1
|
Błaszczyk F, Sosinka A, Wilczek G, Student S, Rost-Roszkowska M. Effect of gluten on the digestive tract and fat body of Telodeinopus aoutii (Diplopoda). J Morphol 2023; 284:e21546. [PMID: 36533734 DOI: 10.1002/jmor.21546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022]
Abstract
Adult specimens or larvae of invertebrates used as food for vertebrates are often maintained close to gluten so they might become vectors for cereal proteins. However, the tissues and internal organs can respond differently in animals with different feeding habits. The midgut epithelium might be a first and sufficient barrier preventing uptake and effects of gluten on the whole body, while the fat body is the main organ that accumulates different xenobiotics. Good models for such research are animals that do not feed on gluten-rich products in their natural environment. The project's goal was to investigate alterations in the midgut epithelium and fat body of the herbivorous millipede Telodeinopus aoutii (Diplopoda) and analyze cell death processes activated by gluten. It enabled us to determine whether changes were intensified or reversed by adaptive mechanisms. Adult specimens were divided into control and experimental animals fed with mushrooms supplemented with gluten and analyzed using transmission electron microscopy, flow cytometry, and confocal microscopy. Two organs were isolated for the qualitative and quantitative analysis: the midgut and the fat body. Our study of the herbivorous T. aoutii which does not naturally feed on gluten containing diet showed that continuous and prolonged gluten feeding activates repair processes that inhibit the processes of cell death (apoptosis and necrosis) and induce an increase in cell viability.
Collapse
Affiliation(s)
- Florentyna Błaszczyk
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Agnieszka Sosinka
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Grażyna Wilczek
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| | - Sebastian Student
- Faculty of Automatic Control, Electronics and Computer Science, Silesian University of Technology, Gliwice, Poland.,Biotechnology Center, Silesian University of Technology, Gliwice, Poland
| | - Magdalena Rost-Roszkowska
- Institute of Biology, Biotechnology and Environmental Protection, University of Silesia in Katowice, Katowice, Poland
| |
Collapse
|
2
|
Segura V, Ruiz-Carnicer Á, Sousa C, Moreno MDL. New Insights into Non-Dietary Treatment in Celiac Disease: Emerging Therapeutic Options. Nutrients 2021; 13:2146. [PMID: 34201435 PMCID: PMC8308370 DOI: 10.3390/nu13072146] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/11/2021] [Accepted: 06/15/2021] [Indexed: 02/07/2023] Open
Abstract
To date, the only treatment for celiac disease (CD) consists of a strict lifelong gluten-free diet (GFD), which has numerous limitations in patients with CD. For this reason, dietary transgressions are frequent, implying intestinal damage and possible long-term complications. There is an unquestionable need for non-dietary alternatives to avoid damage by involuntary contamination or voluntary dietary transgressions. In recent years, different therapies and treatments for CD have been developed and studied based on the degradation of gluten in the intestinal lumen, regulation of the immune response, modulation of intestinal permeability, and induction of immunological tolerance. In this review, therapeutic lines for CD are evaluated with special emphasis on phase III and II clinical trials, some of which have promising results.
Collapse
Affiliation(s)
| | | | | | - María de Lourdes Moreno
- Departamento de Microbiología y Parasitología, Facultad de Farmacia, Universidad de Sevilla, 41012 Sevilla, Spain; (V.S.); (Á.R.-C.); (C.S.)
| |
Collapse
|
3
|
Gluten Degrading Enzymes for Treatment of Celiac Disease. Nutrients 2020; 12:nu12072095. [PMID: 32679754 PMCID: PMC7400306 DOI: 10.3390/nu12072095] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 07/10/2020] [Accepted: 07/10/2020] [Indexed: 02/06/2023] Open
Abstract
Celiac disease (CeD) affects about 1% of most world populations. It presents a wide spectrum of clinical manifestations, ranging from minor symptoms to mild or severe malabsorption, and it may be associated with a wide variety of autoimmune diseases. CeD is triggered and maintained by the ingestion of gluten proteins from wheat and related grains. Gluten peptides that resist gastrointestinal digestion are antigenically presented to gluten specific T cells in the intestinal mucosa via HLA-DQ2 or HLA-DQ8, the necessary genetic predisposition for CeD. To date, there is no effective or approved treatment for CeD other than a strict adherence to a gluten-free diet, which is difficult to maintain in professional or social environments. Moreover, many patients with CeD have active disease despite diet adherence due to a high sensitivity to traces of gluten. Therefore, safe pharmacological treatments that complement the gluten-free diet are urgently needed. Oral enzyme therapy, employing gluten-degrading enzymes, is a promising therapeutic approach. A prerequisite is that such enzymes are active under gastro-duodenal conditions, quickly neutralize the T cell activating gluten peptides and are safe for human consumption. Several enzymes including prolyl endopeptidases, cysteine proteases and subtilisins can cleave the human digestion-resistant gluten peptides in vitro and in vivo. Examples are several prolyl endopeptidases from bacterial sources, subtilisins from Rothia bacteria that are natural oral colonizers and synthetic enzymes with optimized gluten-degrading activities. Without exception, these enzymes must cleave the otherwise unusual glutamine and proline-rich domains characteristic of antigenic gluten peptides. Moreover, they should be stable and active in both the acidic environment of the stomach and under near neutral pH in the duodenum. This review focuses on those enzymes that have been characterized and evaluated for the treatment of CeD, discussing their origin and activities, their clinical evaluation and challenges for therapeutic application. Novel developments include strategies like enteric coating and genetic modification to increase enzyme stability in the digestive tract.
Collapse
|
4
|
Bascuñán KA, Araya M, Roncoroni L, Doneda L, Elli L. Dietary Gluten as a Conditioning Factor of the Gut Microbiota in Celiac Disease. Adv Nutr 2020; 11:160-174. [PMID: 31399743 PMCID: PMC7442381 DOI: 10.1093/advances/nmz080] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 06/12/2019] [Accepted: 07/01/2019] [Indexed: 12/20/2022] Open
Abstract
The gut microbiota plays a relevant role in determining an individual's health status, and the diet is a major factor in modulating the composition and function of gut microbiota. Gluten constitutes an essential dietary component in Western societies and is the environmental trigger of celiac disease. The presence/absence of gluten in the diet can change the diversity and proportions of the microbial communities constituting the gut microbiota. There is an intimate relation between gluten metabolism and celiac disease pathophysiology and gut microbiota; their interrelation defines intestinal health and homeostasis. Environmental factors modify the intestinal microbiota and, in turn, its changes modulate the mucosal and immune responses. Current evidence from studies of young and adult patients with celiac disease increasingly supports that dysbiosis (i.e., compositional and functional alterations of the gut microbiome) is present in celiac disease, but to what extent this is a cause or consequence of the disease and whether the different intestinal diseases (celiac disease, ulcerative colitis, Crohn disease) have specific change patterns is not yet clear. The use of bacterial-origin enzymes that help completion of gluten digestion is of interest because of the potential application as coadjuvant in the current treatment of celiac disease. In this narrative review, we address the current knowledge on the complex interaction between gluten digestion and metabolism, celiac disease, and the intestinal microbiota.
