1
|
Sadeghloo Z, Nabavi-Rad A, Zali MR, Klionsky DJ, Yadegar A. The interplay between probiotics and host autophagy: mechanisms of action and emerging insights. Autophagy 2024:1-23. [PMID: 39291740 DOI: 10.1080/15548627.2024.2403277] [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: 05/07/2024] [Revised: 07/09/2024] [Accepted: 07/10/2024] [Indexed: 09/19/2024] Open
Abstract
Autophagy, a lysosome-dependent protein degradation mechanism, is a highly conserved catabolic process seen in all eukaryotes. This cell protection system, which is present in all tissues and functions at a basic level, can be up- or downregulated in response to various stresses. A disruption in the natural route of the autophagy process is frequently followed by an interruption in the inherent operation of the body's cells and organs. Probiotics are live bacteria that protect the host through various mechanisms. One of the processes through which probiotics exert their beneficial effects on various cells and tissues is autophagy. Autophagy can assist in maintaining host homeostasis by stimulating the immune system and affecting numerous physiological and pathological responses. In this review, we particularly focus on autophagy impairments occurring in several human illnesses and investigate how probiotics affect the autophagy process under various circumstances.Abbreviation: AD: Alzheimer disease; AKT: AKT serine/threonine kinase; AMPK: 5'AMP-activated protein kinase; ATG: autophagy related; CCl4: carbon tetrachloride; CFS: cell-free supernatant; CMA: chaperone-mediated autophagy; CRC: colorectal cancer; EPS: L. plantarum H31 exopolysaccharide; HD: Huntington disease; HFD: high-fat diet; HPV: human papillomavirus; IFNG/IFN-γ: interferon gamma; IL6: interleukin 6; LGG: L. rhamnosus GG; LPS: lipopolysaccharide; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; NAFLD: non-alcoholic fatty liver disease; NASH: non-alcoholic steatohepatitis; PD: Parkinson disease; Pg3G: pelargonidin-3-O-glucoside; PI3K: phosphoinositide 3-kinase; PolyQ: polyglutamine; ROS: reactive oxygen species; SCFAs: short-chain fatty acids; SLAB51: a novel formulation of lactic acid bacteria and bifidobacteria; Slp: surface layer protein (of acidophilus NCFM); SNCA: synuclein alpha; ULK1: unc-51 like autophagy-activating kinase 1; YB: B. longum subsp. infantis YB0411; YFP: yeast fermentate prebiotic.
Collapse
Affiliation(s)
- Zahra Sadeghloo
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Nabavi-Rad
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Zali
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, USA
- Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Abbas Yadegar
- Foodborne and Waterborne Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Borah M, Mandal M, Konwar BK. Characterization of probiotic strains of Bacillus sp. from fermented palm wine (Nypa fructicans sp.) and exploration of cellulolytic potential for use as an addition in animal feed. Int Microbiol 2024:10.1007/s10123-024-00589-5. [PMID: 39289261 DOI: 10.1007/s10123-024-00589-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 08/26/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024]
Abstract
The main objective of this study was to assess cellulolytic probiotic strains from traditional fermented beverages such as palm wine in order to supplement the animal feed and strengthen the gut health of the animal for better digestibility and absorption. In the present study, different types of microbes were isolated from traditionally prepared palm wine and analyzed for their probiotic nature. For any microbe to be probiotic in nature, it has to sustain the harsh conditions of the human gastrointestinal tract such as acid tolerance, bile tolerance at the lower range of pH, and other properties like auto aggregation test, cell surface hydrophobicity test with non-polar hydrocarbons for evaluating its capabilities to adhere to the intestinal cells and antimicrobial nature against pathogens. Bacillus mycoides strain PR04 and Bacillus subtilis strain PR21 were found to be resistant to acid and bile in simulated artificial gastrointestinal tract model, found to be than 55% hydrophobic with xylene and n-hexadecane and also showed antimicrobial activity greater towards pathogenic strains like Pseudomonas aeruginosa, Staphylococcus aureus, Candida albicans, and Salmonella typhimurium respectively. The cellulolytic activity of the isolates PR04 and PR21 was evaluated in (0.2-2) % CMC (carboxymethyl cellulose) plate. Bacillus mycoides PR04 and Bacillus subtilis PR21 could degrade carboxymethyl cellulose, filter paper, and sugarcane bagasse. The degradation of sugarcane bagasse was confirmed by Scanning electron microscopy and filter paper degradation after 4 days of incubation at 37 °C. Cellulase gene of the identified Bacillus sp. strains was amplified by primers CF5'-ACAGGATCCGATGAAAACGGTCAATTTCTATTTT-3' and CR5'-ACTCTCGAGATTGGGTTCTGTTCCCAAT-3'. This study proposes potential probiotic Bacillus mycoides PR04 (Accession no. OR625070) and Bacillus subtilis PR21 (Accession no. OR625072) in the application as an animal feed additive to assist in its digestibility and encourage the gut health.
Collapse
Affiliation(s)
- Munmi Borah
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur-784028, Assam, India.
| | - Manabendra Mandal
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur-784028, Assam, India
| | - Bolin K Konwar
- Department of Molecular Biology and Biotechnology, Tezpur University, Napaam, Tezpur-784028, Assam, India
| |
Collapse
|
3
|
Perna A, Venditti N, Merolla F, Fusco S, Guerra G, Zoroddu S, De Luca A, Bagella L. Nutraceuticals in Pregnancy: A Special Focus on Probiotics. Int J Mol Sci 2024; 25:9688. [PMID: 39273635 PMCID: PMC11395456 DOI: 10.3390/ijms25179688] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Revised: 08/30/2024] [Accepted: 09/04/2024] [Indexed: 09/15/2024] Open
Abstract
The placenta is crucial to fetal development and performs vital functions such as nutrient exchange, waste removal and hormone regulation. Abnormal placental development can lead to conditions such as fetal growth restriction, pre-eclampsia and stillbirth, affecting both immediate and long-term fetal health. Placental development is a highly complex process involving interactions between maternal and fetal components, imprinted genes, signaling pathways, mitochondria, fetal sexomes and environmental factors such as diet, supplementation and exercise. Probiotics have been shown to make a significant contribution to prenatal health, placental health and fetal development, with associations with reduced risk of preterm birth and pre-eclampsia, as well as improvements in maternal health through effects on gut microbiota, lipid metabolism, vaginal infections, gestational diabetes, allergic diseases and inflammation. This review summarizes key studies on the influence of dietary supplementation on placental development, with a focus on the role of probiotics in prenatal health and fetal development.
Collapse
Affiliation(s)
- Angelica Perna
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Via F. De Sanctis, 86100 Campobasso, Italy
| | - Noemi Venditti
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Via F. De Sanctis, 86100 Campobasso, Italy
- UO Laboratory Analysis, Responsible Research Hospital, Largo Agostino Gemelli, 1, 86100 Campobasso, Italy
| | - Francesco Merolla
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Via F. De Sanctis, 86100 Campobasso, Italy
| | - Sabato Fusco
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Via F. De Sanctis, 86100 Campobasso, Italy
| | - Germano Guerra
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Via F. De Sanctis, 86100 Campobasso, Italy
| | - Stefano Zoroddu
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari, Italy
| | - Antonio De Luca
- Department of Mental and Physical Health and Preventive Medicine, Section of Human Anatomy, University of Campania "Luigi Vanvitelli", Via Costantinopoli 16, 80138 Naples, Italy
| | - Luigi Bagella
- Department of Biomedical Sciences, University of Sassari, Viale San Pietro 43/b, 07100 Sassari, Italy
- Sbarro Institute for Cancer Research and Molecular Medicine, Centre for Biotechnology, College of Science and Technology, Temple University, Philadelphia, PA 19122, USA
| |
Collapse
|
4
|
Jankoski PR, Bach E, da Fonseca RN, Hübner S, de Carvalho JB, de Souza da Motta A. Bacillus altitudinis 1.4 genome analysis-functional annotation of probiotic properties and immunomodulatory activity. World J Microbiol Biotechnol 2024; 40:293. [PMID: 39112831 DOI: 10.1007/s11274-024-04096-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 07/26/2024] [Indexed: 10/17/2024]
Abstract
Probiotics are live microorganisms that, when administered in adequate quantities, provide health benefits to the host. In this study, phenotypic and genotypic methods were used to evaluate the probiotic properties of Bacillus altitudinis 1.4. The isolate was sensitive to all antimicrobials tested and presented a positive result in the hemolysis test. B. altitudinis 1.4 spores were more resistant than vegetative cells, when evaluated in simulation of cell viability in the gastrointestinal tract, as well as adhesion to the intestinal mucosa. The isolate was capable of self-aggregation and coaggregation with pathogens such as Escherichia coli ATCC 25922 and Salmonella Enteritidis ATCC 13076. Genomic analysis revealed the presence of genes with probiotic characteristics. From this study it was possible to evaluate the gene expression of pro-inflammatory and anti-inflammatory cytokines for different treatments. Viable vegetative cells of B. altitudinis 1.4 increased the transcription of pro-inflammatory factors, in addition to also increasing the transcription of IL-10, indicating a tendency to stimulate a pro-inflammatory profile. Given the results presented, B. altitudinis 1.4 showed potential to be applied in the incorporation of this microorganism into animal feed, since the spores could tolerate the feed handling and pelletization processes.
