1
|
Midha A, Oser L, Schlosser-Brandenburg J, Laubschat A, Mugo RM, Musimbi ZD, Höfler P, Kundik A, Hayani R, Adjah J, Groenhagen S, Tieke M, Elizalde-Velázquez LE, Kühl AA, Klopfleisch R, Tedin K, Rausch S, Hartmann S. Concurrent Ascaris infection modulates host immunity resulting in impaired control of Salmonella infection in pigs. mSphere 2024; 9:e0047824. [PMID: 39140728 PMCID: PMC11423588 DOI: 10.1128/msphere.00478-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 07/09/2024] [Indexed: 08/15/2024] Open
Abstract
Ascaris is one of the most widespread helminth infections, leading to chronic morbidity in humans and considerable economic losses in pig farming. In addition, pigs are an important reservoir for the zoonotic salmonellosis, where pigs can serve as asymptomatic carriers. Here, we investigated the impact of an ongoing Ascaris infection on the immune response to Salmonella in pigs. We observed higher bacterial burdens in experimentally coinfected pigs compared to pigs infected with Salmonella alone. The impaired control of Salmonella in the coinfected pigs was associated with repressed interferon gamma responses in the small intestine and with the alternative activation of gut macrophages evident in elevated CD206 expression. Ascaris single and coinfection were associated with a rise of CD4-CD8α+FoxP3+ Treg in the lymph nodes draining the small intestine and liver. In addition, macrophages from coinfected pigs showed enhanced susceptibility to Salmonella infection in vitro and the Salmonella-induced monocytosis and tumor necrosis factor alpha production by myeloid cells was repressed in pigs coinfected with Ascaris. Hence, our data indicate that acute Ascaris infection modulates different immune effector functions with important consequences for the control of tissue-invasive coinfecting pathogens.IMPORTANCEIn experimentally infected pigs, we show that an ongoing infection with the parasitic worm Ascaris suum modulates host immunity, and coinfected pigs have higher Salmonella burdens compared to pigs infected with Salmonella alone. Both infections are widespread in pig production and the prevalence of Salmonella is high in endemic regions of human Ascariasis, indicating that this is a clinically meaningful coinfection. We observed the type 2/regulatory immune response to be induced during an Ascaris infection correlates with increased susceptibility of pigs to the concurrent bacterial infection.
Collapse
Affiliation(s)
- Ankur Midha
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Larissa Oser
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Josephine Schlosser-Brandenburg
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Alexandra Laubschat
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Robert M Mugo
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Zaneta D Musimbi
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Philipp Höfler
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Arkadi Kundik
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Rima Hayani
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Joshua Adjah
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Saskia Groenhagen
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Malte Tieke
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Luis E Elizalde-Velázquez
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Anja A Kühl
- Charité Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, iPATH.Berlin, Core unit of Charité, Campus Benjamin Franklin, Berlin, Germany
| | - Robert Klopfleisch
- Department of Veterinary Medicine, Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Karsten Tedin
- Department of Veterinary Medicine, Institute of Microbiology and Epizootics, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Sebastian Rausch
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| | - Susanne Hartmann
- Department of Veterinary Medicine, Institute of Immunology, Centre for Infection Medicine, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
2
|
Sweed D, Mohamady M, Gouda MA, Fayed Y, Saied SA, Elhamed SMA. Does the expression of granzyme B participate in inflammation, fibrosis, and fertility of hydatid cysts? Parasitol Res 2023; 123:22. [PMID: 38072840 PMCID: PMC10710967 DOI: 10.1007/s00436-023-08056-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 10/28/2023] [Indexed: 12/18/2023]
Abstract
Echinococcus granulosus (sensu lato), a cestode that is endemic in Egypt, causes cystic echinococcosis (CE), a significant but neglected zoonotic disease that is prevalent throughout the world. Infected hydatid cysts are classified as fertile or non-fertile based on the presence of protoscoleces; nevertheless, the mechanism of non-fertile CE cysts remains unknown. The study aimed to assess whether granzyme B (GrB) expression and CD4 + /CD8 + could be related to the induction of non-fertile CE cysts. A total of fifty-eight individuals diagnosed with visceral hydatid cysts were selected, and they were further divided according to cyst fertility into fertile and non-fertile. Immunohistochemistry for CD4, CD8, and GrB was done. According to the results, hydatid cysts are common in adults and have no gender preference. The same clinical and laboratory data were shared by patients with fertile and non-fertile cysts (p = 0.186). GrB expression was not impacted by the fibrous deposition inside the hydatid cyst wall (p = 0.85); however, GrB was significantly correlated with the inflammatory density (p = 0.005). GrB expression was also found to be significantly higher in non-fertile cysts (p = 0.04). GrB expression is positively correlated with CD4 and CD8 expression. In conclusion, the expression of GrB in hydatid cysts may exacerbate the inflammatory response and impede cyst fertility while not affecting the fibrous deposition in the cyst wall.
Collapse
Affiliation(s)
- Dina Sweed
- Pathology Department, National Liver Institute, Menoufia University, Shebin Elkom, Menoufia, Egypt.
| | - Mohamed Mohamady
- Pathology Department, National Liver Institute, Menoufia University, Shebin Elkom, Menoufia, Egypt
| | - Marwa A Gouda
- Clinical and Molecular Parasitology Department, National Liver Institute, Shebin Elkom, Menoufia, Egypt
| | - Yahya Fayed
- Hepatopancreatobiliary Surgery Department, National Liver Institute, Menoufia University, Shebin Elkom, Menoufia, Egypt
| | - Sara A Saied
- Clinical Pathology Department, National Liver Institute, Menoufia University, Shebin Elkom, Menoufia, Egypt
| | - Sara M Abd Elhamed
- Pathology Department, National Liver Institute, Menoufia University, Shebin Elkom, Menoufia, Egypt
| |
Collapse
|
3
|
Saad AE, Ashour DS, Rashad E. Immunomodulatory effects of chronic trichinellosis on Toxoplasma gondii RH virulent strain in experimental rats. Pathog Glob Health 2023; 117:417-434. [PMID: 36922743 PMCID: PMC10177679 DOI: 10.1080/20477724.2023.2191233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023] Open
Abstract
Mixed parasitic infections could affect the host immunological responses and re-design the pathogenesis of each other. The impact of Toxoplasma gondii (T. gondii) and Trichinella spiralis (T. spiralis) co-infection on the immune response remains unclear. The objective of the present study was to investigate the possible effect of chronic trichinellosis on the immune response of rats infected with T. gondii virulent RH strain. Animals were divided into four groups: group I: non-infected negative control; group II: infected with T. spiralis; group III: infected with T. gondii and group IV: infected with T. spiralis then infected with T. gondii 35 days post T. spiralis infection (co-infected group). The interaction between T. spiralis and T. gondii was evaluated by histopathological examination of liver and brain tissues, immunohistochemical expression of inducible nitric oxide synthase (iNOS), and β-catenin in the brain tissues, and CD4+ and CD8+ T cells percentages, and tumor necrosis factor (TNF)-alpha expression in the spleen tissues. Along with, splenic interleukin (IL)-4 and IL-10 mRNA expression levels were measured 15 days post-Toxoplasma infection. Our study revealed that prior infection with T. spiralis leads to attenuation of Th1 response against T. gondii, including iNOS, TNF-α, and CD8+ T-cell response with improvement of the histopathological changes in the tissues. In conclusion, in the co-infected rats, a balanced immune response has been developed with the end result, improvement of the histopathological changes in the liver and brain.
Collapse
Affiliation(s)
- Abeer E. Saad
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
- Medical Parasitology sub-unit, Pathology Department, College of Medicine, Jouf University, Sakaka, Saudi Arabia
| | - Dalia S. Ashour
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Eman Rashad
- Cytology and Histology Department, Faculty of Veterinary Medicine, Cairo University, Giza, Egypt
| |
Collapse
|
4
|
Atagozli T, Elliott DE, Ince MN. Helminth Lessons in Inflammatory Bowel Diseases (IBD). Biomedicines 2023; 11:1200. [PMID: 37189818 PMCID: PMC10135676 DOI: 10.3390/biomedicines11041200] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/10/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Helminths are multicellular invertebrates that colonize the gut of many vertebrate animals including humans. This colonization can result in pathology, which requires treatment. It can also lead to a commensal and possibly even a symbiotic relationship where the helminth and the host benefit from each other's presence. Epidemiological data have linked helminth exposure to protection from immune disorders that include a wide range of diseases, such as allergies, autoimmune illnesses, and idiopathic inflammatory disorders of the gut, which are grouped as inflammatory bowel diseases (IBD). Treatment of moderate to severe IBD involves the use of immune modulators and biologics, which can cause life-threatening complications. In this setting, their safety profile makes helminths or helminth products attractive as novel therapeutic approaches to treat IBD or other immune disorders. Helminths stimulate T helper-2 (Th2) and immune regulatory pathways, which are targeted in IBD treatment. Epidemiological explorations, basic science studies, and clinical research on helminths can lead to the development of safe, potent, and novel therapeutic approaches to prevent or treat IBD in addition to other immune disorders.
Collapse
Affiliation(s)
- Tyler Atagozli
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
| | - David E. Elliott
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
- Iowa City Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| | - Mirac Nedim Ince
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Iowa Carver College of Medicine, Iowa City, IA 52246, USA
- Iowa City Veterans Affairs Medical Center, Iowa City, IA 52246, USA
| |
Collapse
|
5
|
Tian Q, Zhang T, Wang L, Ma J, Sun X. Gut dysbiosis contributes to chlamydial induction of hydrosalpinx in the upper genital tract. Front Microbiol 2023; 14:1142283. [PMID: 37125189 PMCID: PMC10133527 DOI: 10.3389/fmicb.2023.1142283] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 03/23/2023] [Indexed: 05/02/2023] Open
Abstract
Chlamydia trachomatis is one of the most common sexually infections that cause infertility, and its genital infection induces tubal adhesion and hydrosalpinx. Intravaginal Chlamydia muridarum infection in mice can induce hydrosalpinx in the upper genital tract and it has been used for studying C. trachomatis pathogenicity. DBA2/J strain mice were known to be resistant to the chlamydial induction of hydrosalpinx. In this study, we took advantage of this feature of DBA2/J mice to evaluate the role of antibiotic induced dysbiosis in chlamydial pathogenicity. Antibiotics (vancomycin and gentamicin) were orally administrated to induce dysbiosis in the gut of DBA2/J mice. The mice with or without antibiotic treatment were evaluated for gut and genital dysbiosis and then intravaginally challenged by C. muridarum. Chlamydial burden was tested and genital pathologies were evaluated. We found that oral antibiotics significantly enhanced chlamydial induction of genital hydrosalpinx. And the antibiotic treatment induced severe dysbiosis in the GI tract, including significantly reduced fecal DNA and increased ratios of firmicutes over bacteroidetes. The oral antibiotic did not alter chlamydial infection or microbiota in the mouse genital tracts. Our study showed that the oral antibiotics-enhanced hydrosalpinx correlated with dysbiosis in gut, providing the evidence for associating gut microbiome with chlamydial genital pathogenicity.
