1
|
Uthailak N, Adisakwattana P, Thiangtrongjit T, Limpanont Y, Chusongsang P, Chusongsang Y, Tanasarnprasert K, Reamtong O. Discovery of Schistosoma mekongi circulating proteins and antigens in infected mouse sera. PLoS One 2022; 17:e0275992. [PMID: 36227939 PMCID: PMC9562170 DOI: 10.1371/journal.pone.0275992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Accepted: 09/27/2022] [Indexed: 11/24/2022] Open
Abstract
Schistosomiasis is a neglected tropical disease caused by an infection of the parasitic flatworms schistosomes. Schistosoma mekongi is a restricted Schistosoma species found near the Mekong River, mainly in southern Laos and northern Cambodia. Because there is no vaccine or effective early diagnosis available for S. mekongi, additional biomarkers are required. In this study, serum biomarkers associated with S. mekongi-infected mice were identified at 14-, 28-, 42-, and 56-days post-infection. Circulating proteins and antigens of S. mekongi in mouse sera were analyzed using mass spectrometry-based proteomics. Serine protease inhibitors and macrophage erythroblast attacher were down-regulated in mouse sera at all infection timepoints. In addition, 54 circulating proteins and 55 antigens of S. mekongi were identified. Notable circulating proteins included kyphoscoliosis peptidase and putative tuberin, and antigens were detected at all four infection timepoints, particularly in the early stages (12 days). The putative tuberin sequence of S. mekongi was highly similar to homologs found in other members of the genus Schistosoma and less similar to human and murine sequences. Our study provided the identity of promising diagnostic biomarkers that could be applicable in early schistosomiasis diagnosis and vaccine development.
Collapse
Affiliation(s)
- Naphatsamon Uthailak
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Poom Adisakwattana
- Department of Helminthology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Tipparat Thiangtrongjit
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yanin Limpanont
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Phiraphol Chusongsang
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Yupa Chusongsang
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Kanthi Tanasarnprasert
- Department of Social and Environmental Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- * E-mail:
| |
Collapse
|
2
|
Bayani M, Kalantari N, Esmailzadeh S, Ghaffari S, Mahjoub S, Ghofrani F, Ghaffari T. An evaluation of the level of testosterone, DHEA and prolactin among Toxoplasma gondii infected and uninfected infertile couples attending to Fatima Al-Zahra infertility treatment center, Babol, Northern Iran. ACTA FACULTATIS MEDICAE NAISSENSIS 2022. [DOI: 10.5937/afmnai39-31982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022] Open
Abstract
Introduction/Aims: Toxoplasmosis modifies various hormones and cytokines in the infected hosts which may result in several disorders. This study was conducted to assess testosterone, DHEA, and prolactin concentration levels among Toxoplasma gondii infected and uninfected infertile couples. Methods: Blood samples were collected and sera were separated. The sera were analyzed for the detection of anti-Toxoplasma (IgG& IgM) antibodies using commercial ELISA kits. The level of DHEA was measured by ELISA and the levels of testosterone and prolactin were evaluated by enzyme-linked fluorescent assay (ELFA, VIDAS). Results: The overall seroprevalence of toxoplasmosis was 58.0% (218/376). Among women, 56.9% (107/188) and 6.5% (7/107) were positive for anti-T. gondii antibodies IgG and IgM, respectively. The IgG and IgM were detected in the sera of 111/188 (59.0%) and 9/111 (8.1%) in male subjects, respectively. A positive association was observed between T. gondii infection and the upper and lower ranges of the normal value of testosterone in males (x 2 = 6.8, p = 0.033) but not in females (x 2 = 0.62, p = 0.99). A positive correlation was seen between toxoplasmosis and the upper and lower ranges of the normal value of prolactin in females (x 2 = 6.5, p = 0.039) but not in male cases (x 2 = 1.06, p = 0.59). Conclusion: Our results demonstrated no statistically significant differences between the level of sexual hormones between Toxoplasma-infected individuals and Toxoplasma-free subjects suffering from infertility. These findings suggest that further studies should be performed on infertile cases with large sample sizes in a case-control format.
