51
|
Mantovani V, Galeotti F, Maccari F, Volpi N. Recent advances on separation and characterization of human milk oligosaccharides. Electrophoresis 2016; 37:1514-24. [PMID: 26801168 DOI: 10.1002/elps.201500477] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 12/03/2015] [Accepted: 12/04/2015] [Indexed: 01/14/2023]
Abstract
Free human milk oligosaccharides (HMOs) are unique due to their highly complex nature and important emerging biological and protective functions during early life such as prebiotic activity, pathogen deflection, and epithelial and immune cell modulation. Moreover, four genetically determined heterogeneous HMO secretory groups are known to be based on their structure and composition. Over the years, several analytical techniques have been applied to characterize and quantitate HMOs, including nuclear magnetic resonance spectroscopy, high-performance liquid chromatography (HPLC), high pH anion-exchange chromatography, off-line and on-line mass spectrometry (MS), and capillary electrophoresis (CE). Even if these techniques have proven to be efficient and simple, most glycans have no significant UV absorption and derivatization with fluorophore groups prior to separation usually results in higher sensitivity and an improved chromatographic/electrophoretic profile. Consequently, the analysis by HPLC/CE of derivatized milk oligosaccharides with different chromophoric active tags has been developed. However, UV or fluorescence detection does not provide specific structural information and this is a key point in particular related to the highly complex nature of the milk glycan mixtures. As a consequence, for a specific determination of complex mixtures of oligomers, analytical separation is usually required with evaluation by means of MS, which has been successfully applied to HMOs, resulting in efficient compositional analysis and profiling in various milk samples. This review aims to give an overview of the current state-of-the-art techniques used in HMO analysis.
Collapse
Affiliation(s)
- Veronica Mantovani
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Fabio Galeotti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Francesca Maccari
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Nicola Volpi
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| |
Collapse
|
52
|
Sherif LS, Raouf RKA, Sayede RME, Wakkadd ASE, Shoaib AR, Ali HM, Refay ASE. Glutathione Transferase as a Potential Marker for Gut Epithelial Injury versus the Protective Role of Breast Milk sIgA in Infants with Rota Virus Gastroenteritis. Open Access Maced J Med Sci 2015; 3:676-80. [PMID: 27275307 PMCID: PMC4877907 DOI: 10.3889/oamjms.2015.125] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2015] [Revised: 11/20/2015] [Accepted: 11/21/2015] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND: Secretory immunoglobulin A (SIgA) plays an important protective role in the recognition and clearance of enteric pathogens. AIM: This study was designed to assess if mucosal integrity “measured by secretory IgA (SIgA)” is a protective factor from more epithelial alteration “measured by glutathione transferase” in infants with Rota gastroenteritis and its relation to infants’ feeding pattern. PATIENTS AND METHODS: This study was conducted on 79 infants aged 6 months and less from those diagnosed as having gastroenteritis and admitted to Gastroenteritis Department in Abo El Rish Pediatric Hospital, Cairo University. Plasma glutathione s-transferases and Stool SIgA were measured using ELISA technique. Rota virus detection was done by Reverse transcriptase PCR. RESULTS: SIgA was found to be significantly positive in exclusive breast fed infants, Glutathione transferase was significantly more frequently positive in Rota positive cases than Rota negative cases by Reverse transcriptase PCR. A significant negative correlation between Glutathione transferase and Secretory IgA was found, (p < 0.05). CONCLUSION: Breast feeding should be encouraged and highly recommended in the first two years of life as it provides Secretory IgA to breast fed infants who in turn protect them against epithelial damage caused by Rota viral gastroenteritis.
Collapse
Affiliation(s)
- Lobna S Sherif
- National Research Centre, Department of Child Health, Giza, Egypt
| | - Randaa K Abdel Raouf
- Institute of Postgraduate Childhood Studies - Department of Medical Studies, Cairo, Egypt
| | | | | | - Ashraf R Shoaib
- National Research Centre, Department of Virology, Giza, Egypt
| | - Hanan M Ali
- National Research Centre, Department of Cell Biology, Giza, Egypt
| | - Amira S El Refay
- National Research Centre, Department of Child Health, Giza, Egypt
| |
Collapse
|
53
|
Abstract
The best-characterized mucosa-associated lymphoid tissue (MALT), and also the most relevant for this review, is the gastrointestinal-associated lymphoid tissue (GALT). The review reviews our understanding of the importance of mucosal immune responses in resisting infections caused by E. coli and Salmonella spp. It focuses on the major human E. coli infections and discusses whether antigen-specific mucosal immune responses are important for resistance against primary infection or reinfection by pathogenic E. coli. It analyzes human data on mucosal immunity against E. coli, a growing body of data of mucosal responses in food production animals and other natural hosts of E. coli, and more recent experimental studies in mice carrying defined deletions in genes encoding specific immunological effectors, to show that there may be considerable conservation of the effective host mucosal immune response against this pathogen. The species Salmonella enterica contains a number of serovars that include pathogens of both humans and animals; these bacteria are frequently host specific and may cause different diseases in different hosts. Ingestion of various Salmonella serovars, such as Typhimurium, results in localized infections of the small intestine leading to gastroenteritis in humans, whereas ingestion of serovar Typhi results in systemic infection and enteric fever. Serovar Typhi infects only humans, and the review discusses the mucosal immune responses against serovar Typhi, focusing on the responses in humans and in the mouse typhoid fever model.
Collapse
|
54
|
Aksoy F, Dogan R, Kocak I, Veyseller B, Ozturan O, Incir S. Effect of Nasal Corticosteroid on Secretory Immunoglobulin A Measured in Rat Nasal Lavage: Experimental Study. Otolaryngol Head Neck Surg 2015; 153:298-301. [PMID: 26084823 DOI: 10.1177/0194599815589073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2015] [Accepted: 05/08/2015] [Indexed: 12/20/2022]
Abstract
OBJECTIVE In this study, we aimed to experimentally investigate the effects of nasal corticosteroids on the levels of secretory immunoglobulin A (sIgA) in nasal mucosa in rats. STUDY DESIGN Prospective, randomized control trial. SETTING Research laboratory. SUBJECT AND METHODS Twenty-four male Sprague Dawley rats were included in our study. The rats were randomized into 3 groups. In group 1, nasal mometasone furoate was applied to the rats for 30 days. Saline was applied to group 2 for 30 days. Group 3 was the control group and received no treatment throughout the study period. Nasal lavage was conducted on both nasal openings of all rats in the 3 groups at the beginning of the study and on days 15 and 30, and the lavage solution (distilled water) was collected by aspiration. RESULTS In group 1, the sIgA value was significantly higher at day 15 than at baseline. No significant difference was found between the sIgA values on day 15 and day 30. In groups 2 and 3, there were no significant differences in sIgA values at baseline, day 15, and day 30. The sIgA value of group 1 on day 15 was significantly higher than the values of groups 2 and 3. The sIgA value of group 1 on day 30 was significantly higher than the values of groups 2 and 3. CONCLUSION Topical corticosteroids (mometasone furoate) applied to the nasal mucosa significantly increase nasal sIgA levels.
Collapse
Affiliation(s)
- Fadlullah Aksoy
- Bezmialem Vakif University, Department of Otorhinolaryngology, Fatih, Istanbul, Turkey
| | - Remzi Dogan
- Bayrampasa State Hospital, Department of Otorhinolaryngology, Bayrampasa, Istanbul, Turkey
| | - Ilker Kocak
- Koç University, Department of Otorhinolaryngology, Sarıyer, Istanbul, Turkey
| | - Bayram Veyseller
- Bezmialem Vakif University, Department of Otorhinolaryngology, Fatih, Istanbul, Turkey
| | - Orhan Ozturan
- Bezmialem Vakif University, Department of Otorhinolaryngology, Fatih, Istanbul, Turkey
| | - Said Incir
- Istanbul University, Cerrahpasa School of Medicine Department of Biochemistry, Fatih, Istanbul, Turkey
| |
Collapse
|
55
|
The impact of phosphorus on the immune system and the intestinal microbiota with special focus on the pig. Nutr Res Rev 2015; 28:67-82. [PMID: 26004147 DOI: 10.1017/s0954422415000049] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
There is increasing interest in dietary ingredients that are appropriate to support digestive and immune functions, but also maintain a stable microbial ecosystem in the gastrointestinal tract (GIT), particularly in weaned pigs. P is an essential nutrient for both microbes and their host, as it is involved, for example, in bone formation, energy metabolism, cellular signalling and stabilisation of cell membranes. Non-ruminant animals have limited access to phytate, the main storage form of P in plant seeds. The release of P bound to phytate requires phytase activity of plant or microbial origin, resulting in the formation of variable phosphorylated inositol phosphates (InsPs). The present review focuses on interactions between variations in dietary P supply, the immune system of the host, and the intestinal microbial ecosystem. Although results on the interaction between P and the immune system are inconsistent, several studies in different species have shown a positive impact of dietary P and phytase addition on the adaptive immune response. Recent studies with pigs suggest that P supply may influence intestinal microbial composition and activity. Individual InsPs or phosphate may also affect properties of pathogenic micro-organisms, such as metabolism or virulence. In conclusion, P may be considered as part of an integrated approach to support immune functions and maintain a stable microbial ecosystem in the GIT, thereby providing a barrier against potential pathogens. Within this regard, differences in phytate-P content and intrinsic phytase activity of plant feedstuffs, as well as the formation of individual InsPs, have to be taken into account.
Collapse
|
56
|
CD69 is the crucial regulator of intestinal inflammation: a new target molecule for IBD treatment? J Immunol Res 2015; 2015:497056. [PMID: 25759842 PMCID: PMC4352431 DOI: 10.1155/2015/497056] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Accepted: 10/07/2014] [Indexed: 12/19/2022] Open
Abstract
CD69 has been identified as an early activation marker of lymphocytes. However, recent work has indicated that CD69 plays an essential role for the regulation of inflammatory processes. Particularly, CD69 is highly expressed by lymphocytes at mucosal sites being constantly exposed to the intestinal microflora (one of the nature's most complex and most densely populated microbial habitats) and food antigens, while only a small number of circulating leukocytes express this molecule. In this review we will discuss the role of CD69 in mucosal tissue and consider CD69 as a potential target for the development of novel treatments of intestinal inflammation.
