1
|
Dilaghi E, Felici E, Lahner E, Pilozzi E, Furio S, Lucchini L, Quatrale G, Piccirillo M, Parisi P, Curto S, Annibale B, Ferretti A, Mennini M, Persechino S, Di Nardo G. Helicobacter Pylori infection in children with inflammatory bowel disease: a prospective multicenter study. BMC Pediatr 2024; 24:417. [PMID: 38951792 PMCID: PMC11218114 DOI: 10.1186/s12887-024-04902-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 06/20/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND The relationship between Helicobacter-pylori(Hp)infection and inflammatory-bowel-disease(IBD) in pediatric-patients remains controversial. We aimed to assess the Hp-infection occurrence in newly-diagnosed pediatric-patients with IBD compared to no-IBD patients. Additionally, we aimed to examine differences in clinical-activity-index(CAI) and endoscopic-severity-score(ESS)between IBD-patients with and without Hp-infection, at baseline and at 1-year-follow-up(FU), after eradication-therapy(ET). METHODS IBD diagnosis was based on Porto-criteria, and all patients underwent gastroscopy at baseline and 1-year FU. For Crohn's-disease(CD) and ulcerative colitis(UC), IBD-CAI and -ESS were classified using PCDAI/SES-CD and PUCAI/UCEIS, respectively. RESULTS 76 IBD-patients were included in the study[35 F(46.1%),median-age 12(range 2-17)]. CD and UC were diagnosed in 29(38.2%) and 45(59.2%)patients, respectively, and unclassified-IBD in two(2.6%)patients. Non-IBD patients were 148[71 F(48.0%),median-age 12(range 1-17)]. Hp-infection at baseline was reported in 7(9.2%) and 18(12.2%)IBD and non-IBD patients, respectively(p = 0.5065). The 7 IBD patients with Hp infection were compared to 69 IBD patients without Hp-infection at baseline evaluation, and no significant differences were reported considering CAI and ESS in these two groups. At 1-year FU, after ET, IBD patients with Hp infection improved, both for CAI and ESS, but statistical significance was not reached. CONCLUSION The occurrence of Hp-infection did not differ between IBD and no-IBD patients. No differences in CAI or ESS were observed at the diagnosis, and after ET no worsening of CAI or ESS was noted at one-year FU, between Hp-positive and -negative IBD patients.
Collapse
Affiliation(s)
- Emanuele Dilaghi
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Rome, Italy
| | - Enrico Felici
- Pediatric and Pediatric Emergency Unit, Children Hospital, AO SS Antonio E Biagio E C. Arrigo, Alessandria, Italy
| | - Edith Lahner
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Rome, Italy
| | - Emanuela Pilozzi
- Department of Clinical and Molecular Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Rome, Italy
| | - Silvia Furio
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Via Grottarossa 1035, Rome, 00189, Italy
| | - Livia Lucchini
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Via Grottarossa 1035, Rome, 00189, Italy
| | - Giovanna Quatrale
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Via Grottarossa 1035, Rome, 00189, Italy
| | - Marisa Piccirillo
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Via Grottarossa 1035, Rome, 00189, Italy
| | - Pasquale Parisi
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Via Grottarossa 1035, Rome, 00189, Italy
| | - Sara Curto
- Pediatric and Pediatric Emergency Unit, Children Hospital, AO SS Antonio E Biagio E C. Arrigo, Alessandria, Italy
| | - Bruno Annibale
- Department of Medical-Surgical Sciences and Translational Medicine, Sant'Andrea Teaching Hospital, Sapienza University of Rome, Rome, Italy
| | - Alessandro Ferretti
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Via Grottarossa 1035, Rome, 00189, Italy
| | - Maurizio Mennini
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Via Grottarossa 1035, Rome, 00189, Italy
| | - Severino Persechino
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Via Grottarossa 1035, Rome, 00189, Italy
| | - Giovanni Di Nardo
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Via Grottarossa 1035, Rome, 00189, Italy.
| |
Collapse
|
2
|
Tang CL, Lian Z, Ding FR, Liang J, Li XY. Schistosoma-related molecules as a new strategy to combat type 1 diabetes through immune regulation. Parasitol Int 2024; 98:102818. [PMID: 37848126 DOI: 10.1016/j.parint.2023.102818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 09/08/2023] [Accepted: 10/12/2023] [Indexed: 10/19/2023]
Abstract
The study of immune regulation mechanisms induced by parasites may help develop new treatment methods for inflammatory diseases including type 1 diabetes, which is related to type 1 immune responses. The negative correlation between schistosomiasis infection and type 1 diabetes has been confirmed, and the mechanism of Schistosoma-mediated prevention of type 1 diabetes may be related to the adaptive and innate immune systems. Schistosoma-related molecules affect immune cell composition and macrophage polarization and stimulate an increase in natural killer T cells. Furthermore, Schistosoma-related molecules can regulate the adaptive immune responses related to the prevention of type 1 diabetes and change the Th1/Th2 and Th17/Treg axis. Our previous review showed the role of regulatory T cells in the protective of type 1 diabetes mediated by Schistosoma. Here, we aim to review the other mechanisms of schistosomiasis infection and Schistosoma-related products in regulating the immune response associated with the treatment of type 1 diabetes.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Wuchang Hospital, Wuhan University of Science and Technology, Wuhan 430063, China
| | - Zhan Lian
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan 430030, China
| | - Fan-Rong Ding
- Wuchang Hospital, Wuhan University of Science and Technology, Wuhan 430063, China
| | - Jun Liang
- Wuhan Pulmonary Hospital, Wuhan Institute for Tuberculosis Control, Wuhan 430030, China.
| | - Xiang-You Li
- Wuchang Hospital, Wuhan University of Science and Technology, Wuhan 430063, China.
| |
Collapse
|
3
|
Tenório MDL, Araujo JMS, de Melo EV, Cazzaniga RA, Aragão ALF, Valois LQ, Severo J, Santos-Filho MAA, Menezes-Silva L, Machado JA, Reed SG, Duthie MS, de Almeida RP, Bezerra-Santos M, de Jesus AR. Association between asthma, rhinitis and atopic dermatitis with leprosy: A case-control study. Indian J Dermatol Venereol Leprol 2023; 89:834-841. [PMID: 37067141 DOI: 10.25259/ijdvl_347_2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 08/01/2022] [Indexed: 02/05/2023]
Abstract
Background Considering the cross-regulation of Th1 and Th2 responses, we hypothesised that atopic diseases (Th2) inhibit the protective Th1 immune response to Mycobacterium leprae and exacerbates leprosy. Objective In this study, we aimed to evaluate the association between leprosy and atopic diseases. Methods To evaluate the association of atopic diseases with leprosy, we conducted a case-control study that included leprosy patients (n = 333) and their household contacts (n = 93). The questionnaire from the International Study of Asthma and Allergies in Childhood, which is validated in several countries for epidemiological diagnosis of atopic diseases, was applied to determine the occurrence of atopic diseases, allergic rhinitis, asthma, and atopic dermatitis among leprosy patients and the household contacts. Results Considering clinical and epidemiological data, among the leprosy group 51.6% (n = 172) were determined to have at least one atopic disease, while atopy was observed less frequently at 40.86% among household contacts (n = 38). When two or more atopic diseases were assessed, the frequency was significantly higher among the leprosy patients than in the household contacts (21.9% vs. 11.8%; P-value = 0.03). Likewise, the frequency of asthma was significantly higher among leprosy patients (21%) than in the household contacts (10.8%; P-value = 0.02). Thus, our analyses revealed an association of atopic diseases with leprosy, with a significant linear increase in the occurrence of leprosy with an increase in the number of atopic diseases (P-value = 0.01). Limitation Due to the difficulties in recruiting household contacts that have prolonged contact with patients, but are not genetically related to the patient, the household contacts group is smaller than the leprosy patient group. Conclusion The data reveal an association between atopic diseases and leprosy outcomes. This knowledge could improve the treatment of leprosy patients with co-incident atopic diseases.
Collapse
Affiliation(s)
- Martha Débora Lira Tenório
- Department of Medicine, Dermathology Clinic, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Jonnia Maria Sherlock Araujo
- Department of Medicine, Dermathology Clinic, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Enaldo Vieira de Melo
- Department of Medicine, Dermathology Clinic, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Rodrigo Anselmo Cazzaniga
- Immunology and Molecular Biology Laboratory, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Ana-Luiza Furtado Aragão
- Department of Medicine, Dermathology Clinic, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Laís Quadros Valois
- Department of Medicine, Dermathology Clinic, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Joanna Severo
- Immunology and Molecular Biology Laboratory, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | | | - Lucas Menezes-Silva
- Immunology and Molecular Biology Laboratory, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Julianne Alves Machado
- Clinical Immunology Clinic, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Steven G Reed
- Department of Vaccines, Host Directed Therapeutics (HDT) Bio Corp, Seattle, Washington, United States
| | - Malcolm S Duthie
- Department of Vaccines, Host Directed Therapeutics (HDT) Bio Corp, Seattle, Washington, United States
| | - Roque Pacheco de Almeida
- Immunology and Molecular Biology Laboratory, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Marcio Bezerra-Santos
- Immunology and Molecular Biology Laboratory, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| | - Amélia Ribeiro de Jesus
- Immunology and Molecular Biology Laboratory, University Hospital, Universidade Federal de Sergipe, Aracaju, SE, Brazil
| |
Collapse
|
4
|
Huang H, Hu D, Chen Z, Xu J, Xu R, Gong Y, Fang Z, Wang T, Chen W. Immunotherapy for type 1 diabetes mellitus by adjuvant-free Schistosoma japonicum-egg tip-loaded asymmetric microneedle patch (STAMP). J Nanobiotechnology 2022; 20:377. [PMID: 35964125 PMCID: PMC9375265 DOI: 10.1186/s12951-022-01581-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022] Open
Abstract
Background Type 1 diabetes mellitus (T1DM) is an autoimmune disease mediated by autoreactive T cells and dominated by Th1 response polarization. Insulin replacement therapy faces great challenges to this autoimmune disease, requiring highly frequent daily administration. Intriguingly, the progression of T1DM has proven to be prevented or attenuated by helminth infection or worm antigens for a relatively long term. However, the inevitable problems of low safety and poor compliance arise from infection with live worms or direct injection of antigens. Microneedles would be a promising candidate for local delivery of intact antigens, thus providing an opportunity for the clinical immunotherapy of parasitic products. Methods We developed a Schistosoma japonicum-egg tip-loaded asymmetric microneedle patch (STAMP) system, which serves as a new strategy to combat TIDM. In order to improve retention time and reduce contamination risk, a specific imperfection was introduced on the STAMP (asymmetric structure), which allows the tip to quickly separate from the base layer, improving reaction time and patient’s comfort. After loading Schistosoma japonicum-egg as the immune regulator, the effects of STAMP on blood glucose control and pancreatic pathological progression improvement were evaluated in vivo. Meanwhile, the immunoregulatory mechanism and biosafety of STAMP were confirmed by histopathology, qRT-PCR, ELISA and Flow cytometric analysis. Results Here, the newly developed STAMP was able to significantly reduce blood glucose and attenuate the pancreatic injury in T1DM mice independent of the adjuvants. The isolated Schistosoma japonicum-eggs micron slowly degraded in the skin and continuously released egg antigen for at least 2 weeks, ensuring localization and safety of antigen stimulation. This phenomenon should be attributed to the shift of Th2 immune response to reduce Th1 polarization. Conclusion Our results exhibited that STAMP could significantly regulate the blood glucose level and attenuate pancreatic pathological injury in T1DM mice by balancing the Th1/Th2 immune responses, which is independent of adjuvants. This technology opens a new window for the application of parasite products in clinical immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1186/s12951-022-01581-9.