Collapse
Affiliation(s)
- Karla A Bascuñán
- Department of Nutrition, School of Medicine, University of Chile, Santiago, Chile
- Centre for the Prevention and Diagnosis of Celiac Disease/Gastroenterology II, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, and Università degli Studi di Milano, Milan, Italy
| | - Magdalena Araya
- Institute of Nutrition and Food Technology (INTA), University of Chile, Santiago, Chile
| | - Leda Roncoroni
- Centre for the Prevention and Diagnosis of Celiac Disease/Gastroenterology II, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, and Università degli Studi di Milano, Milan, Italy
- Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Luisa Doneda
- Department of Biomedical, Surgical, and Dental Sciences, University of Milan, Milan, Italy
| | - Luca Elli
- Centre for the Prevention and Diagnosis of Celiac Disease/Gastroenterology II, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, and Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
5
|
Kaliszewska A, Martinez V, Laparra JM. Proinflammatory responses driven by non-gluten factors are masked when they appear associated to gliadins. Food Chem Toxicol 2016; 95:89-95. [PMID: 27377345 DOI: 10.1016/j.fct.2016.06.030] [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: 06/02/2016] [Revised: 06/29/2016] [Accepted: 06/29/2016] [Indexed: 12/14/2022]
Abstract
Cereal proteins are of clinical interest because of their cytotoxic and immunogenic features being associated to allergic processes and intestinal disorders. In addition to gliadins, there has been suggested an important role for non-gluten modulating factors (nGMF) on the onset of intestinal inflammatory processes. In this study, the amino acid sequences generated after a simulated human gastrointestinal (sGI) digestion of a commercial extract of gliadins (GEF) and the nGMF-enriched fraction (nGEF) obtained from them were characterized (nanoESI-qQTOF). These fractions were fed (20 days) to Wistar rats, sensitized with interferon (IFN)-γ (1000 IU/rat) and further treated with indomethacin (2 mg/kg). The production of inflammatory mediators (ELISAs) and the expression (rt-qPCR) of innate biomarkers was monitored in duodenal tissue sections. There were also evaluated changes in defined leukocyte (flow cytometry) populations quantified in peripheral blood samples. Expected nGMF components, CM3 and 0.19, as well as toxic gliadin-derived peptides were generated after sGI digestion. Rats fed with the nGEF showed higher concentrations of IFNγ, as well as expression levels of TLR-4 and the peroxisome proliferator activated receptor-α in duodenal samples than those animals fed with GEF. Rats fed with GEF showed higher expression levels of the endocannabinoid receptor-1 and an increased CD4(+)CD3(+)Foxp3(+) cell population. The data points out significant different cytotoxic and immunogenic potential of the nGEF when administered independently of the GEF to which are commonly associated. However, proinflammatory responses to nGMF are masked when present associated to gliadins.
Collapse
Affiliation(s)
- A Kaliszewska
- Institute of Animal Reproduction and Food Research of the Polish Academy of Sciences, Department of Immunology and Food Microbiology, Tuwima str 10, 10-748, Olsztyn, Poland
| | - V Martinez
- Immunonutrition and Health Group, Valencian International University, C/ Gorgos 5-7, 46021, Valencia, Spain
| | - J M Laparra
- Immunonutrition and Health Group, Valencian International University, C/ Gorgos 5-7, 46021, Valencia, Spain.
| |
Collapse
|
6
|
Supplementation of Reduced Gluten Barley Diet with Oral Prolyl Endopeptidase Effectively Abrogates Enteropathy-Associated Changes in Gluten-Sensitive Macaques. Nutrients 2016; 8:nu8070401. [PMID: 27367722 PMCID: PMC4963877 DOI: 10.3390/nu8070401] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 06/21/2016] [Accepted: 06/23/2016] [Indexed: 02/07/2023] Open
Abstract
Celiac disease (CD) is an autoimmune disorder that affects approximately three million people in the United States. Furthermore, non-celiac gluten sensitivity (NCGS) affects an estimated additional 6% of the population, e.g., 20 million in the U.S. The only effective treatment of CD and NCGS requires complete removal of gluten sources from the diet. While required adherence to a gluten-free diet (GFD) is extremely difficult to accomplish, efforts to develop additional supportive treatments are needed. To facilitate these efforts, we developed a gluten-sensitive (GS) rhesus macaque model to study the effects of novel therapies. Recently reported results from phase one of this project suggest that partial improvement—but not remission—of gluten-induced disease can be accomplished by 100-fold reduction of dietary gluten, i.e., 200 ppm—by replacement of conventional dietary sources of gluten with a mutant, reduced gluten (RG) barley (lys3a)-derived source. The main focus of this (phase two) study was to determine if the inflammatory effects of the residual gluten in lys3a mutant barley grain could be further reduced by oral supplementation with a prolylendopeptidase (PE). Results reveal that PE supplementation of RG barley diet induces more complete immunological, histopathological and clinical remission than RG barley diet alone. The combined effects of RG barley diet and PE supplementation resulted in a further decrease of inflammatory mediators IFN-γ and TNF secretion by peripheral lymphocytes, as well as decreased plasma anti-gliadin and anti-intestinal tissue transglutaminase (TG2) antibodies, diminished active caspase production in small intestinal mucosa, and eliminated clinical diarrhea—all comparable with a gluten-free diet induced remission. In summary, the beneficial results of a combined RG barley and PE administration in GS macaques may warrant the investigation of similar synergistic approaches.
Collapse
|
7
|
Bruun SW, Josefsen K, Tanassi JT, Marek A, Pedersen MHF, Sidenius U, Haupt-Jorgensen M, Antvorskov JC, Larsen J, Heegaard NH, Buschard K. Large Gliadin Peptides Detected in the Pancreas of NOD and Healthy Mice following Oral Administration. J Diabetes Res 2016; 2016:2424306. [PMID: 27795959 PMCID: PMC5067331 DOI: 10.1155/2016/2424306] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 08/10/2016] [Indexed: 12/20/2022] Open
Abstract
Gluten promotes type 1 diabetes in nonobese diabetic (NOD) mice and likely also in humans. In NOD mice and in non-diabetes-prone mice, it induces inflammation in the pancreatic lymph nodes, suggesting that gluten can initiate inflammation locally. Further, gliadin fragments stimulate insulin secretion from beta cells directly. We hypothesized that gluten fragments may cross the intestinal barrier to be distributed to organs other than the gut. If present in pancreas, gliadin could interact directly with the immune system and the beta cells to initiate diabetes development. We orally and intravenously administered 33-mer and 19-mer gliadin peptide to NOD, BALB/c, and C57BL/6 mice and found that the peptides readily crossed the intestinal barrier in all strains. Several degradation products were found in the pancreas by mass spectroscopy. Notably, the exocrine pancreas incorporated large amounts of radioactive label shortly after administration of the peptides. The study demonstrates that, even in normal animals, large gliadin fragments can reach the pancreas. If applicable to humans, the increased gut permeability in prediabetes and type 1 diabetes patients could expose beta cells directly to gliadin fragments. Here they could initiate inflammation and induce beta cell stress and thus contribute to the development of type 1 diabetes.
Collapse
MESH Headings
- Administration, Oral
- Animals
- Chromatography, Liquid
- Diabetes Mellitus, Type 1/immunology
- Electrophoresis, Polyacrylamide Gel
- Gliadin/immunology
- Gliadin/pharmacokinetics
- Inflammation
- Insulin/metabolism
- Insulin Secretion
- Insulin-Secreting Cells/immunology
- Insulin-Secreting Cells/metabolism
- Intestinal Mucosa/metabolism
- Male
- Mass Spectrometry
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Inbred NOD
- Pancreas/metabolism
- Pancreas, Exocrine/metabolism
- Peptide Fragments/pharmacokinetics
- Permeability
- Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization
Collapse
Affiliation(s)
| | - Knud Josefsen
- The Bartholin Institute, Rigshospitalet, Copenhagen N, Denmark
- *Knud Josefsen:
| | - Julia T. Tanassi
- Clinical Biochemistry, Immunology & Genetics, Statens Serum Institut, Copenhagen S, Denmark
| | - Aleš Marek
- The Hevesy Laboratory, DTU Nutech, Technical University of Denmark, Roskilde, Denmark
- Institute of Organic Chemistry and Biochemistry, Academy of Sciences of the Czech Republic, Prague 6, Czech Republic
| | - Martin H. F. Pedersen
- The Hevesy Laboratory, DTU Nutech, Technical University of Denmark, Roskilde, Denmark
| | - Ulrik Sidenius
- Enzyme Purification and Characterization, Novozymes A/S, Bagsværd, Denmark
| | | | | | - Jesper Larsen
- The Bartholin Institute, Rigshospitalet, Copenhagen N, Denmark
| | - Niels H. Heegaard
- Clinical Biochemistry, Immunology & Genetics, Statens Serum Institut, Copenhagen S, Denmark
| | | |
Collapse
|
8
|
Costes LMM, Meresse B, Cerf-Bensussan N, Samsom JN. The role of animal models in unravelling therapeutic targets in coeliac disease. Best Pract Res Clin Gastroenterol 2015; 29:437-50. [PMID: 26060108 DOI: 10.1016/j.bpg.2015.04.007] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Revised: 04/21/2015] [Accepted: 04/26/2015] [Indexed: 01/31/2023]
Abstract
Coeliac disease is a complex small intestinal enteropathy that develops consequently to a breach of tolerance to gliadin, a storage protein abundantly found in cereals such as wheat, rye and barley. The understanding of the mechanisms underlying the development of coeliac disease in HLA-DQ2 and HLA-DQ8 genetically susceptible individuals has greatly improved during the last decades but so far did not allow to develop curative therapeutics, leaving a long-life gluten free diet as the only treatment option for the patients. In order to bring new therapeutic targets to light and to test the safety and efficacy of putative drugs, animal models recapitulating features of the disease are needed. Here, we will review the existing animal models and the clinical features of coeliac disease they reflect and discuss their relevance for modelling immune pathways that may lead to potential therapeutic approaches.
