1
|
Ma G, Chen Z, Xie Z, Liu J, Xiao X. Mechanisms underlying changes in intestinal permeability during pregnancy and their implications for maternal and infant health. J Reprod Immunol 2025; 168:104423. [PMID: 39793281 DOI: 10.1016/j.jri.2025.104423] [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: 08/31/2024] [Revised: 12/01/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Proper regulation of intestinal permeability is essential for maintaining the integrity of the intestinal mucosal barrier. An abnormal increase in permeability can significantly contribute to the onset and progression of various diseases, including autoimmune disorders, metabolic conditions, allergies, and inflammatory bowel diseases. The potential connection between intestinal permeability and maternal health during pregnancy is increasingly recognized, yet a comprehensive review remains lacking. Pregnancy triggers a series of physiological structural adaptations and significant hormonal fluctuations that collectively contribute to an increase in intestinal permeability. Although an increase in intestinal permeability is typically a normal physiological response during pregnancy, an abnormal rise is associated with immune dysregulation, metabolic disorders, and various pregnancy-related complications, such as recurrent pregnancy loss, gestational diabetes mellitus, overweight and obesity during pregnancy, intrahepatic cholestasis of pregnancy, and preeclampsia. This paper discusses the components of the intestinal mucosal barrier, the concept of intestinal permeability and its measurement methods, and the mechanisms and physiological significance of increased intestinal permeability during pregnancy. It thoroughly explores the association between abnormal intestinal permeability during pregnancy and maternal diseases, aiming to provide evidence for the pathophysiology of disease development in pregnant women. Additionally, the paper examines intervention methods, such as gut microbiota modulation and nutritional interventions, to regulate intestinal permeability during pregnancy, improve immune and metabolic states, and offer feasible strategies for the prevention and adjuvant treatment of clinical pregnancy complications.
Collapse
Affiliation(s)
- Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zhongsheng Chen
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Zhuojun Xie
- General Medicine Department, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - JinXiang Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.
| |
Collapse
|
2
|
Amini-Salehi E, Hassanipour S, Keivanlou MH, Shahdkar M, Orang Goorabzarmakhi M, Vakilpour A, Joukar F, Hashemi M, Sattari N, Javid M, Mansour-Ghanaei F. The impact of gut microbiome-targeted therapy on liver enzymes in patients with nonalcoholic fatty liver disease: an umbrella meta-analysis. Nutr Rev 2024; 82:815-830. [PMID: 37550264 DOI: 10.1093/nutrit/nuad086] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023] Open
Abstract
CONTEXT Nonalcoholic fatty liver disease (NAFLD) is considered the leading cause of chronic liver disease worldwide. To date, no confirmed medication is available for the treatment of NAFLD. Previous studies showed the promising effects of gut microbiome-targeted therapies; however, the results were controversial and the strength of the evidence and their clinical significance remained unclear. OBJECTIVES This umbrella study summarizes the results of meta-analyses investigating the effects of probiotics, prebiotics, and synbiotics on liver enzymes in the NAFLD population. DATA SOURCE A comprehensive search of the PubMed, Scopus, Web of Science, and Cochrane Library databases was done up to December 20, 2022, to find meta-analyses on randomized control trials reporting the effects of gut microbial therapy on patients with NAFLD. DATA EXTRACTION Two independent investigators extracted data on the characteristics of meta-analyses, and any discrepancies were resolved by a third researcher. The AMSTAR2 checklist was used for evaluating the quality of studies. DATA ANALYSIS A final total of 15 studies were included in the analysis. Results showed that microbiome-targeted therapies could significantly reduce levels of alanine aminotransferase (ALT; effect size [ES], -10.21; 95% confidence interval [CI], -13.29, -7.14; P < 0.001), aspartate aminotransferase (AST; ES, -8.86; 95%CI, -11.39, -6.32; P < 0.001), and γ-glutamyltransferase (ES, -5.56; 95%CI, -7.92, -3.31; P < 0.001) in patients with NAFLD. Results of subgroup analysis based on intervention showed probiotics could significantly reduce levels of AST (ES, -8.69; 95%CI, -11.01, -6.37; P < 0.001) and ALT (ES, -9.82; 95%CI, -11.59, -8.05; P < 0.001). Synbiotics could significantly reduce levels of AST (ES, -11.40; 95%CI, -13.91, -8.88; P < 0.001) and ALT (ES, -11.87; 95%CI, -13.80, -9.95; P < 0.001). Prebiotics had no significant effects on AST and ALT levels (ES, -2.96; 95%CI, -8.12, 2.18, P = 0.259; and ES, -4.69; 95%CI, -13.53, 4.15, P = 0.299, respectively). CONCLUSION Gut microbiome-targeted therapies could be a promising therapeutic approach in the improvement of hepatic damage in patients with NAFLD. However, more studies are needed to better determine the best bacterial strains, duration of treatment, and optimum dosage of gut microbiome-targeted therapies in the treatment of the NAFLD population. SYSTEMATIC REVIEW REGISTRATION PROSPERO registration no. CRD42022346998.
Collapse
Affiliation(s)
- Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Soheil Hassanipour
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Milad Shahdkar
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Azin Vakilpour
- School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Farahnaz Joukar
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | - Mohammad Hashemi
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Nazila Sattari
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | - Fariborz Mansour-Ghanaei
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
3
|
Amini-Salehi E, Samethadka Nayak S, Maddineni G, Mahapatro A, Keivanlou MH, Soltani Moghadam S, Vakilpour A, Aleali MS, Joukar F, Hashemi M, Norouzi N, Bakhshi A, Bahrampourian A, Mansour-Ghanaei F, Hassanipour S. Can modulation of gut microbiota affect anthropometric indices in patients with non-alcoholic fatty liver disease? An umbrella meta-analysis of randomized controlled trials. Ann Med Surg (Lond) 2024; 86:2900-2910. [PMID: 38694388 PMCID: PMC11060227 DOI: 10.1097/ms9.0000000000001740] [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: 09/06/2023] [Accepted: 01/08/2024] [Indexed: 05/04/2024] Open
Abstract
Background and aim Modulating the gut microbiota population by administration of probiotics, prebiotics, and synbiotics has shown to have a variety of health benefits in different populations, particularly those with metabolic disorders. Although the promising effects of these compounds have been observed in the management of patients with non-alcoholic fatty liver disease (NAFLD), the exact effects and the mechanisms of action are yet to be understood. In the present study, we aimed to evaluate how gut microbiota modulation affects anthropometric indices of NAFLD patients to achieve a comprehensive summary of current evidence-based knowledge. Methods Two researchers independently searched international databases, including PubMed, Scopus, and Web of Science, from inception to June 2023. Meta-analysis studies that evaluated the effects of probiotics, prebiotics, and synbiotics on patients with NAFLD were entered into our umbrella review. The data regarding anthropometric indices, including body mass index, weight, waist circumference (WC), and waist-to-hip ratio (WHR), were extracted by the investigators. The authors used random effect model for conducting the meta-analysis. Subgroup analysis and sensitivity analysis were also performed. Results A total number of 13 studies were finally included in our study. Based on the final results, BMI was significantly decreased in NAFLD patients by modulation of gut microbiota [effect size (ES): -0.18, 05% CI: -0.25, -0.11, P<0.001]; however, no significant alteration was observed in weight and WC (ES: -1.72, 05% CI: -3.48, 0.03, P=0.055, and ES: -0.24, 05% CI: -0.75, 0.26, P=0.353, respectively). The results of subgroup analysis showed probiotics had the most substantial effect on decreasing BMI (ES: -0.77, 95% CI: -1.16, -0.38, P<0.001) followed by prebiotics (ES: -0.51, 95% CI: -0.76, -0.27, P<0.001) and synbiotics (ES: -0.12, 95% CI: -0.20, -0.04, P=0.001). Conclusion In conclusion, the present umbrella meta-analysis showed that although modulation of gut microbiota by administration of probiotics, prebiotics, and synbiotics had promising effects on BMI, no significant change was observed in the WC and weight of the patients. No sufficient data were available for other anthropometric indices including waist-to-hip ratio and waist-to-height ratio and future meta-analyses should be done in this regard.
Collapse
Affiliation(s)
- Ehsan Amini-Salehi
- Gastrointestinal and Liver Diseases Research Center
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
- Gastrointestinal and Liver Diseases Research Center, Guilan University of Medical Sciences, Rasht, Iran
| | | | | | | | | | | | - Azin Vakilpour
- School of Medicine, Guilan University of Medical Sciences, Rasht
| | | | | | - Mohammad Hashemi
- Student Research Committee, Faculty of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | | | | | | | | | | |
Collapse
|
4
|
Zhu La ALT, Wen Q, Xiao Y, Hu D, Liu D, Guo Y, Hu Y. A New Bacillus velezensis Strain CML532 Improves Chicken Growth Performance and Reduces Intestinal Clostridium perfringens Colonization. Microorganisms 2024; 12:771. [PMID: 38674715 PMCID: PMC11051962 DOI: 10.3390/microorganisms12040771] [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: 02/22/2024] [Revised: 03/29/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Bacillus velezensis has gained increasing recognition as a probiotic for improving animal growth performance and gut health. We identified six B. velezensis strains from sixty Bacillus isolates that were isolated from the cecal samples of fifteen different chicken breeds. We characterized the probiotic properties of these six B. velezensis strains. The effect of a selected strain (B. velezensis CML532) on chicken growth performance under normal feeding and Clostridium perfringens challenge conditions was also evaluated. The results revealed that the six B. velezensis strains differed in their probiotic properties, with strain CML532 exhibiting the highest bile salt and acid tolerance and high-yield enzyme and antibacterial activities. Genomic analyses showed that genes related to amino acid and carbohydrate metabolism, as well as genes related to starch and cellulose hydrolysis, were abundant in strain CML532. Dietary supplementation with strain CML532 promoted chicken growth, improved the gut barrier and absorption function, and modulated the gut microbiota. Under the C. perfringens challenge condition, strain CML532 alleviated intestinal damage, reduced ileal colonization of C. perfringens, and also improved chicken growth performance. Collectively, this study demonstrated that the newly isolated B. velezensis strain is a promising probiotic with beneficial effects on chicken growth performance and gut health.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yongfei Hu
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Beijing 100193, China; (A.L.T.Z.L.); (Q.W.); (Y.X.); (D.H.); (D.L.); (Y.G.)
| |
Collapse
|
5
|
Ajibola OO, Thomas R, Bakare BF. Selected fermented indigenous vegetables and fruits from Malaysia as potential sources of natural probiotics for improving gut health. FOOD SCIENCE AND HUMAN WELLNESS 2023. [DOI: 10.1016/j.fshw.2023.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2023]
|
6
|
Singh TP, Kadyan S, Devi H, Park G, Nagpal R. Gut microbiome as a therapeutic target for liver diseases. Life Sci 2023; 322:121685. [PMID: 37044173 DOI: 10.1016/j.lfs.2023.121685] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/27/2023] [Accepted: 04/05/2023] [Indexed: 04/14/2023]
Abstract
The prominent role of gut in regulating the physiology of different organs in a human body is increasingly acknowledged, to which the bidirectional communication between gut and liver is no exception. Liver health is modulated via different key components of gut-liver axis. The gut-derived products mainly generated from dietary components, microbial metabolites, toxins, or other antigens are sensed and transported to the liver through portal vein to which liver responds by secreting bile acids and antibodies. Therefore, maintaining a healthy gut microbiome can promote homeostasis of this gut-liver axis by regulating the intestinal barrier function and reducing the antigenic molecules. Conversely, liver secretions also regulate the gut microbiome composition. Disturbed homeostasis allows luminal antigens to reach liver leading to impaired liver functioning and instigating liver disorders. The perturbations in gut microbiome, permeability, and bile acid pool have been associated with several liver disorders, although precise mechanisms remain largely unresolved. Herein, we discuss functional fingerprints of a healthy gut-liver axis while contemplating mechanistic understanding of pathophysiology of liver diseases and plausible role of gut dysbiosis in different diseased states of liver. Further, novel therapeutic approaches to prevent the severity of liver disorders are discussed in this review.