Collapse
|
57
|
Milk proteins, peptides, and oligosaccharides: effects against the 21st century disorders. BIOMED RESEARCH INTERNATIONAL 2015; 2015:146840. [PMID: 25789308 PMCID: PMC4350585 DOI: 10.1155/2015/146840] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Accepted: 12/21/2014] [Indexed: 12/17/2022]
Abstract
Milk is the most complete food for mammals, as it supplies all the energy and nutrients needed for the proper growth and development of the neonate. Milk is a source of many bioactive components, which not only help meeting the nutritional requirements of the consumers, but also play a relevant role in preventing various disorders. Milk-derived proteins and peptides have the potential to act as coadjuvants in conventional therapies, addressing cardiovascular diseases, metabolic disorders, intestinal health, and chemopreventive properties. In addition to being a source of proteins and peptides, milk contains complex oligosaccharides that possess important functions related to the newborn's development and health. Some of the health benefits attributed to milk oligosaccharides include prebiotic probifidogenic effects, antiadherence of pathogenic bacteria, and immunomodulation. This review focuses on recent findings demonstrating the biological activities of milk peptides, proteins, and oligosaccharides towards the prevention of diseases of the 21st century. Processing challenges hindering large-scale production and commercialization of those bioactive compounds have been also addressed.
Collapse
|
58
|
Owen JL, Yang T, Mohamadzadeh M. New insights into gastrointestinal anthrax infection. Trends Mol Med 2014; 21:154-63. [PMID: 25577136 DOI: 10.1016/j.molmed.2014.12.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 11/16/2014] [Accepted: 12/11/2014] [Indexed: 02/08/2023]
Abstract
Bacterial infections are the primary cause of gastrointestinal (GI) disorders in both developing and developed countries, and are particularly dangerous for infants and children. Bacillus anthracis is the 'archetype zoonotic' pathogen; no other infectious disease affects such a broad range of species, including humans. Importantly, there are more case reports of GI anthrax infection in children than inhalational disease. Early diagnosis is difficult and widespread systemic disease develops rapidly. This review highlights new findings concerning the roles of the gut epithelia, commensal microbiota, and innate lymphoid cells (ILCs) in initiation of disease and systemic dissemination in animal models of GI anthrax, the understanding of which is crucial to designing alternative therapies that target the establishment of infection.
Collapse
Affiliation(s)
- Jennifer L Owen
- Department of Physiological Sciences, College of Veterinary Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Tao Yang
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32608, USA; Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA
| | - Mansour Mohamadzadeh
- Department of Infectious Diseases and Pathology, University of Florida, Gainesville, FL 32608, USA; Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Florida, Gainesville, FL 32610, USA.
| |
Collapse
|
59
|
Judaki A, Hafeziahmadi M, Yousefi A, Havasian MR, Panahi J, Sayehmiri K, Alizadeh S. Evaluation of dairy allergy among ulcerative colitis patients. Bioinformation 2014; 10:693-6. [PMID: 25512686 PMCID: PMC4261114 DOI: 10.6026/97320630010693] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 11/15/2014] [Indexed: 12/11/2022] Open
Abstract
The intestine is the largest mucosal organ of the body and also the first line immune homeostasis. Inflammatory bowel disease or IBD is divided into ulcerative colitis and Crohn's disease. One of the problems that can occur with UC is dietary allergy to some foods. This study aimed to evaluated the dairy allergy among patients with ulcerative colitis. This study is a Case - control study, that studied 72 patients with Ulcerative Colitis, after recording history of the disease, colonoscopy and confirmed by biopsy and 72 person without history of colitis. In this study, in order to investigate of food allergy, used of the EUROMMUM kit with an international code number DP3420-1601-11E. We used chi-square and Monte Carlo method for analysis of data. Among UC patients, 30.6% mild, 52.8% moderate and 16.6% of cases were in sever stage. 9.7% of them reported a history of abdominal surgery due to disease. According to the chi-square and Monte Carlo methods, dairy allergy (including: cow milk, cow milk UHT and casein) in UC group was significant (P=0.00). This study indicated that there is significant relationship between UC and cow milk, cow milk UHT and casein. UC patients who are allergic to dairy products and the use of dairy products can increase the severity of UC.
Collapse
Affiliation(s)
- Arezo Judaki
- Department of Gastroenterology, Ilam University of Medical Sciences, Ilam/ Iran
| | | | - Atefe Yousefi
- Student Research of Committee, Ilam University of Medical Sciences, Ilam/ Iran
| | | | - Jafar Panahi
- Student Research of Committee, Ilam University of Medical Sciences, Ilam/ Iran
| | - Koroush Sayehmiri
- Center for Prevention of Psychosocial Trauma, Ilam University of Medical Sciences, Ilam/ Iran
| | - Sajjad Alizadeh
- Student Research of Committee, Ilam University of Medical Sciences, Ilam/ Iran
| |
Collapse
|
60
|
Cao S, Feehley TJ, Nagler CR. The role of commensal bacteria in the regulation of sensitization to food allergens. FEBS Lett 2014; 588:4258-66. [PMID: 24791655 PMCID: PMC4216641 DOI: 10.1016/j.febslet.2014.04.026] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2014] [Revised: 04/24/2014] [Accepted: 04/24/2014] [Indexed: 12/23/2022]
Abstract
The prevalence of life-threatening anaphylactic responses to food is rising at an alarming rate. The emerging role of the gut microbiota in regulating food allergen sensitization may help explain this trend. The mechanisms by which commensal bacteria influence sensitization to dietary antigens are only beginning to be explored. We have found that a population of mucosa-associated commensal anaerobes prevents food allergen sensitization by promoting an IL-22-dependent barrier protective immune response that limits the access of food allergens to the systemic circulation. This early response is followed by an adaptive immune response mediated in part by an expansion of Foxp3(+) Tregs that fortifies the tolerogenic milieu needed to maintain non-responsiveness to food. Bacterial metabolites, such as short-chain fatty acids, may contribute to the process through their ability to promote Foxp3(+) Treg differentiation. This work suggests that environmentally induced alterations of the gut microbiota offset the regulatory signals conferred by protective bacterial species to promote aberrant responses to food. Our research presents exciting new possibilities for preventing and treating food allergies based on interventions that modulate the composition of the gut microbiota.
Collapse
Affiliation(s)
- Severine Cao
- Department of Pathology and Committee on Immunology, The University of Chicago, 924 East 57th Street, JFK R120, Chicago, IL 60637, United States
| | - Taylor J Feehley
- Department of Pathology and Committee on Immunology, The University of Chicago, 924 East 57th Street, JFK R120, Chicago, IL 60637, United States
| | - Cathryn R Nagler
- Department of Pathology and Committee on Immunology, The University of Chicago, 924 East 57th Street, JFK R120, Chicago, IL 60637, United States.
| |
Collapse
|
61
|
Ozbılgın ND, Saka OM, Bozkır A. Preparation and in vitro/in vivo evaluation of mucosal adjuvant in situ forming gels with diphtheria toxoid. Drug Deliv 2014; 21:140-7. [PMID: 24559517 DOI: 10.3109/10717544.2013.834754] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Studies on preparation of in situ gel formulations containing diphtheria toxoid as the model active substance and their intranasal administration have been conducted in this study. The objective of mucosal vaccination is to stimulate both systemic and mucosal immune responses. In situ gel formulations were prepared by using, in different ratios, mixtures of Poloxamer 407 and Poloxamer 188 polymers, which gelate in a temperature-dependent manner, and mucoadhesive polymers carbopol 934, hydroxypropyl methyl cellulose, hydroxypropyl cellulose or chitosan. Following pre-formulation studies, F1, F2, F3, F4, F5, F6 and F7 formulations, which gelate at intervals and temperatures in accordance with nasal temperatures, were subjected to more comprehensive studies. For this purpose, organoleptic characteristics of the formulations were identified, their pH and mucoadhesive potencies were measured and rheological behaviors were characterized. Calculated amounts of diphtheria toxoid were added to formulations after optimization of formulations was achieved, and assay and in vitro release studies were carried out. Formulations coded F3 and F7 were considered to be superior to other formulations given the in vitro test results. Therefore, these formulations were tested in guinea pigs to determine immune responses, which they would produce following intranasal and subcutaneous administration. Absorbance values of ELISA tests and antibody neutralization test showed that formulations coded F3 and F7 were unable to stimulate adequate systemic immune response when either of the formulations was administered alone intranasally, whereas F7 resulted in significantly increased neutralizing antibody titers with intranasal administration as a booster dose following subcutaneous administration.
Collapse
Affiliation(s)
- Nalan Deniz Ozbılgın
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Ankara University , Ankara , Turkey
| | | | | |
Collapse
|
62
|
Stefka AT, Feehley T, Tripathi P, Qiu J, McCoy K, Mazmanian SK, Tjota MY, Seo GY, Cao S, Theriault BR, Antonopoulos DA, Zhou L, Chang EB, Fu YX, Nagler CR. Commensal bacteria protect against food allergen sensitization. Proc Natl Acad Sci U S A 2014; 111:13145-50. [PMID: 25157157 PMCID: PMC4246970 DOI: 10.1073/pnas.1412008111] [Citation(s) in RCA: 557] [Impact Index Per Article: 50.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Environmentally induced alterations in the commensal microbiota have been implicated in the increasing prevalence of food allergy. We show here that sensitization to a food allergen is increased in mice that have been treated with antibiotics or are devoid of a commensal microbiota. By selectively colonizing gnotobiotic mice, we demonstrate that the allergy-protective capacity is conferred by a Clostridia-containing microbiota. Microarray analysis of intestinal epithelial cells from gnotobiotic mice revealed a previously unidentified mechanism by which Clostridia regulate innate lymphoid cell function and intestinal epithelial permeability to protect against allergen sensitization. Our findings will inform the development of novel approaches to prevent or treat food allergy based on modulating the composition of the intestinal microbiota.