Collapse
Affiliation(s)
- Léa M M Costes
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus University Medical Center Rotterdam-Sophia Children's Hospital, Rotterdam, The Netherlands.
| | - Bertrand Meresse
- INSERM UMR1163, Laboratory of Intestinal Immunity, Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France.
| | - Nadine Cerf-Bensussan
- INSERM UMR1163, Laboratory of Intestinal Immunity, Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut Imagine, 75015 Paris, France.
| | - Janneke N Samsom
- Laboratory of Pediatrics, Division of Gastroenterology and Nutrition, Erasmus University Medical Center Rotterdam-Sophia Children's Hospital, Rotterdam, The Netherlands.
| |
Collapse
|
9
|
Abstract
Currently, the only effective treatment for celiac disease is a strict lifelong gluten-free diet. However, gluten-free dieting is restrictive, difficult to maintain and nutritionally less than optimal. The improved knowledge on celiac disease pathogenesis has enabled researchers to suggest alternative strategies to treat the disorder. The drug development poses a challenge as any novel drug for celiac disease should be simultaneously effective and as safe as the gluten-free diet. The rationale behind enzyme supplementation therapy as a future treatment option for celiac patients lies in the fact that gluten is only poorly digested by gastrointestinal proteases. Due to incomplete degradation in the gastrointestinal tract, fairly long gluten peptides enter the small-intestinal lumen and come into contact with the mucosal epithelium, and in celiac disease patients this encounter launches deleterious downstream effects. Enzyme supplement therapy using either bacterial or fungal endopeptidases or proteases from germinating cereals has been proposed to promote complete digestion of prolamins and destroy disease-inducing gluten peptides. A major advantage of these glutenases is that they work in the lumen of the small intestine and do not themselves take part in the immunological cascade of events in the lamina propria, thus being unlikely to cause harmful side effects to the host. Studies to test this rationale, e.g. with Aspergillus niger prolyl endoprotease and a combination enzyme product ALV003, are already ongoing. The development of a novel medication for celiac disease is still in its early days, and thus the conventional dietary treatment will hold its place for the time being.
Collapse
|
10
|
The effects of reduced gluten barley diet on humoral and cell-mediated systemic immune responses of gluten-sensitive rhesus macaques. Nutrients 2015; 7:1657-71. [PMID: 25756783 PMCID: PMC4377872 DOI: 10.3390/nu7031657] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2015] [Revised: 02/11/2015] [Accepted: 02/27/2015] [Indexed: 12/20/2022] Open
Abstract
Celiac disease (CD) affects approximately 1% of the general population while an estimated additional 6% suffers from a recently characterized, rapidly emerging, similar disease, referred to as non-celiac gluten sensitivity (NCGS). The only effective treatment of CD and NCGS requires removal of gluten sources from the diet. Since required adherence to a gluten-free diet (GFD) is difficult to accomplish, efforts to develop alternative treatments have been intensifying in recent years. In this study, the non-human primate model of CD/NCGS, e.g., gluten-sensitive rhesus macaque, was utilized with the objective to evaluate the treatment potential of reduced gluten cereals using a reduced gluten (RG; 1% of normal gluten) barley mutant as a model. Conventional and RG barleys were used for the formulation of experimental chows and fed to gluten-sensitive (GS) and control macaques to determine if RG barley causes a remission of dietary gluten-induced clinical and immune responses in GS macaques. The impacts of the RG barley diet were compared with the impacts of the conventional barley-containing chow and the GFD. Although remission of the anti-gliadin antibody (AGA) serum responses and an improvement of clinical diarrhea were noted after switching the conventional to the RG barley diet, production of inflammatory cytokines, e.g., interferon-gamma (IFN-γ), tumor necrosis factor (TNF) and interleukin-8 (IL-8) by peripheral CD4+ T helper lymphocytes, persisted during the RG chow treatment and were partially abolished only upon re-administration of the GFD. It was concluded that the RG barley diet might be used for the partial improvement of gluten-induced disease but its therapeutic value still requires upgrading—by co-administration of additional treatments.
Collapse
|
11
|
Abnormal apical-to-basal transport of dietary ovalbumin by secretory IgA stimulates a mucosal Th1 response. Mucosal Immunol 2014; 7:315-24. [PMID: 23839063 DOI: 10.1038/mi.2013.49] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Accepted: 06/06/2013] [Indexed: 02/04/2023]
Abstract
In celiac disease, enhanced permeability to gliadin peptides can result from their apico-basal transport by secretory immunoglobulin A1 (SIgA1) binding to the CD71 receptor ectopically expressed at the gut epithelial surface. Herein, we have established a mouse model in which there is apico-basal transport of the model antigen ovalbumin (OVA) by specific SIgA1 and have analyzed local T-cell activation. Transgenic DO11.10 mice were grafted with a hybridoma-secreting OVA-specific humanized IgA1, which could bind mouse CD71 and which were released in the intestinal lumen as SIgA. CD71 expression was induced at the gut apical surface by treating the mice with tyrphostin A8. Following gavage of the mice with OVA, OVA-specific CD4⁺ T cells isolated from the mesenteric lymph nodes displayed higher expression of the activation marker CD69 and produced more interferon gamma in mice bearing the hybridoma-secreting OVA-specific IgA1, than in ungrafted mice or in mice grafted with an irrelevant hybridoma. These results indicate that the protective role of SIgA1 might be jeopardized in human pathological conditions associated with ectopic expression of CD71 at the gut surface.
Collapse
|
12
|
Marietta EV, Rubio-Tapia A, Murray JA. Using Animal Models of Celiac Disease to Understand the Role of MHC II. CLINICAL GASTROENTEROLOGY 2014. [DOI: 10.1007/978-1-4614-8560-5_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
13
|
Celiac Disease Resolution After Allogeneic Bone Marrow Transplantation is Associated with Absence of Gliadin-Specific Memory Response by Donor-Derived Intestinal T-cells. J Clin Immunol 2013; 33:1395-402. [DOI: 10.1007/s10875-013-9943-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2013] [Accepted: 09/30/2013] [Indexed: 12/23/2022]
|
14
|
Demin OO, Smirnov SV, Sokolov VV, Cucurull-Sanchez L, Pichardo-Almarza C, Flores MV, Benson N, Demin OV. Modeling of celiac disease immune response and the therapeutic effect of potential drugs. BMC SYSTEMS BIOLOGY 2013; 7:56. [PMID: 23826972 PMCID: PMC3706242 DOI: 10.1186/1752-0509-7-56] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 06/27/2013] [Indexed: 12/22/2022]
Abstract
Background Celiac disease (CD) is an autoimmune disorder that occurs in genetically predisposed people and is caused by a reaction to the gluten protein found in wheat, which leads to intestinal villous atrophy. Currently there is no drug for treatment of CD. The only known treatment is lifelong gluten-free diet. The main aim of this work is to develop a mathematical model of the immune response in CD patients and to predict the efficacy of a transglutaminase-2 (TG-2) inhibitor as a potential drug for treatment of CD. Results A thorough analysis of the developed model provided the following results: 1. TG-2 inhibitor treatment leads to insignificant decrease in antibody levels, and hence remains higher than in healthy individuals. 2. TG-2 inhibitor treatment does not lead to any significant increase in villous area. 3. The model predicts that the most effective treatment of CD would be the use of gluten peptide analogs that antagonize the binding of immunogenic gluten peptides to APC. The model predicts that the treatment of CD by such gluten peptide analogs can lead to a decrease in antibody levels to those of normal healthy people, and to a significant increase in villous area. Conclusions The developed mathematical model of immune response in CD allows prediction of the efficacy of TG-2 inhibitors and other possible drugs for the treatment of CD: their influence on the intestinal villous area and on the antibody levels. The model also allows to understand what processes in the immune response have the strongest influence on the efficacy of different drugs. This model could be applied in the pharmaceutical R&D arena for the design of drugs against autoimmune small intestine disorders and on the design of their corresponding clinical trials.