Collapse
Affiliation(s)
- Haoming Huang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Dian Hu
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhuo Chen
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Jiarong Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Rengui Xu
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Yusheng Gong
- Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Zhengming Fang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China
| | - Ting Wang
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| | - Wei Chen
- National Demonstration Center for Experimental Basic Medical Education, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,Department of Pharmacology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China. .,Hubei Key Laboratory for Drug Target Researches and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, Wuhan, Hubei, 430030, China.
| |
Collapse
|
5
|
Jafari AA, Keikha M, Mirmoeeni S, Rahimi MT, Jafari R. Parasite-based interventions in systemic lupus erythematosus (SLE): A systematic review. Autoimmun Rev 2021; 20:102896. [PMID: 34274545 DOI: 10.1016/j.autrev.2021.102896] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2021] [Accepted: 05/15/2021] [Indexed: 12/23/2022]
Abstract
BACKGROUND The hygiene hypothesis proposed in 1989 expresses that allergic and infectious diseases are inversely related. Accordingly, it has been demonstrated that infection with some microorganisms such as parasites and helminths can provide a potential immunity and prevent the onset of some life-threatening autoimmune diseases like systemic lupus erythematosus (SLE). Therefore, in this comprehensive study, we systematically reviewed and discussed the use of live parasites or parasitic products in the treatment of mouse models of SLE. METHODS The present systematic review was performed using the following search terms: ("systemic lupus erythematosus" OR "SLE" OR "lupus") AND ("parasite" OR "protozoa" OR "helminths" OR "worms" OR "helminth" OR "worm") in PubMed, Scopus, and Web of Science online databases. We included studies reporting the effect of any intervention using parasites or parasitic-based products on animal models of SLE, which were published until January 20th, 2021 without any language or date restrictions. For each included study, we extracted the authors' names, publication year, type of animal, number of groups, types of intervention, sample size, changes in immunologic cells, auto-Abs, cytokines, and blood cells count, urine analysis, histological analysis of kidney/spleen/liver, outcome and survival. (PROSPERO CRD42020160460). RESULTS A total of 17 eligible articles were included in this systematic review. Sixteen out of the 17 studies reported immunomodulating changes in immunologic cells, cytokines, and/or auto-Abs in mouse models of SLE after using parasitic interventions compared to not-infected or control groups. Moreover, 14 studies reported decreased level of proteinuria and/or favorable kidney, liver, or spleen histological changes. CONCLUSION In conclusion, we have demonstrated that parasites like Hymenolepis microstoma, TPC and ES-62 from Acanthocheilonema viteae, Plasmodium chabaudi, Schistosoma mansoni, and Toxoplasma gondii have favorable immunomodulating effects on SLE outcomes in lupus-prone mice.
Collapse
Affiliation(s)
- Amirhossein Azari Jafari
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mojtaba Keikha
- Department of Public Health, Sirjan School of Medical Sciences, Sirjan, Iran
| | | | - Mohammad Taghi Rahimi
- Center for Health Related Social and Behavioral Sciences Research, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Reza Jafari
- School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran.
| |
Collapse
|
6
|
Masamba P, Kappo AP. Immunological and Biochemical Interplay between Cytokines, Oxidative Stress and Schistosomiasis. Int J Mol Sci 2021; 22:ijms22137216. [PMID: 34281269 PMCID: PMC8268096 DOI: 10.3390/ijms22137216] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 06/20/2021] [Accepted: 06/20/2021] [Indexed: 12/17/2022] Open
Abstract
The host–parasite schistosome relationship relies heavily on the interplay between the strategies imposed by the schistosome worm and the defense mechanisms the host uses to counter the line of attack of the parasite. The ultimate goal of the schistosome parasite entails five important steps: evade elimination tactics, survive within the human host, develop into adult forms, propagate in large numbers, and transmit from one host to the next. The aim of the parasitized host on the other hand is either to cure or limit infection. Therefore, it is a battle between two conflicting aspirations. From the host’s standpoint, infection accompanies a plethora of immunological consequences; some are set in place to defend the host, while most end up promoting chronic disease, which ultimately crosses paths with oxidative stress and cancer. Understanding these networks provides attractive opportunities for anti-schistosome therapeutic development. Hence, this review discusses the mechanisms by which schistosomes modulate the human immune response with ultimate links to oxidative stress and genetic instability.
Collapse
|
7
|
de Oliveira Nóbrega CG, do Nascimento WRC, Santos PDA, de Lorena VMB, Medeiros D, Costa VMA, Barbosa CCGS, Solé D, Sarinho ESC, de Souza VMO. Schistosoma mansoni infection is associated with decreased risk of respiratory allergy symptoms and low production of CCL2. Trop Med Int Health 2021; 26:1098-1109. [PMID: 34107115 DOI: 10.1111/tmi.13639] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES We measured the production of cytokines, chemokines and antibodies involved in allergic responses and sCD23 levels during Schistosoma mansoni infection. METHODS Individuals (n = 164) were selected using the ISAAC questionnaire and parasitological exams. The subjects were divided as follows: those infected individuals with allergy-related symptoms (A-I), those with allergy-related symptoms only (A-NI); those only infected (NA-I); and those non-infected individuals without allergy-related symptoms (NA-NI). We used supernatants from cell culture (mitogenic stimulation) to measure cytokine and chemokine levels using cytometric bead arrays. Serum levels of anti-Ascaris lumbricoides (Asc) and anti-Blomia tropicalis IgE were measured using ImmunoCAP, and sCD23 was measured using ELISA. RESULTS Schistosoma mansoni infection was associated with a lower risk of allergy-related symptoms. In A-I, there were higher levels of TNF-α, IL-10, IL-6, IFN-γ and CXCL8 than in NA-NI group, with TNF-α and IL-6 also at higher levels compared to A-NI group. Levels of IL-6, CXCL8, total and anti-Asc IgE, as well as the numbers of eosinophils, were higher in NA-I than in NA-NI, and the antibodies were also lower in A-NI than in NA-I group. In AI and NA-I, there was less production of CCL2 than in NA-NI. There were no differences in the levels of IL-2, IL-4, IL-17, CCL5, sCD23 and anti-Blomia IgE. CONCLUSIONS Patients with allergy-related symptoms and infected (simultaneously) had higher levels of IL-10; due to the infection, there was increased production of IL-6 and CXCL8 and less CCL2. These data may characterize deviation to Th1 or attenuation of the Th2 response in allergy sufferers in areas endemic for schistosomiasis.
Collapse
Affiliation(s)
| | | | | | | | - Décio Medeiros
- Centro de Pesquisa em Alergia e Imunologia Clínica, Hospital das Clínicas, Universidade Federal de Pernambuco, Recife, Brasil
| | - Vláudia Maria Assis Costa
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Recife, Brasil.,Departamento de Medicina Tropical, Universidade Federal de Pernambuco, Recife, Brasil
| | | | - Dirceu Solé
- Divisão de Alergia, Imunologia Clínica e Reumatologia, Departamento de Pediatria da Universidade Federal de São Paulo, São Paulo, Brasil
| | | | - Valdênia Maria Oliveira de Souza
- Setor de Imunologia, Laboratório de Imunopatologia Keizo Asami, Universidade Federal de Pernambuco, Recife, Brasil.,Departamento de Ciências Farmacêuticas, Universidade Federal de Pernambuco, Recife, Brasil
| |
Collapse
|
8
|
Antigenic cross-reactivity between Schistosoma mansoni and allergenic invertebrates putatively due to shared glycanic epitopes. Sci Rep 2020; 10:3350. [PMID: 32099050 PMCID: PMC7042331 DOI: 10.1038/s41598-020-59892-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 01/28/2020] [Indexed: 01/15/2023] Open
Abstract
Previous studies have shown that rabbit IgG antibodies against Schistosoma mansoni egg antigens (SmSEA) cross-react with allergens in natural rubber latex, peanuts and grass and tree pollens. Here we describe antigenic molecules that cross-react with rabbit anti-S. mansoni IgG antibodies in extracts of the house dust mite (HDM) Dermatophagoides farinae, the Australian cockroach (ACR) Periplaneta australasiae and in the venom of the honey bee Apis mellifera (HBV). Tandem mass spectrometry identified the cross-reactive allergens as Der f 15 in HDM, two homologues of the Periplaneta americana cockroach allergen Cr-PI/Per a 3 in ACR and two isoforms of the allergen Api m 1 (phospholipase A2: PLA2) in HBV. Cross-reactive rabbit anti-SmSEA IgG antibodies eluted from the three invertebrate allergens reacted with S. mansoni egg antigens and variably with schistosome cercarial and worm antigens. Treatment of the electroblotted allergens with sodium metaperiodate abrogated most of the cross-reactivity of the rabbit anti-SmSEA antibodies, suggesting it was due to cross-reactive carbohydrate determinants (CCDs). Furthermore, analyses of the allergens’ amino acid sequences indicated that they had potential for both N- and O-linked glycosylation. A potential role for the CCDs shared by the schistosome and invertebrates in inducing an allergy-protective effect, as proposed by the hygiene hypothesis, is discussed.
Collapse
|
9
|
Fu R, Li L, Hu J, Wang Y, Tao J, Liu H, Liu Z, Zhang W. Elevated TIM3 expression of T helper cells affects immune system in patients with myelodysplastic syndrome. J Investig Med 2019; 67:1125-1130. [PMID: 31511310 DOI: 10.1136/jim-2019-001059] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/14/2019] [Indexed: 12/28/2022]
Abstract
T cell immunoglobulin and mucin domain 3 (TIM3) expression is associated with immunosuppression and clinical outcomes in many diseases. However, the specific mechanism of TIM3 in immune system has not been clarified. In order to illustrate the mechanism of TIM3 in immune system, we analyzed the expression, function and regulation of TIM3 in T helper (Th)1 cells, Th2 cells, Th17 cells and regulatory T cells (Treg) through flow cytometry in patients with myelodysplastic syndrom (MDS). Our data showed elevated proportion of Th2 and Treg cells, while the proportion of Th1 and Th17 cells decreased in patients with MDS (p<0.05) and the expression of TIM3 increased in Th1, Th17 and Treg cells in patients with MDS when compared with expression in control patients (p<0.05). The secretion of transforming growth factor-β in TIM3+Treg cells decreased in patients with MDS. These findings suggested that TIM3 might affect immune helper systems by regulating Treg cells and related immune cells. Therefore, studying the role of the TIM3 pathway in MDS is necessary and may help to provide a new way to explore the pathogenesis and treatments of MDS.
Collapse
Affiliation(s)
- Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiaxin Hu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yingshuai Wang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Jinglian Tao
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Hui Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| | - Wei Zhang
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
10
|
Resende SD, Magalhães FC, Rodrigues-Oliveira JL, Castro VN, Souza CSA, Oliveira EJ, Carneiro M, Geiger SM, Negrão-Corrêa DA. Modulation of Allergic Reactivity in Humans Is Dependent on Schistosoma mansoni Parasite Burden, Low Levels of IL-33 or TNF-α and High Levels of IL-10 in Serum. Front Immunol 2019; 9:3158. [PMID: 30713536 PMCID: PMC6345678 DOI: 10.3389/fimmu.2018.03158] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/21/2018] [Indexed: 12/15/2022] Open
Abstract
Helminth infections and allergies are characterized by a predominant type-2 immune response. In schistosomiasis, the Th-2 response is usually accompanied by induction of immunoregulatory mechanisms that contribute to worm survival and less severe schistosomiasis. Although helminth-induced immunomodulatory mechanisms seem to affect atopy, epidemiological studies on the relationship between helminths and allergy have been inconsistent, and data suggest that the modulatory effects may be influenced by helminth species, chronicity of infection, and parasite burden. Here we performed a cross-sectional study to investigate the effects of Schistosoma mansoni parasite burden and immune response on allergic reactivity of individuals living in a schistosomiasis endemic area in Brazil. Fecal samples from the participants were collected for extensive parasitological examinations by spontaneous sedimentation, Kato-Katz, Helmintex and Saline Gradient tests and molecular detection of S. mansoni by qPCR. Additionally, the concentrations of cytokines and chemokines, total IgE and IgE-reactivity to common house dust allergens were quantified from serum samples. IgE reactivity to dust allergens was detected in 47 individuals (23.8%), and 140 individuals (54.4%) were diagnosed with S. mansoni infection. Most of the infected population (108 individuals) presented very low parasite burden (≤12 eggs/g of feces). The frequency and intensity (p ≤ 0.03) of allergic reactivity were lower in S. mansoni-infected compared with non-infected individuals. Multivariable logistic regression models adjusted by age revealed that allergic reactivity was positively associated with low IL-10 response (OR, 4.55, 95% CI, 0.56–7.36) and high concentration of the inflammatory mediators IL-33 (OR, 2.70, 95% CI, 1.02–7.15) or TNF-α (OR, 6.88, 95% CI, 0.32–143.39) in serum, and inversely associated with S. mansoni infection (OR, 0.38, 95% CI, 0.16–0.87). Most importantly, the logistic regression demonstrated that the modulatory effects of Schistosoma infection depend on parasite burden, with individuals infected with ≤12 eggs/g of feces showing allergic IgE-reactivity similar to non-infected individuals Altogether, our data show that immunomodulation of allergic reactivity depends on S. mansoni burden, low type-2 inflammatory response, and high level of IL-10.