Collapse
Affiliation(s)
- Qi Tian
- Department of Obstetrics and Gynecology, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
- National Health Commission Key Laboratory for Birth Defect Research and Prevention, Hunan Provincial Maternal and Child Health Care Hospital, Changsha, Hunan, China
- *Correspondence: Qi Tian,
| | - Tianyuan Zhang
- Key Lab of Molecular Virology and Immunology, Institute Pasteur of Shanghai, Chinese Academy of Sciences, Shanghai, China
- Tianyuan Zhang,
| | - Luying Wang
- Department of Obstetrics and Gynecology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jingyue Ma
- Department of Dermatovenereology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xin Sun
- Department of Obstetrics and Gynecology, Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
6
|
Hou X, Shi Y, Kang X, Rousu Z, Li D, Wang M, Ainiwaer A, Zheng X, Wang M, Jiensihan B, Li L, Li J, Wang H, Zhang C. Echinococcus granulosus: The establishment of the metacestode in the liver is associated with control of the CD4+ T-cell-mediated immune response in patients with cystic echinococcosis and a mouse model. Front Cell Infect Microbiol 2022; 12:983119. [PMID: 36046744 PMCID: PMC9422084 DOI: 10.3389/fcimb.2022.983119] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Accepted: 07/25/2022] [Indexed: 11/16/2022] Open
Abstract
The larval stage of the tapeworm Echinococcus granulosus sensu lato (E. granulosus s.l.) caused a chronic infection, known as cystic echinococcosis (CE), which is a worldwide public health problem. The human secondary CE is caused by the dissemination of protoscoleces (PSCs) when fertile cysts are accidentally ruptured, followed by development of PSCs into new metacestodes. The local immune mechanisms responsible for the establishment and established phases after infection with E. granulosus s.l. are not clear. Here, we showed that T cells were involved in the formation of the immune environment in the liver in CE patients and Echinococcus granulosus sensu strict (E. granulosus s.s.)-infected mice, with CD4+ T cells being the dominant immune cells; this process was closely associated with cyst viability and establishment. Local T2-type responses in the liver were permissive for early infection establishment by E. granulosus s.s. between 4 and 6 weeks in the experimental model. CD4+ T-cell deficiency promoted PSC development into cysts in the liver in E. granulosus s.s.-infected mice. In addition, CD4+ T-cell-mediated cellular immune responses and IL-10-producing CD8+ T cells play a critical role in the establishment phase of secondary E. granulosus s.s. PSC infection. These data contribute to the understanding of local immune responses to CE and the design of new therapies by restoring effective immune responses and blocking evasion mechanisms during the establishment phase of infection.
Collapse
Affiliation(s)
- Xinling Hou
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Yang Shi
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Xuejiao Kang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Zibigu· Rousu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Dewei Li
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Maolin Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, and World Health Organization Collaborating Centre on Prevention and Case Management of Echinococcosis, Urumqi, China
- Department of Hepatic Hydatid and Hepatobiliary Surgery, Digestive and Vascular Surgery Centre, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
| | - Abidan· Ainiwaer
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Xuran Zheng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - MingKun Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Bahejiang· Jiensihan
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, and World Health Organization Collaborating Centre on Prevention and Case Management of Echinococcosis, Urumqi, China
| | - Liang Li
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, and World Health Organization Collaborating Centre on Prevention and Case Management of Echinococcosis, Urumqi, China
| | - Jing Li
- Basic Medical College, Xinjiang Medical University, Urumqi, China
| | - Hui Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Basic Medical College, Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, and World Health Organization Collaborating Centre on Prevention and Case Management of Echinococcosis, Urumqi, China
- *Correspondence: Chuanshan Zhang, ; Hui Wang,
| | - Chuanshan Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi, China
- Basic Medical College, Xinjiang Medical University, Urumqi, China
- Xinjiang Key Laboratory of Echinococcosis, Clinical Medicine Institute, The First Affiliated Hospital of Xinjiang Medical University, and World Health Organization Collaborating Centre on Prevention and Case Management of Echinococcosis, Urumqi, China
- *Correspondence: Chuanshan Zhang, ; Hui Wang,
| |
Collapse
|
7
|
Characterization of pathogenic CD8 + T cells in Chlamydia-infected OT1 mice. Infect Immun 2021; 90:e0045321. [PMID: 34724387 DOI: 10.1128/iai.00453-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chlamydia trachomatis is a leading infectious cause of infertility in women due to its induction of lasting pathology such as hydrosalpinx. Chlamydia muridarum induces mouse hydrosalpinx because C. muridarum can both invade tubal epithelia directly (as a 1st hit) and induce lymphocytes to promote hydrosalpinx indirectly (as a 2nd hit). In the current study, a critical role of CD8+ T cells in chlamydial induction of hydrosalpinx was validated in both wild type C57BL/6J and OT1 transgenic mice. OT1 mice failed to develop hydrosalpinx partially due to the failure of their lymphocytes to recognize chlamydial antigens. CD8+ T cells from naïve C57BL/6J rescued the recipient OT1 mice to develop hydrosalpinx when naïve CD8+ T cells were transferred at the time of infection with Chlamydia. However, when the transfer was delayed for 2 weeks or longer after the chlamydial infection, naïve CD8+ T cells no longer promoted hydrosalpinx. Nevertheless, Chlamydia-immunized CD8+ T cells still promoted significant hydrosalpinx in the recipient OT1 mice even when the transfer was delayed for 3 weeks. Thus, CD8+ T cells must be primed within 2 weeks after chlamydial infection to be pathogenic but once primed, they can promote hydrosalpinx for >3 weeks. However, Chlamydia-primed CD4+ T cells failed to promote chlamydial induction of pathology in OT1 mice. This study has optimized an OT1 mouse-based model for revealing the pathogenic mechanisms of Chlamydia-specific CD8+ T cells.
Collapse
|
8
|
Gastrointestinal Chlamydia-induced CD8 + T cells promote chlamydial pathogenicity in the female upper genital tract. Infect Immun 2021; 89:e0020521. [PMID: 34227838 DOI: 10.1128/iai.00205-21] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Chlamydia is known to both ascend to the upper genital tract and spread to the gastrointestinal tract following intravaginal inoculation. The gastrointestinal Chlamydia was recently reported to promote chlamydial pathogenicity in the genital tract since mice intravaginally inoculated with an attenuated Chlamydia, which alone failed to develop pathology in the genital tract, were restored to develop hydrosalpinx by intragastric co-inoculation with wild type Chlamydia. Gastrointestinal Chlamydia promoted hydrosalpinx via an indirect mechanism since Chlamydia in the gut did not directly spread to the genital tract lumen. In the current study, we further investigated the role of CD8+ T cells in the promotion of hydrosalpinx by gastrointestinal Chlamydia. First, we confirmed that intragastric co-inoculation with wild type Chlamydia promoted hydrosalpinx in mice that were inoculated with an attenuated Chlamydia in the genital tract one week earlier. Second, the promotion of hydrosalpinx by intragastrically co-inoculated Chlamydia was blocked by depleting CD8+ T cells. Third, adoptive transfer of the gastrointestinal Chlamydia-induced CD8+ T cells was sufficient for promoting hydrosalpinx in mice that were intravaginally inoculated with an attenuated Chlamydia. These observations have demonstrated that CD8+ T cells induced by gastrointestinal Chlamydia are both necessary and sufficient for promoting hydrosalpinx in the genital tract. The study has laid a foundation for further revealing the mechanisms by which Chlamydia-induced T lymphocyte responses (as a 2nd hit) promote hydrosalpinx in mice with genital Chlamydia-triggered tubal injury (as a 1st hit), a continuing effort in testing the two-hit hypothesis as a chlamydial pathogenic mechanism.
Collapse
|
9
|
Filbey KJ, Mehta PH, Meijlink KJ, Pellefigues C, Schmidt AJ, Le Gros G. The Gastrointestinal Helminth Heligmosomoides bakeri Suppresses Inflammation in a Model of Contact Hypersensitivity. Front Immunol 2020; 11:950. [PMID: 32508831 PMCID: PMC7249854 DOI: 10.3389/fimmu.2020.00950] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Accepted: 04/22/2020] [Indexed: 01/15/2023] Open
Abstract
Helminths regulate host immune responses to ensure their own long-term survival. Numerous studies have demonstrated that these helminth-induced regulatory mechanisms can also limit host inflammatory responses in several disease models. We used the Heligmosomoides bakeri (Hb) infection model (also known as H. polygyrus or H. polygyrus bakeri in the literature) to test whether such immune regulation affects skin inflammatory responses induced by the model contact sensitiser dibutyl phthalate fluorescein isothiocynate (DBP-FITC). Skin lysates from DBP-FITC-sensitized, Hb-infected mice produced less neutrophil specific chemokines and had significantly reduced levels of skin thickening and cellular inflammatory responses in tissue and draining lymph nodes (LNs) compared to uninfected mice. Hb-induced suppression did not appear to be mediated by regulatory T cells, nor was it due to impaired dendritic cell (DC) activity. Mice cleared of infection remained unresponsive to DBP-FITC sensitization indicating that suppression was not via the secretion of Hb-derived short-lived regulatory molecules, although long-term effects on cells cannot be ruled out. Importantly, similar helminth-induced suppression of inflammation was also seen in the draining LN after intradermal injection of the ubiquitous allergen house dust mite (HDM). These findings demonstrate that Hb infection attenuates skin inflammatory responses by suppressing chemokine production and recruitment of innate cells. These findings further contribute to the growing body of evidence that helminth infection can modulate inflammatory and allergic responses via a number of mechanisms with potential to be exploited in therapeutic and preventative strategies in the future.
Collapse
Affiliation(s)
- Kara J Filbey
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Palak H Mehta
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | | | | | | | - Graham Le Gros
- Malaghan Institute of Medical Research, Wellington, New Zealand
| |
Collapse
|
10
|
Gastrointestinal Coinfection Promotes Chlamydial Pathogenicity in the Genital Tract. Infect Immun 2020; 88:IAI.00905-19. [PMID: 31988173 DOI: 10.1128/iai.00905-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2019] [Accepted: 01/21/2020] [Indexed: 01/11/2023] Open
Abstract
Sexually transmitted Chlamydia, which can cause fibrotic pathology in women's genital tracts, is also frequently detected in the gastrointestinal tract. However, the medical significance of the gastrointestinal Chlamydia remains unclear. A murine Chlamydia readily spreads from the mouse genital tract to the gastrointestinal tract while inducing oviduct fibrotic blockage or hydrosalpinx. We previously proposed a two-hit model in which the mouse gastrointestinal Chlamydia might induce the second hit to promote genital tract pathology, and we are now providing experimental evidence for testing the hypothesis. First, chlamydial mutants that are attenuated in inducing hydrosalpinx in the genital tract also reduce their colonization in the gastrointestinal tract, leading to a better correlation of chlamydial induction of hydrosalpinx with chlamydial colonization in the gastrointestinal tract than in the genital tract. Second, intragastric coinoculation with a wild-type Chlamydia rescued an attenuated Chlamydia mutant to induce hydrosalpinx, while the chlamydial mutant infection in the genital tract alone was unable to induce any significant hydrosalpinx. Finally, the coinoculated gastrointestinal Chlamydia failed to directly spread to the genital tract lumen, suggesting that gastrointestinal Chlamydia may promote genital pathology via an indirect mechanism. Thus, we have demonstrated a significant role of gastrointestinal Chlamydia in promoting pathology in the genital tract possibly via an indirect mechanism. This study provides a novel direction/dimension for further investigating chlamydial pathogenic mechanisms.