Collapse
Affiliation(s)
- Priscila Ribeiro Jankoski
- Instituto de Ciências Básicas da Saúde, Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, sala 216, Porto Alegre, RS, 90050-170, Brazil
| | - Evelise Bach
- Departamento de Genética, Instituto de Biociências (IB), Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, RS, Brazil
| | - Renata Nobre da Fonseca
- Departamento de Veterinária Preventiva, Universidade Federal de Pelotas (UFPel), Pelotas, RS, Brazil
| | - Silvia Hübner
- Departamento de Veterinária Preventiva, Universidade Federal de Pelotas (UFPel), Pelotas, RS, Brazil
| | | | - Amanda de Souza da Motta
- Instituto de Ciências Básicas da Saúde, Departamento de Microbiologia, Imunologia e Parasitologia, Universidade Federal do Rio Grande do Sul, Rua Sarmento Leite 500, sala 216, Porto Alegre, RS, 90050-170, Brazil.
| |
Collapse
|
5
|
Li D, Jiang Y, Cui Z, Ma M, Zhu F, Li G, Yang H, Li S, Zhang T, Chen D, Ma W. Lactobacillus acidophilus protects against Corynebacterium pseudotuberculosis infection by regulating the autophagy of macrophages and maintaining gut microbiota homeostasis in C57BL/6 mice. mSystems 2024; 9:e0048424. [PMID: 38934644 PMCID: PMC11265446 DOI: 10.1128/msystems.00484-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 05/22/2024] [Indexed: 06/28/2024] Open
Abstract
Corynebacterium pseudotuberculosis (C. p), a facultative intracellular bacterium, is an important zoonotic pathogen that causes abscesses and pyogenic granulomas. The relationship between gut microbiota and host health or diseases has received increasing attention. However, the role of gut microbiota in the process of C. p infection is still unclear. In this study, we established a C. p infection model in C57BL/6 mice and examined the impact of preemptive oral administration Lactobacillus acidophilus (L. acidophilus) on infection. Our findings revealed that C. p infection led to pronounced pathological alterations in the liver and kidneys, characterized by abscess formation, intense inflammatory responses, and bacterial overload. Remarkably, these deleterious effects were greatly relieved by oral administration of L. acidophilus before infection with C. p. Additionally, we further found that during C. p infection, peritoneal macrophages (PMs) of mice orally administered with L. acidophilus accumulated more rapidly at sites of infection. Furthermore, our results showed that PMs from mice with oral L. acidophilus administration showed a stronger C. p clearance effect, and this was mediated by high expression of LC3-II protein. Meanwhile, oral administration of L. acidophilus protected the gut microbiota disorder in C57BL/6 mice caused by C. p infection. In summary, our study demonstrates that oral administration of L. acidophilus confers effective protection against C. p infection in C57BL/6 mice by modulating macrophage autophagy, thereby augmenting bacterial clearance and preserving gut microbiota and function stability. These findings position L. acidophilus as a viable probiotic candidate for the clinical prevention of C. p infection. IMPORTANCE Corynebacterium pseudotuberculosis (C. p) is known to induce a range of chronic diseases in both animals and humans. Currently, clinical treatment for C. p infection mainly relies on antibiotic therapy or surgical intervention. However, excessive use of antibiotics may increase the risk of drug-resistant strains, and the effectiveness of treatment remains unsatisfactory. Furthermore, surgical procedures do not completely eradicate pathogens and can easily cause environmental pollution. Probiotic interventions are receiving increasing attention for improving the body's immune system and maintaining health. In this study, we established a C. p infection model in C57BL/6 mice to explore the impact of Lactobacillus acidophilus during C. p infection. Our results showed that L. acidophilus effectively protected against C. p infection by regulating the autophagy of macrophages and maintaining intestinal microbiota homeostasis. This study may provide a new strategy for the prevention of C. p infection.
Collapse
Affiliation(s)
- Dengliang Li
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Yuecai Jiang
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Zhanding Cui
- State Key Laboratory for Animal Disease Control and Prevention, College of Veterinary Medicine, Lanzhou University, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Mengzhen Ma
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Fang Zhu
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Guanhua Li
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Haoyue Yang
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Shaofei Li
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Tianliang Zhang
- Shaanxi Qianyang Saanen dairy goats Development Co., Ltd, Qianyang, Shaanxi, China
| | - Dekun Chen
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| | - Wentao Ma
- Veterinary Immunology Laboratory, College of Veterinary Medicine, Northwest Agriculture and Forestry University, Yangling, Shaanxi, China
| |
Collapse
|
6
|
Jin W, Shen S, Xu X, Xie X, Zhou X, Su X, Wu L, Wang S, Zhang L, Chen B, Yang F. All-in-one hydrogel patches with sprayed bFGF-loaded GelMA microspheres for infected wound healing studies. Int J Pharm 2024; 658:124205. [PMID: 38734278 DOI: 10.1016/j.ijpharm.2024.124205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 04/30/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
The current wound healing process faces numerous challenges such as bacterial infection, inflammation and oxidative stress. However, wound dressings used to promote wound healing, are not well suited to meet the clinical needs. Hyaluronic acid (HA) not only has excellent water absorption and good biocompatibility but facilitates cell function and tissue regeneration. Dopamine, on the other hand, increases the overall viscosity of the hydrogel and possesses antioxidant property. Furthermore, chitosan exhibits outstanding performance in antimicrobial, anti-inflammatory and antioxidant activities. Basic fibroblast growth factor (bFGF) is conducive to cell proliferation and migration, vascular regeneration and wound healing. Hence, we designed an all-in-one hydrogel patch containing dopamine and chitosan framed by hyaluronic acid (HDC) with sprayed gelatin methacryloyl (GelMA) microspheres loaded with bFGF (HDC-bFGF). The hydrogel patch exhibits excellent adhesive, anti-inflammatory, antioxidant and antibacterial properties. In vitro experiments, the HDC-bFGF hydrogel patch not only showed significant inhibitory effect on RAW cell inflammation and Staphylococcus aureus (S. aureus) growth but also effectively scavenged free radicals, in addition to promoting the migration of 3 T3 cells. In the mice acute infected wound model, the HDC-bFGF hydrogel patch adhered to the wound surface greatly accelerated the healing process via its anti-inflammatory and antioxidant activities, bacterial inhibition and pro-vascularization effects. Therefore, the multifunctional HDC-bFGF hydrogel patch holds great promise for clinical application.
