2020] [Accepted: 08/12/2020] [Indexed: 12/31/2022] Open
Abstract
Escherichia coli (E. coli), a main mastitis-causing pathogen in sows, leads to mammary tissue damage. Here, we explored the effects of Lactobacillus johnsonii L531 on attenuating E. coli-induced inflammatory damage in porcine mammary epithelial cells (PMECs). L. johnsonii L531 pretreatment reduced E. coli adhesion to PMECs by competitive exclusion and the production of inhibitory factors and decreased E. coli-induced destruction of cellular morphology and ultrastructure. E. coli induced activation of NLRP3 inflammasome associated with increased expression of NLRP3, ASC, and cleaved caspase-1, however, L. johnsonii L531 inhibited E. coli-induced activation of NLRP3 inflammasome. Up-regulation of interleukin (Il)-1β, Il-6, Il-8, Il-18, tumor necrosis factor alpha, and chemokine Cxcl2 expression after E. coli infection was attenuated by L. johnsonii L531. E. coli infection inhibited autophagy, whereas L. johnsonii L531 reversed the inhibitory effect of E. coli on autophagy by decreasing the expression of autophagic receptor SQSTM1/p62 and increasing the expression of autophagy-related proteins ATG5, ATG16L1, and light chain 3 protein by Western blotting analysis. Our findings suggest that L. johnsonii L531 pretreatment restricts NLRP3 inflammasome activity and induces autophagy through promoting ATG5/ATG16L1-mediated autophagy, thereby protecting against E. coli-induced inflammation and cell damage in PMECs.
Collapse
|
50
|
Maternal Supplementation of Food Ingredient (Prebiotic) or Food Contaminant (Mycotoxin) Influences Mucosal Immune System in Piglets. Nutrients 2020; 12:nu12072115. [PMID: 32708852 PMCID: PMC7400953 DOI: 10.3390/nu12072115] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 07/08/2020] [Accepted: 07/15/2020] [Indexed: 12/29/2022] Open
Abstract
The early life period is crucial for the maturation of the intestinal barrier, its immune system, and a life-long beneficial host-microbiota interaction. The study aims to assess the impact of a beneficial dietary (short-chain fructooligosaccharides, scFOS) supplementation vs. a detrimental dietary environment (such as mycotoxin deoxynivalenol, DON) on offspring intestinal immune system developmental profiles. Sows were given scFOS-supplemented or DON-contaminated diets during the last 4 weeks of gestation, whereas force-feeding piglets with DON was performed during the first week of offspring life. Intestinal antigen-presenting cell (APC) subset frequency was analyzed by flow cytometry in the Peyer's patches and in lamina propria and the responsiveness of intestinal explants to toll-like receptor (TLR) ligands was performed using ELISA and qRT-PCR from post-natal day (PND) 10 until PND90. Perinatal exposure with scFOS did not affect the ontogenesis of APC. While it early induced inflammatory responses in piglets, scFOS further promoted the T regulatory response after TLR activation. Sow and piglet DON contamination decreased CD16+ MHCII+ APC at PND10 in lamina propria associated with IFNγ inflammation and impairment of Treg response. Our study demonstrated that maternal prebiotic supplementation and mycotoxin contamination can modulate the mucosal immune system responsiveness of offspring through different pathways.
Collapse
|