Collapse
|
11
|
Ruiz-Manzano RA, Hernández-Cervantes R, Del Río-Araiza VH, Palacios-Arreola MI, Nava-Castro KE, Morales-Montor J. Immune response to chronic Toxocara canis infection in a mice model. Parasite Immunol 2019; 41:e12672. [PMID: 31557337 DOI: 10.1111/pim.12672] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 09/18/2019] [Accepted: 09/23/2019] [Indexed: 12/13/2022]
Abstract
AIMS The zoonotic nematode Toxocara canis causes larva migrans syndrome that induces an immune response characterized by the production of antibodies and eosinophilia. A Th2 polarization has been associated with the infection, but there are still details of the cellular and humoral immune response that need to be described. Thus, the aim of this study was to describe the systemic host immune response to T canis chronic infection in a mouse model. METHODS AND RESULTS BALB/c mice were inoculated once with 500 T canis embryonated eggs, per os. After 49 days, the amounts of larval found in brain and muscle tissues were statistically two and four times higher, respectively, than the amounts found in lung, liver, kidney or heart tissues. Splenic proportions of F4/80+ cells, as well as B, cytotoxic T and CD4+ Foxp3+ lymphocytes, were statistically higher (P ≤ .05, P ≤ .01, P ≤ .001 and P ≤ .001, respectively) as compared with control mice. In lymph nodes, some of these proportions changed, with the exception of F4/80+ cells. IgG1 levels in infected mice sera were increased. IL-4, IL-10 and VEGF levels were statistically higher in spleen (P ≤ .05, all) and sera (P ≤ .01, P ≤ .05 and P ≤ .05, respectively) in the infected mice. Also, in infected animals, IL-5 serum levels were increased (P ≤ .01). CONCLUSION These results suggest that T canis chronic infection in BALB/c mice results in a type 2 response with an incipient regulatory response.
Collapse
Affiliation(s)
- Rocío Alejandra Ruiz-Manzano
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Rosalía Hernández-Cervantes
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Víctor Hugo Del Río-Araiza
- Departamento de Parasitología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Margarita Isabel Palacios-Arreola
- Departamento de Genotoxicología, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Karen Elizabeth Nava-Castro
- Departamento de Genotoxicología, Centro de Ciencias de la Atmósfera, Universidad Nacional Autónoma de México, Ciudad de México, México
| | - Jorge Morales-Montor
- Departamento de Inmunología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, México
| |
Collapse
|
12
|
Metwali A, Thorne PS, Ince MN, Metwali N, Winckler S, Guan X, Beyatli S, Truscott J, Urban JF, Elliott DE. Recirculating Immunocompetent Cells in Colitic Mice Intensify Their Lung Response to Bacterial Endotoxin. Dig Dis Sci 2018; 63:2930-2939. [PMID: 30022451 PMCID: PMC6182434 DOI: 10.1007/s10620-018-5196-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 07/03/2018] [Indexed: 12/17/2022]
Abstract
BACKGROUND Patients with inflammatory bowel disease have higher incidence of airway hyperresponsiveness compared to the general population. Lung inflammation leading to airway hyperresponsiveness causes illnesses for more than ten percent of the population in USA. AIMS We investigated the lung response to bacterial endotoxin in colitic mice. METHODS Rag-1 mice were transplanted with negatively selected splenic T cells. Some mice groups were treated with NSAID to develop colitis. All mice were treated with bacterial endotoxin and necropsied 3 weeks later. RESULTS Colitic mice developed intensified lung inflammation on day 21 of treatment with bacterial endotoxin. Pulmonary lymphocytes from colitic mice displayed a proinflammatory cytokine profile, expressed high ICAM1 and low FoxP3. CD11c+, CD8+ cells bound and responded to non-systemic antigens from gut-localized microbiota and had higher expression of TLR4. CONCLUSIONS Colitic mice developed exacerbated lung inflammation in response to bacterial endotoxin compared to non-colitic mice. Proinflammatory cytokines from pulmonary lymphocytes induced high expression of ICAM1 and suppressed FoxP3 on CD4+ cells. CD11c+, CD8+ cells binding and responding to gut-localized antigens as well as high expression of TLR4 indicate innate and adaptive lung response to bacterial endotoxin. Inflammatory cells from colons of colitic mice homed in the lungs as well as the intestine suggesting recirculation of sensitized immunocompetent cells. These data support our hypothesis that colitis intensifies lung inflammation.
Collapse
Affiliation(s)
- Ahmed Metwali
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA USA
- Veterans Administration Medical Center, Iowa City, IA USA
| | - Peter S. Thorne
- Department of Occupational and Environmental Health Pulmonary Toxicology Facility, University of Iowa, Iowa City, USA
| | - M. Nedim Ince
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA USA
| | - Nervana Metwali
- Department of Occupational and Environmental Health Pulmonary Toxicology Facility, University of Iowa, Iowa City, USA
| | - Sarah Winckler
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA USA
- Veterans Administration Medical Center, Iowa City, IA USA
| | - Xiaoqun Guan
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA USA
| | - Sonay Beyatli
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA USA
| | - Jamie Truscott
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA USA
| | | | - David E. Elliott
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Iowa, Carver College of Medicine, Iowa City, IA USA
- Veterans Administration Medical Center, Iowa City, IA USA
| |
Collapse
|
13
|
Maruszewska-Cheruiyot M, Donskow-Łysoniewska K, Doligalska M. Helminth Therapy: Advances in the use of Parasitic Worms Against Inflammatory Bowel Diseases and its Challenges. Helminthologia 2018; 55:1-11. [PMID: 31662622 PMCID: PMC6799527 DOI: 10.1515/helm-2017-0048] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Accepted: 08/31/2017] [Indexed: 12/21/2022] Open
Abstract
Development of modern medicine and better living conditions in the 20th century helped in reducing a number of cases of infectious diseases. During the same time, expansion of autoimmunological disorders was noticed. Among other are Inflammatory Bowel Diseases (IBD) including ulcerative colitis and Crohn's disease which are chronic and relapsing inflammation of the gastrointestinal tract. Absence of effective treatment in standard therapies effects the search for alternative opportunities. As per hygienic hypothesis increasing number of cases of autoimmune diseases is as a result of reduced exposure to pathogens, especially parasites. Thus, one of the promising remedial acts against IBD and other allergic and autoimmune disorders is "helminth therapy". Cure with helminths seems to be the most effective therapy of IBD currently proposed. Helminth therapy focuses on advantageous results that have been obtained from the clinical trials, but its mechanisms are still unclear. Explanation of this phenomenon would help to develop new drugs against IBD based on helminth immunomodulatory molecules.
Collapse
Affiliation(s)
- M. Maruszewska-Cheruiyot
- Department of Parasitology, Faculty of Biology University of Warsaw, Miecznikowa 1, 02-096Warsaw, Poland
- E-mail:
| | - K. Donskow-Łysoniewska
- Department of Parasitology, Faculty of Biology University of Warsaw, Miecznikowa 1, 02-096Warsaw, Poland
| | - M. Doligalska
- Department of Parasitology, Faculty of Biology University of Warsaw, Miecznikowa 1, 02-096Warsaw, Poland
| |
Collapse
|
14
|
Zhong G. Chlamydia Spreading from the Genital Tract to the Gastrointestinal Tract - A Two-Hit Hypothesis. Trends Microbiol 2017; 26:611-623. [PMID: 29289422 DOI: 10.1016/j.tim.2017.12.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Revised: 11/21/2017] [Accepted: 12/06/2017] [Indexed: 12/18/2022]
Abstract
Chlamydia trachomatis, a leading bacterial cause of sexually transmitted infection-induced infertility, is frequently detected in the gastrointestinal tract. Chlamydia muridarum, a model pathogen for investigating C. trachomatis pathogenesis, readily spreads from the mouse genital tract to the gastrointestinal tract, establishing long-lasting colonization. C. muridarum mutants, despite their ability to activate acute oviduct inflammation, are attenuated in inducing tubal fibrosis and are no longer able to colonize the gastrointestinal tract, suggesting that the spread of C. muridarum to the gastrointestinal tract may contribute to its pathogenicity in the upper genital tract. However, gastrointestinal C. muridarum cannot directly autoinoculate the genital tract. Both antigen-specific CD8+ T cells and profibrotic cytokines, such as TNFα and IL-13, are essential for C. muridarum to induce tubal fibrosis; this may be induced by the gastrointestinal C. muridarum, as a second hit, to transmucosally convert tubal repairing - initiated by C. muridarum infection of tubal epithelial cells (serving as the first hit) - into pathogenic fibrosis. Testing the two-hit mouse model should both add new knowledge to the growing list of mechanisms by which gastrointestinal microbes contribute to pathologies in extragastrointestinal tissues and provide information for investigating the potential role of gastrointestinal C. trachomatis in human chlamydial pathogenesis.
Collapse
Affiliation(s)
- Guangming Zhong
- Department of Microbiology, Immunology and Molecular Genetics, University of Texas Health, Science Center at San Antonio, 7703 Floyd Curl Drive, San Antonio, TX 78229, USA.
| |
Collapse
|
15
|
Junginger J, Raue K, Wolf K, Janecek E, Stein VM, Tipold A, Günzel-Apel AR, Strube C, Hewicker-Trautwein M. Zoonotic intestinal helminths interact with the canine immune system by modulating T cell responses and preventing dendritic cell maturation. Sci Rep 2017; 7:10310. [PMID: 28871165 PMCID: PMC5583179 DOI: 10.1038/s41598-017-10677-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2016] [Accepted: 08/14/2017] [Indexed: 12/30/2022] Open
Abstract
Parasite co-evolution alongside the mammalian immune system gave rise to several modulatory strategies by which they prevent exaggerated pathology and facilitate a longer worm survival. As little is known about the immunoregulatory potential of the zoonotic canine parasites Ancylostoma caninum and Toxocara canis in the natural host, the present study aimed to investigate whether their larval excretory-secretory (ES) products can modulate the canine immune system. We demonstrated TcES to increase the frequency of CD4+ Foxp3high T cells, while both AcES and TcES were associated with elevated Helios expression in Foxp3high lymphocytes. ES products were further capable of inducing IL-10 production by lymphocytes, which was mainly attributed to CD8+ T cells. ES treatment of PBMCs prior to mitogen stimulation inhibited polyclonal proliferation of CD4+ and CD8+ T cells. Moreover, monocyte-derived ES-pulsed dendritic cells reduced upregulation of MHC-II and CD80 in response to lipopolysaccharide. The data showed that regulation of the canine immune system by A. caninum and T. canis larvae comprises the modification of antigen-specific and polyclonal T cell responses and dendritic cell maturation.