Collapse
Affiliation(s)
- Tejinder Pal Singh
- Department of Dairy Microbiology, College of Dairy Science and Technology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125004, India
| | - Saurabh Kadyan
- Department of Nutrition and Integrative Physiology, College of Health and Human Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Harisha Devi
- Department of Dairy Microbiology, College of Dairy Science and Technology, Lala Lajpat Rai University of Veterinary and Animal Sciences, Hisar, Haryana 125004, India
| | - Gwoncheol Park
- Department of Nutrition and Integrative Physiology, College of Health and Human Sciences, Florida State University, Tallahassee, FL 32306, USA
| | - Ravinder Nagpal
- Department of Nutrition and Integrative Physiology, College of Health and Human Sciences, Florida State University, Tallahassee, FL 32306, USA.
| |
Collapse
|
7
|
Saeed NK, Al-Beltagi M, Bediwy AS, El-Sawaf Y, Toema O. Gut microbiota in various childhood disorders: Implication and indications. World J Gastroenterol 2022; 28:1875-1901. [PMID: 35664966 PMCID: PMC9150060 DOI: 10.3748/wjg.v28.i18.1875] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 03/08/2022] [Accepted: 03/27/2022] [Indexed: 02/06/2023] Open
Abstract
Gut microbiota has a significant role in gut development, maturation, and immune system differentiation. It exerts considerable effects on the child's physical and mental development. The gut microbiota composition and structure depend on many host and microbial factors. The host factors include age, genetic pool, general health, dietary factors, medication use, the intestine's pH, peristalsis, and transit time, mucus secretions, mucous immunoglobulin, and tissue oxidation-reduction potentials. The microbial factors include nutrient availability, bacterial cooperation or antagonism, and bacterial adhesion. Each part of the gut has its microbiota due to its specific characteristics. The gut microbiota interacts with different body parts, affecting the pathogenesis of many local and systemic diseases. Dysbiosis is a common finding in many childhood disorders such as autism, failure to thrive, nutritional disorders, coeliac disease, Necrotizing Enterocolitis, helicobacter pylori infection, functional gastrointestinal disorders of childhood, inflammatory bowel diseases, and many other gastrointestinal disorders. Dysbiosis is also observed in allergic conditions like atopic dermatitis, allergic rhinitis, and asthma. Dysbiosis can also impact the development and the progression of immune disorders and cardiac disorders, including heart failure. Probiotic supplements could provide some help in managing these disorders. However, we are still in need of more studies. In this narrative review, we will shed some light on the role of microbiota in the development and management of common childhood disorders.
Collapse
Affiliation(s)
- Nermin Kamal Saeed
- Medical Microbiology Section, Department of Pathology, Salmaniya Medical Complex, Ministry of Health, Manama 12, Bahrain
- Microbiology Section, Department of Pathology, Irish Royal College of Surgeon, Busaiteen 15503, Bahrain
| | - Mohammed Al-Beltagi
- Department of Pediatrics, University Medical Center, Arabian Gulf University, Dr. Sulaiman Al Habib Medical Group, Manama 26671, Bahrain
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| | - Adel Salah Bediwy
- Department of Chest Disease, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
- Department of Pulmonology, University Medical Center, Arabian Gulf University, Dr. Sulaiman Al Habib Medical Group, Manama 26671, Bahrain
| | - Yasser El-Sawaf
- Department of Tropical Medicine, Faculty of Medicine, Tanta University, Tanta 31527, Egypt
- Department of Gastroenterology, University Medical Center, Arabian Gulf University, Dr. Sulaiman Al-Habib Medical Group, Manama 26671, Bahrain
| | - Osama Toema
- Department of Pediatrics, Faculty of Medicine, Tanta University, Tanta 31511, Egypt
| |
Collapse
|
8
|
Ramírez-Rico G, Drago-Serrano ME, León-Sicairos N, de la Garza M. Lactoferrin: A Nutraceutical with Activity against Colorectal Cancer. Front Pharmacol 2022; 13:855852. [PMID: 35264972 PMCID: PMC8899398 DOI: 10.3389/fphar.2022.855852] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 01/31/2022] [Indexed: 12/29/2022] Open
Abstract
Homeostasis in the human body results from the tight regulation of several events, since too little inflammation disrupts the process of tissue repair and remodeling, whereas too much exerts a collateral effect by causing tissue damage with life-threatening consequences. In some clinical conditions, such as inflammatory bowel disease (IBD), inflammation functions as a double-edged sword by either enabling or inhibiting cancer development and progression. Generally, cancer develops through evasion mechanisms that regulate cell growth, causing a high rate of uncontrolled proliferation, and mechanisms for evading cell death, such as apoptosis. Moreover, chronic inflammation is a factor that contributes to colorectal cancer (CRC), as observed in individuals with IBD; all these conditions favor an increased rate of angiogenesis and eventual metastasis. Lactoferrin (Lf) is a mammalian iron-binding multifunctional glycoprotein regarded as a natural compound that up- and downregulates both humoral and cellular components of immunity involved in regulating the inflammatory response and maintaining gut homeostasis. Human and bovine Lf share high sequence homology and have very similar antimicrobial, anti-inflammatory, and immunomodulatory activities. Bovine Lf from milk is considered a safe molecule and is commercially available in large quantities. This review mainly focuses on the regulatory effects of orally administered bovine Lf on the inflammatory response associated with CRC; this approach indicates that CRC is one of the most frequently diagnosed cancers and affects the intestinal tract with high clinical and epidemiologic relevance. Thus, this review may provide foundations for the potential use of bovine Lf alone or as a natural adjunct agent to increase the effectiveness and reduce the side effects of anticancer chemotherapy.
Collapse
Affiliation(s)
- Gerardo Ramírez-Rico
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional (CINVESTAV-IPN), México City, Mexico
- Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), México City, Mexico
| | - Maria Elisa Drago-Serrano
- Departamento de Sistemas Biológicos, Universidad Autónoma Metropolitana Unidad Xochimilco, Mexico City, Mexico
| | - Nidia León-Sicairos
- Centro de Investigación Aplicada a La Salud Pública (CIASaP), Facultad de Medicina, Universidad Autónoma de Sinaloa, Culiacán, Mexico
- Hospital Pediátrico de Sinaloa, Culiacán, Mexico
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados Del Instituto Politécnico Nacional (CINVESTAV-IPN), México City, Mexico
- *Correspondence: Mireya de la Garza,
| |
Collapse
|
9
|
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: 107] [Impact Index Per Article: 26.8] [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
|
10
|
Gong L, Wang B, Zhou Y, Tang L, Zeng Z, Zhang H, Li W. Protective Effects of Lactobacillus plantarum 16 and Paenibacillus polymyxa 10 Against Clostridium perfringens Infection in Broilers. Front Immunol 2021; 11:628374. [PMID: 33679724 PMCID: PMC7930238 DOI: 10.3389/fimmu.2020.628374] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/31/2020] [Indexed: 12/16/2022] Open
Abstract
This study aimed to investigate the protective effects of Lactobacillus plantarum 16 (Lac16) and Paenibacillus polymyxa 10 (BSC10) against Clostridium perfringens (Cp) infection in broilers. A total of 720 one-day-old chicks were randomly divided into four groups. The control and Cp group were only fed a basal diet, while the two treatment groups received basal diets supplemented with Lac16 (1 × 108 cfu·kg-1) and BSC10 (1 × 108 cfu·kg-1) for 21 days, respectively. On day 1 and days 14 to 20, birds except those in the control group were challenged with 1 × 108 cfu C. perfringens type A strain once a day. The results showed that both Lac16 and BSC10 could ameliorate intestinal structure damage caused by C. perfringens infection. C. perfringens infection induced apoptosis by increasing the expression of Bax and p53 and decreasing Bcl-2 expression and inflammation evidence by higher levels of IFN-γ, IL-6, IL-1β, iNOS, and IL-10 in the ileum mucosa, and NO production in jejunal mucosa, which was reversed by Lac16 and BSC10 treatment except for IL-1β (P < 0.05). Besides, the two probiotics restored the intestinal microbiota imbalance induced by C. perfringens infection, characterized by the reduced Firmicutes and Proteobacteria and the increased Bacteroidetes at the phyla level and decreased Bacteroides fragilis and Gallibacterium anatis at the genus level. The two probiotics also reversed metabolic pathways of the microbiota in C. perfringens-infected broilers, including B-vitamin biosynthesis, peptidoglycan biosynthesis, and pyruvate fermentation to acetate and lactate II pathway. In conclusion, Lac16 and BSC10 can effectively protect broilers against C. perfringens infection through improved composition and metabolic pathways of the intestinal microbiota, intestinal structure, inflammation, and anti-apoptosis.