Collapse
Affiliation(s)
- Wenzhang Jin
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province Wenzhou 325000, PR China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Department of Colorectal Surgery, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310000, PR China
| | - Shuqi Shen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Xiaoniuyue Xu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China; Department of Hand Surgery, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province Wenzhou 325000, PR China
| | - Xueting Xie
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Xingjian Zhou
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Xiang Su
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province Wenzhou 325000, PR China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Lina Wu
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Shunfu Wang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Lijiang Zhang
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China
| | - Bicheng Chen
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China.
| | - Fajing Yang
- Department of Vascular Surgery, The First Affiliated Hospital of Wenzhou Medical University, Zhejiang Province Wenzhou 325000, PR China; Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, PR China.
| |
Collapse
|
7
|
Negi A, Pasam T, Farqadain SM, Mahalaxmi Y, Dandekar MP. In-vitro and preclinical testing of bacillus subtilis UBBS-14 probiotic in rats shows no toxicity. Toxicol Res (Camb) 2024; 13:tfae021. [PMID: 38406637 PMCID: PMC10891425 DOI: 10.1093/toxres/tfae021] [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: 10/24/2023] [Revised: 01/23/2024] [Accepted: 02/06/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Probiotics made from Bacillus subtilis provide a wide spread of health benefits, particularly in the treatment of diarrhea and gastrointestinal problems. Herein, we employed in vitro and in vivo paradigms to assess the potential adverse effects and toxicity of B. subtilis UBBS-14. Materials and methods According to Organization for Economic Co-operation and Development (OECD) 423 and 407 requirements, a preclinical investigation was conducted in male and female Sprague-Dawley rats. Acute toxicity was examined following a single peroral (PO) administration of 5,000 mg/kg body weight (bw) i.e. equivalent to 500 billion colony-forming units (CFU) per kg bw. Single administration of B. subtilis UBBS-14 showed no mortality or adverse effects until the 14-day observation period, indicating LD50 is >5,000 mg/kg bw. Results Incubation of B. subtilis UBBS-14 with Caco2, HT29, and Raw 264.7 cell lines, showed no cytotoxic effects. This probiotic strain was also found responsive to the majority of antibiotics. For a 28-day repeated dose toxicity study, rats were administered 100, 500, and 1,000 mg/kg bw daily once (10, 50, and 100 billion CFU/kg bw/day, respectively) doses of B. subtilis UBBS-14. No notable changes were seen in the morphology, weight, and histopathology of the critical internal organs. The haematological, biochemical, electrolyte (sodium, potassium, chloride, and calcium), and urine analytical results were within the normal range and equivalent to the vehicle-treated group. Conclusion B. subtilis UBBS-14's no-observed-effect level (NOEL) was thus determined to be >1,000 mg/kg bw/day following a 28-day oral dosing.
Collapse
Affiliation(s)
- Ankit Negi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), NH 9, Balanagar Main Rd, Kukatpally Industrial Estate, Balanagar, Hyderabad, Telangana 500037, India
| | - Tulasi Pasam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), NH 9, Balanagar Main Rd, Kukatpally Industrial Estate, Balanagar, Hyderabad, Telangana 500037, India
| | - Syed Muhammad Farqadain
- Unique Biotech Limited, Centre for Research and Development, Hyderabad, Telangana, 500 101, India
| | - Y Mahalaxmi
- Unique Biotech Limited, Centre for Research and Development, Hyderabad, Telangana, 500 101, India
| | - Manoj P Dandekar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), NH 9, Balanagar Main Rd, Kukatpally Industrial Estate, Balanagar, Hyderabad, Telangana 500037, India
| |
Collapse
|
8
|
Khokhlova E, Colom J, Simon A, Mazhar S, García-Lainez G, Llopis S, Gonzalez N, Enrique-López M, Álvarez B, Martorell P, Tortajada M, Deaton J, Rea K. Immunomodulatory and Antioxidant Properties of a Novel Potential Probiotic Bacillus clausii CSI08. Microorganisms 2023; 11:microorganisms11020240. [PMID: 36838205 PMCID: PMC9962608 DOI: 10.3390/microorganisms11020240] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2022] [Revised: 01/10/2023] [Accepted: 01/13/2023] [Indexed: 01/19/2023] Open
Abstract
Spore-forming bacteria of the Bacillus genus have demonstrated potential as probiotics for human use. Bacillus clausii have been recognized as efficacious and safe agents for preventing and treating diarrhea in children and adults, with pronounced immunomodulatory properties during several in vitro and clinical studies. Herein, we characterize the novel strain of B. clausii CSI08 (Munispore®) for probiotic attributes including resistance to gastric acid and bile salts, the ability to suppress the growth of human pathogens, the capacity to assimilate wide range of carbohydrates and to produce potentially beneficial enzymes. Both spores and vegetative cells of this strain were able to adhere to a mucous-producing intestinal cell line and to attenuate the LPS- and Poly I:C-triggered pro-inflammatory cytokine gene expression in HT-29 intestinal cell line. Vegetative cells of B. clausii CSI08 were also able to elicit a robust immune response in U937-derived macrophages. Furthermore, B. clausii CSI08 demonstrated cytoprotective effects in in vitro cell culture and in vivo C. elegans models of oxidative stress. Taken together, these beneficial properties provide strong evidence for B. clausii CSI08 as a promising potential probiotic.
Collapse
Affiliation(s)
- Ekaterina Khokhlova
- Deerland Ireland R&D, Ltd., ADM, Bio-Innovation Unit, Rm. 331 Food Science Building, College Rd., University College Cork, T12 K8AF Cork, Ireland
| | - Joan Colom
- Deerland Ireland R&D, Ltd., ADM, Bio-Innovation Unit, Rm. 331 Food Science Building, College Rd., University College Cork, T12 K8AF Cork, Ireland
| | - Annie Simon
- Deerland Ireland R&D, Ltd., ADM, Bio-Innovation Unit, Rm. 331 Food Science Building, College Rd., University College Cork, T12 K8AF Cork, Ireland
| | - Shahneela Mazhar
- Deerland Ireland R&D, Ltd., ADM, Bio-Innovation Unit, Rm. 331 Food Science Building, College Rd., University College Cork, T12 K8AF Cork, Ireland
| | - Guillermo García-Lainez
- Archer Daniels Midland, Nutrition, Health&Wellness, Biopolis S.L. Parc Científic Universitat de València, C/ Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain
| | - Silvia Llopis
- Archer Daniels Midland, Nutrition, Health&Wellness, Biopolis S.L. Parc Científic Universitat de València, C/ Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain
| | - Nuria Gonzalez
- Archer Daniels Midland, Nutrition, Health&Wellness, Biopolis S.L. Parc Científic Universitat de València, C/ Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain
| | - María Enrique-López
- Archer Daniels Midland, Nutrition, Health&Wellness, Biopolis S.L. Parc Científic Universitat de València, C/ Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain
| | - Beatriz Álvarez
- Archer Daniels Midland, Nutrition, Health&Wellness, Biopolis S.L. Parc Científic Universitat de València, C/ Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain
| | - Patricia Martorell
- Archer Daniels Midland, Nutrition, Health&Wellness, Biopolis S.L. Parc Científic Universitat de València, C/ Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain
| | - Marta Tortajada
- Archer Daniels Midland, Nutrition, Health&Wellness, Biopolis S.L. Parc Científic Universitat de València, C/ Catedrático Agustín Escardino Benlloch, 9, 46980 Paterna, Spain
| | - John Deaton
- Deerland Probiotics & Enzymes, ADM, Science and Technology Department, 3800 Cobb International Blvd., Kennesaw, GA 30152, USA
| | - Kieran Rea
- Deerland Ireland R&D, Ltd., ADM, Bio-Innovation Unit, Rm. 331 Food Science Building, College Rd., University College Cork, T12 K8AF Cork, Ireland
- Correspondence:
| |
Collapse
|
9
|
Qin D, Ma Y, Wang Y, Hou X, Yu L. Contribution of Lactobacilli on Intestinal Mucosal Barrier and Diseases: Perspectives and Challenges of Lactobacillus casei. LIFE (BASEL, SWITZERLAND) 2022; 12:life12111910. [PMID: 36431045 PMCID: PMC9696601 DOI: 10.3390/life12111910] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/13/2022] [Accepted: 11/15/2022] [Indexed: 11/18/2022]
Abstract
The intestine barrier, the front line of normal body defense, relies on its structural integrity, microbial composition and barrier immunity. The intestinal mucosal surface is continuously exposed to a complex and dynamic community of microorganisms. Although it occupies a relatively small proportion of the intestinal microbiota, Lactobacilli has been discovered to have a significant impact on the intestine tract in previous studies. It is undeniable that some Lactobacillus strains present probiotic properties through maintaining the micro-ecological balance via different mechanisms, such as mucosal barrier function and barrier immunity, to prevent infection and even to solve some neurology issues by microbiota-gut-brain/liver/lung axis communication. Notably, not only living cells but also Lactobacillus derivatives (postbiotics: soluble secreted products and para-probiotics: cell structural components) may exert antipathogenic effects and beneficial functions for the gut mucosal barrier. However, substantial research on specific effects, safety and action mechanisms in vivo should be done. In clinical application of humans and animals, there are still doubts about the precise evaluation of Lactobacilli's safety, therapeutic effect, dosage and other aspects. Therefore, we provide an overview of central issues on the impacts of Lactobacillus casei (L. casei) and their products on the intestinal mucosal barrier and some diseases and highlight the urgent need for further studies.