Collapse
Affiliation(s)
- Johannes Junginger
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, D-30559, Hannover, Germany
| | - Katharina Raue
- Institute for Parasitology, Center for Infection Medicine, University of Veterinary Medicine, Bünteweg 17, D-30559, Hannover, Germany
| | - Karola Wolf
- Unit of Reproductive Medicine of Clinics, University of Veterinary Medicine, Bünteweg 15, D-30559, Hannover, Germany.,Small Animal Clinic, University of Veterinary Medicine, Bünteweg 9, D-30559, Hannover, Germany
| | - Elisabeth Janecek
- Institute for Parasitology, Center for Infection Medicine, University of Veterinary Medicine, Bünteweg 17, D-30559, Hannover, Germany
| | - Veronika M Stein
- Small Animal Clinic, University of Veterinary Medicine, Bünteweg 9, D-30559, Hannover, Germany.,Vetsuisse Faculty, University of Bern, Länggassstrasse 128, CH-3012, Bern, Switzerland
| | - Andrea Tipold
- Small Animal Clinic, University of Veterinary Medicine, Bünteweg 9, D-30559, Hannover, Germany
| | - Anne-Rose Günzel-Apel
- Unit of Reproductive Medicine of Clinics, University of Veterinary Medicine, Bünteweg 15, D-30559, Hannover, Germany.,Small Animal Clinic, University of Veterinary Medicine, Bünteweg 9, D-30559, Hannover, Germany
| | - Christina Strube
- Institute for Parasitology, Center for Infection Medicine, University of Veterinary Medicine, Bünteweg 17, D-30559, Hannover, Germany
| | - Marion Hewicker-Trautwein
- Department of Pathology, University of Veterinary Medicine, Bünteweg 17, D-30559, Hannover, Germany.
| |
Collapse
|
16
|
Varyani F, Fleming JO, Maizels RM. Helminths in the gastrointestinal tract as modulators of immunity and pathology. Am J Physiol Gastrointest Liver Physiol 2017; 312:G537-G549. [PMID: 28302598 PMCID: PMC5495915 DOI: 10.1152/ajpgi.00024.2017] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 03/06/2017] [Accepted: 03/12/2017] [Indexed: 01/31/2023]
Abstract
Helminth parasites are highly prevalent in many low- and middle-income countries, in which inflammatory bowel disease and other immunopathologies are less frequent than in the developed world. Many of the most common helminths establish themselves in the gastrointestinal tract and can exert counter-inflammatory influences on the host immune system. For these reasons, interest has arisen as to how parasites may ameliorate intestinal inflammation and whether these organisms, or products they release, could offer future therapies for immune disorders. In this review, we discuss interactions between helminth parasites and the mucosal immune system, as well as the progress being made toward identifying mechanisms and molecular mediators through which it may be possible to attenuate pathology in the intestinal tract.
Collapse
Affiliation(s)
- Fumi Varyani
- 1Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom; ,2Edinburgh Clinical Academic Track, Western General Hospital, University of Edinburgh, Edinburgh, United Kingdom; and
| | - John O. Fleming
- 3Department of Neurology, University of Wisconsin School of Medicine and Public Health, Madison, Wisconsin
| | - Rick M. Maizels
- 1Wellcome Centre for Molecular Parasitology, Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, United Kingdom;
| |
Collapse
|
17
|
Parande Shirvan S, Ebrahimby A, Dousty A, Maleki M, Movassaghi A, Borji H, Haghparast A. Somatic extracts of Marshallagia marshalli downregulate the Th2 associated immune responses in ovalbumin-induced airway inflammation in BALB/c mice. Parasit Vectors 2017; 10:233. [PMID: 28494800 PMCID: PMC5427607 DOI: 10.1186/s13071-017-2159-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Accepted: 04/25/2017] [Indexed: 12/26/2022] Open
Abstract
Background Recently the role of gastrointestinal nematodes in modulating the immune responses in inflammatory and immune-mediated conditions such as allergy and autoimmune diseases has been introduced. This is mainly due to the suppressive effects of somatic and excretory secretory (ES) products of nematodes on the immune responses. In this study, we evaluated the immunomodulatory potentials of somatic products of Marshallagia marshalli, a gastrointestinal nematodes of sheep, to suppress the immune-mediated responses in a murine model of allergic airway inflammation. BALB/c mice were intraperitoneally (IP) sensitized with ovalbumin (OVA)/Alum and then challenged with 1% OVA. Somatic products of M. marshalli were administered during each sensitization. The effects of somatic products on development of allergic airway inflammation were evaluated by analyzing inflammatory cells recruitment, histopathological changes, cytokines production (IL-4, IL-13, IL-10, TGF-β) and serum antibody titers (IgG1, IgG2a). Results Somatic products of M. marshalli were able to suppress the induction of allergic airway inflammation in mice. Modulation of Th2 type responses (IL-4, IL-13, IgG1) via upregulations of IL-10 and TGF-β production was observed after injection of somatic products of M. marshalli. In addition, inflammatory cells infiltration and pathological disorders were significantly diminished following administration of somatic products. Conclusions Our data raised the possibility that helminths could be a potential therapeutic candidate to alleviate the inflammatory conditions in allergic asthma. According to these results, we concluded that M. marshalli may contain immune-modulatory molecules that attenuate allergic airway inflammation via induction of regulatory cytokines. Further investigations are required to identify molecules that might have potentials for development of novel therapeutic targets.
Collapse
Affiliation(s)
- Sima Parande Shirvan
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P. O. Box: 91775-1793, Mashhad, Iran
| | - Azadeh Ebrahimby
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P. O. Box: 91775-1793, Mashhad, Iran
| | - Arezoo Dousty
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P. O. Box: 91775-1793, Mashhad, Iran
| | - Mohsen Maleki
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P. O. Box: 91775-1793, Mashhad, Iran
| | - Ahmadreza Movassaghi
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P. O. Box: 91775-1793, Mashhad, Iran
| | - Hassan Borji
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P. O. Box: 91775-1793, Mashhad, Iran.
| | - Alireza Haghparast
- Department of Pathobiology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P. O. Box: 91775-1793, Mashhad, Iran. .,Division of Biotechnology, Faculty of Veterinary Medicine, Ferdowsi University of Mashhad, P. O. Box: 91775-1793, Mashhad, Iran.
| |
Collapse
|
18
|
Abstract
By reputation, the parasite is a pariah, an unwelcome guest. Infection with helminth parasites evokes stereotypic immune responses in humans and mice that are dominated by T helper (Th)-2 responses; thus, a hypothesis arises that infection with helminths would limit immunopathology in concomitant inflammatory disease. Although infection with some species of helminths can cause devastating disease and affect the course of microbial infections, analyses of rodent models of inflammatory disease reveal that infection with helminth parasites, or treatment with helminth extracts, can limit the severity of autoinflammatory disease, including colitis. Intriguing, but fewer, studies show that adoptive transfer of myeloid immune cells treated with helminth products/extracts in vitro can suppress inflammation. Herein, 3 facets of helminth therapy are reviewed and critiqued: treatment with viable ova or larvae, treatment with crude extracts of the worm or purified molecules, and cellular immunotherapy. The beneficial effect of helminth therapy often converges on the mobilization of IL-10 and regulatory/alternatively activated macrophages, while there are reports on transforming growth factor (TGF)-β, regulatory T cells and dendritic cells, and recent data suggest that helminth-evoked changes in the microbiota should be considered when defining anticolitic mechanisms. We speculate that if the data from animal models translate to humans, noting the heterogeneity therein, then the choice between use of viable helminth ova, helminth extracts/molecules or antigen-pulsed immune cells could be matched to disease management in defined cohorts of patients with inflammatory bowel disease.
Collapse
|
19
|
Weinstock JV, Elliott DE. Helminth infections decrease host susceptibility to immune-mediated diseases. THE JOURNAL OF IMMUNOLOGY 2014; 193:3239-47. [PMID: 25240019 DOI: 10.4049/jimmunol.1400927] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Helminthic infection has become rare in highly industrialized nations. Concurrent with the decline in helminthic infection has been an increase in the prevalence of inflammatory disease. Removal of helminths from our environment and their powerful effects on host immunity may have contributed to this increase. Several helminth species can abrogate disease in murine models of inflammatory bowel disease, type 1 diabetes, multiple sclerosis, and other conditions. Helminths evoke immune regulatory pathways often involving dendritic cells, regulatory T cells, and macrophages that help to control disease. Cytokines, such as IL-4, IL-10, and TGF-β, have a role. Notable is the helminthic modulatory effect on innate immunity, which impedes development of aberrant adaptive immunity. Investigators are identifying key helminth-derived immune modulatory molecules that may have therapeutic usefulness in the control of inflammatory disease.
Collapse
Affiliation(s)
- Joel V Weinstock
- Division of Gastroenterology, Tufts Medical Center, Boston, MA 02111; and
| | - David E Elliott
- Division of Gastroenterology, University of Iowa, Iowa City, IA 52242
| |
Collapse
|
20
|
Adnađević T, Jovanović VM, Blagojević J, Budinski I, Čabrilo B, Bijelić-Čabrilo O, Vujošević M. Possible influence of B chromosomes on genes included in immune response and parasite burden in Apodemus flavicollis. PLoS One 2014; 9:e112260. [PMID: 25372668 PMCID: PMC4221283 DOI: 10.1371/journal.pone.0112260] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2014] [Accepted: 10/02/2014] [Indexed: 11/18/2022] Open
Abstract
Genetic background underlying wild populations immune response to different parasites is still not well understood. We studied immune response to multiple infections and to competition between different parasite species at different developmental stages in population of yellow-necked mouse, Apodemus flavicollis. Quantitative real-time PCR was used to investigate associations of MHC II-DRB, IL-10 and Tgf-β genes expressions with presence of intestinal parasites at different developmental stages. Furthermore, we were interested whether the host related characteristics (sex, age, body condition, presence of B chromosomes or expression of other genes) or characteristics of present parasites (number of adult parasites of each identified species, egg count of each parasite genus, total number of nematode individuals) affect differential expression of the studied genes. A significant invert association between the expression of MHC II-DRB and Tgf-β gene was found, which together with absence of IL-10 association confirmed modified Th2 as the main type of immune response to nematode infections. Effect of recorded parasites and parasite life-cycle stage on expression levels of MHC II-DRB gene was detected only through interactions with host-related characteristics such as sex, age, and the presence of B chromosomes. The presence of B chromosomes is associated with lower expression level of Tgf-β gene. Although the influence of host genetic background on parasite infection has already been well documented, this is the first study in mammals that gave presence of B chromosomes on immune response full consideration.
Collapse
Affiliation(s)
- Tanja Adnađević
- Department of Genetic Research, Institute for Biological Research “Siniša Stanković”, University of Belgrade, Belgrade, Serbia
- * E-mail:
| | - Vladimir M. Jovanović
- Department of Genetic Research, Institute for Biological Research “Siniša Stanković”, University of Belgrade, Belgrade, Serbia
| | - Jelena Blagojević
- Department of Genetic Research, Institute for Biological Research “Siniša Stanković”, University of Belgrade, Belgrade, Serbia
| | - Ivana Budinski
- Department of Genetic Research, Institute for Biological Research “Siniša Stanković”, University of Belgrade, Belgrade, Serbia
| | - Borislav Čabrilo
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - Olivera Bijelić-Čabrilo
- Department of Biology and Ecology, Faculty of Sciences, University of Novi Sad, Novi Sad, Serbia
| | - Mladen Vujošević
- Department of Genetic Research, Institute for Biological Research “Siniša Stanković”, University of Belgrade, Belgrade, Serbia
| |
Collapse
|
21
|
Heylen M, Ruyssers NE, Gielis EM, Vanhomwegen E, Pelckmans PA, Moreels TG, De Man JG, De Winter BY. Of worms, mice and man: an overview of experimental and clinical helminth-based therapy for inflammatory bowel disease. Pharmacol Ther 2014; 143:153-167. [PMID: 24603369 DOI: 10.1016/j.pharmthera.2014.02.011] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2014] [Accepted: 02/25/2014] [Indexed: 12/17/2022]
Abstract
The incidence of inflammatory and autoimmune disorders is highest in well-developed countries which is directly related to their higher hygienic standards: it is suggested that the lack of exposure to helminths contributes to the susceptibility for immune-related diseases. Epidemiological, experimental and clinical data support the idea that helminths provide protection against immune-mediated diseases such as inflammatory bowel disease (IBD). The most likely mechanism for the suppression of immune responses by helminths is the release of helminth-derived immunomodulatory molecules. This article reviews the experimental and clinical studies investigating the therapeutic potential of helminth-based therapy in IBD and also focuses on the current knowledge of its immunomodulatory mechanisms of action highlighting innate as well as adaptive immune mechanisms. Identifying the mechanisms by which these helminths and helminth-derived molecules modulate the immune system will help in creating novel drugs for the treatment of IBD and other disorders that result from an overactive immune response.