Collapse
Affiliation(s)
- Samira D Resende
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Fernanda C Magalhães
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | | | - Vanessa N Castro
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Carolina S A Souza
- Schistosomiasis Laboratory, René Rachou Research Center, Oswaldo Cruz Foundation, Belo Horizonte, Brazil
| | - Edward J Oliveira
- Schistosomiasis Laboratory, René Rachou Research Center, Oswaldo Cruz Foundation, Belo Horizonte, Brazil
| | - Mariângela Carneiro
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Stefan M Geiger
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Deborah A Negrão-Corrêa
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
11
|
Castro VN, Rodrigues JL, Cardoso DT, Resende SD, Magalhães FC, Souza DC, Requeijo MH, Negrão-Corrêa D, Geiger SM. Systemic Cytokine and Chemokine Profiles in Individuals With Schistosoma mansoni Infection and Low Parasite Burden. Front Immunol 2018; 9:2975. [PMID: 30619332 PMCID: PMC6305627 DOI: 10.3389/fimmu.2018.02975] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 12/04/2018] [Indexed: 11/13/2022] Open
Abstract
Intestinal schistosomiasis, caused by the parasitic trematode Schistosoma mansoni, is a chronic disease and the prolonged and continuous exposure to S. mansoni antigens results in a deviation of the host's immune response. For diagnosis, the Kato-Katz (KK) method is recommended, however, this method showed low accuracy in areas of low endemicity. This study aimed to characterize the cytokine and chemokine profile of individuals with an extremely low parasite load (<4 eggs per gram of feces), e.g., individuals who were detected by alternative parasitological methods, such as the saline gradient and/or Helmintex®. In order to search for immunological markers for infection, the immunological profile in serum samples of these individuals was then compared with patients detected with the KK method and with a higher parasite load and with individuals repetitively negative by extensive stool exams. The study was conducted in Northern Minas Gerais in a rural area of the Municipality of Januária. Serum samples of a total of 139 parasitologically well-characterized individuals were assessed for the following immunological markers by commercially available immunoassays: TNF-α, IL-1β, IL-6, IL-17A, IL-5, IL-10, IL-13, IL-33, IL-27, CCL3, CCL5, CXCL10, CCL11, and CCL17. As a result, there were no significant differences in concentrations or frequencies for immunological markers between egg-negative individuals or individuals with ultra-low (<4 epg) or low (4-99 epg) parasite loads. However, we found significant correlations between egg counts and eosinophil counts and between egg counts and IL-1β or TNF-α concentrations. The most striking alterations were found in individuals with the highest parasite load (≥100 epg). They had significantly higher TNF-α concentrations in serum when compared with individuals with a low parasite load (4-99 epg) and CCL17 concentrations were significantly elevated when compared with egg-negative individuals. Radar diagrams of frequencies for cytokine and chemokine responders in each infection group confirmed a distinct profile only in the infection group with highest parasite loads (≥100 epg).
Collapse
Affiliation(s)
- Vanessa N. Castro
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Jailza L. Rodrigues
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Diogo T. Cardoso
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Samira D. Resende
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Fernanda C. Magalhães
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Dayane C. Souza
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Maira H. Requeijo
- Faculdade da Saúde e Ecologia Humana (FASEH) Vespasiano, Belo Horizonte, Brazil
| | - Deborah Negrão-Corrêa
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Stefan M. Geiger
- Department of Parasitology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| |
Collapse
|
12
|
Lacorcia M, Prazeres da Costa CU. Maternal Schistosomiasis: Immunomodulatory Effects With Lasting Impact on Allergy and Vaccine Responses. Front Immunol 2018; 9:2960. [PMID: 30619318 PMCID: PMC6305477 DOI: 10.3389/fimmu.2018.02960] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Accepted: 12/03/2018] [Indexed: 12/14/2022] Open
Abstract
Early exposure to immune stimuli, including maternal infection during the perinatal period, is increasingly recognized to affect immune predisposition during later life. This includes exposure to not only viral and bacterial infection but also parasitic helminths which remain widespread. Noted effects of helminth infection, including altered incidence of atopic inflammation and vaccine responsiveness, support further research into the impact these infections have for skewing immune responses. At the same time, despite a sea of recommendations, clear phenotypic and mechanistic understandings of how environmental perturbations in pregnancy and nursing modify immune predisposition and allergy in offspring remain unrefined. Schistosomes, as strong inducers of type 2 immunity embedded in a rich network of regulatory processes, possess strong abilities to shift inflammatory and allergic diseases in infected hosts, for example by generating feedback loops that impair T cell responses to heterologous antigens. Based on the current literature on schistosomiasis, we explore in this review how maternal schistosome infection could drive changes in immune system development of offspring and how this may lead to identifying factors involved in altering responses to vaccination as well as manifestations of immune disorders including allergy.
Collapse
Affiliation(s)
- Matthew Lacorcia
- Department of Medicine, Institute for Medical Microbiology, Immunology, and Hygiene, Technical University of Munich, Munich, Germany
| | - Clarissa U Prazeres da Costa
- Department of Medicine, Institute for Medical Microbiology, Immunology, and Hygiene, Technical University of Munich, Munich, Germany
| |
Collapse
|
13
|
Igetei JE, El-Faham M, Liddell S, Schramm G, Doenhoff MJ. Antigenic cross-reactivity between Schistosoma mansoni and pollen allergens from the birch tree (Betula verrucosa) and Timothy grass (Phleum pratense): involvement of shared glycan epitopes and implications for the hygiene hypothesis. Int J Parasitol 2018; 48:345-357. [PMID: 29510117 DOI: 10.1016/j.ijpara.2017.12.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/28/2017] [Accepted: 12/05/2017] [Indexed: 01/05/2023]
Abstract
Previous studies have shown that schistosome infection can protect against allergic symptoms, but the underlying mechanisms are still not fully understood. Here we have shown that rabbit IgG antibodies raised against Schistosoma mansoni soluble egg antigens (SmSEA) are cross-reactive with a wide array of molecules in Timothy grass pollen (TGP) and birch tree pollen (BTP). Five of the cross-reactive pollen molecules (two from TGP and three from BTP) were selected randomly and identified by tandem mass spectrometric (TMS) analysis to be, respectively, the TGP allergens Phl p 1 and Phl p 5b, and BTP glutathione S-transferase (GST), and the BTP allergens Bet v 1 and Bet v 6.0102. Rabbit anti-SmSEA IgG antibodies that cross-reacted with each of the five allergens were found to be reactive with three major S. mansoni egg antigens, IPSE/alpha-1, omega-1 and kappa-5. Pairwise alignment of the amino acid sequences of each of the five TMS-identified pollen allergens with each of the three egg antigens revealed a low level of amino acid sequence identity. Further experiments indicated that the schistosome antigen/allergen cross-reactivity was mostly due to similar glycans present in helminths and plants, but not in mammals: so called cross-reactive carbohydrate determinants (CCDs). Previously, CCDs have been implicated in the cross-reactivity between many plants and invertebrates. Furthermore, pollen-induced anti-CCD IgGs have been found in sera of patients undergoing allergen-specific immunotherapy (SIT) and implicated in the treatment of the allergy. Thus, our finding provides not only possible explanations for the allergy-protective effect of helminth/schistosome infections as explained by the hygiene hypothesis, but also a potential starting point for improved SIT.
Collapse
Affiliation(s)
- Joseph E Igetei
- School of Life Sciences, University Park, University of Nottingham, Nottinghamshire NG7 2RD, UK; Department of Animal and Environmental Biology, Faculty of Life Sciences, University of Benin, Benin City, Edo State, Nigeria.
| | - Marwa El-Faham
- School of Life Sciences, University Park, University of Nottingham, Nottinghamshire NG7 2RD, UK; Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Egypt
| | - Susan Liddell
- School of Biosciences, Sutton Bonington Campus, University of Nottingham LE12 5RD, UK
| | - Gabriele Schramm
- Research Center Borstel, Priority Area Asthma and Allergy, Experimental Pneumology, Parkallee 22, D-23845 Borstel, Germany
| | - Michael J Doenhoff
- School of Life Sciences, University Park, University of Nottingham, Nottinghamshire NG7 2RD, UK
| |
Collapse
|
14
|
Tang CL, Liu ZM, Gao YR, Xiong F. Schistosoma Infection and Schistosoma-Derived Products Modulate the Immune Responses Associated with Protection against Type 2 Diabetes. Front Immunol 2018; 8:1990. [PMID: 29387059 PMCID: PMC5776330 DOI: 10.3389/fimmu.2017.01990] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Accepted: 12/21/2017] [Indexed: 12/27/2022] Open
Abstract
Studies on parasite-induced immunoregulatory mechanisms could contribute to the development of new therapies for inflammatory diseases such as type 2 diabetes (T2D), which is a chronic inflammatory disease characterized by persistent elevated glucose levels due to insulin resistance. The association between previous Schistosoma infection and T2D has been confirmed—Schistosoma infection and Schistosoma-derived products modulate the immune system, including innate and acquired immune responses, contributing to T2D disease control. Schistosoma infections and Schistosoma-derived molecules affect the immune cell composition in adipose tissue, dampening inflammation and improving glucose tolerance. This protective role includes the polarization of immune cells to alternatively activated macrophages, dendritic cells, eosinophils, and group 2 innate lymphoid cells. Furthermore, Schistosoma infection and Schistosoma products are effective for the treatment of T2D, as they increase the number of type 2 helper T cells (Th2) and regulatory T cells (Tregs) and decrease type 1 helper T cells (Th1) and type 17 helper T cells (Th17) cells. Thus, our aim was to comprehensively review the mechanism through which Schistosoma infection and Schistosoma products modulate the immune response against T2D.
Collapse
Affiliation(s)
- Chun-Lian Tang
- Department of Science and Education, Wuchang Hospital, Wuhan, China
| | - Zhi-Ming Liu
- Department of Science and Education, Wuchang Hospital, Wuhan, China
| | - Yan Ru Gao
- Medical Department, City College, Wuhan University of Science and Technology, Wuhan, China
| | - Fei Xiong
- The Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
15
|
Lopes DM, de Almeida TVVS, de Souza RDP, Ribeiro LEV, Page B, Fernandes JDS, Carvalho EM, Cardoso LS. Susceptibility of dendritic cells from individuals with schistosomiasis to infection by Leishmania braziliensis. Mol Immunol 2017; 93:173-183. [PMID: 29197260 DOI: 10.1016/j.molimm.2017.11.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/09/2017] [Accepted: 11/18/2017] [Indexed: 12/18/2022]
Abstract
Coinfection with leishmaniasis and schistosomiasis has been associated with increased time to healing of cutaneous lesions of leishmaniasis. The objective of this study was to evaluate the effect of Leishmania braziliensis infection on co-cultures of monocyte-derived dendritic cells (MoDCs) with autologous lymphocytes from patients with schistosomiasis and patients with cutaneous leishmaniasis. MoDCs were differentiated from peripheral blood monocytes, isolated by magnetic beads, infected with L. braziliensis, and co-cultured with autologous lymphocytes. Expression of HLA-DR, CD1a, CD83, CD80, CD86, CD40, and the IL-10 receptor (IL-10R) on MoDCs as well as CD28, CD40L, CD25, and CTLA-4 on lymphocytes were evaluated by flow cytometry. The production of the cytokines IL-10, TNF, IL-12p40, and IFN-γ were evaluated by sandwich ELISA of the culture supernatant. The infectivity evaluation was performed by light microscopy after concentration of cells by cytospin and Giemsa staining. It was observed that the frequency of MoDCs expressing CD83, CD80, and CD86 as well as the MFI of HLA-DR were smaller in the group of patients with schistosomiasis compared to the group of patients with leishmaniasis. On the other hand, the frequency of IL-10R on MoDCs was higher in patients with schistosomiasis than in patients with leishmaniasis. CD4+ and CD8+ T lymphocytes from patients with schistosomiasis presented a lower frequency of CD28 and a higher frequency of CTLA-4 compared to lymphocytes from patients with leishmaniasis. Levels of IL-10 were higher in the supernatants of co-cultures from individuals with schistosomiasis compared to those with leishmaniasis. However, levels of TNF, IL-12p40, and IFN-γ were lower in the group of individuals with schistosomiasis. Regarding the frequency of MoDCs infected by L. braziliensis after 72h in culture, it was observed that higher frequencies of cells from patients with schistosomiasis were infected compared to cells from patients with leishmaniasis. It was concluded that MoDCs from patients with schistosomiasis are more likely to be infected by L. braziliensis, possibly due to a lower degree of activation and a regulatory profile.