Collapse
Affiliation(s)
| | | | | | - Ju Qiu
- Departments of Pathology and Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | - Kathy McCoy
- Department of Clinical Research, University of Bern, 3010 Bern, Switzerland
| | - Sarkis K Mazmanian
- Department of Biology, California Institute of Technology, Pasadena, CA 91125; and
| | | | | | | | | | | | - Liang Zhou
- Departments of Pathology and Microbiology-Immunology, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611
| | | | | | | |
Collapse
|
63
|
Quantification of cell-associated atazanavir, darunavir, lopinavir, ritonavir, and efavirenz concentrations in human mononuclear cell extracts. Antimicrob Agents Chemother 2014; 58:2866-70. [PMID: 24614370 DOI: 10.1128/aac.02551-13] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
An ultrasensitive assay utilizing high-pressure liquid chromatography and mass spectrometry detection was developed and validated for the quantification of the antiretrovirals atazanavir (ATV), darunavir (DRV), lopinavir (LPV), ritonavir (RTV), and efavirenz (EFV) in human mononuclear cell (MNC) extracts. The assay utilizes 20 μl of cellular extract that contains as few as 50,000 MNCs. The analytical range of the assay is 0.0200 to 10.0 fmol/μl for ATV, 0.0500 to 25.0 fmol/μl for DRV, LPV, and RTV, and 0.200 to 100 fmol/μl for EFV. The assay has proven to be a clinically useful tool for investigating antiretroviral drug concentrations in virologic sanctuaries where harvested cell numbers are extremely low. The assay provides a tool for investigators to explore the clinical pharmacology of strategies for prevention, treatment, and cure in pathophysiologically relevant sites.
Collapse
|
64
|
McKim JM. Food additive carrageenan: Part I: A critical review of carrageenanin vitrostudies, potential pitfalls, and implications for human health and safety. Crit Rev Toxicol 2014; 44:211-43. [DOI: 10.3109/10408444.2013.861797] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
65
|
Sitta DLA, Guilherme MR, Garcia FP, Cellet TSP, Nakamura CV, Muniz EC, Rubira AF. Covalent Albumin Microparticles as an Adjuvant for Production of Mucosal Vaccines against Hepatitis B. Biomacromolecules 2013; 14:3231-7. [DOI: 10.1021/bm400859z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Danielly L. A. Sitta
- Grupo de Materiais Poliméricos
e Compósitos, Departamento de Química, Universidade Estadual de Maringá, Avenida Colombo,
5790, CEP 87020-900 Maringá, PR, Brazil
| | - Marcos R. Guilherme
- Grupo de Materiais Poliméricos
e Compósitos, Departamento de Química, Universidade Estadual de Maringá, Avenida Colombo,
5790, CEP 87020-900 Maringá, PR, Brazil
| | - Francielle P. Garcia
- Departamento de Ciências
Básicas da Saúde, Universidade Estadual de Maringá, Avenida Colombo, 5790, CEP 87020-900
Maringá, PR, Brazil
| | - Thelma S. P. Cellet
- Grupo de Materiais Poliméricos
e Compósitos, Departamento de Química, Universidade Estadual de Maringá, Avenida Colombo,
5790, CEP 87020-900 Maringá, PR, Brazil
| | - Celso V. Nakamura
- Departamento de Ciências
Básicas da Saúde, Universidade Estadual de Maringá, Avenida Colombo, 5790, CEP 87020-900
Maringá, PR, Brazil
- Programa de pós-graduação
em Ciências Farmacêuticas, Departamento de Ciências
Básicas da Saúde, Universidade Estadual de Maringá, Avenida Colombo, 5790, CEP 87020-900
Maringá, PR, Brazil
| | - Edvani C. Muniz
- Grupo de Materiais Poliméricos
e Compósitos, Departamento de Química, Universidade Estadual de Maringá, Avenida Colombo,
5790, CEP 87020-900 Maringá, PR, Brazil
| | - Adley F. Rubira
- Grupo de Materiais Poliméricos
e Compósitos, Departamento de Química, Universidade Estadual de Maringá, Avenida Colombo,
5790, CEP 87020-900 Maringá, PR, Brazil
| |
Collapse
|
66
|
Garcia-Garcia E, Galindo-Villegas J, Mulero V. Mucosal immunity in the gut: the non-vertebrate perspective. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2013; 40:278-288. [PMID: 23537860 DOI: 10.1016/j.dci.2013.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2013] [Revised: 03/07/2013] [Accepted: 03/14/2013] [Indexed: 06/02/2023]
Abstract
Much is now known about the vertebrate mechanisms involved in mucosal immunity, and the requirement of commensal microbiota at mucosal surfaces for the proper functioning of the immune system. In comparison, very little is known about the mechanisms of immunity at the barrier epithelia of non-vertebrate organisms. The purpose of this review is to summarize key experimental evidence illustrating how non-vertebrate immune mechanisms at barrier epithelia compare to those of higher vertebrates, using the gut as a model organ. Not only effector mechanisms of gut immunity are similar between vertebrates and non-vertebrates, but it also seems that the proper functioning of non-vertebrate gut defense mechanisms requires the presence of a resident microbiota. As more information becomes available, it will be possible to obtain a more accurate picture of how mucosal immunity has evolved, and how it adapts to the organisms' life styles.
Collapse
Affiliation(s)
- Erick Garcia-Garcia
- Department of Cell Biology and Histology, Faculty of Biology, University of Murcia, Campus Universitario de Espinardo, 30100 Murcia, Spain.
| | | | | |
Collapse
|
67
|
Chandra LC, Traoré D, French C, Marlow D, D’Offay J, Clarke SL, Smith BJ, Kuvibidila S. White button, portabella, and shiitake mushroom supplementation up-regulates interleukin-23 secretion in acute dextran sodium sulfate colitis C57BL/6 mice and murine macrophage J.744.1 cell line. Nutr Res 2013; 33:388-96. [DOI: 10.1016/j.nutres.2013.02.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2012] [Revised: 02/13/2013] [Accepted: 02/20/2013] [Indexed: 01/25/2023]
|
68
|
Hulst M, Smits M, Vastenhouw S, de Wit A, Niewold T, van der Meulen J. Transcription networks responsible for early regulation of Salmonella-induced inflammation in the jejunum of pigs. JOURNAL OF INFLAMMATION-LONDON 2013; 10:18. [PMID: 23590759 PMCID: PMC3637394 DOI: 10.1186/1476-9255-10-18] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 04/10/2013] [Indexed: 05/16/2023]
Abstract
Background The aim of this study was to identify transcription factors/regulators that play a crucial role in steering the (innate) immune response shortly (within a few hours) after the first contact of the intestinal mucosa with an inflammatory mediator, and to test whether the processes regulated by these factors/regulators can be modulated by chemical substances of natural origin. Methods We experimentally induced inflammation by perfusion of surgically applied jejunal loops with Salmonella enterica subspecies enterica serovar Typhimurium DT104 in three pigs. Segments of mock and Salmonella treated loops were dissected after 2, 4 and 8 hours of perfusion. IL8 and IL1-beta mRNA expression levels were measured in mucosal scrapings of all segments. Furthermore, intra-animal microarray comparisons (isogenic) between Salmonella and mock treated segments after 8 hours, and inter-animal comparisons between similar Salmonella-treated loops of each pig at 2 and 4 hours, were performed. Results IL-1beta and IL8 mRNA levels, and intra-animal microarray comparisons at 8 hours between Salmonella and mock treated segments showed that the response-time and type of response to Salmonella was different in all three pigs. This plasticity allowed us to extract a comprehensive set of differentially expressed genes from inter-animal comparisons at 2 and 4 hours. Pathway analysis indicated that many of these genes play a role in induction and/or tempering the inflammatory response in the intestine. Among them a set of transcription factors/regulators known to be involved in regulation of inflammation, but also factors/regulators for which involvement was not expected. Nine out of twenty compounds of natural origin, which according to literature had the potential to modulate the activity of these factors/regulators, were able to stimulate or inhibit a Salmonella-induced mRNA response of inflammatory-reporter genes IL8 and/or nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor alpha in cultured intestinal porcine epithelial cells. Conclusions We describe a set of transcription factors/regulators possibly involved in regulation of “very early” immune mechanism which determines the inflammatory status of the intestine later on. In addition, we show that these mechanisms may be modulated by chemical substances of natural origin.
Collapse
Affiliation(s)
- Marcel Hulst
- Livestock Research of Wageningen University and Research Centre, P,O, Box 65, Lelystad, 8200AB, The Netherlands.
| | | | | | | | | | | |
Collapse
|
69
|
Di Mauro A, Neu J, Riezzo G, Raimondi F, Martinelli D, Francavilla R, Indrio F. Gastrointestinal function development and microbiota. Ital J Pediatr 2013; 39:15. [PMID: 23433508 PMCID: PMC3615966 DOI: 10.1186/1824-7288-39-15] [Citation(s) in RCA: 118] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Accepted: 02/14/2013] [Indexed: 12/15/2022] Open
Abstract
The intestinal microbiota plays an important role in the development of post-natal gastrointestinal functions of the host. Recent advances in our capability to identify microbes and their function offer exciting opportunities to evaluate the complex cross talk between microbiota, intestinal barrier, immune system and the gut-brain axis. This review summarizes these interactions in the early colonization of gastrointestinal tract with a major focus on the role of intestinal microbiota in the pathogenesis of feeding intolerance in preterm newborn. The potential benefit of early probiotic supplementation opens new perspectives in case of altered intestinal colonization at birth as preventive and therapeutic agents.