Collapse
Affiliation(s)
- Marthe Heylen
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Nathalie E Ruyssers
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Els M Gielis
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Els Vanhomwegen
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Paul A Pelckmans
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Division of Gastroenterology & Hepatology, Antwerp, Belgium
| | - Tom G Moreels
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium; Antwerp University Hospital, Division of Gastroenterology & Hepatology, Antwerp, Belgium
| | - Joris G De Man
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium
| | - Benedicte Y De Winter
- Laboratory of Experimental Medicine and Pediatrics, Division of Gastroenterology, University of Antwerp, Antwerp, Belgium.
| |
Collapse
|
22
|
Norman JM, Handley SA, Virgin HW. Kingdom-agnostic metagenomics and the importance of complete characterization of enteric microbial communities. Gastroenterology 2014; 146:1459-69. [PMID: 24508599 PMCID: PMC4009354 DOI: 10.1053/j.gastro.2014.02.001] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 01/27/2014] [Accepted: 02/03/2014] [Indexed: 12/13/2022]
Abstract
Advanced sequencing techniques have shown that bacteria are not the only complex and important microbes in the human intestine. Nonbacterial organisms, particularly the virome and the mycobiome, are important regulators of intestinal immunity and inflammation. The virome is mucosal and systemic; it can alter the host response to bacteria and interact with host genes and bacteria to contribute to disease pathogenesis. The human mycobiome is also complex and can contribute to intestinal inflammation. We review what has recently been learned about the nonbacterial and nonarchaeal microbes in the gastrointestinal tract, discussing their potential effects on health and disease and analytical approaches for their study. Studies of associations between the microbiome and intestinal pathology should incorporate kingdom-agnostic approaches if we are to fully understand intestinal health and disease.
Collapse
Affiliation(s)
| | | | - Herbert W. Virgin
- Reprint requests Address requests for reprints to: Herbert W. Virgin, MD, PhD, Washington University School of Medicine, Box 8118, 660 South Euclid Avenue, St Louis, Missouri 63110.
| |
Collapse
|
23
|
Rausch S, Held J, Fischer A, Heimesaat MM, Kühl AA, Bereswill S, Hartmann S. Small intestinal nematode infection of mice is associated with increased enterobacterial loads alongside the intestinal tract. PLoS One 2013; 8:e74026. [PMID: 24040152 PMCID: PMC3769368 DOI: 10.1371/journal.pone.0074026] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2013] [Accepted: 07/25/2013] [Indexed: 01/04/2023] Open
Abstract
Parasitic nematodes are potent modulators of immune reactivity in mice and men. Intestinal nematodes live in close contact with commensal gut bacteria, provoke biased Th2 immune responses upon infection, and subsequently lead to changes in gut physiology. We hypothesized that murine nematode infection is associated with distinct changes of the intestinal bacterial microbiota composition. We here studied intestinal inflammatory and immune responses in mice following infection with the hookworm Heligmosomoidespolygyrusbakeri and applied cultural and molecular techniques to quantitatively assess intestinal microbiota changes in the ileum, cecum and colon. At day 14 post nematode infection, mice harbored significantly higher numbers of γ-Proteobacteria/Enterobacteriaceae and members of the Bacteroides/Prevotella group in their cecum as compared to uninfected controls. Abundance of Gram-positive species such as Lactobacilli, Clostridia as well as the total bacterial load was not affected by worm infection. The altered microbiota composition was independent of the IL-4/-13 – STAT6 signaling axis, as infected IL-4Rα-/- mice showed a similar increase in enterobacterial loads. In conclusion, infection with an enteric nematode is accompanied by distinct intestinal microbiota changes towards higher abundance of gram-negative commensal species at the small intestinal site of infection (and inflammation), but also in the parasite-free large intestinal tract. Further studies should unravel the impact of nematode-induced microbiota changes in inflammatory bowel disease to allow for a better understanding of how theses parasites interfere with intestinal inflammation and bacterial communities in men.
Collapse
MESH Headings
- Animals
- Bacterial Load
- Cytokines/biosynthesis
- Enterobacteriaceae/classification
- Enterobacteriaceae/genetics
- Enterobacteriaceae/growth & development
- Female
- Interleukin-4 Receptor alpha Subunit/genetics
- Interleukin-4 Receptor alpha Subunit/metabolism
- Intestinal Diseases, Parasitic/immunology
- Intestinal Diseases, Parasitic/microbiology
- Intestinal Mucosa/microbiology
- Intestinal Mucosa/parasitology
- Intestinal Mucosa/pathology
- Intestine, Small/immunology
- Intestine, Small/microbiology
- Intestine, Small/parasitology
- Intestine, Small/pathology
- Mice
- Mice, Knockout
- Microbiota
- Nematode Infections/immunology
- Nematode Infections/microbiology
- Nematode Infections/parasitology
- RNA, Bacterial
- RNA, Ribosomal, 16S
- Signal Transduction
Collapse
Affiliation(s)
- Sebastian Rausch
- Institute of Immunology, Department of Veterinary Medicine, Freie Universität, Berlin, Germany
- * E-mail:
| | - Josephin Held
- Department of Neuropathology, Charité - University Medicine Berlin, Berlin, Germany
| | - André Fischer
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Markus M. Heimesaat
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Anja A. Kühl
- Department of Internal Medicine, Rheumatology and Clinical Immunology/Research Center ImmunoSciences (RCIS), Charité - University Medicine Berlin, Berlin, Germany
| | - Stefan Bereswill
- Department of Microbiology and Hygiene, Charité - University Medicine Berlin, Berlin, Germany
| | - Susanne Hartmann
- Institute of Immunology, Department of Veterinary Medicine, Freie Universität, Berlin, Germany
| |
Collapse
|
24
|
Othman AA, Ashour DS, Mohamed DA. Toxocara-induced hepatic inflammation: Immunohistochemical characterization of lymphocyte subpopulations and Bcl-2 expression. Exp Parasitol 2013; 134:76-81. [DOI: 10.1016/j.exppara.2013.02.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Revised: 01/30/2013] [Accepted: 02/04/2013] [Indexed: 11/25/2022]
|
25
|
Doligalska M, Brodaczewska K, Donskow-Łysoniewska K. The antiapoptotic activity of Heligmosomoides polygyrus antigen fractions. Parasite Immunol 2013; 34:589-603. [PMID: 23009264 DOI: 10.1111/pim.12006] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Accepted: 08/31/2012] [Indexed: 11/30/2022]
Abstract
Our study identified Heligmosomoides polygyrus antigen factors with potential activity for regulation of T-cell proliferation and surviving of CD4(+) CD25(-) , CD4(+) CD25(hi) and CD3(+) CD8(+) cell populations. The antiapoptotic activity of antigenic fractions separated by HPLC was evaluated in vitro after exposure of cells to DEX and rTNF-α. Different populations of cells responded to antigen fractions in distinct pattern; the most sensitive population of cells to H. polygyrus products were CD4(+) CD25(hi) after exposure to DEX and CD3(+) CD8(+) T cells after exposure to rTNF-α. H. polygyrus antigens may influence survival of CD8(+) T cells by regulation of c-FLIP rather than Bcl-2, which affects survival of CD4(+) CD25(hi) Treg cells and CD4(+) T cells. Activation of NF-κB subunits, for example, p50 and p65 was essential for resistance of cells to apoptosis, and antigenic fractions F9 and F17 exerted different effect to F13. The most active fraction in inhibition of apoptosis was F9, which includes Hsp-60, calumenin, ferritin, galectin and thrombospondin. This study may provide new clues for recognition of factors that regulate the immune response during infection and which engage the TNF-α receptor-mediated and the mitochondria-mediated death pathway.
Collapse
Affiliation(s)
- M Doligalska
- Department of Parasitology, University of Warsaw, Warsaw, Poland
| | | | | |
Collapse
|
26
|
Donskow-Łysoniewska K, Majewski P, Brodaczewska K, Jóźwicka K, Doligalska M. Heligmosmoides polygyrus fourth stages induce protection against DSS-induced colitis and change opioid expression in the intestine. Parasite Immunol 2013; 34:536-46. [PMID: 22889318 DOI: 10.1111/pim.12003] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Primary exposure of mice to the nematode Heligmosomoides polygyrus infection reduces inflammation in an experimental model of colitis. The aim of the present investigation was to evaluate whether the reduced inflammation provoked by H. polygyrus L4 larvae in BALB/c mice treated with dextran sulphate sodium is associated with changed expression of opioids in the small intestine and colon. Colitis was induced by 5% Dextran sulphate sodium (DSS) oral administration for 3 days before oral infection with 200 infective larvae (L3) H. polygyrus until the end of the experiment, 6 days post-infection. Clinical disease symptoms were monitored daily. The expressions of proopiomelanocortin POMC1, MOR1 (Oprm1) - opioid receptor and β-endorphin were determined by RT-PCR, Western blot and immunoassay, respectively, in the colon and small intestine of mice. RT-PCR analysis of colon tissues showed up-regulation of the expression of POMC and MOR1 opioid-dependent genes in mice with DSS-induced colitis. H. polygyrus L4 larvae inhibited DSS-induced colitis symptoms that were correlated with increased IL-1β, TNF-α, IL-6, myeloperoxidase (MPO) concentration, macrophages infiltration and MOR1, POMC and β-endorphin increased expression in the small intestine and inhibition of those in the colon.
Collapse
|
27
|
Daniłowicz-Luebert E, Steinfelder S, Kühl AA, Drozdenko G, Lucius R, Worm M, Hamelmann E, Hartmann S. A nematode immunomodulator suppresses grass pollen-specific allergic responses by controlling excessive Th2 inflammation. Int J Parasitol 2013; 43:201-10. [DOI: 10.1016/j.ijpara.2012.10.014] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2012] [Revised: 10/18/2012] [Accepted: 10/24/2012] [Indexed: 11/16/2022]
|
28
|
Nematode asparaginyl-tRNA synthetase resolves intestinal inflammation in mice with T-cell transfer colitis. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2012; 20:276-81. [PMID: 23254300 DOI: 10.1128/cvi.00594-12] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The therapeutic effects of a controlled parasitic nematode infection on the course of inflammatory bowel disease (IBD) have been demonstrated in both animal and human models. However, the inability of individual well-characterized nematode proteins to recreate these beneficial effects has limited the application of component immunotherapy to human disease. The nematodes that cause chronic human lymphatic filariasis, Brugia malayi and Wuchereria bancrofti, are among the parasites that induce immune suppression. Filarial lymphatic pathology has been shown to involve NF-κB pathway-dependent production of vascular endothelial growth factor (VEGF), and stimulation of VEGF expression has also been reported by interleukin 8 (IL-8) via NF-κB pathways. Previously, we have shown that the filarial asparaginyl-tRNA synthetase (rBmAsnRS) interacts with IL-8 receptors using a combination of extracellular loops that differ from those bound by IL-8. To test the hypothesis that rBmAsnRS might induce an anti-inflammatory effect in vivo, we studied the effects of rBmAsnRS in an established murine colitis model using T-cell transfer mice. T-cell transfer colitis mice treated intraperitoneally with 100 μg of rBmAsnRS four times over 2 weeks showed resolution of cellular infiltration in the colonic mucosa, along with induction of a CD8(+) cellular response. In addition, rBmAsnRS induced a rise in IL-10 production from CD3(+) and lipopolysaccharide (LPS)- and cytosine phosphate guanosine (CPG)-stimulated splenic cells. In summary, this work demonstrates a novel anti-inflammatory nematode protein, supports the hygiene hypothesis, and supports continued refinement of alternative immunotherapies for treatment of IBD.