Collapse
Affiliation(s)
- Li Gong
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Institute of Feed Science, Zhejiang University, Hangzhou, China.,School of Life Science and Engineering, Foshan University, Foshan, China
| | - Baikui Wang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Yuanhao Zhou
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Li Tang
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Zihan Zeng
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Institute of Feed Science, Zhejiang University, Hangzhou, China
| | - Huihua Zhang
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Weifen Li
- Key Laboratory of Molecular Animal Nutrition of the Ministry of Education, College of Animal Sciences, Institute of Feed Science, Zhejiang University, Hangzhou, China
| |
Collapse
|
11
|
Yang X, Geng J, Meng H. Glucocorticoid receptor modulates dendritic cell function in ulcerative colitis. Histol Histopathol 2020; 35:1379-1389. [PMID: 32706033 DOI: 10.14670/hh-18-241] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Ulcerative colitis (UC) is a serious form of inflammatory bowel disease (IBD) occurring worldwide. Although anti-TNF therapy is found to be effective in over 70% of patients with UC, nearly one-third are still deprived of effective treatment. Because glucocorticoids (GC) can effectively inhibit granulocyte-recruitment into the mucosa, cytokine secretion and T cell activation, they are used widely in the treatment of UC. However, remission is observed in only 55% of the patients after one year of steroid use due to a condition known as steroid response. Additionally, it has been noted that 20%-40% of the patients with UC do not respond to GC treatment. Researchers have revealed that the number of dendritic cells (DCs) in patients with UC tends to increase in the colonic mucosa. Many studies have determined that the removal of peripheral DCs through the adsorption and separation of granulocytes and monocytes could improve tolerance of the intestine to its symbiotic flora. Based on these results, further insights regarding the beneficial effects of Adacolumn apheresis in patients subjected to this treatment could be revealed. GC can effectively inhibit the activation of DCs by reducing the levels of major histocompatibility complex class II (MHC II) molecules, which is critical for controlling the recruitment of granulocytes. Therefore, alternative biological and new individualized therapies based on these approaches need to be evaluated to counter UC. In this review, progress in research associated with the regulatory effect of glucocorticoid receptors on DCs under conditions of UC is discussed, thus providing insights and identifying potential targets which could be employed in the treatment strategies against UC.
Collapse
Affiliation(s)
- Xinxin Yang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Jingshu Geng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin, China.,Department of Pathology, Harbin Medical University, Harbin, China.
| |
Collapse
|
12
|
Mechanisms Underlying Bone Loss Associated with Gut Inflammation. Int J Mol Sci 2019; 20:ijms20246323. [PMID: 31847438 PMCID: PMC6940820 DOI: 10.3390/ijms20246323] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 11/29/2019] [Accepted: 12/10/2019] [Indexed: 12/11/2022] Open
Abstract
Patients with gastrointestinal diseases frequently suffer from skeletal abnormality, characterized by reduced bone mineral density, increased fracture risk, and/or joint inflammation. This pathological process is characterized by altered immune cell activity and elevated inflammatory cytokines in the bone marrow microenvironment due to disrupted gut immune response. Gastrointestinal disease is recognized as an immune malfunction driven by multiple factors, including cytokines and signaling molecules. However, the mechanism by which intestinal inflammation magnified by gut-residing actors stimulates bone loss remains to be elucidated. In this article, we discuss the main risk factors potentially contributing to intestinal disease-associated bone loss, and summarize current animal models, illustrating gut-bone axis to bridge the gap between intestinal inflammation and skeletal disease.
Collapse
|
13
|
Xie X, Lyu J, Hussain T, Li M. Drug Prevention and Control of Ventilator-Associated Pneumonia. Front Pharmacol 2019; 10:298. [PMID: 31001116 PMCID: PMC6455059 DOI: 10.3389/fphar.2019.00298] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 03/11/2019] [Indexed: 01/10/2023] Open
Abstract
Ventilator-associated pneumonia (VAP) is one of the most prevalent and serious complications of mechanical ventilation, which is considered a common nosocomial infection in critically ill patients. There are some great options for the prevention of VAP: (i) minimize ventilator exposure; (ii) intensive oral care; (iii) aspiration of subglottic secretions; (iv) maintain optimal positioning and encourage mobility; and (v) prophylactic probiotics. Furthermore, clinical management of VAP depends on appropriate antimicrobial therapy, which needs to be selected based on individual patient factors, such as previous antibacterial therapy, history of hospitalization or mechanical ventilation, and bacterial pathogens and antibiotic resistance patterns. In fact, antibiotic resistance has exponentially increased over the last decade, and the isolation of a multidrug-resistant (MDR) pathogen has been identified as an independent predictor of inadequate initial antibiotic therapy and which is significantly associated with increased mortality. Multiple attempts were used in the treatment of VAP, such as novel antibacterial agents, inhaled antibiotics and monoclonal antibodies. In this review, we summarize the current therapeutic options for the prevention and treatment of VAP, aiming to better management of VAP in clinical practice.
Collapse
Affiliation(s)
- Xinming Xie
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jun Lyu
- Clinical Research Center, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Tafseel Hussain
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Manxiang Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
14
|
Slawinska A, Dunislawska A, Plowiec A, Radomska M, Lachmanska J, Siwek M, Tavaniello S, Maiorano G. Modulation of microbial communities and mucosal gene expression in chicken intestines after galactooligosaccharides delivery In Ovo. PLoS One 2019; 14:e0212318. [PMID: 30811518 PMCID: PMC6392319 DOI: 10.1371/journal.pone.0212318] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Accepted: 01/31/2019] [Indexed: 12/21/2022] Open
Abstract
Intestinal mucosa is the interface between the microbial content of the gut and the host's milieu. The goal of this study was to modulate chicken intestinal microflora by in ovo stimulation with galactooligosaccharides (GOS) prebiotic and to demonstrate the molecular responses of the host. The animal trial was performed on meat-type chickens (Ross 308). GOS was delivered by in ovo injection performed into the air cell on day 12 of egg incubation. Analysis of microbial communities and mucosal gene expression was performed at slaughter (day 42 post-hatching). Chyme (for DNA isolation) and intestinal mucosa (for RNA isolation) from four distinct intestinal segments (duodenum, jejunum, ileum, and caecum) was sampled. The relative abundance of Bifidobacterium spp. and Lactobacillus spp. in DNA isolated from chyme samples was determined using qPCR. On the host side, the mRNA expression of 13 genes grouped into two panels was analysed with RT-qPCR. Panel (1) included genes related to intestinal innate immune responses (IL-1β, IL-10 and IL-12p40, AvBD1 and CATHL2). Panel (2) contained genes involved in intestinal barrier function (MUC6, CLDN1 and TJAP1) and nutrients sensing (FFAR2 and FFAR4, GLUT1, GLUT2 and GLUT5). GOS increased the relative abundance of Bifidobacterium in caecum (from 1.3% to 3.9%). Distinct effects of GOS on gene expression were manifested in jejunum and caecum. Cytokine genes (IL-1β, IL-10 and IL-12p40) were up-regulated in the jejunum and caecum of the GOS-treated group. Host defence peptides (AvBD1 and CATHL2) were up-regulated in the caecum of the GOS-treated group. Free fatty acid receptors (FFAR2 and FFAR4) were up-regulated in all three compartments of the intestine (except the duodenum). Glucose transporters were down-regulated in duodenum (GLUT2 and GLUT5) but up-regulated in the hindgut (GLUT1 and GLUT2). In conclusion, GOS delivered in ovo had a bifidogenic effect in adult chickens. It also modulated gene expression related to intestinal immune responses, gut barrier function, and nutrient sensing.
Collapse
Affiliation(s)
- Anna Slawinska
- Department of Animal Biotechnology and Genetics, UTP University of Science and Technology, Bydgoszcz, Poland
- Department of Agricultural, Environmental and Food Sciences, University of Molise, Campobasso, Italy
| | - Aleksandra Dunislawska
- Department of Animal Biotechnology and Genetics, UTP University of Science and Technology, Bydgoszcz, Poland
| | - Arkadiusz Plowiec
- Department of Animal Biotechnology and Genetics, UTP University of Science and Technology, Bydgoszcz, Poland
| | - Malgorzata Radomska
- Department of Animal Biotechnology and Genetics, UTP University of Science and Technology, Bydgoszcz, Poland
| | - Jagoda Lachmanska
- Department of Animal Biotechnology and Genetics, UTP University of Science and Technology, Bydgoszcz, Poland
| | - Maria Siwek
- Department of Animal Biotechnology and Genetics, UTP University of Science and Technology, Bydgoszcz, Poland
| | - Siria Tavaniello
- Department of Agricultural, Environmental and Food Sciences, University of Molise, Campobasso, Italy
| | - Giuseppe Maiorano
- Department of Agricultural, Environmental and Food Sciences, University of Molise, Campobasso, Italy
| |
Collapse
|
15
|
Burger E, Araujo A, López-Yglesias A, Rajala MW, Geng L, Levine B, Hooper LV, Burstein E, Yarovinsky F. Loss of Paneth Cell Autophagy Causes Acute Susceptibility to Toxoplasma gondii-Mediated Inflammation. Cell Host Microbe 2018; 23:177-190.e4. [PMID: 29358083 PMCID: PMC6179445 DOI: 10.1016/j.chom.2018.01.001] [Citation(s) in RCA: 89] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2017] [Revised: 12/04/2017] [Accepted: 01/02/2018] [Indexed: 02/07/2023]
Abstract
The protozoan parasite Toxoplasma gondii triggers severe small intestinal immunopathology characterized by IFN-γ- and intestinal microbiota-mediated inflammation, Paneth cell loss, and bacterial dysbiosis. Paneth cells are a prominent secretory epithelial cell type that resides at the base of intestinal crypts and releases antimicrobial peptides. We demonstrate that the microbiota triggers basal Paneth cell-specific autophagy via induction of IFN-γ, a known trigger of autophagy, to maintain intestinal homeostasis. Deletion of the autophagy protein Atg5 specifically in Paneth cells results in exaggerated intestinal inflammation characterized by complete destruction of the intestinal crypts resembling that seen in pan-epithelial Atg5-deficient mice. Additionally, lack of functional autophagy in Paneth cells within intestinal organoids and T. gondii-infected mice causes increased sensitivity to the proinflammatory cytokine TNF along with increased intestinal permeability, leading to exaggerated microbiota- and IFN-γ-dependent intestinal immunopathology. Thus, Atg5 expression in Paneth cells is essential for tissue protection against cytokine-mediated immunopathology during acute gastrointestinal infection.