Collapse
Affiliation(s)
- Diego Mota Lopes
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT- DT) -CNPQ/MCT, Brazil
| | - Tarcísio Vila Verde S de Almeida
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil; Escola Bahiana de Medicina e Saúde Pública, Salvador, Bahia, Brazil
| | - Robson da Paixão de Souza
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Luís Eduardo Viana Ribeiro
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Brady Page
- Tulane University School of Medicine, New Orleans, LA, USA
| | | | - Edgar M Carvalho
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT- DT) -CNPQ/MCT, Brazil; Instituto Gonçalo Moniz, Fundação Oswaldo Cruz, Salvador, BA, Brazil
| | - Luciana Santos Cardoso
- Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT- DT) -CNPQ/MCT, Brazil; Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, UFBA, Salvador, Bahia, Brazil.
| |
Collapse
|
16
|
Marciani DJ. Effects of immunomodulators on the response induced by vaccines against autoimmune diseases. Autoimmunity 2017; 50:393-402. [PMID: 28906131 DOI: 10.1080/08916934.2017.1373766] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
A promising treatment for T-cell-mediated autoimmune diseases is the induction of immune tolerance by modulating the immune response against self-antigens, an objective that may be achieved by vaccination. There are two main types of vaccines currently under development. The tolerogenic vaccines, composed of proteins formed by a cytokine fused to a self-antigen, which usually induce tolerance by eliminating the T-cells that are immune reactive against the self-antigen. The immunogenic vaccines, comprised of a self-antigen plus a sole Th2 adjuvant either free or conjugated, that alleviate autoimmunity by switching the immune response against the self-antigen, from a damaging pro-inflammatory Th1/Th17 to an anti-inflammatory Th2 immunity. Another type of vaccines is the DNA vaccines, where cells transiently express the self-antigen encoded by DNA, which induces a Th2 immunity. Actually, DNA vaccines can benefit from the presence of an adjuvant that elicits a systemic sole Th2 immunity to enhance the initially weak immune response characteristic of these vaccines. While in the tolerogenic vaccines, cytokines are the endogenous immunomodulators, in the immunogenic vaccines, the adjuvants are exogenous agents that elicit Th2 immunity with a production of anti-inflammatory cytokines and antibodies against the self-antigen. Because the commonly used Th2 adjuvant alum, fails to induce an effective immunity in the elderly population, it is unlikely that it would be widely used. Another Th2 adjuvant, the oil/water emulsions mixed with the antigen, while effective in vaccines against infectious agents, due to potential aldehydes in their formulation may be not suitable for autoimmune vaccines. A unique compound is glatiramer, which seems to be both a random polypeptide antigen and an immune modulator that biases the response to Th2 immunity. Its mechanism of action seems to implicate binding to MHC-II, which alters the outcome of T-cell signaling, leading to anergy. Glatiramer, while effective in the treatment of multiple sclerosis has not shown efficacy in other autoimmune diseases. An important new group of promising sole Th2 adjuvants are the fucosylated glycans, which by binding to DC-SIGN bias dendritic cells to Th2 immunity while inhibiting Th1/Th7 immunities. These glycans are similar to those produced by parasitic helminths to prevent inflammatory responses by mammalian hosts. A novel group of sole Th2 adjuvants are some plant-derived fucosylated triterpene glycosides, which share the immune modulatory properties from the fucosylated glycans. These glycosides have also an aldehyde group that delivers an alternative co-stimulatory signal to T-cells, averting the anergy associated with aging due to the loss of the CD28 receptor on T-cells. Hence, the development of vaccines to treat and/or prevent autoimmune conditions and some proteopathies, will significantly benefit from the availability of new sole Th2 adjuvants that while inducing an anti-inflammatory immunity, they do not abrogate pro-inflammatory Th1/Th17 immunities.
Collapse
|
17
|
Igetei JE, El-Faham M, Liddell S, Doenhoff MJ. Antigenic cross-reactivity between Schistosoma mansoni and peanut: a role for cross-reactive carbohydrate determinants (CCDs) and implications for the hygiene hypothesis. Immunology 2017; 150:506-517. [PMID: 28201853 DOI: 10.1111/imm.12711] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Revised: 12/08/2016] [Accepted: 01/01/2017] [Indexed: 12/21/2022] Open
Abstract
The antigenic reactivity of constituents of Schistosoma mansoni and peanut (Arachis hypogaea) was investigated to determine whether identical antigenic epitopes possessed by both organisms provided a possible explanation for the negative correlation between chronic schistosome infection and atopy to allergens. Aqueous extracts of peanuts were probed in Western immunoblots with rabbit IgG antibodies raised against the egg, cercarial and adult worm stages of S. mansoni. Several molecules in the peanut extract were antigenically reactive with antibodies from the various rabbit anti-schistosome sera. A pair of cross-reactive peanut molecules at ~30 000-33 000 molecular weight was purified and both proteins were identified by mass spectrometric analysis as the peanut allergen Ara h 1. Anti-S. mansoni soluble egg antigen antibodies that were eluted off the peanut molecules reacted with two S. mansoni egg antigens identified by mass spectrometry as IPSE/α-1 and κ-5. Alignments of the amino acid sequences of Ara h 1 and either IPSE/α-1 or κ-5 revealed a low level of peptide sequence identity. Incubation of nitrocellulose paper carrying electrophoresed peanut molecules, six constituents of other allergic plants and S. mansoni egg antigens in a mild solution of sodium metaperiodate before probing with antibodies, inhibited most of the cross-reactivities. The results are consistent with the antigenic cross-reactive epitopes of S. mansoni egg antigens, peanut and other allergic plants being cross-reactive carbohydrate determinants (CCDs). These findings are novel and an explanation based on 'blocking antibodies' could provide an insight for the inverse relationship observed between schistosome infection and allergies.
Collapse
Affiliation(s)
- Joseph E Igetei
- School of Life Sciences, University of Nottingham, Nottingham, UK NG7 2RD.,Department of Animal and Environmental Biology, Faculty of Life Sciences, University of Benin, Benin City, Edo State, Nigeria
| | - Marwa El-Faham
- School of Life Sciences, University of Nottingham, Nottingham, UK NG7 2RD.,Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Susan Liddell
- School of Biosciences, University of Nottingham, Sutton Bonington, UK LE12 5RD
| | - Michael J Doenhoff
- School of Life Sciences, University of Nottingham, Nottingham, UK NG7 2RD
| |
Collapse
|
18
|
Oliveira SC, Figueiredo BC, Cardoso LS, Carvalho EM. A double edged sword: Schistosoma mansoni Sm29 regulates both Th1 and Th2 responses in inflammatory mucosal diseases. Mucosal Immunol 2016; 9:1366-1371. [PMID: 27554296 DOI: 10.1038/mi.2016.69] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Sergio C Oliveira
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação, Salvador, Bahia, Brazil
| | - Barbara C Figueiredo
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.,Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação, Salvador, Bahia, Brazil
| | - Luciana S Cardoso
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação, Salvador, Bahia, Brazil.,Departamento de Análises Clínicas e Toxicológicas, Faculdade de Farmácia, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| | - Edgar M Carvalho
- Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Conselho Nacional de Desenvolvimento Científico e Tecnológico, Ministério de Ciência Tecnologia e Inovação, Salvador, Bahia, Brazil.,Serviço de Imunologia, Hospital Universitário Professor Edgard Santos, Universidade Federal da Bahia, Salvador, Bahia, Brazil
| |
Collapse
|
19
|
Wang X, Wang J, Liang Y, Ni H, Shi L, Xu C, Zhou Y, Su Y, Mou X, Chen D, Mao C. Schistosoma japonicum HSP60-derived peptide SJMHE1 suppresses delayed-type hypersensitivity in a murine model. Parasit Vectors 2016; 9:147. [PMID: 26971312 PMCID: PMC4789290 DOI: 10.1186/s13071-016-1434-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/05/2016] [Indexed: 12/20/2022] Open
Abstract
Background Parasite-derived molecules with immunomodulatory properties, which have been optimised during host-parasite co-evolution, exhibit potential applications as novel immunotherapeutics. We have previously demonstrated that Schistosoma japonicum HSP60-derived peptide SJMHE1 induces CD4+CD25+ regulatory T-cells (Tregs) and that adoptively transferred SJMHE1-induced CD4+CD25+ Tregs inhibit delayed-type hypersensitivity (DTH) in mice. However, multiple concerns regarding this method render this treatment unsuitable. To gain further insights into the potential effects of SJMHE1, we used ovalbumin (OVA)-induced DTH and evaluated the effect of SJMHE1 on DTH mice. Methods BALB/c mice were sensitised with OVA alone or combined with SJMHE1 and then challenged with OVA to induce DTH. We first analysed the potential effects of SJMHE1 by measuring DTH responses, T-cell responses, cytokine secretion, and Treg proportions. We then evaluated the expression levels of IL-10 and TGF-β1 in CD4+CD25+ T-cells during DTH and Treg generation to identify the mechanism by which SJMHE1 suppresses DTH. Results SJMHE1 modulated the effector response against OVA-induced DTH and stimulated the production of the anti-inflammatory cytokines IL-10 and TGF-β1 in immunised mice through a mechanism involving CD4+CD25+ Tregs. SJMHE1-induced CD4+CD25+ Tregs expressed high levels of CTLA-4, IL-10, and TGF-β1, which substantially contributed to the suppressive activity during DTH. The administration of SJMHE1 to DTH in mice led to the expansion of CD4+CD25+ Tregs from CD4+CD25− T-cells in the periphery, which inhibited DTH responses. Conclusions Our study proves that the parasite-driven peptide suppresses DTH in mice, which may confer a new option for inflammation treatment.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China. .,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.
| | - Jun Wang
- Department of Nuclear Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
| | - Yong Liang
- Clinical Laboratory, Huai'an Hospital Affiliated of Xuzhou Medical College, Huaian, Jiangsu, 223300, China
| | - Hongchang Ni
- Department of Nuclear Medicine, The Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, China
| | - Liang Shi
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Chengcheng Xu
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yuepeng Zhou
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Yuting Su
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Xiao Mou
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Deyu Chen
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Chaoming Mao
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China.,Department of Nuclear Medicine and Institute of Oncology, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| |
Collapse
|
20
|
Hussain K, Letley DP, Greenaway AB, Kenefeck R, Winter JA, Tomlinson W, Rhead J, Staples E, Kaneko K, Atherton JC, Robinson K. Helicobacter pylori-Mediated Protection from Allergy Is Associated with IL-10-Secreting Peripheral Blood Regulatory T Cells. Front Immunol 2016; 7:71. [PMID: 27014260 PMCID: PMC4779884 DOI: 10.3389/fimmu.2016.00071] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 02/15/2016] [Indexed: 12/18/2022] Open
Abstract
Helicobacter pylori infections are usually established in early childhood and continuously stimulate immunity, including T-helper 1 (Th1), Th17, and regulatory T-cell (Treg) responses, throughout life. Although known to be the major cause of peptic ulcer disease and gastric cancer, disease occurs in a minority of those who are infected. Recently, there has been much interest in beneficial effects arising from infection with this pathogen. Published data robustly show that the infection is protective against asthma in mouse models. Epidemiological studies show that H. pylori is inversely associated with human allergy and asthma, but there is a paucity of mechanistic data to explain this. Since Th1 and Treg responses are reported to protect against allergic responses, we investigated if there were links between the human systemic Th1 and Treg response to H. pylori and allergen-specific IgE levels. The human cytokine and T-cell responses were examined using peripheral blood mononuclear cells (PBMCs) from 49 infected and 58 uninfected adult patients. Concentrations of total and allergen-specific plasma IgE were determined by ELISA and ImmunoCAP assays. These responses were analyzed according to major virulence factor genotypes of the patients' colonizing H. pylori strains. An in vitro assay was employed, using PBMCs from infected and uninfected donors, to determine the role of Treg cytokines in the suppression of IgE. Significantly higher frequencies of IL-10-secreting CD4(+)CD25(hi) Tregs, but not H. pylori-specific Th1 cells, were present in the peripheral blood of infected patients. Total and allergen-specific IgE concentrations were lower when there was a strong Treg response, and blocking IL-10 in vitro dramatically restored IgE responses. IgE concentrations were also significantly lower when patients were infected with CagA(+) strains or those expressing the more active i1 form of VacA. The systemic IL-10(+) Treg response is therefore likely to play a role in H. pylori-mediated protection against allergy in humans.