Collapse
Affiliation(s)
- Oleg O Demin
- Institute for System Biology SPb, Moscow, Russia.
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Garnier-Lengliné H, Malamut G, Cerf-Bensussan N, Ruemmele FM. [Treatment perspectives]. PATHOLOGIE-BIOLOGIE 2013; 61:e61-e64. [PMID: 22037405 DOI: 10.1016/j.patbio.2011.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Accepted: 03/23/2011] [Indexed: 05/31/2023]
Abstract
Celiac disease (CD) is a chronic inflammatory enteropathy caused by the ingestion of gluten. A safe and efficient but unpleasant treatment exists for CD in form of a strict gluten-free diet. Thus, there is a need for new treatment strategies, which are based on the improved and advanced understanding of the pathophysiology of CD. The first strategy consists in reducing or even eliminating major antigenic motifs in gluten, responsible for the inflammatory reaction. The use of less immunogenic wheat was suggested but this seems rather difficult to realize. However, a complete digestion of the immunogenic parts of gluten looks very promising. This can be obtained by the use of polymers, capable to sequester gluten proteins or even better via the exogenous administration of propyl-endopeptidases, with two different enzymes under development. Another approach could be the use of inhibitors of tissue transglutaminase, a strategy which is under clinical investigation. Alternatively, inhibition of the site of liaison of immunostimulatory peptides with HLA molecules was suggested and is also under investigation in vivo. For patients suffering from refractory sprue, the inhibition of IL15 might be of therapeutic interest with the hope to improve the fatal outcome of many of these patients. However, the ultimate treatment approach is in form of prevention and the role of infectious agents, such as Rotavirus, in disease onset has to be considered.
Collapse
|
16
|
Xu H, Feely SL, Wang X, Liu DX, Borda JT, Dufour J, Li W, Aye PP, Doxiadis GG, Khosla C, Veazey RS, Sestak K. Gluten-sensitive enteropathy coincides with decreased capability of intestinal T cells to secrete IL-17 and IL-22 in a macaque model for celiac disease. Clin Immunol 2013; 147:40-49. [PMID: 23518597 PMCID: PMC3732447 DOI: 10.1016/j.clim.2013.02.012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2012] [Revised: 01/29/2013] [Accepted: 02/20/2013] [Indexed: 12/15/2022]
Abstract
Celiac disease (CD) is an autoimmune disorder caused by intolerance to dietary gluten. The interleukin (IL)-17 and IL-22 function as innate regulators of mucosal integrity. Impaired but not well-understood kinetics of the IL-17/22 secretion was described in celiac patients. Here, the IL-17 and IL-22-producing intestinal cells were studied upon their in vitro stimulation with mitogens in class II major histocompatibility complex-defined, gluten-sensitive rhesus macaques. Pediatric biopsies were collected from distal duodenum during the stages of disease remission and relapse. Regardless of dietary gluten content, IL-17 and IL-22-producing cells consisted of CD4+ and CD8+ T lymphocytes as well as of lineage-negative (Lin-) cells. Upon introduction of dietary gluten, capability of intestinal T cells to secrete IL-17/22 started to decline (p<0.05), which was paralleled with gradual disruption of epithelial integrity. These data indicate that IL-17/22-producing cells play an important role in maintenance of intestinal mucosa in gluten-sensitive primates.
Collapse
Affiliation(s)
- Huanbin Xu
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | - Stephanie L Feely
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA
| | - Xiaolei Wang
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | - David X Liu
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | - Juan T Borda
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | - Jason Dufour
- Division of Veterinary Resources, Tulane National Primate Research Center, Covington, LA, USA
| | - Weiwei Li
- Department of Chemistry and Biochemistry, Stanford University, Stanford, CA, USA
| | - Pyone P Aye
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA; Division of Veterinary Resources, Tulane National Primate Research Center, Covington, LA, USA
| | - Gaby G Doxiadis
- Comparative Genetics and Refinement, Biomedical Primate Research Centre, Rijswijk, The Netherlands
| | - Chaitan Khosla
- Department of Chemistry and Biochemistry, Stanford University, Stanford, CA, USA
| | - Ronald S Veazey
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | - Karol Sestak
- Division of Microbiology, Tulane National Primate Research Center, Covington, LA, USA.
| |
Collapse
|
17
|
Stoven S, Murray JA, Marietta EV. Latest in vitro and in vivo models of celiac disease. Expert Opin Drug Discov 2013; 8:445-57. [PMID: 23293929 DOI: 10.1517/17460441.2013.761203] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Currently, the only treatment for celiac disease is a gluten-free diet, and there is an increased desire for alternative therapies. In vitro and in vivo models of celiac disease have been generated in order to better understand the pathogenesis of celiac disease, and this review will discuss these models as well as the testing of alternative therapies using these models. AREAS COVERED The research discussed describes the different in vitro and in vivo models of celiac disease that currently exist and how they have contributed to our understanding of how gluten can stimulate both innate and adaptive immune responses in celiac patients. We also provide a summary on the alternative therapies that have been tested with these models and discuss whether subsequent clinical trials were done based on these tests done with these models of celiac disease. EXPERT OPINION Only a few of the alternative therapies that have been tested with animal models have gone on to clinical trials; however, those that did go on to clinical trial have provided promising results from a safety standpoint. Further trials are required to determine if some of these therapies may serve as an effective adjunct to a gluten-free diet to alleviate the adverse affects associated with accidental gluten exposure. A "magic-bullet" approach may not be the answer to celiac disease, but possibly a future cocktail of these different therapeutics may allow celiac patients to consume an unrestricted diet.
Collapse
Affiliation(s)
- Samantha Stoven
- Mayo Clinic, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, MN 55905, USA
| | | | | |
Collapse
|
18
|
Lebreton C, Ménard S, Abed J, Moura IC, Coppo R, Dugave C, Monteiro RC, Fricot A, Traore MG, Griffin M, Cellier C, Malamut G, Cerf-Bensussan N, Heyman M. Interactions among secretory immunoglobulin A, CD71, and transglutaminase-2 affect permeability of intestinal epithelial cells to gliadin peptides. Gastroenterology 2012; 143:698-707.e4. [PMID: 22750506 DOI: 10.1053/j.gastro.2012.05.051] [Citation(s) in RCA: 83] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2011] [Revised: 05/01/2012] [Accepted: 05/24/2012] [Indexed: 01/31/2023]
Abstract
BACKGROUND & AIMS The transferrin receptor (CD71) is up-regulated in duodenal biopsy samples from patients with active celiac disease and promotes retrotransport of secretory immunoglobulin A (SIgA)-gliadin complexes. We studied intestinal epithelial cell lines that overexpress CD71 to determine how interactions between SIgA and CD71 promote transepithelial transport of gliadin peptides. METHODS We analyzed duodenal biopsy specimens from 8 adults and 1 child with active celiac disease. Caco-2 and HT29-19A epithelial cell lines were transfected with fluorescence-labeled small interfering RNAs against CD71. Interactions among IgA, CD71, and transglutaminase 2 (Tgase2) were analyzed by flow cytometry, immunoprecipitation, and confocal microscopy. Transcytosis of SIgA-CD71 complexes and intestinal permeability to the gliadin 3H-p31-49 peptide were analyzed in polarized monolayers of Caco-2 cells. RESULTS Using fluorescence resonance energy transfer and in situ proximity ligation assays, we observed physical interactions between SIgA and CD71 or CD71 and Tgase2 at the apical surface of enterocytes in biopsy samples and monolayers of Caco-2 cells. CD71 and Tgase2 were co-precipitated with SIgA, bound to the surface of Caco-2 cells. SIgA-CD71 complexes were internalized and localized in early endosomes and recycling compartments but not in lysosomes. In the presence of celiac IgA or SIgA against p31-49, transport of intact 3H-p31-49 increased significantly across Caco-2 monolayers; this transport was inhibited by soluble CD71 or Tgase2 inhibitors. CONCLUSIONS Upon binding to apical CD71, SIgA (with or without gliadin peptides) enters a recycling pathway and avoids lysosomal degradation; this process allows apical-basal transcytosis of bound peptides. This mechanism is facilitated by Tgase2 and might be involved in the pathogenesis of celiac disease.