Collapse
|
29
|
Abstract
Modern hygienic lifestyles are associated with the emergence of inflammatory bowel disease (IBD) which now afflicts millions of people in highly-developed countries. Meticulous hygiene interrupts conduits of transmission required for ubiquitous exposure to parasitic worms (helminths). We proposed that loss of exposure to helminths permits development of IBD. Early clinical trials suggested that exposure to helminths such as Trichuris suis or Necator americanus can improve IBD. Over the last several years, processes to "medicinalize"T. suis have been developed and use of this helminth is now being studied in large multi-center clinical trials. Concurrently, we and others have identified some of the immune regulatory mechanisms elicited by helminth exposure that suppress inappropriate intestinal inflammation. These efforts could soon result in new therapies for patients with IBD.
Collapse
Affiliation(s)
- Joel V Weinstock
- Division of Gastroenterology, Tufts Medical Center, Boston, MA, USA.
| | | |
Collapse
|
30
|
Abstract
PURPOSE OF REVIEW There is something about living in an industrialized country that dramatically increases the risk of acquiring inflammatory bowel disease (IBD). Loss of routine exposure to parasitic worms (helminths), due to modern highly hygienic life styles, likely contributes to this risk. This article reviews current understanding on how helminths influence intestinal inflammation and mucosal immune responses. RECENT FINDINGS IBD emerges in populations as regions develop socioeconomically and lose exposure to previously ubiquitous helminthic infections. Helminthic infections provided strong selective pressure for the dissemination of gene variants, many of which predispose to development of IBD. In animal models of IBD, helminth colonization suppresses intestinal inflammation through multiple mechanisms including induction of innate and adaptive regulatory circuits. Trials using helminths like hookworm (Necator americanus) or porcine whipworm (Trichuris suis) show that they are safe and may be effective therapies for the control of the aberrant intestinal inflammation seen in Crohn's disease and ulcerative colitis. SUMMARY Evidence is accumulating that highly hygienic living conditions create risk for developing immune-mediated disease such as IBD. To live in their host, helminths have developed the ability to activate cells of innate and adaptive immunity that suppress inflammation. Therapeutic trials using helminths are in progress.
Collapse
|
31
|
Reynolds LA, Filbey KJ, Maizels RM. Immunity to the model intestinal helminth parasite Heligmosomoides polygyrus. Semin Immunopathol 2012; 34:829-46. [PMID: 23053394 PMCID: PMC3496515 DOI: 10.1007/s00281-012-0347-3] [Citation(s) in RCA: 164] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2012] [Accepted: 09/13/2012] [Indexed: 02/07/2023]
Abstract
Heligmosomoides polygyrus is a natural intestinal parasite of mice, which offers an excellent model of the immunology of gastrointestinal helminth infections of humans and livestock. It is able to establish long-term chronic infections in many strains of mice, exerting potent immunomodulatory effects that dampen both protective immunity and bystander reactions to allergens and autoantigens. Immunity to the parasite develops naturally in some mouse strains and can be induced in others through immunization; while the mechanisms of protective immunity are not yet fully defined, both antibodies and a host cellular component are required, with strongest evidence for a role of alternatively activated macrophages. We discuss the balance between resistance and susceptibility in this model system and highlight new themes in innate and adaptive immunity, immunomodulation, and regulation of responsiveness in helminth infection.
Collapse
Affiliation(s)
- Lisa A. Reynolds
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JT UK
| | - Kara J. Filbey
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JT UK
| | - Rick M. Maizels
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JT UK
- Institute of Immunology and Infection Research, University of Edinburgh, West Mains Road, Edinburgh, EH9 3JT UK
| |
Collapse
|
32
|
Hernandez JLR, Leung G, McKay DM. Cestode regulation of inflammation and inflammatory diseases. Int J Parasitol 2012; 43:233-43. [PMID: 23058631 DOI: 10.1016/j.ijpara.2012.09.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Revised: 09/11/2012] [Accepted: 09/12/2012] [Indexed: 12/13/2022]
Abstract
Helminth parasites are masters of immune regulation; a likely prerequisite for long-term survival by circumventing their hosts' attempt to eradicate them. From a translational perspective, knowledge of immune events as a response to infection with a helminth parasite could be used to reduce the intensity of unwanted inflammatory reactions. Substantial data have accumulated showing that inflammatory reactions that promote a variety of auto-inflammatory diseases are dampened as a consequence of infection with helminth parasites, via either the mobilization of an anti-worm spectrum of immune events or by the direct effect of secretory/excretory bioactive immunomodulatory molecules released from the parasite. However, many issues are outstanding in the definition of the mechanism(s) by which infection with helminth parasites can affect the outcome, positively or negatively, of concomitant disease. We focus on a subgroup of this complex group of metazoan parasites, the cestodes, summarizing studies from rodent models that illustrate if, and by what mechanisms, infection with tapeworms ameliorate or exaggerate disease in their host. The ability of infection with cestodes, or other classes of helminth, to worsen a disease course or confer susceptibility to intracellular pathogens should be carefully considered in the context of 'helminth therapy'. In addition, poorly characterised cestode extracts can regulate murine and human immunocyte function, yet the impact of these in the context of autoimmune or allergic diseases is poorly understood. Thus, studies with cestodes, as representative helminths, have helped cement the concept that infection with parasitic helminths can inhibit concomitant disease; however, issues relating to long-term effects, potential side-effects, mixed pathogen infections and purification of immunomodulatory molecules from the parasite remain as challenges that need to be addressed in order to achieve the use of helminths as anti-inflammatory agents for human diseases.
Collapse
Affiliation(s)
- Jose-Luis Reyes Hernandez
- Gastrointestinal Research Group, Department of Physiology and Pharmacology, Calvin, Phoebe and Joan Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, Canada
| | | | | |
Collapse
|
33
|
Abstract
Helminth parasites infect almost one-third of the world's population, primarily in tropical regions. However, regions where helminth parasites are endemic record much lower prevalences of allergies and autoimmune diseases, suggesting that parasites may protect against immunopathological syndromes. Most helminth diseases are spectral in nature, with a large proportion of relatively asymptomatic cases and a subset of patients who develop severe pathologies. The maintenance of the asymptomatic state is now recognized as reflecting an immunoregulatory environment, which may be promoted by parasites, and involves multiple levels of host regulatory cells and cytokines; a breakdown of this regulation is observed in pathological disease. Currently, there is much interest in whether helminth-associated immune regulation may ameliorate allergy and autoimmunity, with investigations in both laboratory models and human trials. Understanding and exploiting the interactions between these parasites and the host regulatory network are therefore likely to highlight new strategies to control both infectious and immunological diseases.
Collapse
|
34
|
Maizels RM, Hewitson JP, Murray J, Harcus YM, Dayer B, Filbey KJ, Grainger JR, McSorley HJ, Reynolds LA, Smith KA. Immune modulation and modulators in Heligmosomoides polygyrus infection. Exp Parasitol 2012; 132:76-89. [PMID: 21875581 PMCID: PMC6485391 DOI: 10.1016/j.exppara.2011.08.011] [Citation(s) in RCA: 94] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2011] [Revised: 08/09/2011] [Accepted: 08/15/2011] [Indexed: 01/12/2023]
Abstract
The intestinal nematode parasite Heligmosomoides polygyrus bakeri exerts widespread immunomodulatory effects on both the innate and adaptive immune system of the host. Infected mice adopt an immunoregulated phenotype, with abated allergic and autoimmune reactions. At the cellular level, infection is accompanied by expanded regulatory T cell populations, skewed dendritic cell and macrophage phenotypes, B cell hyperstimulation and multiple localised changes within the intestinal environment. In most mouse strains, these act to block protective Th2 immunity. The molecular basis of parasite interactions with the host immune system centres upon secreted products termed HES (H. polygyrus excretory-secretory antigen), which include a TGF-β-like ligand that induces de novo regulatory T cells, factors that modify innate inflammatory responses, and molecules that block allergy in vivo. Proteomic and transcriptomic definition of parasite proteins, combined with biochemical identification of immunogenic molecules in resistant mice, will provide new candidate immunomodulators and vaccine antigens for future research.
Collapse
Affiliation(s)
- Rick M Maizels
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3JT, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Blum AM, Hang L, Setiawan T, Urban JP, Stoyanoff KM, Leung J, Weinstock JV. Heligmosomoides polygyrus bakeri induces tolerogenic dendritic cells that block colitis and prevent antigen-specific gut T cell responses. THE JOURNAL OF IMMUNOLOGY 2012; 189:2512-20. [PMID: 22844110 DOI: 10.4049/jimmunol.1102892] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Immunological diseases such as inflammatory bowel disease (IBD) are infrequent in less developed countries, possibly because helminths provide protection by modulating host immunity. In IBD murine models, the helminth Heligmosomoides polygyrus bakeri prevents colitis. It was determined whether H. polygyrus bakeri mediated IBD protection by altering dendritic cell (DC) function. We used a Rag IBD model where animals were reconstituted with IL10⁻/⁻ T cells, making them susceptible to IBD and with OVA Ag-responsive OT2 T cells, allowing study of a gut antigenic response. Intestinal DC from H. polygyrus bakeri-infected Rag mice added to lamina propria mononuclear cells (LPMC) isolated from colitic animals blocked OVA IFN-γ/IL-17 responses in vitro through direct contact with the inflammatory LPMC. DC from uninfected Rag mice displayed no regulatory activity. Transfer of DC from H. polygyrus bakeri-infected mice into Rag mice reconstituted with IL10⁻/⁻ T cells protected animals from IBD, and LPMC from these mice lost OVA responsiveness. After DC transfer, OT2 T cells populated the intestines normally. However, the OT2 T cells were rendered Ag nonresponsive through regulatory action of LPMC non-T cells. The process of regulation appeared to be regulatory T cell independent. Thus, H. polygyrus bakeri modulates intestinal DC function, rendering them tolerogenic. This appears to be an important mechanism through which H. polygyrus bakeri suppresses colitis. IFN-γ and IL-17 are colitogenic. The capacity of these DC to block a gut Ag-specific IFN-γ/IL-17 T cell response also is significant.
Collapse
Affiliation(s)
- Arthur M Blum
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Hepworth MR, Hartmann S. Worming our way closer to the clinic. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2012; 2:187-90. [PMID: 24533279 DOI: 10.1016/j.ijpddr.2012.07.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Revised: 06/28/2012] [Accepted: 07/01/2012] [Indexed: 12/26/2022]
Abstract
In a recent issue of "The International Journal for Parasitology: Drugs and Drug Resistance" Prof. David Pritchard from the University of Nottingham offers his intriguing opinion on the current status of "worm therapy" and outlines future research priorities aimed at bringing this research area closer to the clinic. In this response article we discuss various aspects of the current state of the research field and offer some alternative viewpoints regarding the future of "worm therapy".