Collapse
Affiliation(s)
- Elise Burger
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Alessandra Araujo
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Américo López-Yglesias
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Michael W Rajala
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
| | - Linda Geng
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Beth Levine
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Center for Autophagy Research, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lora V Hooper
- Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Immunology, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ezra Burstein
- Department of Internal Medicine, The University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Felix Yarovinsky
- Center for Vaccine Biology and Immunology, Department of Microbiology and Immunology, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
16
|
Karacaer F, Hamed I, Özogul F, Glew RH, Özcengiz D. The function of probiotics on the treatment of ventilator-associated pneumonia (VAP): facts and gaps. J Med Microbiol 2017; 66:1275-1285. [PMID: 28855004 DOI: 10.1099/jmm.0.000579] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Probiotics have been used for centuries in making fermented dairy products. The health benefits related to probiotics consumption are well recognized and they are generally regarded as safe (GRAS). Their therapeutic effects are due to the production of a variety of antimicrobial compounds, such as short-chain fatty acids, organic acids (such as lactic, acetic, formic, propionic and butyric acids), ethanol, hydrogen peroxide and bacteriocins. Ventilator-associated pneumonia (VAP) is a nosocomial infection associated with high mortality in intensive care units. VAP can result from endotracheal intubation and mechanical ventilation. These interventions increase the risk of infection as patients lose the natural barrier between the oropharynx and the trachea, which in turn facilitates the entry of pathogens through the aspiration of oropharyngeal secretions containing bacteria into the lung. In order to prevent this, probiotics have been used extensively against VAP. This review is an update containing information extracted from recent studies on the use of probiotics to treat VAP. In addition, probiotic safety, the therapeutic properties of probiotics, the probiotic strains used and the action of the probiotics mechanism are reviewed. Furthermore, the therapeutic effects of probiotic treatment procedures for VAP are compared to those of antibiotics. Finally, the influences of bacteriocin on the growth of human pathogens, and the side-effects and limitations of using probiotics for the treatment of VAP are addressed.
Collapse
Affiliation(s)
- Feride Karacaer
- Department of Anaesthesiology and Reanimation, School of Medicine, Cukurova University, Adana, Turkey
| | - Imen Hamed
- Biotechnology Research and Application Centre, Cukurova University, Adana, Turkey
| | - Fatih Özogul
- Department of Seafood Processing Technology, Faculty of Fisheries, Cukurova University, 01330, Adana, Turkey
| | - Robert H Glew
- Department of Surgery, School of Medicine, University of New Mexico, Albuquerque, NM, USA
| | - Dilek Özcengiz
- Department of Anaesthesiology and Reanimation, School of Medicine, Cukurova University, Adana, Turkey
| |
Collapse
|
17
|
Fermented milk with probiotic Lactobacillus rhamnosus S1K3 (MTCC5957) protects mice from salmonella by enhancing immune and nonimmune protection mechanisms at intestinal mucosal level. J Nutr Biochem 2016; 30:62-73. [DOI: 10.1016/j.jnutbio.2015.11.018] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2015] [Revised: 10/26/2015] [Accepted: 11/23/2015] [Indexed: 12/20/2022]
|
18
|
Microbiota and gastrointestinal diseases. An Pediatr (Barc) 2015. [DOI: 10.1016/j.anpede.2015.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
|
19
|
Polanco Allué I. [Microbiota and gastrointestinal diseases]. An Pediatr (Barc) 2015; 83:443.e1-5. [PMID: 26534880 DOI: 10.1016/j.anpedi.2015.07.034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Accepted: 07/29/2015] [Indexed: 02/08/2023] Open
Abstract
The bacterial colonisation is established immediately after birth, through direct contact with maternal microbiota, and may be influenced during lactation. There is emerging evidence indicating that quantitative and qualitative changes on gut microbiota contribute to alterations in the mucosal activation of the immune system, leading to intra- or extra-intestinal diseases. A balance between pathogenic and beneficial microbiota throughout childhood and adolescence is important to gastrointestinal health, including protection against pathogens, inhibition of pathogens, nutrient processing (synthesis of vitamin K), stimulation of angiogenesis, and regulation of host fat storage. Probiotics can promote an intentional modulation of intestinal microbiota favouring the health of the host. A review is presented on the modulation of intestinal microbiota on prevention, and adjuvant treatment of some paediatric gastrointestinal diseases.
Collapse
Affiliation(s)
- I Polanco Allué
- Departamento de Pediatría, Facultad de Medicina, Universidad Autónoma, Madrid, España.
| |
Collapse
|
20
|
Vijaya Kumar B, Vijayendra SVN, Reddy OVS. Trends in dairy and non-dairy probiotic products - a review. JOURNAL OF FOOD SCIENCE AND TECHNOLOGY 2015; 52:6112-24. [PMID: 26396359 PMCID: PMC4573104 DOI: 10.1007/s13197-015-1795-2] [Citation(s) in RCA: 193] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Revised: 02/18/2015] [Accepted: 03/02/2015] [Indexed: 02/07/2023]
Abstract
Health awareness has grown to a greater extent among consumers and they are looking for healthy probiotic counterparts. Keeping in this view, the present review focuses recent developments in dairy and non-dairy probiotic products. All over the world, dairy probiotics are being commercialized in many different forms. However, the allergy and lactose intolerance are the major set-backs to dairy probiotics. Whereas, flavor and refreshing nature are the major advantages of non-dairy drinks, especially fruit juices. Phenotypic and genotypic similarities between dairy and non-dairy probiotics along with the matrix dependency of cell viability and cell functionality are reviewed. The heterogeneous food matrices of non-dairy food carriers are the major constraints for the survival of the probiotics, while the probiotic strains from non-dairy sources are satisfactory. Technological and functional properties, besides the viability of the probiotics used in fermented products of non-dairy origin are extremely important to get a competitive advantage in the world market. The functional attributes of dairy and non-dairy probiotic products are further enhanced by adding prebiotics such as galacto-oligosaccharide, fructo-oligosaccharide and inulin.
Collapse
Affiliation(s)
- Bathal Vijaya Kumar
- />Department of Biotechnology, Sri Venkateswara University, Tirupati, 517 502 India
| | | | | |
Collapse
|
21
|
Abstract
Most of what is known about the pathogenesis of inflammatory bowel disease (IBD) pertains to complex interplay between host genetics, immunity, and environmental factors. Epigenetic modifications play pivotal roles in intestinal immunity and mucosal homeostasis as well as mediating gene-environment interactions. In this article, we provide a historical account of epigenetic research either directly related or pertinent to the pathogenesis and management of IBD. We further collate emerging evidence supporting roles for epigenetic mechanisms in relevant aspects of IBD biology, including deregulated immunity, host-pathogen recognition and mucosal integrity. Finally, we highlight key epigenetic mechanisms that link chronic inflammation to specific IBD comorbidities, including colitis-associated cancer and discuss their potential utility as novel biomarkers or pharmacologic targets in IBD therapy.
Collapse
|
22
|
Kemgang TS, Kapila S, Shanmugam VP, Kapila R. Cross-talk between probiotic lactobacilli and host immune system. J Appl Microbiol 2014; 117:303-19. [PMID: 24738909 DOI: 10.1111/jam.12521] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2014] [Revised: 04/02/2014] [Accepted: 04/08/2014] [Indexed: 12/14/2022]
Abstract
The mechanism by which probiotic lactobacilli affect the immune system is strain specific. As the immune system is a multicompartmental system, each strain has its way to interact with it and induce a visible and quantifiable effect. This review summarizes the interplay existing between the host immune system and probiotic lactobacilli, that is, with emphasis on lactobacilli as a prototype probiotic genus. Several aspects including the bacterial-host cross-talk with the mucosal and systemic immune system are presented, as well as short sections on the competing effect towards pathogenic bacteria and their uses as delivery vehicle for antigens.
Collapse
Affiliation(s)
- T S Kemgang
- Department of Animal Biochemistry, National Dairy Research Institute, Karnal, Haryana, India; Department of Food Science/Nutrition, National School of AgroIndustrial Sciences, University of Ngaoundere, Ngaoundere, Adamaoua, Cameroon
| | | | | | | |
Collapse
|
23
|
Däbritz J. Granulocyte macrophage colony-stimulating factor and the intestinal innate immune cell homeostasis in Crohn's disease. Am J Physiol Gastrointest Liver Physiol 2014; 306:G455-65. [PMID: 24503766 DOI: 10.1152/ajpgi.00409.2013] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Current literature consolidates the view of Crohn's disease (CD) as a form of immunodeficiency highlighting dysregulation of intestinal innate immunity in the pathogenesis of CD. Intestinal macrophages derived from blood monocytes play a key role in sustaining the innate immune homeostasis in the intestine, suggesting that the monocyte/macrophage compartment might be an attractive therapeutic target for the management of CD. Granulocyte macrophage colony-stimulating factor (GM-CSF) is a hematopoietic growth factor that also promotes myeloid cell activation, proliferation, and differentiation. GM-CSF has a protective effect in human CD and mouse models of colitis. However, the role of GM-CSF in immune and inflammatory reactions in the intestine is not well defined. Beneficial effects exerted by GM-CSF during intestinal inflammation could relate to modulation of the mucosal barrier function in the intestine, including epithelial cell proliferation, survival, restitution, and immunomodulatory actions. The aim of this review is to summarize potential mechanistic roles of GM-CSF in intestinal innate immune cell homeostasis and to highlight its central role in maintenance of the intestinal immune barrier in the context of immunodeficiency in CD.
Collapse
Affiliation(s)
- Jan Däbritz
- The Royal Children's Hospital Melbourne, Murdoch Children's Research Institute, Gastrointestinal Research in Inflammation & Pathology, Parkville, Victoria, Australia; University of Melbourne, Melbourne Medical School, Department of Paediatrics, Parkville, Victoria, Australia; University Children's Hospital Münster, Department of Pediatric Rheumatology and Immunology, Münster, Germany; and University of Münster, Interdisciplinary Center for Clinical Research, Münster, Germany
| |
Collapse
|
24
|
Pierre JF, Heneghan AF, Lawson CM, Wischmeyer PE, Kozar RA, Kudsk KA. Pharmaconutrition Review. JPEN J Parenter Enteral Nutr 2013; 37:51S-65S. [DOI: 10.1177/0148607113493326] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Joseph F. Pierre
- Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison
| | - Aaron F. Heneghan
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison
| | - Christy M. Lawson
- Department of Surgery, University of Tennessee Medical Center, Knoxville
| | | | - Rosemary A. Kozar
- Department of Surgery, University of Texas–Houston Health Science Center, Houston
| | - Kenneth A. Kudsk
- Veterans Administration Surgical Services, William S. Middleton Memorial Veterans Hospital, Madison, Wisconsin
- Department of Surgery, School of Medicine and Public Health, University of Wisconsin, Madison
| |
Collapse
|
25
|
Nikolova M, Ambrozova G, Kratchanova M, Denev P, Kussovski V, Ciz M, Lojek A. Effects of pectic polysaccharides isolated from leek on the production of reactive oxygen and nitrogen species by phagocytes. J Med Food 2013; 16:711-8. [PMID: 23905651 DOI: 10.1089/jmf.2012.0234] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
The current survey investigates the effect of four polysaccharides isolated from fresh leek or alcohol insoluble substances (AIS) of leek on the production of reactive oxygen species (ROS) and reactive nitrogen species (RNS) from phagocytes. The ability of the polysaccharides to activate serum complement was also investigated. Despite the lack of antioxidant activity, the pectic polysaccharides significantly decreased the production of ROS by human neutrophils. Polysaccharides isolated from AIS markedly activated RAW 264.7 macrophages for RNS production in a concentration-dependent manner. The Western blot analysis revealed that this effect was due to the stimulation of the inducible nitric oxide synthase protein expression of macrophages. The polysaccharides extracted from AIS with water showed the ability to fix serum complement, especially through the alternative pathway. It was found that the polysaccharide that has the highest complement-fixing effect is characterized by the highest content of uronic acids and the highest molecular weight.