Collapse
Affiliation(s)
- Corinne Lebreton
- INSERM, UMR989, Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut IMAGINE, Paris, France
| | - Sandrine Ménard
- INSERM, UMR989, Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut IMAGINE, Paris, France
| | - Juliette Abed
- INSERM, UMR989, Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut IMAGINE, Paris, France
| | - Ivan Cruz Moura
- INSERM, UMR699, Paris, France; Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Rosanna Coppo
- Medical Nephrology Unit, S. Giovanni Hospital, University of Torino, Torino, Italy
| | - Christophe Dugave
- Commissariat à Énergie Atomique, iBiTecS, Service d'Ingénierie Moléculaire des Protéines, Gif-sur-Yvette, France
| | - Renato C Monteiro
- INSERM, UMR699, Paris, France; Université Paris Diderot-Sorbonne Paris Cité, Paris, France
| | - Aurélie Fricot
- Université Paris Descartes-Sorbonne Paris Cité and Institut IMAGINE, Paris, France; CNRS, UMR8147, Hôpital Européen Georges Pompidou, Paris, France
| | - Meriem Garfa Traore
- Université Paris Descartes-Sorbonne Paris Cité and Institut IMAGINE, Paris, France; IFR 94, Imagery Platform, Hôpital Européen Georges Pompidou, Paris, France
| | - Martin Griffin
- School of Life and Health. Aston University, Birmingham B4 7ET, United Kingdom
| | - Christophe Cellier
- INSERM, UMR989, Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut IMAGINE, Paris, France; APHP, Department of Gastroenterology, Hôpital Européen Georges Pompidou, Paris, France
| | - Georgia Malamut
- INSERM, UMR989, Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut IMAGINE, Paris, France; APHP, Department of Gastroenterology, Hôpital Européen Georges Pompidou, Paris, France
| | - Nadine Cerf-Bensussan
- INSERM, UMR989, Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut IMAGINE, Paris, France.
| | - Martine Heyman
- INSERM, UMR989, Paris, France; Université Paris Descartes-Sorbonne Paris Cité and Institut IMAGINE, Paris, France
| |
Collapse
|
19
|
Marietta EV, Murray JA. Animal models to study gluten sensitivity. Semin Immunopathol 2012; 34:497-511. [PMID: 22572887 PMCID: PMC3410984 DOI: 10.1007/s00281-012-0315-y] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2012] [Accepted: 04/19/2012] [Indexed: 12/13/2022]
Abstract
The initial development and maintenance of tolerance to dietary antigens is a complex process that, when prevented or interrupted, can lead to human disease. Understanding the mechanisms by which tolerance to specific dietary antigens is attained and maintained is crucial to our understanding of the pathogenesis of diseases related to intolerance of specific dietary antigens. Two diseases that are the result of intolerance to a dietary antigen are celiac disease (CD) and dermatitis herpetiformis (DH). Both of these diseases are dependent upon the ingestion of gluten (the protein fraction of wheat, rye, and barley) and manifest in the gastrointestinal tract and skin, respectively. These gluten-sensitive diseases are two examples of how devastating abnormal immune responses to a ubiquitous food can be. The well-recognized risk genotype for both is conferred by either of the HLA class II molecules DQ2 or DQ8. However, only a minority of individuals who carry these molecules will develop either disease. Also of interest is that the age at diagnosis can range from infancy to 70-80 years of age. This would indicate that intolerance to gluten may potentially be the result of two different phenomena. The first would be that, for various reasons, tolerance to gluten never developed in certain individuals, but that for other individuals, prior tolerance to gluten was lost at some point after childhood. Of recent interest is the concept of non-celiac gluten sensitivity, which manifests as chronic digestive or neurologic symptoms due to gluten, but through mechanisms that remain to be elucidated. This review will address how animal models of gluten-sensitive disorders have substantially contributed to a better understanding of how gluten intolerance can arise and cause disease.
Collapse
|
20
|
Abstract
Celiac sprue is an inflammatory disease of the small intestine caused by dietary gluten and treated by adherence to a life-long gluten-free diet. The recent identification of immunodominant gluten peptides, the discovery of their cogent properties, and the elucidation of the mechanisms by which they engender immunopathology in genetically susceptible individuals have advanced our understanding of the molecular pathogenesis of this complex disease, enabling the rational design of new therapeutic strategies. The most clinically advanced of these is oral enzyme therapy, in which enzymes capable of proteolyzing gluten (i.e., glutenases) are delivered to the alimentary tract of a celiac sprue patient to detoxify ingested gluten in situ. In this chapter, we discuss the key challenges for discovery and preclinical development of oral enzyme therapies for celiac sprue. Methods for lead identification, assay development, gram-scale production and formulation, and lead optimization for next-generation proteases are described and critically assessed.
Collapse
Affiliation(s)
- Michael T Bethune
- Division of Biology, California Institute of Technology, Pasadena, California, USA
| | | |
Collapse
|
21
|
Sestak K, Mazumdar K, Midkiff CC, Dufour J, Borda JT, Alvarez X. Recognition of epidermal transglutaminase by IgA and tissue transglutaminase 2 antibodies in a rare case of Rhesus dermatitis. J Vis Exp 2011:3154. [PMID: 22214930 PMCID: PMC3369644 DOI: 10.3791/3154] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Tissue transglutaminase 2 (tTG2) is an intestinal digestive enzyme which deamidates already partially digested dietary gluten e.g. gliadin peptides. In genetically predisposed individuals, tTG2 triggers autoimmune responses that are characterized by the production of tTG2 antibodies and their direct deposition into small intestinal wall. The presence of such antibodies constitutes one of the major hallmarks of the celiac disease (CD). Epidermal transglutaminase (eTG) is another member of the transglutaminase family that can also function as an autoantigen in a small minority of CD patients. In these relatively rare cases, eTG triggers an autoimmune reaction (a skin rash) clinically known as dermatitis herpetiformis (DH). Although the exact mechanism of CD and DH pathogenesis is not well understood, it is known that tTG2 and eTG share antigenic epitopes that can be recognized by serum antibodies from both CD and DH patients. In this study, the confocal microscopy examination of biopsy samples from skin lesions of two rhesus macaques (Macaca mulatta) with dermatitis (Table 1, Fig. 1 and 2) was used to study the affected tissues. In one animal (EM96) a spectral overlap of IgA and tTG2 antibodies (Fig. 3) was demonstrated. The presence of double-positive tTG2+IgA+ cells was focused in the deep epidermis, around the dermal papillae. This is consistent with lesions described in DH patients. When EM96 was placed on a gluten-free diet, the dermatitis, as well as tTG2+IgA+ deposits disappeared and were no longer detectable (Figs. 1-3). Dermatitis reappeared however, based on re-introduction of dietary gluten in EM96 (not shown). In other macaques including animal with unrelated dermatitis, the tTG2+IgA+ deposits were not detected. Gluten-free diet-dependent remission of dermatitis in EM96 together with presence of tTG2+IgA+ cells in its skin suggest an autoimmune, DH-like mechanism for the development of this condition. This is the first report of DH-like dermatitis in any non-human primate.