Collapse
Affiliation(s)
- Matthew R Hepworth
- Institute of Immunology, School of Veterinary Medicine, Free University, Berlin, Germany
| | - Susanne Hartmann
- Institute of Immunology, School of Veterinary Medicine, Free University, Berlin, Germany
| |
Collapse
|
37
|
Korpe PS, Petri WA. Environmental enteropathy: critical implications of a poorly understood condition. Trends Mol Med 2012; 18:328-36. [PMID: 22633998 DOI: 10.1016/j.molmed.2012.04.007] [Citation(s) in RCA: 332] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2012] [Revised: 04/12/2012] [Accepted: 04/20/2012] [Indexed: 12/13/2022]
Abstract
Environmental enteropathy (also called tropical enteropathy) is a subclinical condition caused by constant fecal-oral contamination and resulting in blunting of intestinal villi and intestinal inflammation. Although these histological changes were discovered decades ago, the clinical impact of environmental enteropathy is just starting to be recognized. The failure of nutritional interventions and oral vaccines in the developing world may be attributed to environmental enteropathy, as the intestinal absorptive and immunologic functions are significantly deranged. Here we review the existing literature and examine potential mechanisms of pathogenesis for this poorly understood condition.
Collapse
Affiliation(s)
- Poonum S Korpe
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA 22908-1337, USA
| | | |
Collapse
|
38
|
Braga M, Schiavone C, Di Gioacchino G, De Angelis I, Cavallucci E, Lazzarin F, Petrarca C, Di Gioacchino M. Environment and T regulatory cells in allergy. THE SCIENCE OF THE TOTAL ENVIRONMENT 2012; 423:193-201. [PMID: 20825978 DOI: 10.1016/j.scitotenv.2010.08.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2009] [Revised: 08/09/2010] [Accepted: 08/09/2010] [Indexed: 05/29/2023]
Abstract
The central role of T regulatory cells in the responses against harmless environmental antigens has been confirmed by many studies. Impaired T regulatory cell function is implicated in many pathological conditions, particularly allergic diseases. The "hygiene hypothesis" suggests that infections and infestations may play a protective role for allergy, whereas environmental pollutants favor the development of allergic diseases. Developing countries suffer from a variety of infections and are also facing an increasing diffusion of environmental pollutants. In these countries allergies increase in relation to the spreading use of xenobiotics (pesticides, herbicides, pollution, etc.) with a rate similar to those of developed countries, overcoming the protective effects of infections. We review here the main mechanisms of non-self tolerance, with particular regard to relations between T regulatory cell activity, infections and infestations such as helminthiasis, and exposure to environmental xenobiotics with relevant diffusion in developing countries.
Collapse
Affiliation(s)
- M Braga
- Allergy Unit, Spedali Civili, Piazzale Spedali Civili, 25123 Brescia, Italy.
| | | | | | | | | | | | | | | |
Collapse
|
39
|
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: 10.5] [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
|
40
|
Gravano DM, Vignali DAA. The battle against immunopathology: infectious tolerance mediated by regulatory T cells. Cell Mol Life Sci 2011; 69:1997-2008. [PMID: 22205213 DOI: 10.1007/s00018-011-0907-z] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 12/11/2011] [Accepted: 12/12/2011] [Indexed: 02/07/2023]
Abstract
Infectious tolerance is a process whereby one regulatory lymphoid population confers suppressive capacity on another. Diverse immune responses are induced following infection or inflammatory insult that can protect the host, or potentially cause damage if not properly controlled. Thus, the process of infectious tolerance may be critical in vivo for exerting effective immune control and maintaining immune homeostasis by generating specialized regulatory sub-populations with distinct mechanistic capabilities. Foxp3(+) regulatory T cells (T(regs)) are a central mediator of infectious tolerance through their ability to convert conventional T cells into induced regulatory T cells (iT(regs)) directly by secretion of the suppressive cytokines TGF-β, IL-10, or IL-35, or indirectly via dendritic cells. In this review, we will discuss the mechanisms and cell populations that mediate and contribute to infectious tolerance, with a focus on the intestinal environment, where tolerance induction to foreign material is critical.
Collapse
Affiliation(s)
- David M Gravano
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105-3678, USA
| | | |
Collapse
|
41
|
Axtner J, Sommer S. Heligmosomoides polygyrus infection is associated with lower MHC class II gene expression in Apodemus flavicollis: indication for immune suppression? INFECTION GENETICS AND EVOLUTION 2011; 11:2063-71. [PMID: 21983561 DOI: 10.1016/j.meegid.2011.09.020] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2011] [Revised: 09/22/2011] [Accepted: 09/22/2011] [Indexed: 01/16/2023]
Abstract
Due to their key role in recognizing foreign antigens and triggering the subsequent immune response the genes of the major histocompatibility complex (MHC) provide a potential target for parasites to attack in order to evade detection and expulsion from the host. A diminished MHC gene expression results in less activated T cells and might serve as a gateway for pathogens and parasites. Some parasites are suspected to be immune suppressors and promote co-infections of other parasites even in other parts of the body. In our study we found indications that the gut dwelling nematode Heligmosomoides polygyrus might exert a systemic immunosuppressive effect in yellow-necked mice (Apodemus flavicollis). The amount of hepatic MHC class II DRB gene RNA transcripts in infected mice was negatively associated with infection intensity with H. polygyrus. The hepatic expression of immunosuppressive cytokines, such as transforming growth factor β and interleukin 10 was not associated with H. polygyrus infection. We did not find direct positive associations of H. polygyrus with other helminth species. But the prevalence and infection intensity of the nematodes Syphacia stroma and Trichuris muris were higher in multiple infected individuals. Furthermore, our data indicated antagonistic effects in the helminth community of A. flavicollis as cestode infection correlated negatively with H. polygyrus and helminth species richness. Our study shows that expression analyses of immune relevant genes can also be performed in wildlife, opening new aspects and possibilities for future ecological and evolutionary research.
Collapse
Affiliation(s)
- Jan Axtner
- Evolutionary Genetics, Leibniz Institute for Zoo and Wildlife Research, Alfred-Kowalke-Str 15, 10315 Berlin, Germany
| | | |
Collapse
|
42
|
Hewitson JP, Filbey KJ, Grainger JR, Dowle AA, Pearson M, Murray J, Harcus Y, Maizels RM. Heligmosomoides polygyrus elicits a dominant nonprotective antibody response directed against restricted glycan and peptide epitopes. THE JOURNAL OF IMMUNOLOGY 2011; 187:4764-77. [PMID: 21964031 DOI: 10.4049/jimmunol.1004140] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Heligmosomoides polygyrus is a widely used gastrointestinal helminth model of long-term chronic infection in mice, which has not been well-characterized at the antigenic level. We now identify the major targets of the murine primary Ab response as a subset of the secreted products in H. polygyrus excretory-secretory (HES) Ag. An immunodominant epitope is an O-linked glycan (named glycan A) carried on three highly expressed HES glycoproteins (venom allergen Ancylostoma-secreted protein-like [VAL]-1, -2, and -5), which stimulates only IgM Abs, is exposed on the adult worm surface, and is poorly represented in somatic parasite extracts. A second carbohydrate epitope (glycan B), present on both a non-protein high molecular mass component and a 65-kDa molecule, is widely distributed in adult somatic tissues. Whereas the high molecular mass component and 65-kDa molecules bear phosphorylcholine, the glycan B epitope itself is not phosphorylcholine. Class-switched IgG1 Abs are found to glycan B, but the dominant primary IgG1 response is to the polypeptides of VAL proteins, including also VAL-3 and VAL-4. Secondary Ab responses include the same specificities while also recognizing VAL-7. Although vaccination with HES conferred complete protection against challenge H. polygyrus infection, mAbs raised against each of the glycan epitopes and against VAL-1, VAL-2, and VAL-4 proteins were unable to do so, even though these specificities (with the exception of VAL-2) are also secreted by tissue-phase L4 larvae. The primary immune response in susceptible mice is, therefore, dominated by nonprotective Abs against a small subset of antigenic epitopes, raising the possibility that these act as decoy specificities that generate ineffective humoral immunity.
Collapse
Affiliation(s)
- James P Hewitson
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3JT, United Kingdom
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Early peritoneal immune response during Echinococcus granulosus establishment displays a biphasic behavior. PLoS Negl Trop Dis 2011; 5:e1293. [PMID: 21912714 PMCID: PMC3166041 DOI: 10.1371/journal.pntd.0001293] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Accepted: 07/14/2011] [Indexed: 12/31/2022] Open
Abstract
Background Cystic echinococcosis is a worldwide distributed helminth zoonosis caused by the larval stage of Echinococcus granulosus. Human secondary cystic echinococcosis is caused by dissemination of protoscoleces after accidental rupture of fertile cysts and is due to protoscoleces ability to develop into new metacestodes. In the experimental model of secondary cystic echinococcosis mice react against protoscoleces producing inefficient immune responses, allowing parasites to develop into cysts. Although the chronic phase of infection has been analyzed in depth, early immune responses at the site of infection establishment, e.g., peritoneal cavity, have not been well studied. Because during early stages of infection parasites are thought to be more susceptible to immune attack, this work focused on the study of cellular and molecular events triggered early in the peritoneal cavity of infected mice. Principal Findings Data obtained showed disparate behaviors among subpopulations within the peritoneal lymphoid compartment. Regarding B cells, there is an active molecular process of plasma cell differentiation accompanied by significant local production of specific IgM and IgG2b antibodies. In addition, peritoneal NK cells showed a rapid increase with a significant percentage of activated cells. Peritoneal T cells showed a substantial increase, with predominance in CD4+ T lymphocytes. There was also a local increase in Treg cells. Finally, cytokine response showed local biphasic kinetics: an early predominant induction of Th1-type cytokines (IFN-γ, IL-2 and IL-15), followed by a shift toward a Th2-type profile (IL-4, IL-5, IL-6, IL-10 and IL-13). Conclusions Results reported here open new ways to investigate the involvement of immune effectors players in E. granulosus establishment, and also in the sequential promotion of Th1- toward Th2-type responses in experimental secondary cystic echinococcosis. These data would be relevant for designing rational therapies based on stimulation of effective responses and blockade of evasion mechanisms. Cystic echinococcosis is a zoonotic disease caused by the larval stage of the cestode Echinococcus granulosus and shows a cosmopolitan distribution with a worldwide prevalence of roughly 6 million infected people. Human cystic echinococcosis can develop in two types of infection. Primary infection occurs by ingestion of oncospheres, while secondary infection is caused by dissemination of protoscoleces after accidental rupture of fertile cysts. Murine experimental secondary infection in Balb/c mice is the current model to study E. granulosus-host interaction. Secondary infection can be divided into two stages: an early stage in which protoscoleces develop into hydatid cysts (infection establishment) and a later stage in which already differentiated cysts grow and eventually become fertile cysts (chronic infection). During infection establishment parasites are more susceptible to immune attack, thus our study focused on the immunological phenomena triggered early in the peritoneal cavity of experimentally infected mice. Our results suggest that early and local Th2-type responses are permissive for infection establishment.