Collapse
Affiliation(s)
- Mariana Nikolova
- Center of Phytochemistry, Institute of Organic Chemistry, Bulgarian Academy of Sciences, Plovdiv, Bulgaria
| | | | | | | | | | | | | |
Collapse
|
26
|
Theodorakopoulou M, Perros E, Giamarellos-Bourboulis EJ, Dimopoulos G. Controversies in the management of the critically ill: the role of probiotics. Int J Antimicrob Agents 2013; 42 Suppl:S41-4. [PMID: 23664676 DOI: 10.1016/j.ijantimicag.2013.04.010] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Probiotics are commercially available, viable, non-pathogenic micro-organisms that, when ingested in sufficient quantities, exert a health benefit to the host derived through modification of the gut flora, local release of antimicrobial factors, maintenance of integrity of the gut barrier, competition for epithelial adherence, prevention of bacterial translocation, and modulation of the local immune response. In critically ill patients, probiotics appear to lead to decreased susceptibility to antibiotic-associated diarrhoea, Clostridium difficile infections, ventilator-associated pneumonia, necrotising enterocolitis, acute severe pancreatitis, sepsis and multiple organ dysfunction syndrome as well as a shortened duration of infections. Current scientific evidence supporting the use of probiotics is not conclusive and is mainly derived from single-centre, not very well designed trials that are limited by many factors including small sample sizes, heterogeneity in the probiotic strains used, effectiveness of the combined strains, optimum dose regimens, frequency and duration of administration, and certainly incomplete knowledge of the mechanism of action of each strain. Probiotics appear to be well tolerated, whilst adverse events are very rare. The most commonly reported adverse events include bacteraemia, fungaemia and sepsis. At present, based on the available evidence and although helpful and relatively safe for certain disease conditions, routine use of probiotics in the critically ill is not recommended.
Collapse
|
27
|
Dietary n-3 polyunsaturated fatty acids (PUFA) decrease obesity-associated Th17 cell-mediated inflammation during colitis. PLoS One 2012; 7:e49739. [PMID: 23166761 PMCID: PMC3500317 DOI: 10.1371/journal.pone.0049739] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Accepted: 10/11/2012] [Indexed: 01/04/2023] Open
Abstract
Clinical and experimental evidence suggests that obesity-associated inflammation increases disease activity during colitis, attributed in part to the effects of Th17 cells. Using a model of concurrent obesity and colitis, we monitored changes in critical immune cell subsets and inflammatory biomarker expression in three key tissues: visceral adipose tissue, colon (local inflammatory site) and spleen (systemic inflammatory site), and we hypothesized that n-3 PUFA would reduce the percentage of inflammatory immune cell subsets and suppress inflammatory gene expression, thereby improving the disease phenotype. Obesity was induced in C57BL/6 mice by feeding a high fat (HF) diet (59.2% kcal) alone or an isocaloric HF diet supplemented with fish oil (HF-FO) for 12 weeks. Colitis was induced via a 2.5% trinitrobenzene sulfonic acid (TNBS) enema. The HF-FO diet improved the obese phenotype by reducing i) serum hormone concentrations (leptin and resistin), ii) adipose tissue mRNA expression of inflammatory cytokines (MCP-1, IFNγ, IL-6, IL17F and IL-21) and iii) total (F4/80⁺ CD11b⁺) and inflammatory adipose tissue M1 (F4/80⁺ CD11c⁺) macrophage content compared to HF (P<0.05). In addition, the HF-FO diet reduced both colitis-associated disease severity and colonic mRNA expression of the Th17 cell master transcription factor (RORγτ) and critical cytokines (IL-6, IL-17A, IL-17F, IL-21, IL-23 and IFNγ) versus HF (P<0.05). Compared to HF, the percentage of both splenic Th17 and Th1 cells were reduced by the HF-FO group (P<0.05). Under ex vivo polarizing conditions, the percentage of HF-FO derived CD4⁺ T cells that reached Th17 cell effector status was suppressed (P = 0.05). Collectively, these results indicate that n-3 PUFA suppress Th1/Th17 cells and inflammatory macrophage subsets and reconfigure the inflammatory gene expression profile in diverse tissue sites in obese mice following the induction of colitis.
Collapse
|
28
|
Abstract
The bacterial colonization is defined immediately after birth, through direct contact with maternal microbiota and may be influenced during lactation. There is emerging evidence indicating that quantitative and qualitative changes on gut microbiota contribute to alterations in the mucosal activation of immune system leading to intra- or extra-intestinal diseases. A balance between pathogenic and beneficial microbiota throughout childhood and adolescence is important to gastrointestinal health, including protection against pathogens, inhibition of pathogens, nutrient processing (synthesis of vitamin K), stimulation of angiogenesis, and regulation of host fat storage. Probiotics can promote an intentional modulation of intestinal microbiota favoring the health of the host. This paper is a review about modulation of intestinal microbiota on prevention and adjuvant treatment of pediatric gastrointestinal diseases.
Collapse
|
29
|
Bamias G, Corridoni D, Pizarro TT, Cominelli F. New insights into the dichotomous role of innate cytokines in gut homeostasis and inflammation. Cytokine 2012; 59:451-459. [PMID: 22795953 PMCID: PMC3652608 DOI: 10.1016/j.cyto.2012.06.014] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Accepted: 06/04/2012] [Indexed: 02/08/2023]
Abstract
In addition to their well-known role in acute injury and chronic inflammation, "innate" cytokines play an important role in health and the maintenance of normal immune homeostasis. This group includes the prototypic cytokines IL-1 and TNFα, as well as several other members belonging to the IL-1 and TNF family, such as IL-18, IL-33, IL-36-38, and TL1A. The dichotomous role of these cytokines has been best characterized in the intestine where innate cytokines may play both a protective and a pro-inflammatory role, depending upon the immmunological status of the host or the type and phase of the inflammatory process. This new information has produced novel pathogenetic hypotheses that have important translational implications both in regard to the prevention and treatment of chronic intestinal inflammation, including Crohn's disease and ulcerative colitis, the two major forms of inflammatory bowel disease. This review will discuss and summarize current data regarding the role of IL-1, TNFα, and their family members in regulating gut mucosal homeostasis and chronic intestinal inflammation.
Collapse
Affiliation(s)
- Giorgos Bamias
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44122, USA
- GI Division-1 Department of Propaedeutic and Internal Medicine, “Laikon” General Hospital, Athens University Medical School, Athens 11527, Greece
| | - Daniele Corridoni
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44122, USA
- Department of Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| | - Theresa T. Pizarro
- Department of Pathology, Case Western Reserve University, Cleveland, OH 44122, USA
- Department of Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44122, USA
- Department of Digestive Health Research Center, Case Western Reserve University, Cleveland, OH 44122, USA
| |
Collapse
|
30
|
Effects of alanyl-glutamine dipeptide on the expression of colon-inflammatory mediators during the recovery phase of colitis induced by dextran sulfate sodium. Eur J Nutr 2012; 52:1089-98. [PMID: 22847641 DOI: 10.1007/s00394-012-0416-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Accepted: 06/27/2012] [Indexed: 12/13/2022]
Abstract
PURPOSE Glutamine (Gln) is a nutrient with immunomodulatory effects in metabolic stressed conditions. This study investigated the effects of Gln on colonic-inflammatory-mediator expression and mucosal repair in mice with dextran sulfate sodium (DSS)-induced colitis. METHODS C57BL/6 mice received distilled water containing 3 % DSS for 5 d to induce colitis. One of the DSS-treated groups was intraperitoneally injected with an alanyl (Ala)-Gln solution 3 days before (G-DSS) while the other group was administered Ala-Gln 3 days after colitis (DSS-G) was induced. The Ala-Gln solution provided 0.5 g Gln/kg/d. The saline-DSS group (S-DSS) received an identical amount of saline before and after colitis was induced to serve as a positive control. RESULTS The S-DSS group had a shorter colon length, higher plasma haptoglobin level, and more-severe colon inflammation. Also, the toll-like receptor (TLR)4 level, nuclear factor (NF)-κB activation, and inflammatory cytokine gene expression in the colon were higher than those of the normal control group. Gln administration either before or after colitis suppressed TLR4 protein levels, decreased plasma haptoglobin, and reduced colon inflammation. Histological inflammatory scores were also lowered. Compared to the post-colitis Gln group, preventive use of Gln had higher colon length, expressions of mucin 2, trefoil factor 3, and heat shock protein 72 genes were also upregulated in the colon. CONCLUSIONS These results suggest that Gln administered either before or after the colitis mitigated inflammation of colitis that was not observed in group without Gln injection. Prophylactic treatment with Gln had more-beneficial effects on reducing inflammatory markers and enhancing the recovery of mucosa in DSS-induced colitis.
Collapse
|
31
|
Abstract
Probiotics are living microorganisms that, when ingested in adequate amounts, provide benefits to the host. The benefits include either a shortened duration of infections or decreased susceptibility to pathogens. Proposed mechanisms of beneficial effects include improving gastrointestinal barrier function, modification of the gut flora by inducing host cell antimicrobial peptides and/or local release of probiotic antimicrobial factors, competition for epithelial adherence, and immunomodulation. With increasing intensive care unit (ICU) antibacterial resistance rates and fewer new antibiotics in the research pipeline, focus has been shifted to non-antibiotic approaches for the prevention and treatment of nosocomial infections. Probiotics offer promise to ICU patients for the prevention of antibiotic-associated diarrhea, Clostridium difficile infections, multiple organ dysfunction syndrome, and ventilator-associated pneumonia. Our current understanding of probiotics is confounded by inconsistency in probiotic strains studied, optimal dosages, study durations, and suboptimal sample sizes. Although probiotics are generally safe in the critically ill, adverse event monitoring must be rigorous in these vulnerable patients. Delineation of clinical differences of various effective probiotic strains, their mechanisms of action, and optimal dosing regimens will better establish the role of probiotics in various disorders. However, probiotic research will likely be hindered in the future given a recent ruling by the U.S. Food and Drug Administration.