Collapse
Affiliation(s)
- Karol Sestak
- Division of Microbiology, Tulane National Primate Research Center
| | | | | | | | | | | |
Collapse
|
22
|
Marietta EV, David CS, Murray JA. Important lessons derived from animal models of celiac disease. Int Rev Immunol 2011; 30:197-206. [PMID: 21787225 DOI: 10.3109/08830185.2011.598978] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Several animal models have been recently developed to recapitulate various components of the complex process that is celiac disease. In addition to the increasing diversity of murine models there are now monkey models of celiac disease. Mouse strains and protocols have been developed that are now just beginning to address the complex interactions among the innate and adaptive immune responses to gluten, as well as gluten-dependent autoimmunity in celiac disease. The most important conclusion that these models have provided us with so far is that while all three components (innate gluten sensitivity, adaptive gluten sensitivity, and autoimmunity) are independent phenomena, all are necessary for celiac disease to develop.
Collapse
Affiliation(s)
- E V Marietta
- Department of Immunology, Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine, Rochester, Minnesota 55905, USA
| | | | | |
Collapse
|
23
|
Lübbing N, Barone MV, Rudloff S, Troncone R, Auricchio S, Zimmer KP. Correction of gliadin transport within enterocytes through celiac disease serum. Pediatr Res 2011; 70:357-62. [PMID: 21705964 DOI: 10.1203/pdr.0b013e31822a31e7] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Celiac disease (CD) is caused by loss of tolerance toward gluten and related cereal products. The delivery of gliadin peptides (GP) to HLA-DR-positive late endosomes (LE) of enterocytes is required for antigen presentation and tolerance generation. We hypothesized that anti-gliadin antibodies in CD serum modify gliadin transport into LE within enterocytes. CD and control duodenal biopsies were incubated with digests of gluten as well as with serum of CD patients. Lissamin-labeled GP AA31-43 and AA56-68 were endocytozed by Caco-2 cells with serum of CD- or control patients. Colocalization of gliadin with the LE marker LAMP-2 and cathepsin D was determined and quantified on immunofluorescence and immunoelectron microscopical level. Up to 13% of internalized gliadin was located in LE of CD biopsies incubated with CD serum compared with less than 4% in CD biopsies without CD serum as well as in control biopsies. In Caco-2 cells, the colocalization coefficient of GP AA31-43 and LE was 0.82 with CD serum, 0.42 with control serum, and 0.48 with culture medium. Incubation with CD serum can direct GP AA31-43 into LE of enterocytes which is required for antigen presentation.
Collapse
Affiliation(s)
- Nico Lübbing
- Department of Pediatrics, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | | | | | | | | | | |
Collapse
|
24
|
Marietta EV, Rashtak S, Pittelkow MR. Experiences with animal models of dermatitis herpetiformis: a review. Autoimmunity 2011; 45:81-90. [PMID: 21929335 DOI: 10.3109/08916934.2011.606449] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Dermatitis herpetiformis (DH) is caused by the consumption of gluten, which is also the trigger for celiac disease. DH is currently considered to be the skin manifestation of celiac disease, as both diseases have some degree of gluten-sensitive enteropathy. The human leukocyte antigens class II genes, DQ2 and DQ8, are tightly associated with both diseases, and there is an increased level of anti-gliadin antibodies in both diseases. Animal models of gluten sensitivity have been used to better understand the pathogenesis of both diseases. This paper describes these different models and discusses how certain elements of these models contribute to the development of DH.
Collapse
Affiliation(s)
- Eric V Marietta
- Department of Immunology, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | | | | |
Collapse
|
25
|
Sestak K, Conroy L, Aye PP, Mehra S, Doxiadis GG, Kaushal D. Improved xenobiotic metabolism and reduced susceptibility to cancer in gluten-sensitive macaques upon introduction of a gluten-free diet. PLoS One 2011; 6:e18648. [PMID: 21533263 PMCID: PMC3075256 DOI: 10.1371/journal.pone.0018648] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Accepted: 03/11/2011] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND A non-human primate (NHP) model of gluten sensitivity was employed to study the gene perturbations associated with dietary gluten changes in small intestinal tissues from gluten-sensitive rhesus macaques (Macaca mulatta). METHODOLOGY Stages of remission and relapse were accomplished in gluten-sensitive animals by administration of gluten-free (GFD) and gluten-containing (GD) diets, as described previously. Pin-head-sized biopsies, obtained non-invasively by pediatric endoscope from duodenum while on GFD or GD, were used for preparation of total RNA and gene profiling, using the commercial Rhesus Macaque Microarray (Agilent Technologies),targeting expression of over 20,000 genes. PRINCIPAL FINDINGS When compared with normal healthy control, gluten-sensitive macaques showed differential gene expressions induced by GD. While observed gene perturbations were classified into one of 12 overlapping categories--cancer, metabolism, digestive tract function, immune response, cell growth, signal transduction, autoimmunity, detoxification of xenobiotics, apoptosis, actin-collagen deposition, neuronal and unknown function--this study focused on cancer-related gene networks such as cytochrome P450 family (detoxification function) and actin-collagen-matrix metalloproteinases (MMP) genes. CONCLUSIONS/SIGNIFICANCE A loss of detoxification function paralleled with necessity to metabolize carcinogens was revealed in gluten-sensitive animals while on GD. An increase in cancer-promoting factors and a simultaneous decrease in cancer-preventing factors associated with altered expression of actin-collagen-MMP gene network were noted. In addition, gluten-sensitive macaques showed reduced number of differentially expressed genes including the cancer-associated ones upon withdrawal of dietary gluten. Taken together, these findings indicate potentially expanded utility of gluten-sensitive rhesus macaques in cancer research.
Collapse
Affiliation(s)
- Karol Sestak
- Division of Microbiology, Tulane National Primate Research Center, Covington, Louisiana, United States of America.
| | | | | | | | | | | |
Collapse
|
26
|
Greco L, Gobbetti M, Auricchio R, Di Mase R, Landolfo F, Paparo F, Di Cagno R, De Angelis M, Rizzello CG, Cassone A, Terrone G, Timpone L, D'Aniello M, Maglio M, Troncone R, Auricchio S. Safety for patients with celiac disease of baked goods made of wheat flour hydrolyzed during food processing. Clin Gastroenterol Hepatol 2011; 9:24-9. [PMID: 20951830 DOI: 10.1016/j.cgh.2010.09.025] [Citation(s) in RCA: 79] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2009] [Revised: 09/21/2010] [Accepted: 09/27/2010] [Indexed: 02/07/2023]
Abstract
BACKGROUND & AIMS Celiac disease (CD) is characterized by an inflammatory response to wheat gluten, rye, and barley proteins. Fermentation of wheat flour with sourdough lactobacilli and fungal proteases decreases the concentration of gluten. We evaluated the safety of daily administration of baked goods made from this hydrolyzed form of wheat flour to patients with CD. METHODS Patients were randomly assigned to consumption of 200 g per day of natural flour baked goods (NFBG) (80,127 ppm gluten; n = 6), extensively hydrolyzed flour baked goods (S1BG) (2480 ppm residual gluten; n = 2), or fully hydrolyzed baked goods (S2BG) (8 ppm residual gluten; n = 5) for 60 days. RESULTS Two of the 6 patients who consumed NFBG discontinued the challenge because of symptoms; all had increased levels of anti-tissue transglutaminase (tTG) antibodies and small bowel deterioration. The 2 patients who ate the S1BG goods had no clinical complaints but developed subtotal atrophy. The 5 patients who ate the S2BG had no clinical complaints; their levels of anti-tTG antibodies did not increase, and their Marsh grades of small intestinal mucosa did not change. CONCLUSIONS A 60-day diet of baked goods made from hydrolyzed wheat flour, manufactured with sourdough lactobacilli and fungal proteases, was not toxic to patients with CD. A combined analysis of serologic, morphometric, and immunohistochemical parameters is the most accurate method to assess new therapies for this disorder.