Collapse
|
3
|
Immunosuppression in Malaria: Do Plasmodium falciparum Parasites Hijack the Host? Pathogens 2021; 10:pathogens10101277. [PMID: 34684226 PMCID: PMC8536967 DOI: 10.3390/pathogens10101277] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Malaria reflects not only a state of immune activation, but also a state of general immune defect or immunosuppression, of complex etiology that can last longer than the actual episode. Inhabitants of malaria-endemic regions with lifelong exposure to the parasite show an exhausted or immune regulatory profile compared to non- or minimally exposed subjects. Several studies and experiments to identify and characterize the cause of this malaria-related immunosuppression have shown that malaria suppresses humoral and cellular responses to both homologous (Plasmodium) and heterologous antigens (e.g., vaccines). However, neither the underlying mechanisms nor the relative involvement of different types of immune cells in immunosuppression during malaria is well understood. Moreover, the implication of the parasite during the different stages of the modulation of immunity has not been addressed in detail. There is growing evidence of a role of immune regulators and cellular components in malaria that may lead to immunosuppression that needs further research. In this review, we summarize the current evidence on how malaria parasites may directly and indirectly induce immunosuppression and investigate the potential role of specific cell types, effector molecules and other immunoregulatory factors.
Collapse
|
4
|
Jeong MJ, Kang SA, Choi JH, Lee DI, Yu HS. Extracellular vesicles of Echinococcus granulosus have therapeutic effects in allergic airway inflammation. Parasite Immunol 2021; 43:e12872. [PMID: 34174101 DOI: 10.1111/pim.12872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 06/02/2021] [Accepted: 06/18/2021] [Indexed: 11/30/2022]
Abstract
BACKGROUND Previous studies have shown that Echinococcus granulosus cystic fluid can alleviate Th2 allergic airway inflammatory responses by increasing the number of CD4+ CD25+ Foxp3+ T (regulatory T; Treg) cells. Parasite-derived extracellular vesicles (EV) are known to not only promote parasite infection by communicating between parasites but also regulate the inflammatory response by acting as an immunomodulatory agent in the host. METHODS To evaluate the effect of EV extracted from the cystic fluid of E. granulosus on allergic airway inflammation, gene expression was investigated after administering EV to mouse lung epithelial cells (MLE-12) following 2 h of pretreatment with Aspergillus proteins. An allergic airway inflammation animal model was used to investigate the regulation of the inflammatory response by EV and induced with ovalbumin. RESULTS EV treatment significantly reduced airway resistance and the number of eosinophils and other immune cells in the bronchoalveolar lavage fluid and Th2- and Th17-related cytokine levels. EV pretreatment decreased the number of IL-4+ CD4+ T cells and increased the number of Treg cells in the lung-draining lymph nodes and spleen. CONCLUSIONS Echinococcus granulosus cystic fluid derived EV ameliorated Th2 allergic airway inflammatory through Treg cells, similar to whole cystic fluid treatment. Thus, EV may be important immunomodulatory molecules in cystic fluid.