Collapse
Affiliation(s)
- Da Qin
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yixuan Ma
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Yanhong Wang
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
| | - Xilin Hou
- Colleges of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Correspondence: (X.H.); (L.Y.); Tel.: +86-4596-819-290 (X.H. & L.Y.); Fax: +86-4596-819-292 (X.H. & L.Y.)
| | - Liyun Yu
- Heilongjiang Provincial Key Laboratory of Environmental Microbiology and Recycling of Argo-Waste in Cold Region, College of Life Science and Biotechnology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Colleges of Animal Science and Technology, Heilongjiang Bayi Agricultural University, Daqing 163319, China
- Correspondence: (X.H.); (L.Y.); Tel.: +86-4596-819-290 (X.H. & L.Y.); Fax: +86-4596-819-292 (X.H. & L.Y.)
| |
Collapse
|
10
|
Lactiplantibacillus plantarum LOC1 Isolated from Fresh Tea Leaves Modulates Macrophage Response to TLR4 Activation. Foods 2022. [DOI: 10.3390/foods11203257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Previously, we demonstrated that Lactiplantibacillus plantarum LOC1, originally isolated from fresh tea leaves, was able to improve epithelial barrier integrity in in vitro models, suggesting that this strain is an interesting probiotic candidate. In this work, we aimed to continue characterizing the potential probiotic properties of the LOC1 strain, focusing on its immunomodulatory properties in the context of innate immunity triggered by Toll-like receptor 4 (TLR4) activation. These studies were complemented by comparative and functional genomics analysis to characterize the bacterial genes involved in the immunomodulatory capacity. We carried out a transcriptomic study to evaluate the effect of L. plantarum LOC1 on the response of murine macrophages (RAW264.7 cells) to the activation of TLR4. We demonstrated that L. plantarum LOC1 exerts a modulatory effect on lipopolysaccharide (LPS)-induced inflammation, resulting in a differential regulation of immune factor expression in macrophages. The LOC1 strain markedly reduced the LPS-induced expression of some inflammatory cytokines (IL-1β, IL-12, and CSF2) and chemokines (CCL17, CCL28, CXCL3, CXCL13, CXCL1, and CX3CL1), while it significantly increased the expression of other cytokines (TNF-α, IL-6, IL-18, IFN-β, IFN-γ, and CSF3), chemokines (IL-15 and CXCL9), and activation markers (H2-k1, H2-M3, CD80, and CD86) in RAW macrophages. Our results show that L. plantarum LOC1 would enhance the intrinsic functions of macrophages, promoting their protective effects mediated by the stimulation of the Th1 response without affecting the regulatory mechanisms that help control inflammation. In addition, we sequenced the LOC1 genome and performed a genomic characterization. Genomic comparative analysis with the well-known immunomodulatory strains WCSF1 and CRL1506 demonstrated that L. plantarum LOC1 possess a set of adhesion factors and genes involved in the biosynthesis of teichoic acids and lipoproteins that could be involved in its immunomodulatory capacity. The results of this work can contribute to the development of immune-related functional foods containing L. plantarum LOC1.
Collapse
|
11
|
Wong-Chew RM, de Castro JAA, Morelli L, Perez M, Ozen M. Gut immune homeostasis: the immunomodulatory role of Bacillus clausii, from basic to clinical evidence. Expert Rev Clin Immunol 2022; 18:717-729. [PMID: 35674642 DOI: 10.1080/1744666x.2022.2085559] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION The gut microbiota affects the development of the gut immune system in early life. Perturbations to microbiota structure and composition during this period can have long-term consequences on the health of the individual, through its effects on the immune system. Research in the last few decades has shown that probiotic administration can reverse these effects in strain- and environment-specific ways. Bacillus clausii (B. clausii) has been in use for many decades as a safe and efficacious probiotic, but its mode of action has not yet been completely elucidated. AREAS COVERED In this review, we discuss how the gut immune system works, the factors that affect its functioning, and the plethora of research highlighting its role in various diseases. We also discuss the known modes of action of Bacillus probiotics, and highlight the preclinical and clinical evidence that reveal how B. clausii acts to bolster gut defense. EXPERT OPINION We anticipate that the treatment and/or prevention of dysbiosis will be central to managing human health and disease in the future. Discovering the pathophysiology of autoimmune diseases, infections, allergies, and some cancers will aid our understanding of the key role played by microbial communities in these diseases.
Collapse
Affiliation(s)
- Rosa María Wong-Chew
- Facultad de Medicina, División de Investigación, Universidad Nacional Autónoma de México, Coyoacán, Cdmx
| | - Jo-Anne A de Castro
- Department of Pediatrics de la Salle Medical and Health Sciences Institute (DLSMHSI), Dasmariñas Cavite, Philippines; Department of Microbiology and Parasitology, Pamantasan ng Lunsod ng Maynila (PLM), College of Medicine Intramuros, Manila, Philippines
| | - Lorenzo Morelli
- Faculty of Agriculture, Food and Environmental Sciences, Università Cattolica del Sacro Cuore Piacenza - Cremona, Italy
| | | | - Metehan Ozen
- Division of Pediatric Infectious Diseases, Acıbadem Mehmet Ali Aydınlar University, School of Medicine, Istanbul Turkey
| |
Collapse
|
12
|
Current Progress and Future Perspectives on the Use of Bacillus clausii. Microorganisms 2022; 10:microorganisms10061246. [PMID: 35744764 PMCID: PMC9230978 DOI: 10.3390/microorganisms10061246] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/30/2022] [Accepted: 05/31/2022] [Indexed: 12/12/2022] Open
Abstract
Bacillus clausii is a probiotic that benefits human health. Its key characteristics include the ability to form spores; the resulting tolerance to heat, acid, and salt ensures safe passage through the human gastrointestinal tract with no loss of cells. Although B. clausii has been widely used for many decades, the beneficial properties of other probiotics, such as Lactobacillus spp. and Bifidobacterium spp., are better disseminated in the literature. In this review, we summarize the physiological, antimicrobial, and immunomodulatory properties of probiotic B. clausii strains. We also describe findings from studies that have investigated B. clausii probiotics from the perspective of quality and safety. We highlight innovative properties based on biochemical investigations of non-probiotic strains of B. clausii, revealing that B. clausii may have further health benefits in other therapeutic areas.