Collapse
Affiliation(s)
- Khiyam Hussain
- Nottingham Digestive Diseases Biomedical Research Unit, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK; Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Darren P Letley
- Nottingham Digestive Diseases Biomedical Research Unit, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK; Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - A Borgel Greenaway
- Nottingham Digestive Diseases Biomedical Research Unit, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK; Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Rupert Kenefeck
- Nottingham Digestive Diseases Biomedical Research Unit, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK; Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Jody A Winter
- Nottingham Digestive Diseases Biomedical Research Unit, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK; Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - William Tomlinson
- Nottingham Digestive Diseases Biomedical Research Unit, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK; Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Joanne Rhead
- Nottingham Digestive Diseases Biomedical Research Unit, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK; Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Emily Staples
- Nottingham Digestive Diseases Biomedical Research Unit, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK; Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Kazuyo Kaneko
- Nottingham Digestive Diseases Biomedical Research Unit, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK; Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - John C Atherton
- Nottingham Digestive Diseases Biomedical Research Unit, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK; Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| | - Karen Robinson
- Nottingham Digestive Diseases Biomedical Research Unit, Queen's Medical Centre, Nottingham University Hospitals, Nottingham, UK; Centre for Biomolecular Sciences, University of Nottingham, Nottingham, UK
| |
Collapse
|
21
|
Robinson K. Helicobacter pylori-Mediated Protection against Extra-Gastric Immune and Inflammatory Disorders: The Evidence and Controversies. Diseases 2015; 3:34-55. [PMID: 28943607 PMCID: PMC5548235 DOI: 10.3390/diseases3020034] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 03/03/2015] [Accepted: 03/05/2015] [Indexed: 12/11/2022] Open
Abstract
A large number of studies link H. pylori infection with a reduced risk of developing extra-gastric conditions such as allergy, asthma, inflammatory bowel disease, coeliac disease and multiple sclerosis. The strength of the evidence for these protective associations is quite variable, and published studies often do not agree. This review article discusses some of the reasons for these discrepancies, and the difficulties faced when designing studies. Examples of some protective disease associations are described in detail, where the evidence is most abundant and thought to be more reliable. The most convincing of these are supported by published mechanistic data, for example with animal models, or incidence of disease exacerbation in humans following H. pylori eradication. Although controversial, this field is very important as the prevalence of H. pylori is decreasing throughout the world whilst many chronic diseases are becoming more common. These trends are likely to continue in the future, therefore it is important that we fully understand if and how H. pylori confers protection.
Collapse
Affiliation(s)
- Karen Robinson
- Nottingham Digestive Diseases Biomedical Research Unit, School of Medicine, University of Nottingham, Nottingham NG7 2RD, UK.
| |
Collapse
|
22
|
Afifi MA, Jiman-Fatani AA, El Saadany S, Fouad MA. Parasites-allergy paradox: Disease mediators or therapeutic modulators. J Microsc Ultrastruct 2015; 3:53-61. [PMID: 30023182 PMCID: PMC6014186 DOI: 10.1016/j.jmau.2015.03.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2015] [Accepted: 03/09/2015] [Indexed: 02/07/2023] Open
Abstract
The noticeable phenomenon of an increased frequency of immune-inflammatory disorders, in the industrialized world, has led to the implication of parasitic infections in the pathophysiology of these diseases. Most of the studies investigated the infection connection to allergy have centered on helminthes. Parasitic helminthes are a group of metazoans that are evolutionary diverse, yet converge to evolve common modes of immunomodulation. Helminth immunoregulation is mainly mediated by a regulatory response including Treg and Breg cells with alternatively-activated macrophages. There is increasing evidence for a causal relationship between helminth infection and allergic hyporesponsiveness, however, conflicting data are still generating. The helminth immunoregulation seems to be species-specific and phase-specific. It depends on the stage of the clinical disease which correlates with a corresponding parasitic stage (egg, larva or mature adult). Here, we review the cellular and molecular mechanisms utilized by helminthes to manipulate the immune system and the consequent bystander immunomodulatory responses toward environmental allergens. We especially focus on parasitic species and molecules involved in the modulation of allergic disorders and summarize the experimental and clinical trials using them as therapeutic agents. We also discuss the potentials and obstacles, for helminthes and/or their derived molecules, to emerge as novel therapeutic modalities.
Collapse
Affiliation(s)
- Mohammed A. Afifi
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
- Corresponding author at: Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, P.O. Box 80205, Jeddah 21589, Saudi Arabia. Tel.: +966 569722590. E-mail address: (M.A. Afifi)
| | - Asif A. Jiman-Fatani
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sherif El Saadany
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mahmoud A. Fouad
- Department of Medical Microbiology and Parasitology, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
23
|
Hasby EA, Hasby Saad MA, Shohieb Z, El Noby K. FoxP3+ T regulatory cells and immunomodulation after Schistosoma mansoni egg antigen immunization in experimental model of inflammatory bowel disease. Cell Immunol 2015; 295:67-76. [PMID: 25766778 DOI: 10.1016/j.cellimm.2015.02.013] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Revised: 02/18/2015] [Accepted: 02/19/2015] [Indexed: 02/05/2023]
Abstract
To assess the effect of Schistosoma mansoni egg antigen immunization on the immunomodulation in dextran sodium sulfate (DSS) induced colitis as an experimental model of IBD in comparison to non immunization and healthy control. The study was performed on 180 mice; 25 healthy control, 15 to identify the inflammatory peak of DSS, 25 received DSS for 7 days; 90 infected with S. mansoni cercariae to collect eggs for antigen preparation, and 25 immunized with the prepared antigen then received DSS course. Disease activity index, macroscopic & microscopic inflammatory scores, FoxP3+ T regulatory cell count, myeloperoxidase activity, and Th1/Th2 cytokine profile were compared in studied groups. Immunization induced both FoxP3+ T(regs) and Th2 cytokines to establish a state of immune homeostasis and create a quiescent steadier immune response to DSS. S. mansoni egg antigen succeeded in acting like a prophylactic helminthic therapy as it has a profitable modulatory effect on DSS-induced colitis model.
Collapse
Affiliation(s)
- Eiman A Hasby
- Pathology Department, Tanta Faculty of Medicine, Egypt.
| | | | - Zeinab Shohieb
- Medical Parasitology Department, Tanta Faculty of Medicine, Egypt
| | - Kholoud El Noby
- Medical Parasitology Department, Tanta Faculty of Medicine, Egypt
| |
Collapse
|
24
|
Odegaard JI, Hsieh MH. Immune responses to Schistosoma haematobium infection. Parasite Immunol 2014; 36:428-38. [PMID: 25201406 DOI: 10.1111/pim.12084] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Accepted: 10/20/2013] [Indexed: 02/02/2023]
Abstract
Urogenital schistosomiasis is one of the greatest single infectious sources of human morbidity and mortality known. Through a complex cycle of infection, migration and eventual maturation and mating, S. haematobium (the aetiological agent of urogenital schistosomiasis) deposits highly immunogenic eggs within the bladder and other pelvic organs, activating a wide range of immune programs that determine both infection outcome as well as downstream immunopathology. In this review, we discuss the experimental and observational bases for our current understanding of these immune programs, focusing specifically on how the balance of type 1 and type 2 responses governs subsequent immunopathology and clinical outcome.
Collapse
Affiliation(s)
- J I Odegaard
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | | |
Collapse
|
25
|
Wang H, Li J, Pu H, Hasan B, Ma J, Jones MK, Zheng K, Zhang X, Ma H, McManus DP, Lin R, Wen H, Zhang W. Echinococcus granulosus infection reduces airway inflammation of mice likely through enhancing IL-10 and down-regulation of IL-5 and IL-17A. Parasit Vectors 2014; 7:522. [PMID: 25409540 PMCID: PMC4256745 DOI: 10.1186/s13071-014-0522-6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 11/05/2014] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Cystic echinococcosis (CE) is a near cosmopolitan zoonosis caused by the larval stage of the dog tapeworm Echinococcus granulosus. E. granulosus infection induces a polarized T-helper type 2 (Th2) systematic immune response in its intermediate hosts. However, it is not known whether the infection modulates lung inflammation by regulating local immune response. In this study, we examined the effects of E. granulosus infection on mouse ovalbumin (OVA)-induced asthma model. METHODS BALB/c mice were intraperitoneally transplanted with 50 small E. granulosus cysts cultured in vitro. At 3 months post-inoculation, the mice were sensitized and challenged with ovalbumin (OVA). For histopathological studies, hematoxylin eosin and periodic acid schiff staining was used to examine the inflammatory cells infiltration and goblet cells hyperplasia, respectively. Cytokine levels were measured by mouse cytometric bead array (CBA) Kit and quantitative RT-PCR and other molecular biological approaches. Airway hyperresponsiveness was assessed in response to increasing doses of methacholine. Serum immunoglobulins were determined by ELISA. RESULTS E. granulosus infection significantly increased Th2 and Treg cytokine levels in serum and lung tissues, but down-regulated the expression of IL-5 in the lungs and IL-17A in serum and lung tissues of asthmatic mice sensitized and challenged with OVA. Histological staining of lung tissues showed that E. granulosus infection significantly reduced the severity of OVA-induced airway inflammation including reduction of eosinophil cell infiltration and mucus production. The E. granulosus infection also reduced eosinophil accumulation induced by OVA in bronchoalveolar lavage fluid (BALF) and also ameliorated airway hyperresponsiveness, a hallmark symptom of asthma. CONCLUSIONS E. granulosus infection remarkably reduces the severity of OVA-induced airway inflammation likely through enhancing IL-10 and down-regulation of IL-5 and IL-17A.
Collapse
Affiliation(s)
- Hui Wang
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| | - Jun Li
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China. .,Department of Immunology, Xinjiang Medical University, Urumqi, 830011, Xinjiang, China.
| | - Hongwei Pu
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| | - Bilal Hasan
- Laboratory of Respiratory Physiology and Pathology, Traditional Chinese Medicine Hospital affiliated with Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| | - Jinfeng Ma
- Department of Epidemiology and Health statistics, School of Public Health Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| | - Malcolm K Jones
- School of Veterinary Sciences, The University of Queensland, Queensland, Warrego Highway, Gatton, Qld, 4343, Australia.
| | - Kan Zheng
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| | - Xue Zhang
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| | - Haimei Ma
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| | - Donald P McManus
- Molecular Parasitology Laboratory, Infectious Diseases Division, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| | - Renyong Lin
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| | - Hao Wen
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| | - Wenbao Zhang
- State Key Laboratory Incubation Base of Xinjiang Major Diseases Research, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang, 830054, China.
| |
Collapse
|
26
|
Abstract
Even at a time when HIV/AIDS and immunosuppressive therapy have increased the number of individuals living with significant immunocompromise, diabetes mellitus (DM) remains a major comorbid disorder for several rare but potentially lethal infections, including rhino-orbital-cerebral mucormycosis and malignant external otitis. DM is also a commonly associated condition in patients with nontropical pyomyositis, pyogenic spinal infections, Listeria meningitis, and blastomycosis. As West Nile virus spread to and across North America over a decade ago, DM appeared in many series as a risk factor for death or neuroinvasive disease. More recently, in several large international population-based studies, DM was identified as a risk factor for herpes zoster. The relationships among infection, DM, and the nervous system are multidirectional. Viral infections have been implicated in the pathogenesis of type 1 and type 2 DM, while parasitic infections have been hypothesized to protect against autoimmune disorders, including type 1 DM. DM-related neurologic disease can predispose to systemic infection - polyneuropathy is the predominant risk factor for diabetic foot infection. Because prognosis for many neurologic infections depends on timely institution of antimicrobial and sometimes surgical therapy, neurologists caring for diabetic patients should be familiar with the clinical features of the neuroinfectious syndromes associated with DM.
Collapse
Affiliation(s)
- Cheryl A Jay
- Department of Neurology, University of California San Francisco and Neurology Service, San Francisco General Hospital, San Francisco, CA, USA.
| | - Marylou V Solbrig
- Departments of Internal Medicine (Neurology) and Medical Microbiology, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
27
|
Campolina SS, Araujo MSS, Rezende TMRL, Matoso L, Quites HFO, Teixeira-Carvalho A, Martins-Filho OA, Gazzinelli A, Correa-Oliveira R. Effective anthelmintic therapy of residents living in endemic area of high prevalence for Hookworm and Schistosoma mansoni infections enhances the levels of allergy risk factor anti-Der p1 IgE. RESULTS IN IMMUNOLOGY 2013; 5:6-12. [PMID: 25905031 PMCID: PMC4404415 DOI: 10.1016/j.rinim.2013.11.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
In this work were investigated the relationship between Hookworm/Schistosoma mansoni infections and allergy related risk factors in two endemic areas with distinct prevalence of infections and co-infection. The intensity of infections, eosinophilia, allergy risk factors, infections status and anti-Der p1 IgE levels before and 2 years (population 1) and 3 years (population 2) after anthelmintic treatment, were evaluated. It was observed that the population with lower prevalence and intensity of infection (population 2) had lower eosinophils counts (>600/mm3) and higher animal contact than the population with higher parasites intensity (population 1). After anthelmintic treatment the intensity of S. mansoni single infection decreased, but no changes were observed in Hookworm and co-infected individuals. The anthelmintic treatment also enhanced anti-Der p1 IgE optical density in ELISA on the subgroups that became negative for helminth infection regardless of their previous infection condition in population 1. Facing that, we evaluated the anti-Der p1 IgE reactivity index, and the ratio (after/before treatment) was significantly higher in patients co-infected before treatment. On the other hand, no association between anti-Der p1 IgE reactivity index and the intensity of infections were observed. In conclusion, effective anthelmintic therapy of subjects from endemic areas with high prevalence of Hookworm and S. mansoni infections enhances anti-Der p1 IgE levels.