Collapse
|
44
|
Hewitson JP, Harcus Y, Murray J, van Agtmaal M, Filbey KJ, Grainger JR, Bridgett S, Blaxter ML, Ashton PD, Ashford D, Curwen RS, Wilson RA, Dowle AA, Maizels RM. Proteomic analysis of secretory products from the model gastrointestinal nematode Heligmosomoides polygyrus reveals dominance of venom allergen-like (VAL) proteins. J Proteomics 2011; 74:1573-94. [PMID: 21722761 PMCID: PMC4794625 DOI: 10.1016/j.jprot.2011.06.002] [Citation(s) in RCA: 124] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2011] [Revised: 05/20/2011] [Accepted: 06/05/2011] [Indexed: 01/25/2023]
Abstract
The intestinal helminth parasite, Heligmosomoides polygyrus bakeri offers a tractable experimental model for human hookworm infections such as Ancylostoma duodenale and veterinary parasites such as Haemonchus contortus. Parasite excretory-secretory (ES) products represent the major focus for immunological and biochemical analyses, and contain immunomodulatory molecules responsible for nematode immune evasion. In a proteomic analysis of adult H. polygyrus secretions (termed HES) matched to an extensive transcriptomic dataset, we identified 374 HES proteins by LC-MS/MS, which were distinct from those in somatic extract HEx, comprising 446 identified proteins, confirming selective export of ES proteins. The predominant secreted protein families were proteases (astacins and other metalloproteases, aspartic, cysteine and serine-type proteases), lysozymes, apyrases and acetylcholinesterases. The most abundant products were members of the highly divergent venom allergen-like (VAL) family, related to Ancylostoma secreted protein (ASP); 25 homologues were identified, with VAL-1 and -2 also shown to be associated with the parasite surface. The dominance of VAL proteins is similar to profiles reported for Ancylostoma and Haemonchus ES products. Overall, this study shows that the secretions of H. polygyrus closely parallel those of clinically important GI nematodes, confirming the value of this parasite as a model of helminth infection.
Collapse
Affiliation(s)
- James P. Hewitson
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JT, UK
| | - Yvonne Harcus
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JT, UK
| | - Janice Murray
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JT, UK
| | - Maaike van Agtmaal
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JT, UK
| | - Kara J. Filbey
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JT, UK
| | | | | | | | | | | | | | | | | | - Rick M. Maizels
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh, EH9 3JT, UK
| |
Collapse
|
45
|
Donskow K, Drela N, Doligalska M. Heligmosomoides bakeri antigen rescues CD4-positive T cells from glucocorticoid-induced apoptosis by Bcl-2 protein expression. Parasite Immunol 2011; 33:158-69. [PMID: 21306399 DOI: 10.1111/j.1365-3024.2010.01262.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Heligmosomoides bakeri infection in mice is associated with a dominant CD4(+) T-cell response and with the activity of natural Treg cells with CD4(+) CD25(+) phenotype. The polarization of Th2 T-cell phenotype and the increase in the CD4(+) CD25(+) T cell population are regulated by glucocorticoids that induce apoptosis in CD4(+) CD25(-) T cells and inhibit apoptosis in CD4(+) CD25(+) T cells. However, exposure of mice to H. bakeri antigen induces a high glucocorticoid concentration in serum and a reduction in the number of CD4-positive; CD4(+) CD25(-) and CD4(+) CD25(+) apoptotic T cells in mesenteric lymph node cells. In this study to evaluate the in vitro effect of the anti-apoptotic property of H. bakeri antigen on T cells, apoptosis of these cells was induced by glucocorticoids-dexamethasone (Dex). Excretory-secretory (ES) antigen of the nematode prevented Dex-induced apoptosis in CD4-positive T cells with CD4(+) CD25(-) and CD4(+) CD25(High) phenotype by Bcl-2 protein expression. Contrary to the effect on CD4-positive T cells, survival of CD8(+) T cells was not connected with expression of Bcl-2 protein. This suggest that H. bakeri antigen modulates CD4-positive T cell sensitivity to glucocorticoid-induced apoptosis by induction of Bcl-2 protein.
Collapse
Affiliation(s)
- K Donskow
- Department of Parasitology, University of Warsaw, Warsaw, Poland.
| | | | | |
Collapse
|
46
|
Abstract
Infectious agents have intimately co-evolved with the host immune system, acquiring a portfolio of highly sophisticated mechanisms to modulate immunity. Among the common strategies developed by viruses, bacteria, protozoa, helminths, and fungi is the manipulation of the regulatory T cell network in order to favor pathogen survival and transmission. Treg activity also benefits the host in many circumstances by controlling immunopathogenic reactions to infection. Interestingly, some pathogens are able to directly induce the conversion of naive T cells into suppressive Foxp3-expressing Tregs, while others activate pre-existing natural Tregs, in both cases repressing pathogen-specific effector responses. However, Tregs can also act to promote immunity in certain settings, such as in initial stages of infection when effector cells must access the site of infection, and subsequently in ensuring generation of effector memory. Notably, there is little current information on whether infections selectively drive pathogen-specific Tregs, and if so whether these cells are also reactive to self-antigens. Further analysis of specificity, together with a clearer picture of the relative dynamics of Treg subsets over the course of disease, should lead to rational strategies for immune intervention to optimize immunity and eliminate infection.
Collapse
|
47
|
Grainger JR, Smith KA, Hewitson JP, McSorley HJ, Harcus Y, Filbey KJ, Finney CAM, Greenwood EJD, Knox DP, Wilson MS, Belkaid Y, Rudensky AY, Maizels RM. Helminth secretions induce de novo T cell Foxp3 expression and regulatory function through the TGF-β pathway. ACTA ACUST UNITED AC 2010; 207:2331-41. [PMID: 20876311 PMCID: PMC2964568 DOI: 10.1084/jem.20101074] [Citation(s) in RCA: 386] [Impact Index Per Article: 25.7] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Foxp3-expressing regulatory T (T reg) cells have been implicated in parasite-driven inhibition of host immunity during chronic infection. We addressed whether parasites can directly induce T reg cells. Foxp3 expression was stimulated in naive Foxp3⁻ T cells in mice infected with the intestinal helminth Heligmosomoides polygyrus. In vitro, parasite-secreted proteins (termed H. polygyrus excretory-secretory antigen [HES]) induced de novo Foxp3 expression in fluorescence-sorted Foxp3⁻ splenocytes from Foxp3-green fluorescent protein reporter mice. HES-induced T reg cells suppressed both in vitro effector cell proliferation and in vivo allergic airway inflammation. HES ligated the transforming growth factor (TGF) β receptor and promoted Smad2/3 phosphorylation. Foxp3 induction by HES was lost in dominant-negative TGF-βRII cells and was abolished by the TGF-β signaling inhibitor SB431542. This inhibitor also reduced worm burdens in H. polygyrus-infected mice. HES induced IL-17 in the presence of IL-6 but did not promote Th1 or Th2 development under any conditions. Importantly, antibody to mammalian TGF-β did not recognize HES, whereas antisera that inhibited HES did not affect TGF-β. Foxp3 was also induced by secreted products of Teladorsagia circumcincta, a related nematode which is widespread in ruminant animals. We have therefore identified a novel pathway through which helminth parasites may stimulate T reg cells, which is likely to be a key part of the parasite's immunological relationship with the host.
Collapse
Affiliation(s)
- John R Grainger
- Institute of Immunology and Infection Research, University of Edinburgh, Edinburgh EH9 3JT, Scotland, UK
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Hang L, Setiawan T, Blum AM, Urban J, Stoyanoff K, Arihiro S, Reinecker HC, Weinstock JV. Heligmosomoides polygyrus infection can inhibit colitis through direct interaction with innate immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2010; 185:3184-9. [PMID: 20702728 PMCID: PMC2948844 DOI: 10.4049/jimmunol.1000941] [Citation(s) in RCA: 71] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Less developed countries have a low incidence of immunological diseases like inflammatory bowel disease (IBD), perhaps prevented by the high prevalence of helminth infections in their populations. In the Rag IL-10(-/-) T cell transfer model of colitis, Heligmosomoides polygyrus, an intestinal helminth, prevents and reverses intestinal inflammation. This model of colitis was used to explore the importance of innate immunity in H. polygyrus protection from IBD. Rag mice briefly exposed to H. polygyrus before reconstitution with IL-10(-/-) colitogenic T cells are protected from colitis. Exposure to H. polygyrus before introduction of IL-10(-/-) and OT2 T cells reduced the capacity of the intestinal mucosa to make IFN-gamma and IL-17 after either anti-CD3 mAb or OVA stimulation. This depressed cytokine response was evident even in the absence of colitis, suggesting that the downmodulation in proinflammatory cytokine secretion was not just secondary to improvement in intestinal inflammation. Following H. polygyrus infection, dendritic cells (DCs) from the lamina propria of Rag mice displayed decreased expression of CD80 and CD86, and heightened expression of plasmacytoid dendritic cell Ag-1 and CD40. They were also less responsive to lamina proprias, producing less IL-12p40 and IL-10. Also diminished was their capacity to present OVA to OT2 T cells. These experiments infer that H. polygyrus does not require direct interactions with T or B cells to render animals resistant to colitis. DCs have an important role in driving both murine and human IBD. Data suggest that phenotypic alternations in mucosal DC function are part of the regulatory process.
Collapse
Affiliation(s)
- Long Hang
- Division of Gastroenterology-Hepatology, Department of Internal Medicine, Tufts Medical Center, Boston, MA 02111, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Cruz-Chan JV, Rosado-Vallado M, Dumonteil E. Malaria vaccine efficacy: overcoming the helminth hurdle. Expert Rev Vaccines 2010; 9:707-11. [PMID: 20624043 DOI: 10.1586/erv.10.63] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Several studies have documented that helminth infections can interfere with the development of the immune response of vaccines against different diseases, although some results have been contradictory. The mechanisms involved in the inhibition of the immune response to vaccination by helminth are still unclear, and murine models of helminth-malaria coinfections have proven helpful in investigating some aspects of the interactions involved. The study evaluated here focuses on the effect of helminth infection in mice on the immunogenicity and protective efficacy of two distinct malaria vaccine candidates, a transmission-blocking DNA vaccine based on Pfs25 antigen and a pre-erythrocytic vaccine based on irradiated sporozoites. Interestingly, the authors found that helminth infection dramatically reduced DNA vaccine immunogenicity, while immunization with irradiated sporozoites was able to induce a high level of antibodies and protection, independently of helminth infection. Immune suppression by helminth infection affected all IgG isotypes, suggesting no particular polarization of the immune response, but the generation of memory B cells was not affected. It will be of key interest to understand the mechanisms underlying the efficacy of the sporozoite vaccine, and its ability to overcome helminth immunosuppression, as this may help in the design of more effective vaccines.
Collapse
Affiliation(s)
- Julio Vladimir Cruz-Chan
- Laboratorio de Parasitología, Centro de Investigaciones Regionales 'Dr. Hideyo Noguchi', Universidad Autónoma de Yucatán, Ave. Itzaes #490 x 59, 97000 Mérida, Yucatan, Mexico
| | | | | |
Collapse
|
50
|
Rausch S, Held J, Stange J, Lendner M, Hepworth MR, Klotz C, Lucius R, Pogonka T, Hartmann S. A matter of timing: Early, not chronic phase intestinal nematode infection restrains control of a concurrent enteric protozoan infection. Eur J Immunol 2010; 40:2804-15. [DOI: 10.1002/eji.201040306] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|