Collapse
Affiliation(s)
- Lee E Morrow
- Division of Pulmonary and Critical Care Medicine, Creighton University Medical Center, Omaha, NE 68131, USA.
| | | | | |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW To summarize the existing data regarding the use of probiotics, prebiotics, and synbiotics in select disorders encountered in the intensive care unit setting. RECENT FINDINGS Recent systematic reviews and meta-analyses have more rigorously aggregated the fragmented primary data which suffers from multiple limitations. SUMMARY Probiotics are living microorganisms which, when ingested in adequate amounts, provide health benefits to the host. The mechanisms of these benefits include improved gastrointestinal barrier function, modification of the gut flora by inducing host cell antimicrobial peptides, releasing probiotic antimicrobial factors, competing for epithelial adherence, and immunomodulation to the advantage of the host. In the intensive care unit, probiotics appear to provide benefits in antibiotic-associated diarrhea, ventilator-associated pneumonia, and necrotizing enterocolitis. With increasing rates of antibiotic resistance among common nosocomial pathogens and fewer new antibiotics in the research pipeline, increasing attention has been placed on nonantibiotic approaches to the prevention and treatment of nosocomial infections. Existing studies of probiotics in critically ill patients are limited by heterogeneity in probiotic strains, dosages, duration of administration, and small sample sizes. Although probiotics are generally well tolerated and adverse events are very rare, the results of the PROPATRIA (Probiotics Prophylaxis in Patients with Predicted Severe Acute Pancreatitis) trial highlight the need for meticulous attention to safety monitoring. Better identification of the ideal characteristics of effective probiotics coupled with improved understanding of mechanisms of action will help to delineate the true beneficial effects of probiotics in various disorders.
Collapse
|
33
|
Abstract
PURPOSE OF REVIEW To summarize novel insights into the immunological mechanisms of sublingual immunotherapy (SLIT). Within the recent decades, several alternative noninvasive allergen application strategies have been investigated in allergen-specific immunotherapy (AIT), of which intra-oral allergen application to sublingual mucosa has been proven to be well tolerated and effective. RECENT FINDINGS To date, SLIT is widely accepted by most allergists as an alternative option to conventional subcutaneous immunotherapy (SCIT). Although detailed immunological mechanisms remain to be elucidated, much scientific effort has been made to shed some light on local and systemic immunological responses to SLIT in mice as well as humans. Only a few studies focused on the detailed mechanisms following allergen application to the oral mucosa as part of the sophisticated mucosal immunological network. Within this network, the pro-tolerogenic properties of local antigen-presenting cells (APCs) such as dendritic cells - which are able to enforce tolerogenic mechanisms and to induce T-cell immune responses - play a central role. Further on, basic research focused not only on the immune response in nasal and bronchial mucosa but also on the systemic T-cell immune response. SUMMARY Thus, much exiting data have been published providing a better understanding of immunological features of SLIT but far more investigations are necessary to uncover further exciting details on the key mechanisms of SLIT.
Collapse
|
34
|
Sánchez-Miralles A, Castellanos G, Badenes R, Conejero R. [Abdominal compartment syndrome and acute intestinal distress syndrome]. Med Intensiva 2012; 37:99-109. [PMID: 22244213 DOI: 10.1016/j.medin.2011.11.019] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2011] [Revised: 11/18/2011] [Accepted: 11/22/2011] [Indexed: 12/19/2022]
Abstract
Seriously ill patients frequently present intra-abdominal hypertension (IAH) and abdominal compartment syndrome (ACS) as complications, and the associated mortality is very high. This review offers an update on the most controversial aspects of these entities: factors favoring their appearance, the most common causes, prognosis, and methods of measuring intra-abdominal pressure (IAP), physiopathological consequences in relation to the different organs and systems, and the currently accepted treatment measures (medical and/or surgical). Simultaneously to the strictly physical mechanisms of injury, such as direct compression of intra-abdominal organs and vessels, the transmission of IAP to other compartments, and the drop in cardiac output, a series of immune-inflammatory mediators generated in the intestine itself may also intervene. Hypoperfusion, sustained ischemia and the ischemia-reperfusion phenomenon, would act upon the microbiota, intestinal epithelium and intestinal immune system, triggering a systemic inflammatory response and multiorgan dysfunction that appears in the final stages of ACS.
Collapse
Affiliation(s)
- A Sánchez-Miralles
- Servicio de Medicina Intensiva, Hospital Universitario San Juan de Alicante, Alicante, España
| | | | | | | |
Collapse
|
35
|
Amalaradjou MAR, Bhunia AK. Modern approaches in probiotics research to control foodborne pathogens. ADVANCES IN FOOD AND NUTRITION RESEARCH 2012; 67:185-239. [PMID: 23034117 PMCID: PMC7150249 DOI: 10.1016/b978-0-12-394598-3.00005-8] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Foodborne illness is a serious public health concern. There are over 200 known microbial, chemical, and physical agents that are known to cause foodborne illness. Efforts are made for improved detection, control and prevention of foodborne pathogen in food, and pathogen associated diseases in the host. Several commonly used approaches to control foodborne pathogens include antibiotics, natural antimicrobials, bacteriophages, bacteriocins, ionizing radiations, and heat. In addition, probiotics offer a potential intervention strategy for the prevention and control of foodborne infections. This review focuses on the use of probiotics and bioengineered probiotics to control foodborne pathogens, their antimicrobial actions, and their delivery strategies. Although probiotics have been demonstrated to be effective in antagonizing foodborne pathogens, challenges exist in the characterization and elucidation of underlying molecular mechanisms of action and in the development of potential delivery strategies that could maintain the viability and functionality of the probiotic in the target organ.
Collapse
|
36
|
Monk JM, Jia Q, Callaway E, Weeks B, Alaniz RC, McMurray DN, Chapkin RS. Th17 cell accumulation is decreased during chronic experimental colitis by (n-3) PUFA in Fat-1 mice. J Nutr 2012; 142:117-24. [PMID: 22131549 PMCID: PMC3237233 DOI: 10.3945/jn.111.147058] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
During colon inflammation, Th17 cells and immunosuppressive regulatory T cells (Treg) are thought to play promotive and preventative roles, respectively. Dietary (n-3) PUFA favorably modulate intestinal inflammation in part by downregulating T-cell activation and functionality. We used the Fat-1 mouse, a genetic model that synthesizes long-chain (n-3) PUFA de novo, to test the hypothesis that (n-3) PUFA protect against colonic inflammation by modulating the polarization of Treg and Th17 cells during colitis. Male and female wild-type (WT) and Fat-1 mice were administered dextran sodium sulfate (DSS) in the drinking water (2.5%) to induce acute (5 d DSS) or chronic (3 cycles DSS) colitis and the percentage of Treg and Th17 cells residing locally [colonic lamina propria (cLP)] and systemically (spleen) was determined by flow cytometry. The percentage of Treg in either tissue site was unaffected by genotype (P > 0.05); however, during chronic colitis, the percentage of Th17 cells residing in both the spleen and cLP was lower in Fat-1 mice compared to WT mice (P < 0.05). Colonic mucosal mRNA expression of critical Th17 cell cytokines and chemokine receptors (IL-17F, IL-21, and CCR6) were lower, whereas expression of the Th17 cell suppressive cytokine, IL-27, was greater in Fat-1 mice compared to WT mice during chronic colitis (P < 0.05). Moreover, colon histological scores were improved in Fat-1 mice (P < 0.05). Collectively, these results demonstrate for the first time, to our knowledge, that (n-3) PUFA can modulate the colonic mucosal microenvironment to suppress Th17 cell accumulation and inflammatory damage following the induction of chronic colitis.
Collapse
Affiliation(s)
- Jennifer M. Monk
- Program in Integrative Nutrition and Complex Diseases,Intercollegiate Faculty of Nutrition
| | - Qian Jia
- Program in Integrative Nutrition and Complex Diseases,Intercollegiate Faculty of Nutrition
| | - Evelyn Callaway
- Program in Integrative Nutrition and Complex Diseases,Intercollegiate Faculty of Nutrition
| | - Brad Weeks
- Department of Veterinary Pathobiology, and
| | - Robert C. Alaniz
- Department of Microbial and Molecular Pathogenesis, Texas A&M University System Health Science Center, Texas A&M University, College Station, TX
| | - David N. McMurray
- Program in Integrative Nutrition and Complex Diseases,Intercollegiate Faculty of Nutrition,Department of Microbial and Molecular Pathogenesis, Texas A&M University System Health Science Center, Texas A&M University, College Station, TX
| | - Robert S. Chapkin
- Program in Integrative Nutrition and Complex Diseases,Intercollegiate Faculty of Nutrition,To whom correspondence should be addressed. E-mail:
| |
Collapse
|
37
|
Damjanovich L, Volkó J, Forgács A, Hohenberger W, Bene L. Crohn's disease alters MHC-rafts in CD4+ T-cells. Cytometry A 2011; 81:149-64. [PMID: 22128034 DOI: 10.1002/cyto.a.21173] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2011] [Revised: 10/28/2011] [Accepted: 11/01/2011] [Indexed: 11/09/2022]
Abstract
Clusters of MHCI, ICAM-1, CD44, CD59, IL-2R, and IL-15R molecules have been studied on the surface of CD4(+) T-cells from peripheral blood and lymph nodes of patients in Crohn's disease and healthy individuals as controls by using a dual-laser flow cytometric fluorescence resonance energy transfer (FRET) technique and fluorescently stained Fabs. When cells from patients in Crohn's disease are compared to those of controls, the surface expression level for the MHCI reduced by ∼45%, for CD44 enhanced by ∼100%, and for IL-2Rα, IL-15Rα, and common γ(c) enhanced by ∼50%, ∼70%, and ∼130%, respectively. Efficiencies of FRET monitoring homoassociation for the MHCI and CD44 reduced, that for IL-2Rα enhanced. While efficiencies of FRET monitoring the association of γ(c) and ICAM-1 with the MHCI reduced, those monitoring association of IL-2/15Rα, CD44, and CD59 with MHCI enhanced. Efficiencies of FRET measured between the MHCI and IL-2Rα, IL-15Rα differently enhanced to the advantage of IL-15Rα, the one measured between γ(c) and IL-2Rα reduced, suggesting modulations in the strength of interaction of MHCI with IL-2R, IL-15R, and γ(c). The increases in density of surface bound cTx and in the associations of the receptors with the G(M1)-ganglioside lipid molecules suggest stronger lipid raft interactions of the receptors. The observed alterations of MHC-rafts in Crohn's disease--summarized in models of receptor patterns of diseased and control cells--may have functional consequences regarding signaling by the raft components.