Collapse
Affiliation(s)
- Antonio Di Mauro
- Department of Pediatrics, University of Bari, Policlinico, Piazza G Cesare, Bari, Italy
| | | | | | | | | | | | | |
Collapse
|
70
|
Yamamoto H, Oda M, Nakano M, Watanabe N, Yabiku K, Shibutani M, Inoue M, Imagawa H, Nagahama M, Himeno S, Setsu K, Sakurai J, Nishizawa M. Development of vizantin, a safe immunostimulant, based on the structure-activity relationship of trehalose-6,6'-dicorynomycolate. J Med Chem 2012; 56:381-5. [PMID: 23210481 DOI: 10.1021/jm3016443] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Vizantin, 6,6'-bis-O-(3-nonyldodecanoyl)-α,α'-trehalose, was developed as a safe immunostimulator on the basis of a structure-activity relationship (SAR) study with trehalose 6,6'-dicorynomycolate (TDCM). It was possible to synthesize vizantin on a large scale more easily than in the case of TDCM, and the compound exhibited more potent prophylactic effect on experimental lung metastasis of B16-F0 melanoma cells. Because vizantin stimulated human macrophages, it is a promising candidate for clinical application.
Collapse
Affiliation(s)
- Hirofumi Yamamoto
- Faculty of Pharmaceutical Sciences, Tokushima Bunri University, Yamashiro-cho, Tokushima 770-8514, Japan.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
71
|
Panzan MQ, Mattar R, Maganhin CC, Simões RDS, Rossi AGZ, Motta ELAD, Baracat EC, Soares JM. Evaluation of FAS and caspase-3 in the endometrial tissue of patients with idiopathic infertility and recurrent pregnancy loss. Eur J Obstet Gynecol Reprod Biol 2012; 167:47-52. [PMID: 23218678 DOI: 10.1016/j.ejogrb.2012.10.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 07/27/2012] [Accepted: 10/23/2012] [Indexed: 10/27/2022]
Abstract
OBJECTIVE To investigate expression of BcL-2, FAS, FAS ligand (FASL) and cleaved caspase-3 in the endometrial tissue of women with idiopathic infertility (with two consecutive failed cycles of in vitro fertilization) and women with idiopathic recurrent pregnancy loss. The control group consisted of fertile women. STUDY DESIGN Endometrial tissue samples from fertile women (n=25), women with idiopathic infertility (n=25) and women with idiopathic recurrent pregnancy loss (n=25) were collected on the seventh or eighth postovulatory day of their menstrual cycles for evaluation. Expression of BcL-2, FAS, FASL and cleaved caspase-3 was assessed using immunohistochemical methods. RESULTS Expression of BcL-2 and FAS was significantly higher and lower, respectively, in the women with idiopathic infertility than in the other groups (p<0.01). Expression of cleaved caspase-3 was significantly lower in the women with idiopathic infertility than in the other groups (p<0.01). Expression of FASL was similar in all three groups. CONCLUSION Disturbances in endometrial apoptosis may be a contributing factor in patients with idiopathic infertility and recurrent pregnancy loss.
Collapse
Affiliation(s)
- Michele Q Panzan
- Department of Gynaecology, Federal University of São Paulo, São Paulo, Brazil
| | | | | | | | | | | | | | | |
Collapse
|
72
|
Abstract
An intricate network of innate and immune cells and their derived mediators function in unison to protect us from toxic elements and infectious microbial diseases that are encountered in our environment. This vast network operates efficiently by use of a single cell epithelium in, for example, the gastrointestinal (GI) and upper respiratory (UR) tracts, fortified by adjoining cells and lymphoid tissues that protect its integrity. Perturbations certainly occur, sometimes resulting in inflammatory diseases or infections that can be debilitating and life threatening. For example, allergies in the eyes, skin, nose, and the UR or digestive tracts are common. Likewise, genetic background and environmental microbial encounters can lead to inflammatory bowel diseases (IBDs). This mucosal immune system (MIS) in both health and disease is currently under intense investigation worldwide by scientists with diverse expertise and interests. Despite this activity, there are numerous questions remaining that will require detailed answers in order to use the MIS to our advantage. In this issue of PLOS Biology, a research article describes a multi-scale in vivo systems approach to determine precisely how the gut epithelium responds to an inflammatory cytokine, tumor necrosis factor-alpha (TNF-α), given by the intravenous route. This article reveals a previously unknown pathway in which several cell types and their secreted mediators work in unison to prevent epithelial cell death in the mouse small intestine. The results of this interesting study illustrate how in vivo systems biology approaches can be used to unravel the complex mechanisms used to protect the host from its environment.
Collapse
Affiliation(s)
- Jerry R McGhee
- Department of Pediatric Dentistry, The School of Dentistry, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America.
| | | |
Collapse
|
73
|
Arnold M, Dai Y, Tso P, Langhans W. Meal-contingent intestinal lymph sampling from awake, unrestrained rats. Am J Physiol Regul Integr Comp Physiol 2012; 302:R1365-71. [PMID: 22513747 DOI: 10.1152/ajpregu.00497.2011] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Standard procedures for intestinal lymph collection involve continuous, quantitative drainage of the lymph fluid in anesthetized or restrained animals that are often euthanized within 48 h. We here describe a novel technique for the nonocclusive cannulation of the major intestinal lymph duct in rats that allows for repetitive in vivo sampling of intestinal lymph from unrestrained, awake, and ad libitum-fed animals. The distinctive feature of this novel technique is that a 5- to 7-mm long piece of Vialon tubing (OD/ID: 0.8/0.7 mm) with a small hole in its wall is first implanted into the major intestinal lymph duct for stabilization. The tapered tip (OD: ≈0.1 mm) of the catheter is then inserted into the hole of the tubing and fixed in place with a polyamid suture and a drop of tissue glue. In our hands, catheters implanted this way remain patent for up to 6 wk after surgery. In an initial experiment we collected lymph from six adult rats before (0) and 15, 30, 45, 60, 75, 90, 120, and 180 min (120 μl, each) after the onset of isocaloric (12.5 kcal) low-fat (LF) or high-fat (HF) test meals and measured active glucagon-like peptide-1 (GLP-1). Intestinal lymphatic GLP-1 concentration increased (P < 0.05) from ≈4 pmol/l (0 min) to a peak of 33 ± 6 (means ± SE) or 22 ± 4 pmol/l at 15 (HF) or 30 min (LF) after meal onset and gradually returned to baseline levels by 180 min. With this new technique fewer animals are required to generate physiologically relevant data for various aspects of gastrointestinal physiology that involve the lymphatic system. Furthermore, the advantage of this system is that the animal can act as its own control when the effect of different experimental protocols is tested.
Collapse
Affiliation(s)
- Myrtha Arnold
- Physiology and Behavior Laboratory, Institute of Food, Nutrition and Health, ETH Zurich, Schorenstr. 16, 8603 Schwerzenbach, Switzerland.
| | | | | | | |
Collapse
|
74
|
Mangold A, Lebherz D, Papay P, Liepert J, Hlavin G, Lichtenberger C, Adami A, Zimmermann M, Klaus D, Reinisch W, Ankersmit HJ. Anti-Gal titers in healthy adults and inflammatory bowel disease patients. Transplant Proc 2012; 43:3964-8. [PMID: 22172880 DOI: 10.1016/j.transproceed.2011.09.074] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2011] [Accepted: 09/20/2011] [Indexed: 12/20/2022]
Abstract
INTRODUCTION ALPHA-GAL is a glycoconjugate present on cell membranes of mammals and bacteria but not humans who display anti-Gal antibodies (AB) in high titers provoked by the commensal gut flora. In the present study, we sought to determine the longitudinal course of alpha-Gal specific AB titers of all isotypes over 8 weeks among healthy adult subjects. Furthermore, we hypothesized that inflammatory bowel disease (IBD) patients display increased anti-Gal titers. MATERIALS AND METHODS We drew serum from healthy probands (n=20) weekly for 8 weeks and obtained plasma samples of from patients suffering from Crohn's disease (n=20) and ulcerative colitis (n=20). We measured anti-Gal ABs of all isotypes and total immunoglobulin (Ig) content using an enzyme-linked immunosorbent assay technique. For statistical evaluation of the longitudinal titers, we calculated confidence intervals for the slopes of a random intercept model, comparing variances between and within the probands. For group comparisons, we performed paired student t-tests and Pearson correlations. RESULTS Alpha-Gal specific IgG, IgM, IgD, and IgA titers remained unvaried within a narrow range upon longitudinal observation. Most probands did not display alpha-Gal specific IgE ABs. Crohn's disease patients showed highly increased alpha-Gal-specific IgA titers compared with control subjects (P<.01). CONCLUSION Apart from IgE, alpha-Gal-specific ABs of all isotypes remained constant over longer time periods in healthy subjects. Thus, significant titer changes actually represent increased antigen exposure and a specific anti-alpha-Gal response. Crohn's disease patients display increased anti-Gal IgA titers compared with healthy controls, which reflects a chronically impaired mucosal gut barrier in this patient cohort.