Collapse
Affiliation(s)
- Sabrina S Campolina
- Laboratório de Imunologia Celular e Molecular, Centro de Pesquisas René Rachou FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Marcio S S Araujo
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Tércia M R L Rezende
- Faculdade de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Leonardo Matoso
- Laboratório de Imunologia Celular e Molecular, Centro de Pesquisas René Rachou FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Humberto F O Quites
- Faculdade de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Andréa Teixeira-Carvalho
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Olindo A Martins-Filho
- Laboratório de Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil
| | - Andrea Gazzinelli
- Faculdade de Enfermagem, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil ; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Belo Horizonte, Minas Gerais, Brazil
| | - Rodrigo Correa-Oliveira
- Laboratório de Imunologia Celular e Molecular, Centro de Pesquisas René Rachou FIOCRUZ-Minas, Belo Horizonte, Minas Gerais, Brazil ; Instituto Nacional de Ciência e Tecnologia em Doenças Tropicais (INCT-DT), Belo Horizonte, Minas Gerais, Brazil
| |
Collapse
|
28
|
Tweyongyere R, Naniima P, Mawa PA, Jones FM, Webb EL, Cose S, Dunne DW, Elliott AM. Effect of maternal Schistosoma mansoni infection and praziquantel treatment during pregnancy on Schistosoma mansoni infection and immune responsiveness among offspring at age five years. PLoS Negl Trop Dis 2013; 7:e2501. [PMID: 24147175 PMCID: PMC3798616 DOI: 10.1371/journal.pntd.0002501] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Accepted: 09/10/2013] [Indexed: 11/18/2022] Open
Abstract
Introduction Offspring of Schistosoma mansoni-infected women in schistosomiasis-endemic areas may be sensitised in-utero. This may influence their immune responsiveness to schistosome infection and schistosomiasis-associated morbidity. Effects of praziquantel treatment of S. mansoni during pregnancy on risk of S. mansoni infection among offspring, and on their immune responsiveness when they become exposed to S. mansoni, are unknown. Here we examined effects of praziquantel treatment of S. mansoni during pregnancy on prevalence of S. mansoni and immune responsiveness among offspring at age five years. Methods In a trial in Uganda (ISRCTN32849447, http://www.controlled-trials.com/ISRCTN32849447/elliott), offspring of women treated with praziquantel or placebo during pregnancy were examined for S. mansoni infection and for cytokine and antibody responses to SWA and SEA, as well as for T cell expression of FoxP3, at age five years. Results Of the 1343 children examined, 32 (2.4%) had S. mansoni infection at age five years based on a single stool sample. Infection prevalence did not differ between children of treated or untreated mothers. Cytokine (IFNγ, IL-5, IL-10 and IL-13) and antibody (IgG1, Ig4 and IgE) responses to SWA and SEA, and FoxP3 expression, were higher among infected than uninfected children. Praziquantel treatment of S. mansoni during pregnancy had no effect on immune responses, with the exception of IL-10 responses to SWA, which was higher in offspring of women that received praziquantel during pregnancy than those who did not. Conclusion We found no evidence that maternal S. mansoni infection and its treatment during pregnancy influence prevalence and intensity of S. mansoni infection or effector immune response to S. mansoni infection among offspring at age five years, but the observed effects on IL-10 responses to SWA suggest that maternal S. mansoni and its treatment during pregnancy may affect immunoregulatory responsiveness in childhood schistosomiasis. This might have implications for pathogenesis of the disease. Infections with the blood fluke Schistosoma mansoni that cause schistosomiasis (also called Bilharzia) were not usually treated during pregnancy until 2002, but in 2002 a World Health Organization (WHO) team of experts recommended that praziquantel treatment of S. mansoni during pregnancy should be done. However, there was limited information on the effects of maternal S. mansoni infection and treatment during pregnancy on the outcomes in the offspring. We conducted a study in the Entebbe peninsula within Lake Victoria in Uganda to examine whether maternal S. mansoni infection or its treatment during pregnancy may have effects on the children's susceptibility to the infection. The children were examined at age five years old for the level of S. mansoni infection and for immune responses to schistosomes. At five years old few of the children in our study cohort were infected with S. mansoni. Our findings suggest that maternal infection with, or praziquantel treatment of S. mansoni during pregnancy did not influence the level of S. mansoni infection among the offspring. However our findings suggest an influence on regulation of the body's immune responses to schistosomes, which may have some effect on the progress of disease manifestations. This is an issue that needs further investigation.
Collapse
Affiliation(s)
- Robert Tweyongyere
- Department of Veterinary Pharmacy, Clinical and Comparative Medicine, Makerere University, Kampala, Uganda
- Uganda Virus Research Institute, Entebbe, Uganda
- * E-mail:
| | | | | | - Frances M. Jones
- Department of Pathology, Cambridge University, Cambridge, United Kingdom
| | - Emily L. Webb
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Stephen Cose
- Uganda Virus Research Institute, Entebbe, Uganda
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - David W. Dunne
- Department of Pathology, Cambridge University, Cambridge, United Kingdom
| | - Alison M. Elliott
- Uganda Virus Research Institute, Entebbe, Uganda
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
29
|
Lundy SK, Lukacs NW. Chronic schistosome infection leads to modulation of granuloma formation and systemic immune suppression. Front Immunol 2013; 4:39. [PMID: 23429492 PMCID: PMC3576626 DOI: 10.3389/fimmu.2013.00039] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/31/2013] [Indexed: 11/13/2022] Open
Abstract
Schistosome worms have been infecting humans for millennia, but it is only in the last half century that we have begun to understand the complexities of this inter-relationship. As our sophistication about the inner workings of every aspect of the immune system has increased, it has also become obvious that schistosome infections have broad ranging effects on nearly all of the innate and adaptive immune response mechanisms. Selective pressures on both the worms and their hosts, has no doubt led to co-evolution of protective mechanisms, particularly those that favor granuloma formation around schistosome eggs and immune suppression during chronic infection. The immune modulatory effects that chronic schistosome infection and egg deposition elicit have been intensely studied, not only because of their major implications to public health issues, but also due to the emerging evidence that schistosome infection may protect humans from severe allergies and autoimmunity. Mouse models of schistosome infection have been extremely valuable for studying immune modulation and regulation, and in the discovery of novel aspects of immunity. A progression of immune reactions occurs during granuloma formation ranging from innate inflammation, to activation of each branch of adaptive immune response, and culminating in systemic immune suppression and granuloma fibrosis. Although molecular factors from schistosome eggs have been identified as mediators of immune modulation and suppressive functions of T and B cells, much work is still needed to define the mechanisms of the immune alteration and determine whether therapies for asthma or autoimmunity could be developed from these pathways.
Collapse
Affiliation(s)
- Steven K Lundy
- Graduate Training Program in Immunology, University of Michigan Medical School Ann Arbor, MI, USA ; Department of Internal Medicine-Rheumatology, University of Michigan Medical School Ann Arbor, MI, USA
| | | |
Collapse
|
30
|
Changes in T-Cell and Monocyte Phenotypes In Vitro by Schistosoma mansoni Antigens in Cutaneous Leishmaniasis Patients. J Parasitol Res 2012; 2012:520308. [PMID: 23209879 PMCID: PMC3504418 DOI: 10.1155/2012/520308] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2012] [Revised: 09/24/2012] [Accepted: 10/08/2012] [Indexed: 12/14/2022] Open
Abstract
High levels of proinflammatory cytokines such as IFN-γ and TNF are associated with tissue lesions in cutaneous leishmaniasis (CL). We previously demonstrated that Schistosoma mansoni antigens downmodulate the in vitro cytokine response in CL. In the current study we evaluated whether S. mansoni antigens alter monocyte and T-lymphocyte phenotypes in leishmaniasis. Peripheral blood mononuclear cells of CL patients were cultured with L. braziliensis antigen in the presence or absence of the S. mansoni antigens rSm29, rSmTSP-2- and PIII. Cells were stained with fluorochrome conjugated antibodies and analyzed by flow cytometry. The addition of rSm29 to the cultures decreased the expression of HLA-DR in nonclassical (CD14+CD16++) monocytes, while the addition of PIII diminished the expression of this molecule in classical (CD14++CD16−) and intermediate (CD14++CD16+) monocytes. The addition of PIII and rSmTSP-2 resulted in downmodulation of CD80 expression in nonclassical and CD86 expression in intermediate monocytes, respectively. These two antigens increased the expression of CTLA-4 in CD4+ T cells and they also expanded the frequency of CD4+CD25highFoxp3+ T cells. Taken together, we show that S. mansoni antigens, mainly rSmTSP-2 and PIII, are able to decrease the activation status of monocytes and also to upregulate the expression of modulatory molecules in T lymphocytes.
Collapse
|
31
|
Elliott DE, Weinstock JV. Helminth-host immunological interactions: prevention and control of immune-mediated diseases. Ann N Y Acad Sci 2012; 1247:83-96. [PMID: 22239614 DOI: 10.1111/j.1749-6632.2011.06292.x] [Citation(s) in RCA: 136] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Exposure to commensal and pathogenic organisms strongly influences our immune system. Exposure to helminths was frequent before humans constructed their current highly hygienic environment. Today, in highly industrialized countries, contact between humans and helminths is rare. Congruent with the decline in helminth infections is an increase in the prevalence of autoimmune and inflammatory disease. It is possible that exclusion of helminths from the environment has permitted the emergence of immune-mediated disease. We review the protective effects of helminths on expression of inflammatory bowel disease, multiple sclerosis, and animal models of these and other inflammatory diseases. We also review the immune pathways altered by helminths that may afford protection from these illnesses. Helminth exposure tends to inhibit IFN-γ and IL-17 production, promote IL-4, IL-10, and TGF-β release, induce CD4(+) T cell Foxp3 expression, and generate regulatory macrophages, dendritic cells, and B cells. Helminths enable protective pathways that may vary by specific species and disease model. Helminths or their products likely have therapeutic potential to control or prevent immune-mediated illness.
Collapse
Affiliation(s)
- David E Elliott
- Division of Gastroenterology, University of Iowa, Iowa City, Iowa, USA
| | | |
Collapse
|
32
|
Reda ES, Ouhtit A, Abdeen SH, El-Shabasy EA. Structural changes of Schistosoma mansoni adult worms recovered from C57BL/6 mice treated with radiation-attenuated vaccine and/or praziquantel against infection. Parasitol Res 2011; 110:979-92. [PMID: 21826487 DOI: 10.1007/s00436-011-2583-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2011] [Accepted: 07/27/2011] [Indexed: 12/18/2022]
Abstract
Although the current treatment of schistosomiasis relies largely on praziquantel (PZQ), it has not significantly reduced the overall number of disease cases, perhaps due to inevitable resistance to PZQ. Previous studies showed that radiation-attenuated vaccine gives protection levels for Schistosoma mansoni in host various species. In the present study, we evaluated the effect of various vaccination strategies in C57BL/6 mice, including single or multiple vaccination strategy, subcurative dose (20 mg/kg) of PZQ, and a combination of single vaccination with subcurative dose of PZQ. Groups of five mice were sacrificed postinfection in 42 days and schistosomes were collected by perfusion and examined by scanning electron microscopy. Treatment either with subcurative dose of PZQ or with a single vaccination of attenuated cercariae (500 per mouse), caused significant reduction in total worm burden, hepatic and intestinal ova counts 43.03%, 73.2%, 59.5% and 37.97%, 52.02%, 26.3%, respectively. Furthermore, tegumental changes were observed, including severe swelling, fusion of tegumental folds, vesicle formation, and loss or shortening of the spines on the tubercles. However, multiple vaccination strategy resulted in much higher reduction in total worm burden, hepatic and intestinal ova count. However, multiple vaccination strategy resulted in high reduction of worm burden, hepatic and intestinal ova counts 72.5%, 90.7%, 65.79%, respectively, and further causing swollen, disruption of tubercles teguments and erosion, extensive peeling, fusion of tegumental folds. Our findings suggest that multiple vaccination strategy is the most effective strategy to clear schistosomal infection, indicating its potential in guiding the design of appropriate therapeutic strategy against schistosomes.