Collapse
Affiliation(s)
- László Damjanovich
- Department of Surgery, Medical and Health Science Center, University of Debrecen, Debrecen, Hungary
| | | | | | | | | |
Collapse
|
38
|
Dietary supplementation with omega-3-PUFA-rich fish oil reduces signs of food allergy in ovalbumin-sensitized mice. Clin Dev Immunol 2011; 2012:236564. [PMID: 22162714 PMCID: PMC3227513 DOI: 10.1155/2012/236564] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2011] [Revised: 08/18/2011] [Accepted: 09/07/2011] [Indexed: 12/18/2022]
Abstract
We investigated the effect of dietary supplementation with n-3 PUFA (fish oil source) in an experimental model of food allergy. Mice were sensitized (allergic group) or not (nonallergic group) with OVA and were fed with OVA diet to induce allergy signals. Mice were fed with regular diet in which 7% of lipid content was provided by soybean (5% of n-3 PUFA) or fish (25% of n-3 PUFA) oil. Allergic group mice had increased serum levels of antiovalbumin IgE and IgG1 and changes in small intestine, characterized by an increased edema, number of rolling leukocytes in microcirculation, eosinophil infiltration, mucus production, and Paneth cell degranulation, in comparison to non-allergic group. All these inflammatory parameters were reduced in mice fed high-n-3-PUFA diet. Our data together suggest that diet supplementation with n-3 PUFA from fish oil may consist of a valid adjuvant in food allergy treatment.
Collapse
|
39
|
Barbosa T, Rescigno M. Host-bacteria interactions in the intestine: homeostasis to chronic inflammation. WILEY INTERDISCIPLINARY REVIEWS-SYSTEMS BIOLOGY AND MEDICINE 2011; 2:80-97. [PMID: 20836012 DOI: 10.1002/wsbm.48] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the past decade it has become clear that the gut constitutes an important frontier of the body, which not only regulates the selective entry of nutrients while keeping vigilant against pathogens but also is largely responsible for shaping the immune response to educate the organism to recognize self from non-self. The very notion of self has undergone a dramatic change, with the acknowledgment that our 'selves' include a plethora of microbial species that actively participate in our body's homeostasis. The immune system continuously adapts to the microbiota in a cyclic, dynamic cross talk where intestinal epithelial cells play an important role in instructing noninflammatory responses for a steady-state control of bacterial growth, or triggering inflammatory mechanisms that can clear the gut from harmful invaders. The system is complex and robust in the sense that many players with partially overlapping roles act to keep the integrity of the intestinal mucosal barrier. Failure of these mechanisms involves genetic and environmental triggers and leads to inflammatory bowel disease. In this review, we seek to collect the state-of-the-art knowledge about how host and microbiota interact to promote gut homeostasis and provide evidences of malfunctioning of the described mechanisms in human inflammatory bowel disease.
Collapse
Affiliation(s)
- Theolis Barbosa
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| | - Maria Rescigno
- Department of Experimental Oncology, European Institute of Oncology, Milan, Italy
| |
Collapse
|
40
|
Wilson MS, Ramalingam TR, Rivollier A, Shenderov K, Mentink-Kane MM, Madala SK, Cheever AW, Artis D, Kelsall BL, Wynn TA. Colitis and intestinal inflammation in IL10-/- mice results from IL-13Rα2-mediated attenuation of IL-13 activity. Gastroenterology 2011; 140:254-64. [PMID: 20951137 PMCID: PMC3006653 DOI: 10.1053/j.gastro.2010.09.047] [Citation(s) in RCA: 78] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/28/2010] [Revised: 09/08/2010] [Accepted: 09/29/2010] [Indexed: 12/16/2022]
Abstract
BACKGROUND & AIMS The cytokine interleukin (IL)-10 is required to maintain immune homeostasis in the gastrointestinal tract. IL-10 null mice spontaneously develop colitis or are more susceptible to induction of colitis by infections, drugs, and autoimmune reactions. IL-13 regulates inflammatory conditions; its activity might be compromised by the IL-13 decoy receptor (IL-13Rα2). METHODS We examined the roles of IL-13 and IL-13Rα2 in intestinal inflammation in mice. To study the function of IL-13Rα2, il10(-/-) mice were crossed with il13rα2(-/-) to generate il10(-/-)il13rα2(-/-) double knockout (dKO) mice. Colitis was induced with the gastrointestinal toxin piroxicam or Trichuris muris infection. RESULTS Induction of colitis by interferon (IFN)-γ or IL-17 in IL-10 null mice requires IL-13Rα2. Following exposure of il10(-/-) mice to piroxicam or infection with T muris, production of IL-13Rα2 increased, resulting in decreased IL-13 bioactivity and increased inflammation in response to IFN-γ or IL-17A. In contrast to il10(-/-) mice, dKO mice were resistant to piroxicam-induced colitis; they also developed less severe colitis during chronic infection with T muris infection. In both models, resistance to IFN-γ and IL-17-mediated intestinal inflammation was associated with increased IL-13 activity. Susceptibility to colitis was restored when the dKO mice were injected with monoclonal antibodies against IL-13, confirming its protective role. CONCLUSIONS Colitis and intestinal inflammation in IL10(-/-) mice results from IL-13Rα2-mediated attenuation of IL-13 activity. In the absence of IL-13Rα2, IL-13 suppresses proinflammatory Th1 and Th17 responses. Reagents that block the IL-13 decoy receptor IL-13Rα2 might be developed for inflammatory bowel disease associated with increased levels of IFN-γ and IL-17.
Collapse
Affiliation(s)
- Mark S. Wilson
- Immunopathogensis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS 50 South Drive, Bethesda, MD 20892
| | - Thirumalai R. Ramalingam
- Immunopathogensis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS 50 South Drive, Bethesda, MD 20892
| | - Aymeric Rivollier
- Laboratory of Mucosal Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS 50 South Drive, Bethesda, MD 20892
| | - Kevin Shenderov
- Immunobiology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS 50 South Drive, Bethesda, MD 20892
| | - Margaret M. Mentink-Kane
- Immunopathogensis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS 50 South Drive, Bethesda, MD 20892
| | - Satish K. Madala
- Immunopathogensis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS 50 South Drive, Bethesda, MD 20892
| | | | - David Artis
- Department of Pathobiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Brian L. Kelsall
- Laboratory of Mucosal Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS 50 South Drive, Bethesda, MD 20892
| | - Thomas A. Wynn
- Immunopathogensis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, DHHS 50 South Drive, Bethesda, MD 20892
| |
Collapse
|
41
|
Niess JH. Role of gut-resident dendritic cells in inflammatory bowel disease. Expert Rev Clin Immunol 2010; 5:451-61. [PMID: 20477041 DOI: 10.1586/eci.09.20] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The gastrointestinal immune system, innate and adaptive, is continuously exposed to challenges provided by the enteric flora. In most cases, the result of mucosal immune responses is the development of tolerance. Mucosal dendritic cells initiate and regulate local immune responses. Uncontrolled local immune responses are thought to be a major factor in the development of inflammatory bowel disease, such as Crohn's disease and ulcerative colitis. This review will discuss the function of dendritic cells in the recognition of the enteric flora and their role in the development of intestinal inflammation.
Collapse
Affiliation(s)
- Jan Hendrik Niess
- Department of Internal Medicine I, Ulm University, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| |
Collapse
|
42
|
Ohland CL, Macnaughton WK. Probiotic bacteria and intestinal epithelial barrier function. Am J Physiol Gastrointest Liver Physiol 2010; 298:G807-19. [PMID: 20299599 DOI: 10.1152/ajpgi.00243.2009] [Citation(s) in RCA: 501] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The intestinal tract is a diverse microenvironment where more than 500 species of bacteria thrive. A single layer of epithelium is all that separates these commensal microorganisms and pathogens from the underlying immune cells, and thus epithelial barrier function is a key component in the arsenal of defense mechanisms required to prevent infection and inflammation. The epithelial barrier consists of a dense mucous layer containing secretory IgA and antimicrobial peptides as well as dynamic junctional complexes that regulate permeability between cells. Probiotics are live microorganisms that confer benefit to the host and that have been suggested to ameliorate or prevent diseases including antibiotic-associated diarrhea, irritable bowel syndrome, and inflammatory bowel disease. Probiotics likely function through enhancement of barrier function, immunomodulation, and competitive adherence to the mucus and epithelium. This review summarizes the evidence about effects of the many available probiotics with an emphasis on intestinal barrier function and the mechanisms affected by probiotics.
Collapse
Affiliation(s)
- Christina L Ohland
- Department of Physiology and Pharmacology, Univ. of Calgary, 3330 Hospital Dr. NW, Calgary, AB, Canada T2N 4N1
| | | |
Collapse
|
43
|
Novak N, Koch S, Allam JP, Bieber T. Dendritic cells: bridging innate and adaptive immunity in atopic dermatitis. J Allergy Clin Immunol 2010; 125:50-9. [PMID: 20109736 DOI: 10.1016/j.jaci.2009.11.019] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2009] [Revised: 11/04/2009] [Accepted: 11/16/2009] [Indexed: 12/17/2022]
Abstract
Much knowledge has been gained about the multifaceted functions of dendritic cells (DCs). The central role of various DC subtypes as bridges between innate and adaptive immunity has become more and more evident. However, a high number of differences exist in the expression of pattern-recognition receptors, the first sensors of the innate immune system, in particular Toll-like receptors (TLRs) by distinct DC subtypes (including myeloid and plasmacytoid DCs), their maturation stage, and tissue distribution, as well as state of health or disease. Furthermore, a plethora of variations in human and murine model systems have to be considered. This review sheds some light on this complex and rapidly growing field. It summarizes the most recent findings and deals with the role of TLR-expressing DCs as promoters of chronic inflammatory immune responses in patients with atopic dermatitis, as well as tolerogenic pathways. Therefore TLR-bearing DCs represent promising targets, which might help to improve tolerance induction during immunotherapeutic approaches in the future.