Collapse
Affiliation(s)
- A Mangold
- Department of Thoracic Surgery, Medical University of Vienna, Vienna, Austria
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
75
|
Babiuch K, Gottschaldt M, Werz O, Schubert US. Particulate transepithelial drug carriers: barriers and functional polymers. RSC Adv 2012. [DOI: 10.1039/c2ra20726e] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
|
76
|
Nagler C, Wroblewska J. TL we meet again. Nat Immunol 2011; 12:1027-8. [PMID: 22012434 DOI: 10.1038/ni.2138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
77
|
Luyer MDP, Habes Q, Hak RV, Buurman W. Nutritional stimulation of the autonomic nervous system. World J Gastroenterol 2011; 17:3859-63. [PMID: 22025873 PMCID: PMC3198014 DOI: 10.3748/wjg.v17.i34.3859] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2010] [Revised: 06/21/2011] [Accepted: 06/28/2011] [Indexed: 02/06/2023] Open
Abstract
Disturbance of the inflammatory response in the gut is important in several clinical diseases ranging from inflammatory bowel disease to postoperative ileus. Several feedback mechanisms exist that control the inflammatory cascade and avoid collateral damage. In the gastrointestinal tract, it is of particular importance to control the immune response to maintain the balance that allows dietary uptake and utilization of nutrients on one hand, while preventing invasion of bacteria and toxins on the other hand. The process of digestion and absorption of nutrients requires a relative hyporesponsiveness of the immune cells in the gut to luminal contents which is not yet fully understood. Recently, the autonomic nervous system has been identified as an important pathway to control local and systemic inflammation and gut barrier integrity. Activation of the pathway is possible via electrical or via pharmacological interventions, but is also achieved in a physiological manner by ingestion of dietary lipids. Administration of dietary lipids has been shown to be very effective in reducing the inflammatory cascade and maintaining intestinal barrier integrity in several experimental studies. This beneficial effect of nutrition on the inflammatory response and intestinal barrier integrity opens new therapeutic opportunities for treatment of certain gastrointestinal disorders. Furthermore, this neural feedback mechanism provides more insight in the relative hyporesponsiveness of the immune cells in the gut. Here, we will discuss the regulatory function of the autonomic nervous system on the inflammatory response and gut barrier function and the potential benefit in a clinical setting.
Collapse
|
78
|
Odumosu O, Nicholas D, Payne K, Langridge W. Cholera toxin B subunit linked to glutamic acid decarboxylase suppresses dendritic cell maturation and function. Vaccine 2011; 29:8451-8. [PMID: 21807047 DOI: 10.1016/j.vaccine.2011.07.077] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2011] [Revised: 06/29/2011] [Accepted: 07/18/2011] [Indexed: 12/16/2022]
Abstract
Dendritic cells are the largest population of antigen presenting cells in the body. One of their main functions is to regulate the delicate balance between immunity and tolerance responsible for maintenance of immunological homeostasis. Disruption of this delicate balance often results in chronic inflammation responsible for initiation of organ specific autoimmune diseases such as rheumatoid arthritis, multiple sclerosis and type I diabetes. The cholera toxin B subunit (CTB) is a weak mucosal adjuvant known for its ability to stimulate immunity to antigenic proteins. However, conjugation of CTB to many autoantigens can induce immunological tolerance resulting in suppression of autoimmunity. In this study, we examined whether linkage of CTB to a 5kDa C-terminal protein fragment of the major diabetes autoantigen glutamic acid decarboxylase (GAD(35)), can block dendritic cell (DC) functions such as biosynthesis of co-stimulatory factor proteins CD86, CD83, CD80 and CD40 and secretion of inflammatory cytokines. The results of human umbilical cord blood monocyte-derived DC-GAD(35) autoantigen incubation experiments showed that inoculation of immature DCs (iDCs), with CTB-GAD(35) protein dramatically suppressed levels of CD86, CD83, CD80 and CD40 co-stimulatory factor protein biosynthesis in comparison with GAD(35) alone inoculated iDCs. Surprisingly, incubation of iDCs in the presence of the CTB-autoantigen and the strong immunostimulatory molecules PMA and Ionomycin revealed that CTB-GAD(35) was capable of arresting PMA+Ionomycin induced DC maturation. Consistent with this finding, CTB-GAD(35) mediated suppression of DC maturation was accompanied by a dramatic decrease in the secretion of the pro-inflammatory cytokines IL-12/23p40 and IL-6 and a significant increase in secretion of the immunosuppressive cytokine IL-10. Taken together, our experimental data suggest that linkage of the weak adjuvant CTB to the dominant type 1 diabetes autoantigen GAD strongly inhibits DC maturation through the down regulation of major co-stimulatory factors and inflammatory cytokine biosynthesis. These results emphasize the possibility that CTB-autoantigen fusion proteins enhance DC priming of naïve Th0 cell development in the direction of immunosuppressive T lymphocytes. The immunological phenomena observed here establish a basis for improvement of adjuvant augmented multi-component subunit vaccine strategies capable of complete suppression of organ-specific autoimmune diseases in vivo.
Collapse
Affiliation(s)
- Oludare Odumosu
- Center for Health Disparities and Molecular Medicine, Loma Linda University, Loma Linda, CA 92354, USA.
| | | | | | | |
Collapse
|
79
|
Heterologous prime-boost oral immunization with GK-1 peptide from Taenia crassiceps cysticerci induces protective immunity. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2011; 18:1067-76. [PMID: 21593234 DOI: 10.1128/cvi.05030-11] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Oral immunization is a goal in vaccine development, particularly for pathogens that enter the host through the mucosal system. This study was designed to explore the immunogenic properties of the Taenia crassiceps protective peptide GK-1 administered orally. Mice were orally immunized with the synthetic GK-1 peptide in its linear form with or without the Brucella lumazine synthase (BLS) protein adjuvant or as a chimera recombinantly bound to BLS (BLS-GK-1). Mice were boosted twice with GK-1 only at 15-day intervals. A significant rate of protection of 64.7% was achieved in GK-1-immunized mice, and that rate significantly increased to 91.8 and 96% when mice were primed with GK-1 coadministered with BLS as an adjuvant and BLS as a carrier, respectively. Specific antibodies and T cell activation and proliferation accompanied the protection induced, revealing the potent immunogenicity of GK-1. Through immunohistochemical studies, GK-1 was detected in T and B cell zones of the Peyer's patches (PP) and mesenteric lymph nodes. In the latter, abundant proliferating cells were detected by 5'-bromo-2'-deoxyuridine incorporation. No proliferation was detected in PP. Altogether, these results portray the potent immunogenic properties of GK-1 administered orally and reinforce the usefulness of BLS as an adjuvant and adequate vaccine delivery system for oral vaccines.
Collapse
|
80
|
Zivkovic AM, Barile D. Bovine milk as a source of functional oligosaccharides for improving human health. Adv Nutr 2011; 2:284-9. [PMID: 22332060 PMCID: PMC3090169 DOI: 10.3945/an.111.000455] [Citation(s) in RCA: 116] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Human milk oligosaccharides are complex sugars that function as selective growth substrates for specific beneficial bacteria in the gastrointestinal system. Bovine milk is a potentially excellent source of commercially viable analogs of these unique molecules. However, bovine milk has a much lower concentration of these oligosaccharides than human milk, and the majority of the molecules are simpler in structure than those found in human milk. Specific structural characteristics of milk-derived oligosaccharides are crucial to their ability to selectively enrich beneficial bacteria while inhibiting or being less than ideal substrates for undesirable and pathogenic bacteria. Thus, if bovine milk products are to provide human milk-like benefits, it is important to identify specific dairy streams that can be processed commercially and cost-effectively and that can yield specific oligosaccharide compositions that will be beneficial as new food ingredients or supplements to improve human health. Whey streams have the potential to be commercially viable sources of complex oligosaccharides that have the structural resemblance and diversity of the bioactive oligosaccharides in human milk. With further refinements to dairy stream processing techniques and functional testing to identify streams that are particularly suitable for enriching beneficial intestinal bacteria, the future of oligosaccharides isolated from dairy streams as a food category with substantiated health claims is promising.
Collapse
|
81
|
Noguchi K, Okumura F, Takahashi N, Kataoka A, Kamiyama T, Todo S, Hatakeyama S. TRIM40 promotes neddylation of IKKγ and is downregulated in gastrointestinal cancers. Carcinogenesis 2011; 32:995-1004. [PMID: 21474709 DOI: 10.1093/carcin/bgr068] [Citation(s) in RCA: 103] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Gastrointestinal neoplasia seems to be a common consequence of chronic inflammation in the gastrointestinal epithelium. Nuclear factor-kappaB (NF-κB) is an important transcription factor for carcinogenesis in chronic inflammatory diseases and plays a key role in promoting inflammation-associated carcinoma in the gastrointestinal tract. Activation of NF-κB is regulated by several posttranslational modifications including phosphorylation, ubiquitination and neddylation. In this study, we showed that tripartite motif (TRIM) 40 is highly expressed in the gastrointestinal tract and that TRIM40 physically binds to Nedd8, which is conjugated to target proteins by neddylation. We also found that TRIM40 promotes the neddylation of inhibitor of nuclear factor kappaB kinase subunit gamma, which is a crucial regulator for NF-κB activation, and consequently causes inhibition of NF-κB activity, whereas a dominant-negative mutant of TRIM40 lacking the RING domain does not inhibit NF-κB activity. Knockdown of TRIM40 in the small intestinal epithelial cell line IEC-6 caused NF-κB activation followed by increased cell growth. In addition, we found that TRIM40 is highly expressed in normal gastrointestinal epithelia but that TRIM40 is downregulated in gastrointestinal carcinomas and chronic inflammatory lesions of the gastrointestinal tract. These findings suggest that TRIM40 inhibits NF-κB activity via neddylation of inhibitor of nuclear factor kappaB kinase subunit gamma and that TRIM40 prevents inflammation-associated carcinogenesis in the gastrointestinal tract.
Collapse
Affiliation(s)
- Keita Noguchi
- Department of Biochemistry, Hokkaido University Graduate School of Medicine, N15, W7, Kita-ku, Sapporo, Hokkaido 060-8638, Japan
| | | | | | | | | | | | | |
Collapse
|
82
|
Sawa S, Lochner M, Satoh-Takayama N, Dulauroy S, Bérard M, Kleinschek M, Cua D, Di Santo JP, Eberl G. RORγt+ innate lymphoid cells regulate intestinal homeostasis by integrating negative signals from the symbiotic microbiota. Nat Immunol 2011; 12:320-6. [PMID: 21336274 DOI: 10.1038/ni.2002] [Citation(s) in RCA: 494] [Impact Index Per Article: 35.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2010] [Accepted: 02/01/2011] [Indexed: 12/12/2022]
Abstract
Lymphoid cells that express the nuclear hormone receptor RORγt are involved in containment of the large intestinal microbiota and defense against pathogens through the production of interleukin 17 (IL-17) and IL-22. They include adaptive IL-17-producing helper T cells (T(H)17 cells), as well as innate lymphoid cells (ILCs) such as lymphoid tissue-inducer (LTi) cells and IL-22-producing NKp46+ cells. Here we show that in contrast to T(H)17 cells, both types of RORγt+ ILCs constitutively produced most of the intestinal IL-22 and that the symbiotic microbiota repressed this function through epithelial expression of IL-25. This function was greater in the absence of adaptive immunity and was fully restored and required after epithelial damage, which demonstrates a central role for RORγt+ ILCs in intestinal homeostasis. Our data identify a finely tuned equilibrium among intestinal symbionts, adaptive immunity and RORγt+ ILCs.