Collapse
Affiliation(s)
- Luigi Greco
- Department of Pediatrics and European Laboratory for the Study of Food Induced Diseases, University of Naples, Federico II, Naples, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Mazumdar K, Alvarez X, Borda JT, Dufour J, Martin E, Bethune MT, Khosla C, Sestak K. Visualization of transepithelial passage of the immunogenic 33-residue peptide from alpha-2 gliadin in gluten-sensitive macaques. PLoS One 2010; 5:e10228. [PMID: 20419103 PMCID: PMC2856682 DOI: 10.1371/journal.pone.0010228] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Accepted: 03/26/2010] [Indexed: 01/08/2023] Open
Abstract
Background Based on clinical, histopathological and serological similarities to human celiac disease (CD), we recently established the rhesus macaque model of gluten sensitivity. In this study, we further characterized this condition based on presence of anti-tissue transglutaminase 2 (TG2) antibodies, increased intestinal permeability and transepithelial transport of a proteolytically resistant, immunotoxic, 33-residue peptide from α2-gliadin in the distal duodenum of gluten-sensitive macaques. Methodology/Principal Findings Six rhesus macaques were selected for study from a pool of 500, including two healthy controls and four gluten-sensitive animals with elevated anti-gliadin or anti-TG2 antibodies as well as history of non-infectious chronic diarrhea. Pediatric endoscope-guided pinch biopsies were collected from each animal's distal duodenum following administration of a gluten-containing diet (GD) and again after remission by gluten-free diet (GFD). Control biopsies always showed normal villous architecture, whereas gluten-sensitive animals on GD exhibited histopathology ranging from mild lymphocytic infiltration to villous atrophy, typical of human CD. Immunofluorescent microscopic analysis of biopsies revealed IgG+ and IgA+ plasma-like cells producing antibodies that colocalized with TG2 in gluten-sensitive macaques only. Following instillation in vivo, the Cy-3-labeled 33-residue gluten peptide colocalized with the brush border protein villin in all animals. In a substantially enteropathic macaque with “leaky” duodenum, the peptide penetrated beneath the epithelium into the lamina propria. Conclusions/Significance The rhesus macaque model of gluten sensitivity not only resembles the histopathology of CD but it also may provide a model for studying intestinal permeability in states of epithelial integrity and disrepair.
Collapse
Affiliation(s)
- Kaushiki Mazumdar
- Division of Microbiology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Xavier Alvarez
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Juan T. Borda
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Jason Dufour
- Division of Veterinary Medicine, Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Edith Martin
- Department of Chemistry, Stanford University, Stanford, California, United States of America
| | - Michael T. Bethune
- Department of Biochemistry, Stanford University, Stanford, California, United States of America
| | - Chaitan Khosla
- Department of Chemistry, Stanford University, Stanford, California, United States of America
- Department of Biochemistry, Stanford University, Stanford, California, United States of America
- Department of Chemical Engineering, Stanford University, Stanford, California, United States of America
| | - Karol Sestak
- Division of Microbiology, Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- * E-mail:
| |
Collapse
|
28
|
De Angelis M, Cassone A, Rizzello CG, Gagliardi F, Minervini F, Calasso M, Di Cagno R, Francavilla R, Gobbetti M. Mechanism of degradation of immunogenic gluten epitopes from Triticum turgidum L. var. durum by sourdough lactobacilli and fungal proteases. Appl Environ Microbiol 2010; 76:508-518. [PMID: 19948868 PMCID: PMC2805216 DOI: 10.1128/aem.01630-09] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2009] [Accepted: 11/15/2009] [Indexed: 12/31/2022] Open
Abstract
As shown by R5 antibody-based sandwich and competitive enzyme-linked immunosorbent assay (ELISA), selected sourdough lactobacilli, in combination with fungal proteases, hydrolyzed gluten (72 h at 37 degrees C) of various cultivars of Triticum turgidum L. var. durum to less than 20 ppm. Complementary electrophoretic, chromatography, and mass spectrometry techniques were used to characterize the gluten and epitope hydrolysis. Nine peptidases were partially purified from the pooled cytoplasmic extract of the sourdough lactobacilli and used to hydrolyze the 33-mer epitope, the most immunogenic peptide generated during digestion of Triticum species. At least three peptidases (general aminopeptidase type N [PepN], X-prolyl dipeptidyl aminopeptidase [PepX], and endopeptidase PepO) were necessary to detoxify the 33-mer without generation of related immunogenic epitopes. After 14 h of incubation, the combination of all or at least six different peptidases totally hydrolyzed the 33-mer (200 mM) into free amino acids. The same results were found for other immunogenic epitopes, such as fragments 57-68 of alpha 9-gliadin, 62-75 of A-gliadin, and 134-153 of gamma-gliadin. When peptidases were used for fermentation of durum wheat semolina, they caused the hydrolysis of gluten to ca. 2 ppm. The in vivo digestion was simulated, and proteins/peptides extracted from pepsin-trypsin (PT) digestion of durum wheat semolina fermented with selected sourdough lactobacilli induced the expression of gamma interferon and interleukin 2 at levels comparable to those of the negative control. Durum wheat semolina fermented with sourdough lactobacilli was freeze-dried and used for making Italian-type pasta. The scores for cooking and sensory properties for this pasta were higher that those of conventional gluten-free pasta.
Collapse
Affiliation(s)
- Maria De Angelis
- Department of Plant Protection and Applied Microbiology, University of Bari, 70126 Bari, Italy, Department of Pediatrics, University of Bari, Bari, Italy
| | - Angela Cassone
- Department of Plant Protection and Applied Microbiology, University of Bari, 70126 Bari, Italy, Department of Pediatrics, University of Bari, Bari, Italy
| | - Carlo G. Rizzello
- Department of Plant Protection and Applied Microbiology, University of Bari, 70126 Bari, Italy, Department of Pediatrics, University of Bari, Bari, Italy
| | - Francesca Gagliardi
- Department of Plant Protection and Applied Microbiology, University of Bari, 70126 Bari, Italy, Department of Pediatrics, University of Bari, Bari, Italy
| | - Fabio Minervini
- Department of Plant Protection and Applied Microbiology, University of Bari, 70126 Bari, Italy, Department of Pediatrics, University of Bari, Bari, Italy
| | - Maria Calasso
- Department of Plant Protection and Applied Microbiology, University of Bari, 70126 Bari, Italy, Department of Pediatrics, University of Bari, Bari, Italy
| | - Raffaella Di Cagno
- Department of Plant Protection and Applied Microbiology, University of Bari, 70126 Bari, Italy, Department of Pediatrics, University of Bari, Bari, Italy
| | - Ruggero Francavilla
- Department of Plant Protection and Applied Microbiology, University of Bari, 70126 Bari, Italy, Department of Pediatrics, University of Bari, Bari, Italy
| | - Marco Gobbetti
- Department of Plant Protection and Applied Microbiology, University of Bari, 70126 Bari, Italy, Department of Pediatrics, University of Bari, Bari, Italy
| |
Collapse
|
29
|
Bethune MT, Crespo-Bosque M, Bergseng E, Mazumdar K, Doyle L, Sestak K, Sollid LM, Khosla C. Noninflammatory gluten peptide analogs as biomarkers for celiac sprue. ACTA ACUST UNITED AC 2009; 16:868-81. [PMID: 19716477 DOI: 10.1016/j.chembiol.2009.07.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2009] [Revised: 07/17/2009] [Accepted: 07/20/2009] [Indexed: 02/09/2023]
Abstract
New tools are needed for managing celiac sprue, a lifelong immune disease of the small intestine. Ongoing drug trials are also prompting a search for noninvasive biomarkers of gluten-induced intestinal change. We have synthesized and characterized noninflammatory gluten peptide analogs in which key Gln residues are replaced by Asn or His. Like their proinflammatory counterparts, these biomarkers are resistant to gastrointestinal proteases, susceptible to glutenases, and permeable across enterocyte barriers. Unlike gluten peptides, however, they are not appreciably recognized by transglutaminase, HLA-DQ2, or disease-specific T cells. In vitro and animal studies show that the biomarkers can detect intestinal permeability changes as well as glutenase-catalyzed gastric detoxification of gluten. Accordingly, controlled clinical studies are warranted to evaluate the use of these peptides as probes for abnormal intestinal permeability in celiac patients and for glutenase efficacy in clinical trials and practice.