Collapse
Affiliation(s)
- Natalija Novak
- Department of Dermatology and Allergy, University of Bonn, D-53105 Bonn, Germany
| | | | | | | |
Collapse
|
44
|
Landraud L, Brisse S. Enterobacteriaceae. Infect Dis (Lond) 2010. [DOI: 10.1016/b978-0-323-04579-7.00169-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
45
|
Willing BP, Gill N, Finlay BB. The role of the immune system in regulating the microbiota. Gut Microbes 2010; 1:213-223. [PMID: 21327028 PMCID: PMC3023603 DOI: 10.4161/gmic.1.4.12520] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2009] [Revised: 01/18/2010] [Accepted: 03/04/2010] [Indexed: 02/03/2023] Open
Abstract
A diverse population of bacteria, archaea and fungi, collectively known as the microbiota, abounds within the gastrointestinal tract of the mammalian host. This microbial population makes many important contributions to host physiology through inter-kingdom signalling and by providing nutrients that have both local and systemic effects. In a healthy state the overall host-microbial interaction is symbiotic; however, a growing number of diseases have been associated with a dysregulated microbiota. To avoid these consequences, the host exerts substantial effort to maintain proper regulation of the microbiota with respect to localization and composition. Although important to maintaining microbial balance, the host immune response can also be the cause of a disrupted microbiota, contributing to disease severity. Here, we discuss the role of the host in both maintaining and disrupting a balanced gastrointestinal microbiota.
Collapse
Affiliation(s)
- Benjamin P Willing
- Michael Smith Laboratories; University of British Columbia; Vancouver, BC Canada
| | - Navkiran Gill
- Michael Smith Laboratories; University of British Columbia; Vancouver, BC Canada
| | - B Brett Finlay
- Michael Smith Laboratories; University of British Columbia; Vancouver, BC Canada,Department of Microbiology and Immunology; University of British Columbia; Vancouver, BC Canada,Department of Biochemistry and Molecular Biology; University of British Columbia; Vancouver, BC Canada
| |
Collapse
|
46
|
Langlois C, Gendron FP. Promoting MPhi transepithelial migration by stimulating the epithelial cell P2Y(2) receptor. Eur J Immunol 2009; 39:2895-905. [PMID: 19634190 DOI: 10.1002/eji.200939369] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
In intestine, neutrophils are recruited in response to bacterial infiltration and their anti-cellular activities contribute to inflammatory bowel diseases. In contrast, little is known regarding the recruitment of MPhi to the intestinal epithelium. Extracellular adenosine and uridine 5'-triphosphate (ATP and UTP) can function as leukocyte chemoattractants. We investigated the effects of these nucleotides on the ability of intestinal epithelial cells (IEC) to promote MPhi transepithelial migration and adhesion. ATP and UTP promoted the migration of neutrophil-like PLB-985 cells and MPhi across a Caco-2 monolayer. The MPhi-like U-937 cells adhered to nucleotide-stimulated IEC monolayers. In mice with intestinal inflammation, there were infiltrating CD68(+) MPhi in the colonic epithelium and CD68(+) MPhi present at the apical surface of colonocytes. We determined that ATP and UTP activated the P2Y(2) receptor P (P2Y(2)R) to increase ICAM-1 expression, which mediated the adhesion of MPhi to the apical surface of IEC. Intriguingly, stimulation of IEC with nucleotides did not increase the adhesion of neutrophils. However, in the presence of adherent MPhi, there was adhesion of neutrophils, suggesting that MPhi may serve as anchors for neutrophil adhesion. These studies provide insight into the inflammatory mechanisms that contribute to inflammatory bowel diseases and identify potential therapeutic targets for the treatment of gastrointestinal disorders.
Collapse
Affiliation(s)
- Christine Langlois
- Canadian Institutes of Health Research Team on the Digestive Epithelium, Département d'anatomie et de biologie cellulaire, Faculté de médecine et des sciences de la santé, Université de Sherbrooke, Sherbrooke, Québec, J1H5N4, Canada
| | | |
Collapse
|
47
|
Gorfu G, Rivera-Nieves J, Ley K. Role of β7 Integrins in Intestinal Lymphocyte Homing and Retention. Curr Mol Med 2009; 9:836-50. [DOI: 10.2174/156652409789105525] [Citation(s) in RCA: 244] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
48
|
Fortin G, Raymond M, Van VQ, Rubio M, Gautier P, Sarfati M, Franchimont D. A role for CD47 in the development of experimental colitis mediated by SIRPalpha+CD103- dendritic cells. ACTA ACUST UNITED AC 2009; 206:1995-2011. [PMID: 19703989 PMCID: PMC2737153 DOI: 10.1084/jem.20082805] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Mesenteric lymph node (mLN) CD103 (alphaE integrin)(+) dendritic cells (DCs) induce regulatory T cells and gut tolerance. However, the function of intestinal CD103(-) DCs remains to be clarified. CD47 is the ligand of signal regulatory protein alpha (SIRPalpha) and promotes SIRPalpha(+) myeloid cell migration. We first show that mucosal CD103(-) DCs selectively express SIRPalpha and that their frequency was augmented in the lamina propria and mLNs of mice that developed Th17-biased colitis in response to trinitrobenzene sulfonic acid. In contrast, the percentage of SIRPalpha(+)CD103(-) DCs and Th17 responses were decreased in CD47-deficient (CD47 knockout [KO]) mice, which remained protected from colitis. We next demonstrate that transferring wild-type (WT), but not CD47 KO, SIRPalpha(+)CD103(-) DCs in CD47 KO mice elicited severe Th17-associated wasting disease. CD47 expression was required on the SIRPalpha(+)CD103(-) DCs for efficient trafficking to mLNs in vivo, whereas it was dispensable on both DCs and T cells for Th17 polarization in vitro. Finally, administration of a CD47-Fc molecule resulted in reduced SIRPalpha(+)CD103(-) DC-mediated Th17 responses and the protection of WT mice from colitis. We thus propose SIRPalpha(+)CD103(-) DCs as a pathogenic DC subset that drives Th17-biased responses and colitis, and the CD47-SIRPalpha axis as a potential therapeutic target for inflammatory bowel disease.
Collapse
Affiliation(s)
- Genevieve Fortin
- Research Institute of McGill University Health Centre, McGill University, Montreal H3H 2R9, Canada
| | | | | | | | | | | | | |
Collapse
|
49
|
Srinivasan A, Nanton M, Griffin A, McSorley SJ. Culling of activated CD4 T cells during typhoid is driven by Salmonella virulence genes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2009; 182:7838-45. [PMID: 19494308 PMCID: PMC2731968 DOI: 10.4049/jimmunol.0900382] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Pathogen-specific CD4 T cells are activated within a few hours of oral Salmonella infection and are essential for protective immunity. However, CD4 T cells do not participate in bacterial clearance until several weeks after infection, suggesting that Salmonella can inhibit or evade CD4 T cells that are activated at early time points. Here, we describe the progressive culling of initially activated CD4 T cells in Salmonella-infected mice. Loss of activated CD4 T cells was independent of early instructional programming, T cell precursor frequency, and Ag availability. In contrast, apoptosis of Ag-specific CD4 T cells was actively induced by live bacteria in a process that required Salmonella pathogenicity island-2 and correlated with increased expression of PD-L1. These data demonstrate efficient culling of initially activated Ag-specific CD4 cells by a microbial pathogen and document a novel strategy for bacterial immune evasion.
Collapse
Affiliation(s)
- Aparna Srinivasan
- Department of Medicine, Division of Gastroenterology, Center for Infectious Diseases and Microbiology Translational Research, McGuire Translational Research Facility, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
50
|
Ungaro R, Fukata M, Hsu D, Hernandez Y, Breglio K, Chen A, Xu R, Sotolongo J, Espana C, Zaias J, Elson G, Mayer L, Kosco-Vilbois M, Abreu MT. A novel Toll-like receptor 4 antagonist antibody ameliorates inflammation but impairs mucosal healing in murine colitis. Am J Physiol Gastrointest Liver Physiol 2009; 296:G1167-79. [PMID: 19359427 PMCID: PMC2697943 DOI: 10.1152/ajpgi.90496.2008] [Citation(s) in RCA: 130] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Dysregulated innate immune responses to commensal bacteria contribute to the development of inflammatory bowel disease (IBD). TLR4 is overexpressed in the intestinal mucosa of IBD patients and may contribute to uncontrolled inflammation. However, TLR4 is also an important mediator of intestinal repair. The aim of this study is to examine the effect of a TLR4 antagonist on inflammation and intestinal repair in two murine models of IBD. Colitis was induced in C57BL/6J mice with dextran sodium sulfate (DSS) or by transferring CD45Rb(hi) T cells into RAG1-/- mice. An antibody (Ab) against the TLR4/MD-2 complex or isotype control Ab was administered intraperitoneally during DSS treatment, recovery from DSS colitis, or induction of colitis in RAG1-/- mice. Colitis severity was assessed by disease activity index (DAI) and histology. The effect of the Ab on the inflammatory infiltrate was determined by cell isolation and immunohistochemistry. Mucosal expression of inflammatory mediators was analyzed by real-time PCR and ELISA. Blocking TLR4 at the beginning of DSS administration delayed the development of colitis with significantly lower DAI scores. Anti-TLR4 Ab treatment decreased macrophage and dendritic cell infiltrate and reduced mucosal expression of CCL2, CCL20, TNF-alpha, and IL-6. Anti-TLR4 Ab treatment during recovery from DSS colitis resulted in defective mucosal healing with lower expression of COX-2, PGE(2), and amphiregulin. In contrast, TLR4 blockade had minimal efficacy in ameliorating inflammation in the adoptive transfer model of chronic colitis. Our findings suggest that anti-TLR4 therapy may decrease inflammation in IBD but may also interfere with colonic mucosal healing.
Collapse
Affiliation(s)
- Ryan Ungaro
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Masayuki Fukata
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - David Hsu
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Yasmin Hernandez
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Keith Breglio
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Anli Chen
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Ruliang Xu
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - John Sotolongo
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Cecillia Espana
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Julia Zaias
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Greg Elson
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Lloyd Mayer
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Marie Kosco-Vilbois
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| | - Maria T. Abreu
- Division of Gastroenterology, Department of Medicine, and Veterinary Resources, University of Miami Miller School of Medicine, Miami, Florida; Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, Department of Pathology, Mount Sinai School of Medicine, New York, New York; and Novimmune, Geneva, Switzerland
| |
Collapse
|