Collapse
Affiliation(s)
- Shinichiro Sawa
- Institut Pasteur, Lymphoid Tissue Development Unit, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
83
|
|
84
|
Derikx JPM, Luyer MDP, Heineman E, Buurman WA. Non-invasive markers of gut wall integrity in health and disease. World J Gastroenterol 2011. [PMID: 21072889 DOI: 10.3748/wjg.v16.i43.5272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The intestinal mucosa is responsible for the absorption of nutrients from the lumen and for the separation of the potentially toxic luminal content (external environment) from the host (internal environment). Disruption of this delicate balance at the mucosal interface is the basis for numerous (intestinal) diseases. Experimental animal studies have shown that gut wall integrity loss is involved in the development of various inflammatory syndromes, including post-operative or post-traumatic systemic inflammatory response syndrome, sepsis, and multiple organ failure. Assessment of gut wall integrity in clinical practice is still a challenge, as it is difficult to evaluate the condition of the gut non-invasively with currently available diagnostic tools. Moreover, non-invasive, rapid diagnostic means to assess intestinal condition are needed to evaluate the effects of treatment of intestinal disorders. This review provides a survey of non-invasive tests and newly identified markers that can be used to assess gut wall integrity.
Collapse
|
85
|
Schirbel A, Fiocchi C. Targeting the innate immune system in pediatric inflammatory bowel disease. Expert Rev Gastroenterol Hepatol 2011; 5:33-41. [PMID: 21309670 DOI: 10.1586/egh.10.76] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The pathogenesis of inflammatory bowel disease (IBD) is complex and involves both innate and adaptive immune responses. This article addresses, in a selective and speculative fashion, the topic of how the various components of the intestinal innate immune system can be manipulated for the purpose of developing new therapeutic approaches. These various components include: agents that stimulate mucosal innate immune responses, such as food components and the gut microbiota; cells that directly respond to these stimuli, including epithelial cells, macrophages and dendritic cells; and molecules that mediate innate immune responses, such as Toll-like receptors and protein kinases. Downregulation of excessive innate immune responses makes therapeutic sense in both pediatric and adult IBD. However, because IBD is complex and characteristically chronic, major alterations of adaptive immunity are also involved in the mediation of inflammation. Thus, novel and truly effective approaches to treat IBD will undoubtedly require intervening in the innate as well as the adaptive branches of the mucosal immunity.
Collapse
Affiliation(s)
- Anja Schirbel
- Department of Hepatology and Gastroenterology, Campus Virchow-Klinikum, Charité Universitätsmedizin, Berlin, Germany
| | | |
Collapse
|
86
|
Broquet AH, Hirata Y, McAllister CS, Kagnoff MF. RIG-I/MDA5/MAVS are required to signal a protective IFN response in rotavirus-infected intestinal epithelium. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2011; 186:1618-26. [PMID: 21187438 DOI: 10.4049/jimmunol.1002862] [Citation(s) in RCA: 170] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Rotavirus is a dsRNA virus that infects epithelial cells that line the surface of the small intestine. It causes severe diarrheal illness in children and ∼500,000 deaths per year worldwide. We studied the mechanisms by which intestinal epithelial cells (IECs) sense rotavirus infection and signal IFN-β production, and investigated the importance of IFN-β production by IECs for controlling rotavirus production by intestinal epithelium and virus excretion in the feces. In contrast with most RNA viruses, which interact with either retinoic acid-inducible gene I (RIG-I) or melanoma differentiation-associated gene 5 (MDA5) inside cells, rotavirus was sensed by both RIG-I and MDA5, alone and in combination. Rotavirus did not signal IFN-β through either of the dsRNA sensors TLR3 or dsRNA-activated protein kinase (PKR). Silencing RIG-I or MDA5, or their common adaptor protein mitochondrial antiviral signaling protein (MAVS), significantly decreased IFN-β production and increased rotavirus titers in infected IECs. Overexpression of laboratory of genetics and physiology 2, a RIG-I-like receptor that interacts with viral RNA but lacks the caspase activation and recruitment domains required for signaling through MAVS, significantly decreased IFN-β production and increased rotavirus titers in infected IECs. Rotavirus-infected mice lacking MAVS, but not those lacking TLR3, TRIF, or PKR, produced significantly less IFN-β and increased amounts of virus in the intestinal epithelium, and shed increased quantities of virus in the feces. We conclude that RIG-I or MDA5 signaling through MAVS is required for the activation of IFN-β production by rotavirus-infected IECs and has a functionally important role in determining the magnitude of rotavirus replication in the intestinal epithelium.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/deficiency
- Adaptor Proteins, Signal Transducing/physiology
- Animals
- Cell Line
- Chlorocebus aethiops
- DEAD Box Protein 58
- DEAD-box RNA Helicases/deficiency
- DEAD-box RNA Helicases/physiology
- HT29 Cells
- Humans
- Interferon-Induced Helicase, IFIH1
- Interferon-beta/biosynthesis
- Interferon-beta/physiology
- Intestinal Mucosa/enzymology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/virology
- Membrane Proteins/deficiency
- Membrane Proteins/physiology
- Mice
- Mice, 129 Strain
- Mice, Inbred C57BL
- Mice, Knockout
- Nerve Tissue Proteins/deficiency
- Nerve Tissue Proteins/physiology
- RNA Helicases/genetics
- RNA Helicases/physiology
- RNA, Viral/biosynthesis
- RNA, Viral/genetics
- Receptors, Cell Surface
- Receptors, Immunologic
- Response Elements/immunology
- Rotavirus/genetics
- Rotavirus/immunology
- Signal Transduction/genetics
- Signal Transduction/immunology
- Virus Replication/genetics
- Virus Replication/immunology
Collapse
Affiliation(s)
- Alexis H Broquet
- Laboratory of Mucosal Immunology, Department of Medicine, University of California, San Diego, La Jolla, CA 92093, USA
| | | | | | | |
Collapse
|
87
|
Liu C, Duan ML, Li A, Weng YB, Wang BE, Zhang SW. Effects of treatment with Tongfu Granules on intestinal cytokine levels in rats with endotoxemia. Shijie Huaren Xiaohua Zazhi 2010; 18:3515-3519. [DOI: 10.11569/wcjd.v18.i33.3515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To observe the changes in the levels of intestinal cytokines in rats with endotoxemia and to investigate the effects of treatment with Tongfu Granules on intestinal cytokine levels.
METHODS: One hundred and twenty male Wistar rats were randomly divided into three groups: control group, endotoxemia group and treatment group. Endotoxemia was induced by a single injection of LPS via the caudal vein. The levels of intestinal cytokines were measured by ELISA and radioimmunoassay.
RESULTS: The level of IFN-γ at 24 h was significantly lower in the endotoxemia group than in the control and treatment groups (32.93 ng/L ± 13.17 ng/L, 37.14 ng/L ± 6.70 ng/L vs 23.21 ng/L ± 8.65 ng/L, both P < 0.05). The level of IFN-γ at 12 h was significantly higher in the treatment group than in the control group (39.96 ng/L ± 8.26 ng/L vs 29.64 ng/L ± 10.78 ng/L, P < 0.05). The level of IL-4 significantly decreased in the endotoxemia group at 6 h but increased at 24 h compared with the control and treatment groups (115.56 ng/L ± 17.67 ng/L, 124.39 ng/L ± 26.02 ng/L vs 83.36 ng/L ± 27.24 ng/L, both P < 0.05 or 0.01). IL-4 level at 24 h was significantly higher in the endotoxemia group than in the control group (107.96 ng/L ± 10.86 ng/L vs 92.21 ng/L ± 17.62 ng/L, P < 0.05). The levels of Foxp3 at 12 and 24 h were significantly higher in the endotoxemia group than in the control group (0.28 g/L ± 0.037 g/L vs 0.24 g/L ± 0.041 g/L; 0.26 g/L ± 0.029 g/L vs 0.22 g/L ± 0.041 g/L, both P < 0.05). The level of Foxp3 at 24 h was significantly lower in the treatment group than in the endotoxemia group (0.23 g/L ± 0.030 g/L vs 0.26 g/L ± 0.029 g/L, P < 0.05). The levels of sIgA at 2, 12 and 24 h was significantly lower in the endotoxemia group than in the control and treatment groups (2.43 × 10-11 ± 0.23 × 10-11, 2.18 × 10-11 ± 0.27 × 10-11vs 1.66 × 10-11 ± 0.25 × 10-11; 2.76 × 10-11 ± 0.22 × 10-11, 2.64 × 10-11 ± 0.22 × 10-11vs 2.09 × 10-11 ± 0.20 × 10-11; 2.48 × 10-11 ± 0.31 × 10-11, 2.25 × 10-11 ± 0.44 × 10-11vs 1.88 × 10-11 ± 0.45 × 10-11, all P < 0.05). The level of sIgA at 2 h was significantly lower in the treatment group than in the control group.
CONCLUSION: Small intestinal mucosa injury and deregulated inflammatory reaction can result in intestinal immunodepression in rats with endotoxemia. Treatment with Tongfu Granules can protect intestinal immune function in rats with endotoxemia by reducing small intestinal mucosa injury and bidirectionally regulating inflammatory reaction.