Collapse
|
13
|
Todorov SD, Ivanova IV, Popov I, Weeks R, Chikindas ML. Bacillus spore-forming probiotics: benefits with concerns? Crit Rev Microbiol 2021; 48:513-530. [PMID: 34620036 DOI: 10.1080/1040841x.2021.1983517] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Representatives of the genus Bacillus are multifunctional microorganisms with a broad range of applications in both traditional fermentation and modern biotechnological processes. Bacillus spp. has several beneficial properties. They serve as starter cultures for various traditional fermented foods and are important biotechnological producers of enzymes, antibiotics, and bioactive peptides. They are also used as probiotics for humans, in veterinary medicine, and as feed additives for animals of agricultural importance. The beneficial effects of bacilli are well-reported and broadly acknowledged. However, with a better understanding of their positive role, many questions have been raised regarding their safety and the relevance of spore formation in the practical application of this group of microorganisms. What is the role of Bacillus spp. in the human microbial consortium? When and why did they start colonizing the gastrointestinal tract (GIT) of humans and other animals? Can spore-forming probiotics be considered as truly beneficial organisms, or should they still be approached with caution and regarded as "benefits with concerns"? In this review, we not only hope to answer the above questions but to expand the scope of the conversation surrounding bacilli probiotics.
Collapse
Affiliation(s)
| | - Iskra Vitanova Ivanova
- Department of General and Applied Microbiology, Faculty of Biology, Sofia University St. Kliment Ohridski, Sofia, Bulgaria
| | - Igor Popov
- Center for Agrobiotechnology, Don State Technical University, Rostov-on-Don, Russia
| | - Richard Weeks
- Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA
| | - Michael Leonidas Chikindas
- Center for Agrobiotechnology, Don State Technical University, Rostov-on-Don, Russia.,Health Promoting Naturals Laboratory, School of Environmental and Biological Sciences, Rutgers State University, New Brunswick, NJ, USA.,I.M. Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
14
|
Benítez-Cabello A, Torres-Maravilla E, Bermúdez-Humarán L, Langella P, Martín R, Jiménez-Díaz R, Arroyo-López FN. Probiotic Properties of Lactobacillus Strains Isolated from Table Olive Biofilms. Probiotics Antimicrob Proteins 2021; 12:1071-1082. [PMID: 31788768 DOI: 10.1007/s12602-019-09604-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
In this work, 16 strains with promising probiotic characteristics belonging to the Lactobacillus pentosus (13) and Lactobacillus plantarum (3) species and isolated from table olive biofilms were tested for adherence to cell lines and to solvents, immunomodulatory, and anti-proliferative properties on epithelial human cellular lines. Most Lactobacillus strains were able to regulate the production of cytokines by stimulating the production of pro-inflammatory (IL-6) and anti-inflammatory (IL-10) interleukins on macrophages, and by suppressing the secretion of IL-8 on HT-29 TNF-α-induced model. Lactobacillus strains also showed anti-proliferative activity on the HT-29 cell line. No clear relation was found between adhesion to solvents and adhesion to HT-29 human cell line. Lactobacillus pentosus LPG1, which showed the best anti-inflammatory and immunomodulatory properties, was then tested in a dinitro-benzene sulfonic acid (DNBS)-induced chronic colitis murine model. As a measure of the inflammation, gut permeability and weight loss, as well as cytokine profiles, were determined. Lactobacillus pentosus LPG1 improved mice health as observed by a significant reduction of weight loss, gut permeability, and beneficial cytokine modulation. Macroscopic scores and tissue damage were also lower in mice administered with LPG1 with respect to the DNBS-treated group. These results showed that L. pentosus LPG1 isolated from plant could have potential as probiotic for use as an anti-inflammatory and immunomodulatory agent for patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Antonio Benítez-Cabello
- Food Biotechnology Department, Instituto de la Grasa (CSIC), Ctra. Utrera km 1, Building 46, 41013, Seville, Spain.,INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, 78350, Jouy-en-Josas, France
| | - Edgar Torres-Maravilla
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, 78350, Jouy-en-Josas, France
| | - Luis Bermúdez-Humarán
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, 78350, Jouy-en-Josas, France
| | - Philippe Langella
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, 78350, Jouy-en-Josas, France
| | - Rebeca Martín
- INRA, Commensal and Probiotics-Host Interactions Laboratory, UMR 1319 Micalis, 78350, Jouy-en-Josas, France
| | - Rufino Jiménez-Díaz
- Food Biotechnology Department, Instituto de la Grasa (CSIC), Ctra. Utrera km 1, Building 46, 41013, Seville, Spain
| | - Francisco Noé Arroyo-López
- Food Biotechnology Department, Instituto de la Grasa (CSIC), Ctra. Utrera km 1, Building 46, 41013, Seville, Spain.
| |
Collapse
|
15
|
Guha D, Mukherjee R, Aich P. Effects of two potential probiotic Lactobacillus bacteria on adipogenesis in vitro. Life Sci 2021; 278:119538. [PMID: 33932443 DOI: 10.1016/j.lfs.2021.119538] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/31/2021] [Accepted: 04/07/2021] [Indexed: 12/12/2022]
Abstract
AIMS Overweight is a major global health problem. Various methodologies to get rid of the extra fat are available, but usually, those are associated with adverse side effects. Probiotics, on the contrary, seem to have the potential to help reduce fat accumulation without much apparent adversity. In this study, we have evaluated a pair of well-documented probiotics for their anti-obesogenic effects. MAIN METHODS We used strains of Lactobacillus acidophilus (LA) and a cocktail (LDB-ST) of Lactobacillus delbruckei sp. bulgaricus (LDB) and Streptococcus thermophilus (ST) in this study. The murine pre-adipocyte cell line 3T3-L1 was terminally differentiated to matured adipocytes to use as a model to evaluate the bacteria's anti-obesogenic effects. The optimal dose for treatment of both the probiotics was determined using a cell viability assay. We assessed the probiotic internalization potential of differentiated 3T3-L1 cells by flow cytometry, fluorescence microscopy, and cell lysis method. We determined the lipolytic and anti-adipogenic potential of probiotics by intracellular lipid staining, spectrophotometry, and gene expression analysis. KEY FINDINGS Both probiotics were effective lipolytic agents as revealed by reducing cellular lipids and down-regulation of mammalian adipogenesis marker genes in terminally differentiated 3T3-L1 cells. SIGNIFICANCE Previous studies from our group had proven the immune-modulatory properties of these probiotics on an immune-biased mouse model. The present study demonstrates LA and LDB-ST to be effective against adipogenesis. Further in vivo studies will be conducted to strengthen this claim.
Collapse
Affiliation(s)
- Dipanjan Guha
- School of Biological Sciences, National Institute of Science Education and Research (NISER), P.O. - Bhimpur-Padanpur, Jatni - 752050, Dist.-Khurda, Odisha, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai 400 094, India
| | - Raktim Mukherjee
- School of Biological Sciences, National Institute of Science Education and Research (NISER), P.O. - Bhimpur-Padanpur, Jatni - 752050, Dist.-Khurda, Odisha, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai 400 094, India
| | - Palok Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), P.O. - Bhimpur-Padanpur, Jatni - 752050, Dist.-Khurda, Odisha, India; Homi Bhabha National Institute (HBNI), Training School Complex, Anushaktinagar, Mumbai 400 094, India.
| |
Collapse
|
16
|
Ray P, Pandey U, Aich P. Comparative analysis of beneficial effects of vancomycin treatment on Th1- and Th2-biased mice and the role of gut microbiota. J Appl Microbiol 2020; 130:1337-1356. [PMID: 32955795 DOI: 10.1111/jam.14853] [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] [Received: 04/19/2020] [Revised: 08/18/2020] [Accepted: 09/07/2020] [Indexed: 12/22/2022]
Abstract
AIMS The aim was to understand the time-dependent antibiotic-induced perturbation pattern of gut microbiota and its effect on the innate immune and metabolic profile of the host. METHODS AND RESULTS Vancomycin was administered at 50 mg kg-1 of body weight twice daily for six consecutive days to perturb the gut microbiota of C57BL/6 (Th1-biased) and BALB/c (Th2-biased) mice. Following treatment with vancomycin, we observed a reduction in the abundance of phyla Firmicutes and Bacteroides and an increase in Proteobacteria in the gut for both strains of mice following treatment with vancomycin till day 4. Abundance of Akkermansia muciniphila of Verrucomicrobia phylum also increased, from day 5 onwards following vancomycin treatment. The time-dependent variation of gut microbiota was associated with increased (i) expression of toll-like receptors and inflammatory genes such as TNF-α, IL-6, and IL-17, (ii) gut barrier permeability and (iii) blood glucose level of the host. The results also showed that (i) transplantation of cecal microbiota from vancomycin-treated day 6 mice to day 3 vancomycin-treated mice helped in restoring blood glucose level in C57BL/6 mice and (ii) short-chain fatty acids like acetate, butyrate and propionate changed with the alteration of gut microbiota to induce differential regulation of host immune response. CONCLUSIONS The current results revealed that an increase in A. muciniphila led to decreased inflammation and increased rate of glucose tolerance in the host. The treatment, with vancomycin till day 4, increased expression of inflammatory genes. The continuation of vancomycin for two more days reversed the effects. The effects were significantly more in C57BL/6 than BALB/c mice. SIGNIFICANCE AND IMPACT OF THE STUDY The current study established that the treatment with vancomycin till day 4 increased pathogenic bacteria but day 5 onwards provided significant health-related benefits to the host by increasing A. muciniphila more in C57BL/6 than BALB/c mice.