Collapse
Affiliation(s)
- Mi Jin Jeong
- Department of Parasitology and Tropical Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Shin Ae Kang
- Department of Parasitology and Tropical Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Jun Ho Choi
- Department of Parasitology and Tropical Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Da In Lee
- Department of Parasitology and Tropical Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea
| | - Hak Sun Yu
- Department of Parasitology and Tropical Medicine, School of Medicine, Pusan National University, Yangsan, Republic of Korea.,Research Institute for Convergence of Biomedical Science and Technology, Pusan National University Yangsan Hospital, Yangsan, Republic of Korea
| |
Collapse
|
5
|
Sibomana JP, Campeche A, Carvalho-Filho RJ, Correa RA, Duani H, Pacheco Guimaraes V, Hilton JF, Kassa B, Kumar R, Lee MH, Loureiro CMC, Mazimba S, Mickael C, Oliveira RKF, Ota-Arakaki JS, Rezende CF, Silva LCS, Sinkala E, Ahmed HY, Graham BB. Schistosomiasis Pulmonary Arterial Hypertension. Front Immunol 2020; 11:608883. [PMID: 33362796 PMCID: PMC7758287 DOI: 10.3389/fimmu.2020.608883] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/03/2020] [Indexed: 12/18/2022] Open
Abstract
Pulmonary arterial hypertension (PAH) is a disease of the lung blood vessels that results in right heart failure. PAH is thought to occur in about 5% to 10% of patients with hepatosplenic schistosomiasis, particularly due to S. mansoni. The lung blood vessel injury may result from a combination of embolization of eggs through portocaval shunts into the lungs causing localized Type 2 inflammatory response and vessel remodeling, triggering of autonomous pathology that becomes independent of the antigen, and high cardiac output as seen in portopulmonary hypertension. The condition is likely underdiagnosed as there is little systematic screening, and risk factors for developing PAH are not known. Screening is done by echocardiography, and formal diagnosis requires invasive right heart catheterization. Patients with Schistosoma-associated PAH show reduced functional capacity and can be treated with pulmonary vasodilators, which improves symptoms and may improve survival. There are animal models of this disease that might help in understanding disease pathogenesis and identify novel targets to screen and treatment. Pathogenic mechanisms include Type 2 immunity and activation and signaling in the TGF-β pathway. There are still major uncertainties regarding Schistosoma-associated PAH development, course and treatment.
Collapse
Affiliation(s)
- Jean Pierre Sibomana
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Tikur Anbessa Specialized Hospital, College of Health Sciences, University of Addis Ababa, Addis Ababa, Ethiopia
- Department of Medicine, Butare University Teaching Hospital, College of Medicine and Health Sciences, University of Rwanda, Kigali, Rwanda
| | - Aloma Campeche
- Division of Gastroenterology, Department of Medicine, Santa Casa Hospital, Salvador, Bahia, Brazil
| | - Roberto J. Carvalho-Filho
- Division of Gastroenterology, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Ricardo Amorim Correa
- Internal Medicine/Pulmonary Division, Medical School, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Helena Duani
- Internal Medicine/Infectious Diseases Division, Medical School, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Virginia Pacheco Guimaraes
- Pulmonary Department, Hospital Júlia Kubistchek, Fundação Hospitalar of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Joan F. Hilton
- Department of Epidemiology and Biostatistics, University of California San Francisco, San Francisco, CA, United States
| | - Biruk Kassa
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- Department of Medicine, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Rahul Kumar
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- Department of Medicine, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | - Michael H. Lee
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- Department of Medicine, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| | | | - Sula Mazimba
- Division of Cardiology, Department of Medicine, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Claudia Mickael
- Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, United States
| | - Rudolf K. F. Oliveira
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Jaquelina S. Ota-Arakaki
- Division of Respiratory Diseases, Department of Medicine, Federal University of São Paulo, São Paulo, Brazil
| | - Camila Farnese Rezende
- Pulmonary Medicine, Hospital das Clinicas, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Luciana C. S. Silva