Collapse
|
88
|
Derikx JPM, Luyer MDP, Heineman E, Buurman WA. Non-invasive markers of gut wall integrity in health and disease. World J Gastroenterol 2010; 16:5272-9. [PMID: 21072889 PMCID: PMC2980675 DOI: 10.3748/wjg.v16.i42.5272] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The intestinal mucosa is responsible for the absorption of nutrients from the lumen and for the separation of the potentially toxic luminal content (external environment) from the host (internal environment). Disruption of this delicate balance at the mucosal interface is the basis for numerous (intestinal) diseases. Experimental animal studies have shown that gut wall integrity loss is involved in the development of various inflammatory syndromes, including post-operative or post-traumatic systemic inflammatory response syndrome, sepsis, and multiple organ failure. Assessment of gut wall integrity in clinical practice is still a challenge, as it is difficult to evaluate the condition of the gut non-invasively with currently available diagnostic tools. Moreover, non-invasive, rapid diagnostic means to assess intestinal condition are needed to evaluate the effects of treatment of intestinal disorders. This review provides a survey of non-invasive tests and newly identified markers that can be used to assess gut wall integrity.
Collapse
|
89
|
Corthësy B. Secretory immunoglobulin A: well beyond immune exclusion at mucosal surfaces. Immunopharmacol Immunotoxicol 2010; 31:174-9. [PMID: 19514992 DOI: 10.1080/08923970802438441] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
At mucosal surfaces, secretory IgA (SIgA) antibodies serve as the first line of defense against microorganisms through a mechanism called immune exclusion that prevents interaction of neutralized antigens with the epithelium. In addition, SIgA plays a role in the immune balance of the epithelial barrier through selective adhesion to M cells in intestinal Peyer's patches. This mediates the transepithelial retro-transport of the antibody and associated antigens from the intestinal lumen to underlying gut-associated organized lymphoid tissue. In Peyer's patches, SIgA-based immune complexes are internalized by underlying antigen-presenting cells, leaving the antigen with masked epitopes, a form that limits the risk of overwhelming the local immune protection system with danger signals. This translates into the onset of mucosal and systemic responses associated with production of anti-inflammatory cytokines and limited activation of antigen-presenting cells. In the gastrointestinal tract, SIgA exhibits thus properties of a neutralizing agent (immune exclusion) and of an immunopotentiator inducing effector immune responses in a noninflammatory context favorable to preserve local homeostasis.
Collapse
Affiliation(s)
- Blaise Corthësy
- R & D Laboratory, Division of Immunology and Allergy, University State Hospital (CHUV), Lausanne, Switzerland.
| |
Collapse
|
90
|
He X, McMahon S, Henderson TD, Griffey SM, Cheng LW. Ricin toxicokinetics and its sensitive detection in mouse sera or feces using immuno-PCR. PLoS One 2010; 5:e12858. [PMID: 20877567 PMCID: PMC2943921 DOI: 10.1371/journal.pone.0012858] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2010] [Accepted: 08/23/2010] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Ricin (also called RCA-II or RCA(60)), one of the most potent toxins and documented bioweapons, is derived from castor beans of Ricinus communis. Several in vitro methods have been designed for ricin detection in complex food matrices in the event of intentional contamination. Recently, a novel Immuno-PCR (IPCR) assay was developed with a limit of detection of 10 fg/ml in a buffer matrix and about 10-1000-fold greater sensitivity than other methods in various food matrices. METHODS AND FINDINGS In order to devise a better diagnostic test for ricin, the IPCR assay was adapted for the detection of ricin in biological samples collected from mice after intoxication. The limit of detection in both mouse sera and feces was as low as 1 pg/ml. Using the mouse intravenous (iv) model for ricin intoxication, a biphasic half-life of ricin, with a rapid t(1/2)α of 4 min and a slower t(1/2)β of 86 min were observed. The molecular biodistribution time for ricin following oral ingestion was estimated using an antibody neutralization assay. Ricin was detected in the blood stream starting at approximately 6-7 h post- oral intoxication. Whole animal histopathological analysis was performed on mice treated orally or systemically with ricin. Severe lesions were observed in the pancreas, spleen and intestinal mesenteric lymph nodes, but no severe pathology in other major organs was observed. CONCLUSIONS The determination of in vivo toxicokinetics and pathological effects of ricin following systemic and oral intoxication provide a better understanding of the etiology of intoxication and will help in the future design of more effective diagnostic and therapeutic methods.
Collapse
Affiliation(s)
- Xiaohua He
- Foodborne Contaminants Research Unit, Western Regional Research Center, United States Department of Agriculture, Agricultural Research Service, Albany, California, United States of America
| | - Stephanie McMahon
- Foodborne Contaminants Research Unit, Western Regional Research Center, United States Department of Agriculture, Agricultural Research Service, Albany, California, United States of America
| | - Thomas D. Henderson
- Foodborne Contaminants Research Unit, Western Regional Research Center, United States Department of Agriculture, Agricultural Research Service, Albany, California, United States of America
| | - Stephen M. Griffey
- Comparative Pathology Lab, School of Veterinary Medicine, University of California Davis, Davis, California, United States of America
| | - Luisa W. Cheng
- Foodborne Contaminants Research Unit, Western Regional Research Center, United States Department of Agriculture, Agricultural Research Service, Albany, California, United States of America
| |
Collapse
|
91
|
Abstract
Diet and exercise are primary strategies recommended for the control of the obesity epidemic. Considerable attention is being paid to the effect of both on the immune system. However, little research has been done on the effect of diet, nutrients or exercise on the mucosal immune system. The gastrointestinal tract (gut) is not only responsible for the entry of nutrients into the organism, but also for triggering the primary immune response to orally ingested antigens. The gut-associated lymphoid tissue contains a large amount of immune cells, disseminated all along the intestine in Peyer's patches and lamina propria. Specific nutrients or their combinations, as well as the microflora, are capable of modulating the immune system through cell activation, production of signalling molecules or gene expression. We have observed an increase in T-cells as well as a decrease in B-cells from Peyer's patches, induced by diets high in fats or carbohydrates in Balb/c mice. It has also been demonstrated that exercise modulates the immune system, where moderate levels may improve its function by increasing the proliferation of lymphocytes from various sites, including gut-associated lymphoid tissue, whereas exhaustive acute exercise may cause immunosuppression. High-fat diets combined with exercise are able to induce an increase in CD3+ lymphocytes due to increased CD8+ cells and a decrease in B-cells. Explanations and consequences of the effects of diet and exercise on the gut mucosal immunity are still being explored.
Collapse
|
92
|
Mathias A, Duc M, Favre L, Benyacoub J, Blum S, Corthésy B. Potentiation of polarized intestinal Caco-2 cell responsiveness to probiotics complexed with secretory IgA. J Biol Chem 2010; 285:33906-13. [PMID: 20729211 DOI: 10.1074/jbc.m110.135111] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The precise mechanisms underlying the interaction between intestinal bacteria and the host epithelium lead to multiple consequences that remain poorly understood at the molecular level. Deciphering such events can provide valuable information as to the mode of action of commensal and probiotic microorganisms in the gastrointestinal environment. Potential roles of such microorganisms along the privileged target represented by the mucosal immune system include maturation prior, during and after weaning, and the reduction of inflammatory reactions in pathogenic conditions. Using human intestinal epithelial Caco-2 cell grown as polarized monolayers, we found that association of a Lactobacillus or a Bifidobacterium with nonspecific secretory IgA (SIgA) enhanced probiotic adhesion by a factor of 3.4-fold or more. Bacteria alone or in complex with SIgA reinforced transepithelial electrical resistance, a phenomenon coupled with increased phosphorylation of tight junction proteins zonula occludens-1 and occludin. In contrast, association with SIgA resulted in both enhanced level of nuclear translocation of NF-κB and production of epithelial polymeric Ig receptor as compared with bacteria alone. Moreover, thymic stromal lymphopoietin production was increased upon exposure to bacteria and further enhanced with SIgA-based complexes, whereas the level of pro-inflammatory epithelial cell mediators remained unaffected. Interestingly, SIgA-mediated potentiation of the Caco-2 cell responsiveness to the two probiotics tested involved Fab-independent interaction with the bacteria. These findings add to the multiple functions of SIgA and underscore a novel role of the antibody in interaction with intestinal bacteria.
Collapse
Affiliation(s)
- Amandine Mathias
- R&D Laboratory of the Division of Immunology and Allergy, Centre Hospitalier Universitaire Vaudois, Rue du Bugnon, 1011 Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
93
|
Suenaert P, Maerten P, Van Assche G, Van Driessche W, Geboes K, Bulteel V, Simaels J, Augustijns P, Ceuppens JL, Rutgeerts P, Perrier C. Effects of T cell-induced colonic inflammation on epithelial barrier function. Inflamm Bowel Dis 2010; 16:1322-31. [PMID: 20155850 DOI: 10.1002/ibd.21211] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Epithelial barrier disturbance is thought to contribute to the pathogenesis of inflammatory bowel diseases; however, it remains unclear whether it is a primary defect participating to the onset of inflammation or only a consequence of sustained inflammation. METHODS A time course study of epithelial barrier functions and immune mediators was performed in the CD4(+)CD45RB(hi) T cell transfer model of colitis using Ussing chambers. RESULTS In nonreconstituted severe combined immunodeficiency (SCID) mice, no epithelial dysfunction was observed. However, after transfer of CD4(+)CD45RB(hi) T cells or total CD4(+) T cells, colon of SCID mice displayed a decreased epithelial resistance, even before overt microscopic inflammation had occurred. Sustained colitis of CD4(+)CD45RB(hi) T cell reconstituted mice was also associated with enhanced subepithelial resistance, enhanced paracellular permeability, and decreased net ion transport. All these reflect a disturbance of barrier function and may contribute to diarrhea. Epithelial resistance was positively correlated with interleukin 10 (IL-10) and transforming growth factor beta (TGF-beta) levels and net ion transport inversely correlated with tumor necrosis factor alpha (TNF-alpha) levels, pointing to the protective effect of IL-10 and TGF-beta and to a damaging effect of TNF-alpha. Indomethacin, a nonselective COX inhibitor, decreased epithelial resistance independent of T cells and inflammation, but its effect was more pronounced in inflamed colon. CONCLUSIONS Induction of colitis by transfer of CD4(+)CD45RB(hi) T cells in SCID mice leads to changes in the colonic epithelium before colitis develops. Decreased epithelium resistance might contribute to the development of colitis; however, it is not sufficient to lead to chronic inflammation.