Collapse
Affiliation(s)
- P Ray
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, Khurdha, Odisha, India
| | - U Pandey
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, Khurdha, Odisha, India
| | - P Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, Khurdha, Odisha, India
| |
Collapse
|
17
|
Effects of an orally supplemented probiotic on the autophagy protein LC3 and Beclin1 in placentas undergoing spontaneous delivery during normal pregnancy. BMC Pregnancy Childbirth 2020; 20:216. [PMID: 32295534 PMCID: PMC7161261 DOI: 10.1186/s12884-020-02905-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Accepted: 03/27/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Probiotic supplementation has been shown to be beneficial and is now widely promoted as an auxiliary medicine for maternal health, but the underlying mechanism is still unclear. Thus, this study aimed to explore the effects of probiotic supplementation on the placental autophagy-related proteins LC3 and Beclin1. METHOD A population-based cohort of specimens was collected under sterile conditions from 37 healthy nulliparous pregnant women who underwent systemic examination and delivered at the First Affiliated Hospital of Jinan University (Guangzhou, China). At 32 weeks of gestation, the pregnant women in the probiotic group were orally supplemented with golden bifid, and the pregnant women in the control group received no probiotic. Pregnant women with pregnancy-associated complications were excluded in the follow-up period, and 25 pregnant women undergoing spontaneous delivery were enrolled. The placental tissue specimens were collected at term. Western blotting was used to detect the protein expression, and qRT-PCR was used to detect the mRNA expression of the placental autophagy-related proteins LC3 and Beclin1. RESULTS ①There was no significant difference in the expression levels of either LC3 or Beclin1 protein between the two groups (P > 0.05). ②Probiotic supplementation induced a modest but not significant decrease in the content of LC3-mRNA with a significant decrease in the content of Beclin1-mRNA (P < 0.05). CONCLUSION Our study indicates that probiotic supplementation may reduce Beclin1-mRNA levels.
Collapse
|
18
|
Pradhan B, Guha D, Naik AK, Banerjee A, Tambat S, Chawla S, Senapati S, Aich P. Probiotics L. acidophilus and B. clausii Modulate Gut Microbiota in Th1- and Th2-Biased Mice to Ameliorate Salmonella Typhimurium-Induced Diarrhea. Probiotics Antimicrob Proteins 2020; 11:887-904. [PMID: 29909486 DOI: 10.1007/s12602-018-9436-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Gut microbiota play important role in maintaining health. Probiotics are believed to augment it further. We aimed at comparing effects of probiotics, Lactobacillus acidophilus (LA) and Bacillus clausii (BC) (a) on the gut microbiota abundance and diversity and (b) their contributions to control intestinal dysbiosis and inflammation in Th1- and Th2-biased mice following Salmonella infection. We report how could gut microbiota and the differential immune bias (Th1 or Th2) of the host regulate host responses when challenged with Salmonella typhimurium in the presence and absence of either of the probiotics. LA was found to be effective in ameliorating the microbial dysbiosis and inflammation caused by Salmonella infection, in Th1 (C57BL/6) and Th2 (BALB/c)-biased mouse. BC was able to ameliorate Salmonella-induced dysbiosis and inflammation in Th2 but not in Th1-biased mouse. These results may support probiotics LA as a treatment option in the case of Salmonella infection.
Collapse
Affiliation(s)
- Biswaranjan Pradhan
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Jatni, Khurdha, Odisha, 752050, India
- S. K. Dash Center of Excellence of Biosciences and Engineering & Technology (SKBET), Indian Institute of Technology Bhubaneswar, Bhubaneswar, Odisha, India
| | - Dipanjan Guha
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Jatni, Khurdha, Odisha, 752050, India
| | - Aman Kumar Naik
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Jatni, Khurdha, Odisha, 752050, India
| | - Arka Banerjee
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Jatni, Khurdha, Odisha, 752050, India
- Biozentrum der Universität Basel, 50-70 Klingelbergstrasse, 4056, Basel, Switzerland
| | - Subodh Tambat
- Bionivid Technology Private Limited, 209, 4th Cross Rd, B Channasandra, East of NGEF Layout, Kasturi Nagar, Bengaluru, Karnataka, 560043, India
| | - Saurabh Chawla
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Jatni, Khurdha, Odisha, 752050, India
| | - Shantibhusan Senapati
- Institute of Life Sciences, Nalco Square, Chandrasekharpur, Bhubaneswar, Odisha, 751023, India
| | - Palok Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Jatni, Khurdha, Odisha, 752050, India.
| |
Collapse
|
19
|
Khatri I, Sharma G, Subramanian S. Composite genome sequence of Bacillus clausii, a probiotic commercially available as Enterogermina ®, and insights into its probiotic properties. BMC Microbiol 2019; 19:307. [PMID: 31888501 PMCID: PMC6937992 DOI: 10.1186/s12866-019-1680-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/11/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Some of the spore-forming strains of Bacillus probiotics are marketed commercially as they survive harsh gastrointestinal conditions and bestow health benefits to the host. RESULTS We report the composite genome of Bacillus clausii ENTPro from a commercially available probiotic Enterogermina® and compare it with the genomes of other Bacillus probiotics. We find that the members of B. clausii species harbor high heterogeneity at the species as well as genus level. The genes conferring resistance to chloramphenicol, streptomycin, rifampicin, and tetracycline in the B. clausii ENTPro strain could be identified. The genes coding for the bacteriocin gallidermin, which prevents biofilm formation in the pathogens Staphylococcus aureus and S. epidermidis, were also identified. KEGG Pathway analysis suggested that the folate biosynthesis pathway, which depicts one of the important roles of probiotics in the host, is conserved completely in B. subtilis and minimally in B. clausii and other probiotics. CONCLUSIONS We identified various antibiotic resistance, bacteriocins, stress-related, and adhesion-related domains, and industrially-relevant pathways, in the genomes of these probiotic bacteria that are likely to help them survive in the harsh gastrointestinal tract, facilitating adhesion to host epithelial cells, persistence during antibiotic treatment and combating bacterial infections.