Collapse
Affiliation(s)
- Enayat S Reda
- Department of Zoology, Faculty of Science, Mansoura University, Mansoura, Egypt.
| | | | | | | |
Collapse
|
33
|
Monteiro L, Souza-Machado A, Menezes C, Melo A. Association between allergies and multiple sclerosis: a systematic review and meta-analysis. Acta Neurol Scand 2011; 123:1-7. [PMID: 20456246 DOI: 10.1111/j.1600-0404.2010.01355.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Multiple sclerosis (MS) is an inflammatory disease of the central nervous system that expresses a typical type 1 immune response (Th1). Allergies, on the other hand, present with high levels of type 2 (Th2) cytokines. Some authors observed that Th1 and Th2 diseases could coexist in the same subject. Besides its biological plausibility, the association between MS and allergies remains controversial. The aim of this systematic review and meta-analysis was to determine if there is an association between allergic diseases and MS. All clinical and epidemiological studies on patients with MS published up to July 2009, that assessed the association between allergic diseases and MS were reviewed. A total of 1010 articles were retrieved from search, and ten epidemiological studies were included in the analysis. The results showed that there is no evidence supporting an association between allergic diseases (OR: 0.91; CI 95%: 0.68-1.23), asthma (OR: 0.83; CI 95%: 0.48-1.44), allergic rhinitis (OR: 0.81; CI 95%: 0.59-1.12), eczema (OR: 0.93; CI 95%: 0.71-1.23) and MS. Additional prospective studies in this field might help to elucidate the nature of these associations.
Collapse
Affiliation(s)
- L Monteiro
- Division of Neurology and Epidemiology, Federal University of Bahia, Brazil.
| | | | | | | |
Collapse
|
34
|
Bodammer P, Waitz G, Loebermann M, Holtfreter MC, Maletzki C, Krueger MR, Nizze H, Emmrich J, Reisinger EC. Schistosoma mansoni infection but not egg antigen promotes recovery from colitis in outbred NMRI mice. Dig Dis Sci 2011; 56:70-8. [PMID: 20428947 DOI: 10.1007/s10620-010-1237-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2009] [Accepted: 04/06/2010] [Indexed: 12/12/2022]
Abstract
BACKGROUND The ability of intestinal helminths to manipulate the immune system of their host towards a Th2 response has been proposed to modulate auto-immune and allergic diseases. AIMS This initial study investigated the anti-inflammatory potential of S. mansoni and soluble egg antigen (SEA) in a murine model of colitis. METHODS Colitis was induced in female NMRI mice by 5% dextran sulfate sodium (DSS) for 7 days, either 9 weeks post-infection with S. mansoni or during treatment with SEA. In addition to clinical signs of colitis, colon histology, immunohistochemistry, and flow cytometry of leukocytes were performed. Colon cytokines were measured using a quantitative real-time technique. RESULTS Infection with cercariae of S. mansoni attenuated DSS-induced colitis. Clinical symptoms such as weight loss and shortening of colon length were significantly prevented. Histological scores and cell infiltration were affected and expression of pro-inflammatory cytokines in the colons of infected DSS colitis mice was reduced. In contrast, application of SEA failed to improve colitis, even though some findings like earlier manifestation of inflammation and local induction of Th2 cytokines were similar to the effects of cercarial infection. CONCLUSIONS The results presented here suggest that SEA treatment could not protect mice from acute colitis. However, both infection with S. mansoni and injection of SEA affect mucosal immune responses.
Collapse
Affiliation(s)
- Peggy Bodammer
- Division of Tropical Medicine and Infectious Diseases, Department of Internal Medicine, University of Rostock, Rostock, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Reyes JL, Espinoza-Jiménez AF, González MI, Verdin L, Terrazas LI. Taenia crassiceps infection abrogates experimental autoimmune encephalomyelitis. Cell Immunol 2010; 267:77-87. [PMID: 21185554 DOI: 10.1016/j.cellimm.2010.11.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2010] [Revised: 11/18/2010] [Accepted: 11/29/2010] [Indexed: 12/19/2022]
Abstract
Helminth infections induce strong immunoregulation that can modulate subsequent pathogenic challenges. Taenia crassiceps causes a chronic infection that induces a Th2-biased response and modulates the host cellular immune response, including reduced lymphoproliferation in response to mitogens, impaired antigen presentation and the recruitment of suppressive alternatively activated macrophages (AAMФ). In this study, we aimed to evaluate the ability of T. crassiceps to reduce the severity of experimental autoimmune encephalomyelitis (EAE). Only 50% of T. crassiceps-infected mice displayed EAE symptoms, which were significantly less severe than uninfected mice. This effect was associated with both decreased MOG-specific splenocyte proliferation and IL-17 production and limited leukocyte infiltration into the spinal cord. Infection with T. crassiceps induced an anti-inflammatory cytokine microenvironment, including decreased TNF-α production and high MOG-specific production of IL-4 and IL-10. While the mRNA expression of TNF-α and iNOS was lower in the brain of T. crassiceps-infected mice with EAE, markers for AAMФ were highly expressed. Furthermore, in these mice, there was reduced entry of CD3(+)Foxp3(-) cells into the brain. The T. crassiceps-induced immune regulation decreased EAE severity by dampening T cell activation, proliferation and migration to the CNS.
Collapse
Affiliation(s)
- José L Reyes
- Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla, Edo, México 54090, Mexico
| | | | | | | | | |
Collapse
|
36
|
Wang X, Zhou S, Chi Y, Wen X, Hoellwarth J, He L, Liu F, Wu C, Dhesi S, Zhao J, Hu W, Su C. CD4+CD25+ Treg induction by an HSP60-derived peptide SJMHE1 from Schistosoma japonicum is TLR2 dependent. Eur J Immunol 2010; 39:3052-65. [PMID: 19882655 DOI: 10.1002/eji.200939335] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Chronic schistosome infection results in the suppression of host immune responses, allowing long-term schistosome survival and restricting pathology. Current theories suggest that Treg play an important role in this regulation. However, the mechanism of Treg induction during schistosome infection is still unknown. The aim of this study was to determine the mechanism behind the induction of CD4(+)CD25(+) T cells by Schistosoma japonicum HSP60 (SjHSP60)-derived peptide SJMHE1 as well as to elucidate the cellular and molecular basis for the induction of CD4(+)CD25(+) T cells during S. japonicum infection. Mice immunized with SJMHE1 or spleen and LN cells from naïve mice pretreated with SJMHE1 in vitro all displayed an increase in CD4(+)CD25(+) T-cell populations. Release of IL-10 and TGF-beta by SJMHE1 stimulation may contribute to suppression. Adoptively transferred SJMHE1-induced CD4(+)CD25(+) T cells inhibited delayed-type hypersensitivity in BALB/c mice. Additionally, SJMHE1-treated APC were tolerogenic and induced CD4(+) cells to differentiate into suppressive CD4(+)CD25(+) Treg. Furthermore, our data support a role for TLR2 in SJMHE1-mediated CD4(+)CD25(+) Treg induction. These findings provide the basis for a more complete understanding of the S. japonicum-host interactions that contribute to host homeostatic mechanisms, preventing an excessive immune response.
Collapse
Affiliation(s)
- Xuefeng Wang
- Department of Pathogen Biology & Immunology, Department of Pharmacology, Jiangsu Key Laboratory of Pathogen Biology, Nanjing Medical University, Nanjing, Jiangsu, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Regulation of type 1 diabetes, tuberculosis, and asthma by parasites. J Mol Med (Berl) 2009; 88:27-38. [PMID: 19844667 DOI: 10.1007/s00109-009-0546-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Revised: 07/27/2009] [Accepted: 09/18/2009] [Indexed: 10/20/2022]
Abstract
Helminth infection is a worldwide health problem. In addition to directly causing disease, helminthic infection also affects the incidence and progression of other diseases by exerting immune modulatory effects. In animal models, infection with helminthic parasites can prevent autoimmune diseases and allergic inflammatory diseases, but worsens protective immunity to certain infectious pathogens. In this review, we summarize current findings regarding the effects of helminth infection on type 1 diabetes, tuberculosis, and asthma and discuss possible mechanisms through which helminthic parasites modulate host immunity. Investigating these mechanisms could lead to treatment strategies that specifically modulate the immune response as well as address fundamental questions in immunobiology.
Collapse
|
38
|
Osada Y, Shimizu S, Kumagai T, Yamada S, Kanazawa T. Schistosoma mansoni infection reduces severity of collagen-induced arthritis via down-regulation of pro-inflammatory mediators. Int J Parasitol 2008; 39:457-64. [PMID: 18835272 DOI: 10.1016/j.ijpara.2008.08.007] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2008] [Revised: 08/01/2008] [Accepted: 08/19/2008] [Indexed: 01/06/2023]
Abstract
Various experimental and epidemiological studies have demonstrated that helminth infections affect outcomes of allergic or autoimmune disorders. Here, we examined the effects of Schistosoma mansoni infection on mouse collagen-induced arthritis, one of the most widely used animal models for rheumatoid arthritis. Male DBA/1 mice were infected with S. mansoni 2 weeks prior to being immunized with type II collagen (IIC). Cytokine mRNA expression in mouse paws, cytokine production by ConA-stimulated spleen cells, and anti-IIC antibodies were evaluated in addition to the severity of arthritis. S. mansoni infection significantly reduced the severity of arthritis. Anti-IIC IgG and IgG2a levels were lower in infected than uninfected mice. With regard to cytokine producing potentials in the infected mice, the down-regulation of Th1 (IFNgamma) and pro-inflammatory cytokines (TNFalpha and IL-17A), and up-regulation of Th2 (IL-4) and an anti-inflammatory cytokine (IL-10) were observed.In addition, real-time PCR revealed that the augmentation of pro-inflammatory mediators such as IL-1 beta, IL-6 and receptor activator of NFkappaB ligand in inflamed paws was abrogated by S. mansoni infection [corrected]. In conclusion, schistosome infection reduced the severity of autoimmune arthritis via systemic and local suppression of pro-inflammatory mediators, suggesting the potential of parasite-derived materials as therapeutic agents against rheumatoid arthritis.
Collapse
Affiliation(s)
- Yoshio Osada
- Department of Immunology and Parasitology, University of Occupational and Environmental Health, Yahatanishi-ku, Kitakyushu, Japan.
| | | | | | | | | |
Collapse
|
39
|
Mo HM, Lei JH, Jiang ZW, Wang CZ, Cheng YL, Li YL, Liu WQ. Schistosoma japonicum infection modulates the development of allergen-induced airway inflammation in mice. Parasitol Res 2008; 103:1183-9. [PMID: 18654798 DOI: 10.1007/s00436-008-1114-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Accepted: 06/30/2008] [Indexed: 01/08/2023]
Abstract
Asthma, a chronic inflammatory disorder of the airways, is coordinated by Th2 cells in both human asthmatics and animal models of allergic asthma. It has been shown that helminth infections including Schistosoma mansoni may modulate atopic diseases including asthma. In the present study, BALB/c mice were infected with bisexual and unisexual (male) S. japonicum, respectively, prior to ovalbumin (OVA) sensitization and challenge. Compared to mice with OVA sensitization/challenge alone, S. japonicum infection led to a significant decrease of eosinophil accumulation in bronchoalveolar lavage fluid (BALF) collected 48 h postchallenge, as well as to a marked reduction in inflammatory cell infiltration around the airways and pulmonary blood vessels. Compared to OVA-immunized uninfected mice, the level of OVA-specific serum IgE as well as interleukin (IL)-4 and IL-5 in BALF were reduced, but IL-10 was strongly elevated in mice with preexisting S. japonicum infection prior to OVA immunization. These results suggest that both bisexual and male S. japonicum infections may modulate the development of allergic asthma.
Collapse
Affiliation(s)
- Hong-mei Mo
- Department of Parasitology, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan, 430030, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
40
|
Protective effect and granuloma down-modulation promoted by RP44 antigen a fructose 1,6 bisphosphate aldolase of Schistosoma mansoni. Immunobiology 2008; 213:437-46. [DOI: 10.1016/j.imbio.2007.10.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 09/11/2007] [Accepted: 10/15/2007] [Indexed: 11/20/2022]
|
41
|
Decreased basal non-insulin-stimulated glucose uptake by diaphragm in streptozotocin-induced diabetic mice infected with Schistosoma mansoni. Parasitol Res 2008; 103:595-601. [PMID: 18509678 DOI: 10.1007/s00436-008-1016-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2008] [Accepted: 04/22/2008] [Indexed: 10/22/2022]
Abstract
It had been suggested that chronic exposure to Schistosoma mansoni prevents the onset of Th1-mediated diseases such as diabetes mellitus. The present study was carried out on four groups of mice: (1) control group, (2) group infected with S. mansoni, (3) group injected with streptozotocin to induce diabetes, and (4) group infected and then 3 months postinfection injected with streptozotocin. No differences were detected between the infected non-diabetic and infected diabetic groups regarding worm burden, tissue egg count, and oogram. At the same time, results showed a reducing effect of S. mansoni infection on the rate of glucose uptake by the diaphragm with reduction in glycogen content of soleus muscle. This an important issue since skeletal muscle is the primary site for insulin-stimulated glucose disposal. In conclusion, because of the detected depressed peripheral glucose uptake by the diaphragm, the protecting effect of helminths infection in diabetes should be reconsidered, to be able to devise therapeutic strategies for the treatment of autoimmune diseases.