Collapse
Affiliation(s)
- Peter Suenaert
- Division of Gastroenterology, University Hospital, Leuven, Belgium
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
94
|
Corthésy B. Role of secretory immunoglobulin A and secretory component in the protection of mucosal surfaces. Future Microbiol 2010; 5:817-29. [PMID: 20441552 DOI: 10.2217/fmb.10.39] [Citation(s) in RCA: 86] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The contribution of secretory immunoglobulin A (SIgA) antibodies in the defense of mucosal epithelia plays an important role in preventing pathogen adhesion to host cells, therefore blocking dissemination and further infection. This mechanism, referred to as immune exclusion, represents the dominant mode of action of the antibody. However, SIgA antibodies combine multiple facets, which together confer properties extending from intracellular and serosal neutralization of antigens, activation of non-inflammatory pathways and homeostatic control of the endogenous microbiota. The sum of these features suggests that future opportunities for translational application from research-based knowledge to clinics include the mucosal delivery of bioactive antibodies capable of preserving immunoreactivity in the lung, gastrointestinal tract, the genito-urinary tract for the treatment of infections. This article covers topics dealing with the structure of SIgA, the dissection of its mode of action in epithelia lining different mucosal surfaces and its potential in immunotherapy against infectious pathogens.
Collapse
Affiliation(s)
- Blaise Corthésy
- R&D Laboratory of the Department of Immunology & Allergy, University State Hospital (CHUV), Rue du Bugnon 46, 1011 Lausanne, Switzerland.
| |
Collapse
|
95
|
|
96
|
Masopust D, Choo D, Vezys V, Wherry EJ, Duraiswamy J, Akondy R, Wang J, Casey KA, Barber DL, Kawamura KS, Fraser KA, Webby RJ, Brinkmann V, Butcher EC, Newell KA, Ahmed R. Dynamic T cell migration program provides resident memory within intestinal epithelium. J Exp Med 2010; 207:553-64. [PMID: 20156972 PMCID: PMC2839151 DOI: 10.1084/jem.20090858] [Citation(s) in RCA: 494] [Impact Index Per Article: 32.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2009] [Accepted: 01/13/2010] [Indexed: 01/01/2023] Open
Abstract
Migration to intestinal mucosa putatively depends on local activation because gastrointestinal lymphoid tissue induces expression of intestinal homing molecules, whereas skin-draining lymph nodes do not. This paradigm is difficult to reconcile with reports of intestinal T cell responses after alternative routes of immunization. We reconcile this discrepancy by demonstrating that activation within spleen results in intermediate induction of homing potential to the intestinal mucosa. We further demonstrate that memory T cells within small intestine epithelium do not routinely recirculate with memory T cells in other tissues, and we provide evidence that homing is similarly dynamic in humans after subcutaneous live yellow fever vaccine immunization. These data explain why systemic immunization routes induce local cell-mediated immunity within the intestine and indicate that this tissue must be seeded with memory T cell precursors shortly after activation.
Collapse
Affiliation(s)
- David Masopust
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Daniel Choo
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Vaiva Vezys
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - E. John Wherry
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Jaikumar Duraiswamy
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Rama Akondy
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Jun Wang
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Kerry A. Casey
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Daniel L. Barber
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Kim S. Kawamura
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Kathryn A. Fraser
- Department of Microbiology, Center for Immunology, University of Minnesota, Minneapolis, MN 55455
| | - Richard J. Webby
- Department of Infectious Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105
| | - Volker Brinkmann
- Autoimmunity, Transplantation, and Inflammation, Novartis Institutes for Biomedical Research, CH-4002 Basel, Switzerland
| | - Eugene C. Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305
| | - Kenneth A. Newell
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| | - Rafi Ahmed
- Emory Vaccine Center and Department of Microbiology and Immunology and Department of Pathology, Emory University School of Medicine, Atlanta GA 30322
| |
Collapse
|
97
|
Protective effect of glutamine-enriched early enteral nutrition on intestinal mucosal barrier injury after liver transplantation in rats. Am J Surg 2010; 199:35-42. [PMID: 20103064 DOI: 10.1016/j.amjsurg.2008.11.039] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2008] [Revised: 11/28/2008] [Accepted: 11/28/2008] [Indexed: 01/25/2023]
Abstract
BACKGROUND The effect of glutamine-enriched early enteral nutrition (Gln-EEN) on intestinal mucosal barrier injury after liver transplantation (LT) remains uncertain. METHODS The Wistar-to-Wistar rat LT model was used to explore the protective effect of Gln-EEN. Morphologic changes of intestinal mucosa, levels of intestinal malondialdehyde and secretory immunoglobulin (sIgA), plasma endotoxin, D-lactic acid, serum tumor necrosis factor-alpha (TNF-alpha), rates of bacterial translocation, and expression of intestinal nuclear factor-kappaB, TNF-alpha, and intercellular adhesion molecule-1 were determined. RESULTS After LT, intestinal mucosa was damaged seriously. At 12, 24, and 48 hours posttransplantation, levels of intestinal sIgA were decreased; levels of malondialdehyde, endotoxin, D-lactic acid, and TNF-alpha, the ratio of bacterial translocation, and the expression of intestinal nuclear factor-kappaB, TNF-alpha, and intercellular adhesion molecule-1 all were increased. However, changes in earlier-mentioned parameters in recipients treated with Gln-EEN were attenuated remarkably at 24 to 48 hours. CONCLUSIONS Our data show that Gln-EEN is a potent protectant against intestinal mucosal barrier injury after LT.
Collapse
|
98
|
Cell-secreted Gp96-Ig-peptide complexes induce lamina propria and intraepithelial CD8+ cytotoxic T lymphocytes in the intestinal mucosa. Mucosal Immunol 2010; 3:182-92. [PMID: 19924120 DOI: 10.1038/mi.2009.127] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Induction of mucosal immunity is critical for protection from enteric pathogens. Heat shock protein gp96 is one of the primary peptide and protein chaperones located in the endoplasmic reticulum. We reported previously that a cell-secreted gp96-Ig fusion protein (gp96-Ig) mediated strong systemic, antigen-specific CD8-CTL expansion in vivo. We now evaluate the mucosal immune response to stimulation by secreted gp96 using allogeneic NIH-3T3 transfected with ovalbumin (OVA) and gp96-Ig. A single intraperitoneal NIH-3T3-OVA-gp96-Ig immunization caused significant homing of OVA-specific TCR transgenic CD8 cells (OT-I) to Peyer's patches, to the intraepithelial compartment and to the lamina propria. Intraperitoneal immunization with cells secreting gp96-Ig provided stronger mucosal immunity than the same dose instilled vaginally or rectally or injected subcutaneously or intradermally. Our results provide the first evidence that cell-based gp96-Ig-secreting vaccines may serve as a potent modality to induce mucosal immunity.
Collapse
|
99
|
Borges O, Lebre F, Bento D, Borchard G, Junginger HE. Mucosal vaccines: recent progress in understanding the natural barriers. Pharm Res 2010; 27:211-23. [PMID: 19953309 DOI: 10.1007/s11095-009-0011-3] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2009] [Accepted: 11/12/2009] [Indexed: 10/24/2022]
Abstract
It has long been known that protection against pathogens invading the organism via mucosal surfaces correlates better with the presence of specific antibodies in local secretions than with serum antibodies. The most effective way to induce mucosal immunity is to administer antigens directly to the mucosal surface. The development of vaccines for mucosal application requires antigen delivery systems and immunopotentiators that efficiently facilitate the presentation of the antigen to the mucosal immune system. This review provides an overview of the events within mucosal tissues that lead to protective mucosal immune responses. The understanding of those biological mechanisms, together with knowledge of the technology of vaccines and adjuvants, provides guidance on important technical aspects of mucosal vaccine design. Not being exhaustive, this review also provides information related to modern adjuvants, including polymeric delivery systems and immunopotentiators.
Collapse
Affiliation(s)
- Olga Borges
- Centre for Neuroscience and Cell Biology & Faculty of Pharmacy, University of Coimbra, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, 3000-548, Coimbra, Portugal.
| | | | | | | | | |
Collapse
|
100
|
Srinivasan N. Telling apart friend from foe: discriminating between commensals and pathogens at mucosal sites. Innate Immun 2010; 16:391-404. [PMID: 20083498 DOI: 10.1177/1753425909357577] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
From the moment we are born, we are exposed to a vast variety of microbes. The intestine in particular is perhaps inhabited by the largest number of microbes, consisting of both established commensals as well as sporadic pathogens. Mucosal surfaces form an important barrier against microbial invasion. Together with the physical barrier that they provide, mucosal surfaces also rely on innate immune functions to sense luminal microbes and signal accordingly to generate protective immune responses. However, since innate immune recognition is microbial specific and antigen-independent, the contact with both beneficial commensals and harmful pathogens creates the need for discrimination between the two. The mechanisms governing the ability of the mucosal immune system to discriminate between commensals and pathogens have long been unclear; however, recent discoveries have shed some light on this distinction. This review will summarize the current theories put forth to explain how the mucosal immune system maintains tolerance towards commensals while retaining the ability to mount inflammatory responses against pathogens.
Collapse
Affiliation(s)
- Naren Srinivasan
- Sir William Dunn School of Pathology, University of Oxford, Oxford, UK.
| |
Collapse
|