Collapse
Affiliation(s)
- Indu Khatri
- CSIR-Institute of Microbial Technology, Sector-39A, Chandigarh, 160036, India.,Leiden University Medical Center, Leiden, the Netherlands
| | - Gaurav Sharma
- CSIR-Institute of Microbial Technology, Sector-39A, Chandigarh, 160036, India.,Institute of Bioinformatics and Applied Biotechnology, Bengaluru, Karnataka, India
| | | |
Collapse
|
20
|
A probiotic formulation containing Lactobacillus bulgaricus DWT1 inhibits tumor growth by activating pro-inflammatory responses in macrophages. J Funct Foods 2019. [DOI: 10.1016/j.jff.2019.03.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
21
|
Chen X, Fu Y, Wang L, Qian W, Zheng F, Hou X. Bifidobacterium longum and VSL#3 ® amelioration of TNBS-induced colitis associated with reduced HMGB1 and epithelial barrier impairment. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2019; 92:77-86. [PMID: 30227219 DOI: 10.1016/j.dci.2018.09.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2018] [Revised: 09/11/2018] [Accepted: 09/11/2018] [Indexed: 06/08/2023]
Abstract
Probiotics are a beneficial treatment for inflammatory bowel disease (IBD). However, studies comparing the effects of similar doses of single and mixed probiotics on IBD are scarce. High mobility group box 1 (HMGB1) is an important proinflammatory mediator involved IBD development. The present study assessed fecal HMGB1 levels in IBD patients and compared the effects of similar doses of Bifidobacterium longum (Bif) versus VSL#3® on HMGB1 levels in 2,4,6-trinitrobenzene sulfonic acid (TNBS)-induced murine colitis. Twenty-four mice were divided into four treatment groups (n = 6 per group): ethanol (control), TNBS, TNBS + Bif, and TNBS + VSL#3®. Bif and VSL#3® (4 × 109 CFU/dose) were administered daily by intragastric gavage, beginning 3 d before TNBS treatment, for a total of 7 d. Fecal HMGB1 levels were higher in both active IBD patients and TNBS-induced colitis mice versus their respective controls. Both Bif and VSL#3® improved intestinal inflammation and fecal microbiota imbalance in TNBS-induced colitis mice. Both treatments also reduced serum and fecal HMGB1 levels as well as increased expression of zonula occludins-1, occludin, and claudin-1 in colon tissues. In Caco-2 cells, HMGB1 reduced transepithelial electrical resistance, zonula occludins-1 protein expression, and increased paracellular permeability of FITC-dextran; the opposite was found with both probiotic treatments. These findings suggest Bif and VSL#3® have similar beneficial effects on TNBS-induced colitis, possibly through inhibition of HMGB1 release and subsequent HMGB1-mediated gut barrier dysfunction. The present study provides novel insights into probiotic treatment of IBD.
Collapse
Affiliation(s)
- Xiaohong Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Fu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lingli Wang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Qian
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang Zheng
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaohua Hou
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
22
|
Jessie Lau LY, Chye FY. Antagonistic effects of Lactobacillus plantarum 0612 on the adhesion of selected foodborne enteropathogens in various colonic environments. Food Control 2018. [DOI: 10.1016/j.foodcont.2018.04.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
23
|
Archer AC, Kurrey NK, Halami PM. In vitro adhesion and anti-inflammatory properties of native Lactobacillus fermentum and Lactobacillus delbrueckii spp. J Appl Microbiol 2018. [PMID: 29537703 DOI: 10.1111/jam.13757] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS This study aimed at characterizing the adhesion and immune-stimulatory properties of native probiotic Lactobacillus fermentum (MCC 2759 and MCC 2760) and Lactobacillus delbrueckii MCC 2775. METHODS AND RESULTS Adhesion of the strains was assessed in Caco-2 and HT-29 cell lines. Expression of adhesion and immune markers were evaluated in Caco-2 cells by real-time qPCR. The cultures displayed >80% of adhesion to both cell lines and also induced the expression of mucin-binding protein (mub) gene in the presence of mucin, bile and pancreatin. Adhesion was mediated by carbohydrate and proteinaceous factors. The cultures stimulated the expression of inflammatory cytokines in Caco-2 cells. However, pro-inflammatory genes were down-regulated upon challenge with lipopolysaccharide and IL-10 was up-regulated by the cultures. Cell wall extract of L. fermentum MCC 2760 induced the expression of IL-6 by 5·47-fold, whereas crude culture filtrate enhanced the expression of IL-10 by 14·87-fold compared to LPS control. CONCLUSIONS The bacterial cultures exhibited strong adhesion and anti-inflammatory properties. SIGNIFICANCE AND IMPACT OF THE STUDY This is the first report to reveal the role of adhesion markers of L. fermentum and L. delbrueckii by qPCR. The strain-specific anti-inflammatory property of native cultures may be useful to alleviate inflammatory conditions and develop a target-based probiotic.
Collapse
Affiliation(s)
- A C Archer
- Microbiology and Fermentation Technology Department, CSIR-Central Food Technological Research Institute, Mysore, Karnataka, India
| | - N K Kurrey
- Department of Biochemistry, CSIR-Central Food Technological Research Institute, Mysore, Karnataka, India
| | - P M Halami
- Microbiology and Fermentation Technology Department, CSIR-Central Food Technological Research Institute, Mysore, Karnataka, India
| |
Collapse
|
24
|
Matsubara VH, Ishikawa KH, Ando-Suguimoto ES, Bueno-Silva B, Nakamae AEM, Mayer MPA. Probiotic Bacteria Alter Pattern-Recognition Receptor Expression and Cytokine Profile in a Human Macrophage Model Challenged with Candida albicans and Lipopolysaccharide. Front Microbiol 2017; 8:2280. [PMID: 29238325 PMCID: PMC5712552 DOI: 10.3389/fmicb.2017.02280] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2017] [Accepted: 11/06/2017] [Indexed: 01/11/2023] Open
Abstract
Probiotics are live microorganisms that confer benefits to the host health. The infection rate of potentially pathogenic organisms such as Candida albicans, the most common agent associated with mucosal candidiasis, can be reduced by probiotics. However, the mechanisms by which the probiotics interfere with the immune system are largely unknown. We evaluated the effect of probiotic bacteria on C. albicans challenged human macrophages. Macrophages were pretreated with lactobacilli alone (Lactobacillus rhamnosus LR32, Lactobacillus casei L324m, or Lactobacillus acidophilus NCFM) or associated with Escherichia coli lipopolysaccharide (LPS), followed by the challenge with C. albicans or LPS in a co-culture assay. The expression of pattern-recognition receptors genes (CLE7A, TLR2, and TLR4) was determined by RT-qPCR, and dectin-1 reduced levels were confirmed by flow cytometry. The cytokine profile was determined by ELISA using the macrophage cell supernatant. Overall probiotic lactobacilli down-regulated the transcription of CLEC7A (p < 0.05), resulting in the decreased expression of dectin-1 on probiotic pretreated macrophages. The tested Lactobacillus species down-regulated TLR4, and increased TLR2 mRNA levels in macrophages challenged with C. albicans. The cytokines profile of macrophages challenged with C. albicans or LPS were altered by the probiotics, which generally led to increased levels of IL-10 and IL-1β, and reduction of IL-12 production by macrophages (p < 0.05). Our data suggest that probiotic lactobacilli impair the recognition of PAMPs by macrophages, and alter the production of pro/anti-inflammatory cytokines, thus modulating inflammation.
Collapse
Affiliation(s)
- Victor H Matsubara
- Dental School, Oral Health Centre of Western Australia, The University of Western Australia, Perth, WA, Australia.,Laboratory of Oral Microbiology, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Department of Prosthodontics, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Karin H Ishikawa
- Laboratory of Oral Microbiology, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Ellen S Ando-Suguimoto
- Laboratory of Oral Microbiology, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Bruno Bueno-Silva
- Laboratory of Oral Microbiology, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.,Dental Division Research, Guarulhos University, Guarulhos, Brazil
| | - Atlas E M Nakamae
- Department of Prosthodontics, School of Dentistry, University of São Paulo, São Paulo, Brazil
| | - Marcia P A Mayer
- Laboratory of Oral Microbiology, Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
25
|
Pradhan B, Guha D, Murmu KC, Sur A, Ray P, Das D, Aich P. Comparative efficacy analysis of anti-microbial peptides, LL-37 and indolicidin upon conjugation with CNT, in human monocytes. J Nanobiotechnology 2017; 15:44. [PMID: 28606090 PMCID: PMC5469186 DOI: 10.1186/s12951-017-0278-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2017] [Accepted: 06/02/2017] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Antimicrobial peptides (AMPs) have the potential to serve as an alternative to antibiotic. AMPs usually exert bactericidal activity via direct killing of microbial pathogens. Reports have proposed that by harnessing innate immune activation, AMPs can regulate pathogen invasion and may control infection. It has been reported that AMPs could be utilized to activate the innate mucosal immune response in order to eliminate pathogenic infections. This way of controlling pathogen infection, by activating host immunity, confers the potential to the select AMPs to alleviate the problem of antibiotic resistance. Among various AMPs tested LL-37 and indolicidin, showed promise to be potential candidates for eliciting enhanced host innate immune responses. LL-37 and indolicidin had exhibited substantial innate immune activation in both human and murine macrophages. Dosage for each of the AMPs, however, was high with adverse side effects. RESULTS In this study, we reported that upon conjugation with carbon nanotubes (CNT), each AMP remained biologically functional at a concentration that was 1000-fold less than the dosage required for free AMP to remain active in the cells. CONCLUSIONS Current study also revealed that while indolicidin induced signalling events mediated through the TNFRSF1A pathway in THP1 cells, followed by activation of NFκB and c-JUN pathways, treatment of cells with LL-37 induced signalling events by activating IL1R, with subsequent activation of NFκB and NFAT2. Thp1 cells, primed with CNT conjugated LL-37 or indolicidin, are protected against Salmonella typhimurium infection at 16 h post challenge.