Collapse
|
30
|
Marietta EV, Schuppan D, Murray JA. In vitroand in vivomodels of celiac disease. Expert Opin Drug Discov 2009; 4:1113-1123. [DOI: 10.1517/17460440903307417] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
31
|
Iismaa SE, Mearns BM, Lorand L, Graham RM. Transglutaminases and disease: lessons from genetically engineered mouse models and inherited disorders. Physiol Rev 2009; 89:991-1023. [PMID: 19584319 DOI: 10.1152/physrev.00044.2008] [Citation(s) in RCA: 272] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The human transglutaminase (TG) family consists of a structural protein, protein 4.2, that lacks catalytic activity, and eight zymogens/enzymes, designated factor XIII-A (FXIII-A) and TG1-7, that catalyze three types of posttranslational modification reactions: transamidation, esterification, and hydrolysis. These reactions are essential for biological processes such as blood coagulation, skin barrier formation, and extracellular matrix assembly but can also contribute to the pathophysiology of various inflammatory, autoimmune, and degenerative conditions. Some members of the TG family, for example, TG2, can participate in biological processes through actions unrelated to transamidase catalytic activity. We present here a comprehensive review of recent insights into the physiology and pathophysiology of TG family members that have come from studies of genetically engineered mouse models and/or inherited disorders. The review focuses on FXIII-A, TG1, TG2, TG5, and protein 4.2, as mice deficient in TG3, TG4, TG6, or TG7 have not yet been reported, nor have mutations in these proteins been linked to human disease.
Collapse
Affiliation(s)
- Siiri E Iismaa
- Molecular Cardiology and Biophysics Division, Victor Chang Cardiac Research Institute and Universityof New South Wales, Sydney, New South Wales 2010, Australia
| | | | | | | |
Collapse
|
32
|
Malamut G, Meresse B, Cellier C, Cerf-Bensussan N. La maladie cœliaque en 2009 : un futur sans régime ? ACTA ACUST UNITED AC 2009; 33:635-47. [DOI: 10.1016/j.gcb.2009.07.007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
33
|
Bethune MT, Siegel M, Howles-Banerji S, Khosla C. Interferon-gamma released by gluten-stimulated celiac disease-specific intestinal T cells enhances the transepithelial flux of gluten peptides. J Pharmacol Exp Ther 2009; 329:657-68. [PMID: 19218531 PMCID: PMC2672868 DOI: 10.1124/jpet.108.148007] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2008] [Accepted: 02/12/2009] [Indexed: 01/24/2023] Open
Abstract
Celiac sprue is a T-cell-mediated enteropathy elicited in genetically susceptible individuals by dietary gluten proteins. To initiate and propagate inflammation, proteolytically resistant gluten peptides must be translocated across the small intestinal epithelium and presented to DQ2-restricted T cells, but the effectors enabling this translocation under normal and inflammatory conditions are not well understood. We demonstrate that a fluorescently labeled antigenic 33-mer gluten peptide is translocated intact across a T84 cultured epithelial cell monolayer and that preincubation of the monolayer with media from gluten-stimulated, celiac patient-derived intestinal T cells enhances the apical-to-basolateral flux of this peptide in a dose-dependent, saturable manner. The permeability-enhancing activity of activated T-cell media is inhibited by blocking antibodies against either interferon-gamma or its receptor and is recapitulated using recombinant interferon-gamma. At saturating levels of interferon-gamma, activated T-cell media does not further increase transepithelial peptide flux, indicating the primacy of interferon-gamma as an effector of increased epithelial permeability during inflammation. Reducing the assay temperature to 4 degrees C reverses the effect of interferon-gamma but does not reduce basal peptide flux occurring in the absence of interferon-gamma, suggesting active transcellular transport of intact peptides is increased during inflammation. A panel of disease-relevant gluten peptides exhibited an inverse correlation between size and transepithelial flux but no apparent sequence constraints. Anti-interferon-gamma therapy may mitigate the vicious cycle of gluten-induced interferon-gamma secretion and interferon-gamma-mediated enhancement of gluten peptide flux but is unlikely to prevent translocation of gluten peptides in the absence of inflammatory conditions.
Collapse
Affiliation(s)
- Michael T Bethune
- Department of Biochemistry, Stanford University, Stanford, CA 94305, USA
| | | | | | | |
Collapse
|
34
|
Celiac disease: from oral tolerance to intestinal inflammation, autoimmunity and lymphomagenesis. Mucosal Immunol 2009; 2:8-23. [PMID: 19079330 DOI: 10.1038/mi.2008.75] [Citation(s) in RCA: 93] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Celiac disease is a multifactorial disorder and provides a privileged model to decipher how the interplay between environmental and genetic factors can alter mucosal tolerance to a food antigen, lead to chronic intestinal inflammation, and ultimately promote T-cell lymphomagenesis. Here we summarize how HLA-DQ2/8 molecules, the main genetic risk factor for this disease can orchestrate a CD4(+) T-cell adaptive immune response against gluten, and discuss recent data which shed light on the innate and adaptive immune stimuli that collaborate to induce a proinflammatory TH1 response, a massive expansion of intraepithelial lymphocytes, and a cytolytic attack of the epithelium. The intestinal immune response driven in genetically predisposed patients by chronic exposure to gluten emerges as the pathological counterpart of normal acute intestinal responses to intracellular pathogens.
Collapse
|
35
|
Bethune MT, Borda JT, Ribka E, Liu MX, Phillippi-Falkenstein K, Jandacek RJ, Doxiadis GGM, Gray GM, Khosla C, Sestak K. A non-human primate model for gluten sensitivity. PLoS One 2008; 3:e1614. [PMID: 18286171 PMCID: PMC2229647 DOI: 10.1371/journal.pone.0001614] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2007] [Accepted: 01/23/2008] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND AIMS Gluten sensitivity is widespread among humans. For example, in celiac disease patients, an inflammatory response to dietary gluten leads to enteropathy, malabsorption, circulating antibodies against gluten and transglutaminase 2, and clinical symptoms such as diarrhea. There is a growing need in fundamental and translational research for animal models that exhibit aspects of human gluten sensitivity. METHODS Using ELISA-based antibody assays, we screened a population of captive rhesus macaques with chronic diarrhea of non-infectious origin to estimate the incidence of gluten sensitivity. A selected animal with elevated anti-gliadin antibodies and a matched control were extensively studied through alternating periods of gluten-free diet and gluten challenge. Blinded clinical and histological evaluations were conducted to seek evidence for gluten sensitivity. RESULTS When fed with a gluten-containing diet, gluten-sensitive macaques showed signs and symptoms of celiac disease including chronic diarrhea, malabsorptive steatorrhea, intestinal lesions and anti-gliadin antibodies. A gluten-free diet reversed these clinical, histological and serological features, while reintroduction of dietary gluten caused rapid relapse. CONCLUSIONS Gluten-sensitive rhesus macaques may be an attractive resource for investigating both the pathogenesis and the treatment of celiac disease.
Collapse
Affiliation(s)
- Michael T. Bethune
- Department of Biochemistry, Stanford University, Stanford, California, United States of America
| | - Juan T. Borda
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Erin Ribka
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | - Michael-Xun Liu
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
| | | | - Ronald J. Jandacek
- Department of Pathology and Laboratory Medicine, University of Cincinnati, Cincinnati, Ohio, United States of America
| | | | - Gary M. Gray
- Department of Medicine, Stanford University, Stanford, California, United States of America
| | - Chaitan Khosla
- Department of Biochemistry, Stanford University, Stanford, California, United States of America
- Department of Chemistry, Stanford University, Stanford, California, United States of America
- Department of Chemical Engineering, Stanford University, Stanford, California, United States of America
| | - Karol Sestak
- Tulane National Primate Research Center, Covington, Louisiana, United States of America
- Department of Microbiology and Immunology, Tulane University School of Medicine, New Orleans, Louisiana, United States of America
- *E-mail:
| |
Collapse
|