Collapse
|
42
|
Mutapi F, Winborn G, Midzi N, Taylor M, Mduluza T, Maizels RM. Cytokine responses to Schistosoma haematobium in a Zimbabwean population: contrasting profiles for IFN-gamma, IL-4, IL-5 and IL-10 with age. BMC Infect Dis 2007; 7:139. [PMID: 18045464 PMCID: PMC2222613 DOI: 10.1186/1471-2334-7-139] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Accepted: 11/28/2007] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND The rate of development of parasite-specific immune responses can be studied by following their age profiles in exposed and infected hosts. This study determined the cytokine-age profiles of Zimbabweans resident in a Schistosoma haematobium endemic area and further investigated the relationship between the cytokine responses and infection intensity. METHODS Schistosome adult worm antigen-specific IFN-gamma, IL-4, IL-5 and IL-10 cytokine responses elicited from whole blood cultures were studied in 190 Zimbabweans exposed to S. haematobium infection (aged 6 to 40 years old). The cytokines were measured using capture ELISAs and the data thus obtained together with S. haematobium egg count data from urine assays were analysed using a combination of parametric and nonparametric statistical approaches. RESULTS Age profiles of schistosome infection in the study population showed that infection rose to peak in childhood (11-12 years) followed by a sharp decline in infection intensity while prevalence fell more gradually. Mean infection intensity was 37 eggs/10 ml urine (SE 6.19 eggs/10 ml urine) while infection prevalence was 54.7%. Measurements of parasite-specific cytokine responses showed that IL-4, IL-5 and IL-10 but not IFN-gamma followed distinct age-profiles. Parasite-specific IL-10 production developed early, peaking in the youngest age group and declining thereafter; while IL-4 and IL-5 responses were slower to develop with a later peak. High IL-10 producers were likely to be egg positive with IL-10 production increasing with increasing infection intensity. Furthermore people producing high levels of IL-10 produced little or no IL-5, suggesting that IL-10 may be involved in the regulation of IL-5 levels. IL-4 and IFN-gamma did not show a significant relationship with infection status or intensity and were positively associated with each other. CONCLUSION Taken together, these results show that the IL-10 responses develop early compared to the IL-5 response and may be down-modulating immunopathological responses that occur during the early phase of infection. The results further support current suggestions that the Th1/Th2 dichotomy does not sufficiently explain susceptibility or resistance to schistosome infection.
Collapse
Affiliation(s)
- Francisca Mutapi
- Institute of Immunology & Infection Research, School of Biological Sciences, University of Edinburgh, Ashworth Laboratories, King's Buildings, West Mains Rd, Edinburgh, EH9 3JT, UK.
| | | | | | | | | | | |
Collapse
|
43
|
Abouel-Nour MF, Lotfy M, Attallah AM, Doughty BL. Schistosoma mansoni major egg antigen Smp40: molecular modeling and potential immunoreactivity for anti-pathology vaccine development. Mem Inst Oswaldo Cruz 2007; 101:365-72. [PMID: 16951805 DOI: 10.1590/s0074-02762006000400004] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2005] [Accepted: 05/10/2006] [Indexed: 11/22/2022] Open
Abstract
The pathogenesis of Schistosoma mansoni infection is largely determined by host T-cell mediated immune responses such as the granulomatous response to tissue deposited eggs and subsequent fibrosis. The major egg antigens have a valuable role in desensitizing the CD4+ Th cells that mediate granuloma formation, which may prevent or ameliorate clinical signs of schistosomiasis.S. mansoni major egg antigen Smp40 was expressed and completely purified. It was found that the expressed Smp40 reacts specifically with anti-Smp40 monoclonal antibody in Western blotting. Three-dimensional structure was elucidated based on the similarity of Smp40 with the small heat shock protein coded in the protein database as 1SHS as a template in the molecular modeling. It was figured out that the C-terminal of the Smp40 protein (residues 130 onward) contains two alpha crystallin domains. The fold consists of eight beta strands sandwiched in two sheets forming Greek key. The purified Smp40 was used for in vitro stimulation of peripheral blood mononuclear cells from patients infected with S. mansoni using phytohemagglutinin mitogen as a positive control. The obtained results showed that there is no statistical difference in interferon-g, interleukin (IL)-4 and IL-13 levels obtained with Smp40 stimulation compared with the control group (P > 0.05 for each). On the other hand, there were significant differences after Smp40 stimulation in IL-5 (P = 0.006) and IL-10 levels (P < 0.001) compared with the control group. Gaining the knowledge by reviewing the literature, it was found that the overall pattern of cytokine profile obtained with Smp40 stimulation is reported to be associated with reduced collagen deposition, decreased fibrosis, and granuloma formation inhibition. This may reflect its future prospect as a leading anti-pathology schistosomal vaccine candidate.
Collapse
|
44
|
Elliott DE, Summers RW, Weinstock JV. Helminths as governors of immune-mediated inflammation. Int J Parasitol 2006; 37:457-64. [PMID: 17313951 DOI: 10.1016/j.ijpara.2006.12.009] [Citation(s) in RCA: 128] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Revised: 11/28/2006] [Accepted: 12/06/2006] [Indexed: 12/11/2022]
Abstract
Immune-mediated diseases (e.g. inflammatory bowel disease, asthma, multiple sclerosis and autoimmune diabetes) are increasing in prevalence and emerge as populations adopt meticulously hygienic lifestyles. This change in lifestyles precludes exposure to helminths (parasitic worms). Loss of natural helminth exposure removes a previously universal Th2 and regulatory immune biasing imparted by these organisms. Helminths protect animals from developing immune-mediated diseases (colitis, reactive airway disease, encephalitis and diabetes). Clinical trials show that exposure to helminths can reduce disease activity in patients with ulcerative colitis or Crohn's disease. This paper summarises work by multiple groups demonstrating that colonization with helminths alters immune reactivity and protects against disease from dysregulated inflammation.
Collapse
Affiliation(s)
- David E Elliott
- Department of Internal Medicine, University of Iowa, Roy J. and Lucille A. Carver College of Medicine, 200 Hawkins Drive, Iowa City, IA 52242-1009, USA.
| | | | | |
Collapse
|
45
|
McConchie BW, Norris HH, Bundoc VG, Trivedi S, Boesen A, Urban JF, Keane-Myers AM. Ascaris suum-derived products suppress mucosal allergic inflammation in an interleukin-10-independent manner via interference with dendritic cell function. Infect Immun 2006; 74:6632-41. [PMID: 16966410 PMCID: PMC1698059 DOI: 10.1128/iai.00720-06] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We have previously demonstrated that protection from allergic inflammation by Ascaris suum infection was characterized by a global increase in interleukin-10 (IL-10) and the development of protective CD4(+)/CD25(+) T cells (L. Schopf, S. Luccioli, V. Bundoc, P. Justice, C. C. Chan, B. J. Wetzel, H. H. Norris, J. F. Urban, Jr., and A. Keane-Myers, Investig. Ophthalmol. Vis. Sci. 46:2772-2780, 2005). Here, we used A. suum pseudocoelomic fluid (PCF) in lieu of infection to define molecular mechanisms of allergic protection in a mouse model of allergic inflammation. Mice were sensitized with ragweed (RW) and PCF (RW/PCF), PCF alone, or RW alone and then challenged intratracheally, intranasally, and supraocularly with RW. Histological examination of the eyes and lungs, analysis of the bronchoalveolar lavage fluid (BALF), and characterization of ex vivo cytokine responses were performed to determine allergic inflammatory responses. RW/PCF-treated mice had suppressed allergic immune responses compared to mice given RW alone. To investigate whether IL-10 was involved in PCF-mediated allergic protection, similar experiments were performed using mice genetically deficient for IL-10. Persistent protection from allergic disease was observed in the absence of IL-10, indicating the primary mechanism of PCF protection is IL-10 independent. Ex vivo and in vitro analysis of PCF-treated dendritic cells (DC) demonstrated reduced activation receptor expression and cytokine production in response to either RW or lipopolysaccharide stimulation. These findings extend previous studies that showed infection with A. suum alters expression of allergic disease and suggest that PCF can contribute to this effect by interference with DC function.
Collapse
Affiliation(s)
- Brittany W McConchie
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, Twinbrook II Room 125, 12441 Parklawn Drive, Rockville, MD 20852, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Negrão-Corrêa D, Pinho V, Souza DG, Pereira ATM, Fernandes A, Scheuermann K, Souza ALS, Teixeira MM. Expression of IL-4 receptor on non-bone marrow-derived cells is necessary for the timely elimination of Strongyloides venezuelensis in mice, but not for intestinal IL-4 production. Int J Parasitol 2006; 36:1185-95. [PMID: 16793046 DOI: 10.1016/j.ijpara.2006.05.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2006] [Revised: 04/12/2006] [Accepted: 05/01/2006] [Indexed: 10/24/2022]
Abstract
In rodents and in humans, Strongyloides infection induces an immune response which is predominantly Th2 in nature. In an attempt to understand the role of the IL-4R/STAT6 signaling pathway, the pathway activated by the Th2 cytokines IL-4 and IL-13, in the induction of protection during Strongyloides venezuelensis infection, we have carried out experiments in mice lacking the IL-4Ralpha chain. Experiments were also carried out in STAT6 (STAT6(-/-)) and IL-12-deficient (IL-12(-/-)) mice for comparison. There was enhancement of IL-13 and abolition of IFN-gamma production in the small intestine of 7 day-infected IL-12(-/-) animals but worm elimination proceeded with very similar kinetics to those of wild-type mice. In IL-4Ralpha- or STAT6-deficient mice, there was a delay in parasite elimination and a large number of S. venezuelensis adult worms was still present in the small intestine 14 days after infection. Moreover, IgE production was completely abolished in IL-4Ralpha- or STAT6-deficient mice but tissue eosinophilia was normally induced by the parasite infection in deficient mice. Bone marrow transfer experiments showed that worm elimination occurred when a functional IL-4 receptor was present only in non-bone marrow-derived cells but not when IL-4R was only expressed in bone marrow cells. The induction of IL-4, but not IL-13, occurred independently of IL-4R. We believe these results are the first direct evidence that the mechanism responsible for the timely elimination of S. venezuelensis is dependent on the activation of IL-4R and STAT6. Moreover, a functional protective response is dependent on the expression of IL-4Ralpha on non-bone marrow-derived cells.
Collapse
Affiliation(s)
- Deborah Negrão-Corrêa
- Departmento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Campus Pampulha, Belo Horizonte, MG, Brazil.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
McKay DM. The beneficial helminth parasite? Parasitology 2005; 132:1-12. [PMID: 16393348 DOI: 10.1017/s003118200500884x] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2005] [Revised: 06/06/2005] [Accepted: 07/19/2005] [Indexed: 12/25/2022]
Abstract
There is unequivocal evidence that parasites influence the immune activity of their hosts, and many of the classical examples of this are drawn from assessment of helminth infections of their mammalian hosts. Thus, helminth infections can impact on the induction or course of other diseases that the host might be subjected to. Epidemiological studies demonstrate that world regions with high rates of helminth infections consistently have reduced incidences of autoimmune and other allergic/inflammatory-type conditions. Here I review and assess the possible ways by which helminth infections can block or modulate concomitant disease processes. There is much to be learned from careful analysis of immuno-regulation in helminth-infected rodents and from an understanding of the immune status of acutely and chronically infected humans. The ultimate reward from this type of investigation will likely be a more comprehensive knowledge of immunity, novel ways to intervene in the immune response to alleviate autoimmune and allergic diseases (growing concerns in economically developed areas), and perhaps the development of helminth therapy for patients suffering from specific inflammatory, autoimmune or allergic disorders.
Collapse
Affiliation(s)
- D M McKay
- Intestinal Disease Research Programme, McMaster University, Hamilton, Ontario, Canada.
| |
Collapse
|
48
|
Morley J. Inflammation and asthma therapy: a false dawn. Pulm Pharmacol Ther 2005; 19:200-4. [PMID: 16011901 DOI: 10.1016/j.pupt.2005.05.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2005] [Accepted: 05/12/2005] [Indexed: 11/22/2022]
Affiliation(s)
- J Morley
- Kings College Sackler Institute of pulmonary Pharmacology, Guys Campus Fifth floor Hodgkin, London, UK.
| |
Collapse
|