Collapse
Affiliation(s)
- Biswaranjan Pradhan
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Khurdha, Jatni, Odisha, 752050, India
| | - Dipanjan Guha
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Khurdha, Jatni, Odisha, 752050, India
| | - Krushna Chandra Murmu
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Khurdha, Jatni, Odisha, 752050, India
| | - Abhinav Sur
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Khurdha, Jatni, Odisha, 752050, India
| | - Pratikshya Ray
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Khurdha, Jatni, Odisha, 752050, India
| | - Debashmita Das
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Khurdha, Jatni, Odisha, 752050, India
| | - Palok Aich
- School of Biological Sciences, National Institute of Science Education and Research (NISER), HBNI, P.O. Bhimpur-Padanpur, Khurdha, Jatni, Odisha, 752050, India.
| |
Collapse
|
26
|
Wu Y, Wang Y, Zou H, Wang B, Sun Q, Fu A, Wang Y, Wang Y, Xu X, Li W. Probiotic Bacillus amyloliquefaciens SC06 Induces Autophagy to Protect against Pathogens in Macrophages. Front Microbiol 2017; 8:469. [PMID: 28382029 PMCID: PMC5360707 DOI: 10.3389/fmicb.2017.00469] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2016] [Accepted: 03/07/2017] [Indexed: 01/25/2023] Open
Abstract
Probiotics are increasingly applied in popularity in both humans and animals. Decades of research has revealed their beneficial effects, including the immune modulation in intestinal pathogens inhibition. Autophagy—a cellular process that involves the delivery of cytoplasmic proteins and organelles to the lysosome for degradation and recirculation—is essential to protect cells against bacterial pathogens. However, the mechanism of probiotics-mediated autophagy and its role in the elimination of pathogens are still unknown. Here, we evaluated Bacillus amyloliquefaciens SC06 (Ba)-induced autophagy and its antibacterial activity against Escherichia coli (E. coli) in murine macrophage cell line RAW264.7 cells. Western blotting and confocal laser scanning analysis showed that Ba activated autophagy in a dose- and time-dependent manner. Ba-induced autophagy was found to play a role in the elimination of intracellular bacteria when RAW264.7 cells were challenged with E. coli. Ba induced autophagy by increasing the expression of Beclin1 and Atg5-Atg12-Atg16 complex, but not the AKT/mTOR signaling pathway. Moreover, Ba pretreatment attenuated the activation of JNK in RAW264.7 cells during E. coli infection, further indicating a protective role for probiotics via modulating macrophage immunity. The above findings highlight a novel mechanism underlying the antibacterial activity of probiotics. This study enriches the current knowledge on probiotics-mediated autophagy, and provides a new perspective on the prevention of bacterial infection in intestine, which further the application of probiotics in food products.
Collapse
Affiliation(s)
- Yanping Wu
- Key Laboratory of Molecular Animal Nutrition of Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University Hangzhou, China
| | - Yang Wang
- Key Laboratory of Molecular Animal Nutrition of Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University Hangzhou, China
| | - Hai Zou
- Department of Cardiology, Zhejiang Provincial People's Hospital Hangzhou, China
| | - Baikui Wang
- Key Laboratory of Molecular Animal Nutrition of Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University Hangzhou, China
| | - Qiming Sun
- Department of Biochemistry, School of Medicine, Zhejiang University Hangzhou, China
| | - Aikun Fu
- Key Laboratory of Molecular Animal Nutrition of Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University Hangzhou, China
| | - Yuanyuan Wang
- Key Laboratory of Molecular Animal Nutrition of Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University Hangzhou, China
| | - Yibing Wang
- Key Laboratory of Molecular Animal Nutrition of Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University Hangzhou, China
| | - Xiaogang Xu
- Key Laboratory of Molecular Animal Nutrition of Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University Hangzhou, China
| | - Weifen Li
- Key Laboratory of Molecular Animal Nutrition of Ministry of Education, Institute of Feed Science, College of Animal Sciences, Zhejiang University Hangzhou, China
| |
Collapse
|
27
|
Patel B, Kumar P, Banerjee R, Basu M, Pal A, Samanta M, Das S. Lactobacillus acidophilus attenuates Aeromonas hydrophila induced cytotoxicity in catla thymus macrophages by modulating oxidative stress and inflammation. Mol Immunol 2016; 75:69-83. [PMID: 27262084 DOI: 10.1016/j.molimm.2016.05.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 05/16/2016] [Accepted: 05/16/2016] [Indexed: 01/16/2023]
Abstract
The pathogenesis of Aeromonas hydrophila, a potent fish pathogen, is attributed to its ability to cause motile aeromonad septicaemia leading to apoptosis in a myriad of fish species, including freshwater carp Catla catla. However, the underlying mechanism of antagonistic activity of probiotics against A. hydrophila induced apoptosis is not elucidated due to lack of appropriate in-vitro models. This study reported that the exposure of catla thymus macrophages (CTM) to A. hydrophila markedly induced cellular injuries as evidenced by elevated levels of reactive oxygen species (ROS), reactive nitrogen species (RNS), increased apoptosis, DNA damage and decreased cellular viability. Flow cytometry analysis and Annexin-V/propidium iodide assay further confirmed increased ROS positive cells leading to cell death after infection. The quantitative real-time PCR analysis, also revealed upregulation of inducible nitric-oxide synthase (iNOS), pro-inflammatory cytokine (TNFα), cyclooxygenase2 (COX-2) and downregulation of anti-inflammatory cytokine (IL-10). Pretreatment of cells with probiotic, Lactobacillus acidophilus attenuated A. hydrophila induced apoptosis as evident from the decrease in the levels of ROS, RNS and DNA damage. Significant increase (P≤0.05) in expression of TNFα and IL-10 and decrease in iNOS and COX-2 was observed on probiotic stimulation. In-vivo study using catla fingerlings confirmed similar pattern of ROS, iNOS, NO production and cytokine expression in thymus. This study provides a comprehensive insight into the mechanistic basis of L. acidophilus induced macrophage mediated inflammatory response against A. hydrophila in CTM cells. Further, it speculates the possibility of using cost-effective in-vitro models for screening probiotic candidates of therapeutic potential in aquaculture industry.
Collapse
Affiliation(s)
- Bhakti Patel
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, 769 008, Odisha, India
| | - Premranjan Kumar
- School of Biotechnology, KIIT University, Bhubaneswar, 751024, Odisha, India
| | - Rajanya Banerjee
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, 769 008, Odisha, India
| | - Madhubanti Basu
- Fish Health Management Division, Central Institute of Freshwater Aquaculture, Kausalyanga, Bhubaneswar, 751002, Odisha, India
| | - Arttatrana Pal
- School of Biotechnology, KIIT University, Bhubaneswar, 751024, Odisha, India
| | - Mrinal Samanta
- Fish Health Management Division, Central Institute of Freshwater Aquaculture, Kausalyanga, Bhubaneswar, 751002, Odisha, India
| | - Surajit Das
- Laboratory of Environmental Microbiology and Ecology (LEnME), Department of Life Science, National Institute of Technology, Rourkela, 769 008, Odisha, India.
| |
Collapse
|