- Internal Medicine Department, Medical School, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Edford Sinkala
- Hepatology Clinic, Department of Medicine, University of Zambia Teaching Hospital, Lusaka, Zambia
| | - Hanan Yusuf Ahmed
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Tikur Anbessa Specialized Hospital, College of Health Sciences, University of Addis Ababa, Addis Ababa, Ethiopia
| | - Brian B. Graham
- Department of Medicine, University of California San Francisco, San Francisco, CA, United States
- Department of Medicine, Zuckerberg San Francisco General Hospital, San Francisco, CA, United States
| |
Collapse
|
6
|
Qian L, Liu Y, Wang S, Gong W, Jia X, Liu L, Ye F, Ding J, Xu Y, Fu Y, Tian F. NKG2D ligand RAE1ε induces generation and enhances the inhibitor function of myeloid-derived suppressor cells in mice. J Cell Mol Med 2017; 21:2046-2054. [PMID: 28276625 PMCID: PMC5571551 DOI: 10.1111/jcmm.13124] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2016] [Accepted: 01/13/2017] [Indexed: 01/06/2023] Open
Abstract
Expression of surface NKG2D ligands on tumour cells, which activates nature killer (NK) cells and CD8+ T cells, is crucial in antitumour immunity. Some types of tumours have evolved mechanisms to suppress NKG2D‐mediated immune cell activation, such as tumour‐derived soluble NKG2D ligands or sustained NKG2D ligands produced by tumours down‐regulate the expression of NKG2D on NK cells and CD8+ T cells. Here, we report that surface NKG2D ligand RAE1ε on tumour cells induces CD11b+Gr‐1+ myeloid‐derived suppressor cell (MDSC) via NKG2D in vitro and in vivo. MDSCs induced by RAE1ε display a robust induction of IL‐10 and arginase, and these MDSCs show greater suppressive activity by inhibiting antigen‐non‐specific CD8+ T‐cell proliferation. Consistently, upon adoptive transfer, MDSCs induced by RAE1ε significantly promote CT26 tumour growth in IL‐10‐ and arginase‐dependent manners. RAE1ε moves cytokine balance towards Th2 but not Th1 in vivo. Furthermore, RAE1ε enhances inhibitory function of CT26‐derived MDSCs and promotes IL‐4 rather than IFN‐γ production from CT26‐derived MDSCs through NKG2D in vitro. Our study has demonstrated a novel mechanism for NKG2D ligand+ tumour cells escaping from immunosurveillance by facilitating the proliferation and the inhibitory function of MDSCs.
Collapse
Affiliation(s)
- Li Qian
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China.,Translational Medicine Research Institute of Yangzhou University, Yangzhou, China.,Jiangsu Key Laboratory of Zoonosis/Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou, China
| | - Yang Liu
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China.,Translational Medicine Research Institute of Yangzhou University, Yangzhou, China
| | - Shaoqing Wang
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China.,Translational Medicine Research Institute of Yangzhou University, Yangzhou, China
| | - Weijuan Gong
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | - Xiaoqin Jia
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | - Lu Liu
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | - Feng Ye
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | - Jingjuan Ding
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | - Yuwei Xu
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | - Yi Fu
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China
| | - Fang Tian
- Department of Immunology, School of Medicine, Yangzhou University, Yangzhou, China.,Translational Medicine Research Institute of Yangzhou University, Yangzhou, China
| |
Collapse
|
7
|
Ke XD, Shen S, Song LJ, Yu CX, Kikuchi M, Hirayama K, Gao H, Wang J, Yin X, Yao Y, Liu Q, Zhou W. Characterization of Schistosoma japonicum CP1412 protein as a novel member of the ribonuclease T2 molecule family with immune regulatory function. Parasit Vectors 2017; 10:89. [PMID: 28212670 PMCID: PMC5316207 DOI: 10.1186/s13071-016-1962-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Accepted: 12/29/2016] [Indexed: 01/05/2023] Open
Abstract
Background Schistosome infection typically induces a polarized Th2 type host immune response. As egg antigen molecules play key roles in this immunoregulatory process, clarifying their functions in schistosomiasis would facilitate the development of vaccine and immunotherapeutic methods. Schistosoma japonicum (Sj) CP1412 (GenBank: AY57074.1) has been identified as a new member of the RNase T2 family with immune regulatory functions. Methods The expression plasmid Sj CP1412-pET28a was constructed and transformed into bacteria for production of recombinant Sj CP1412 protein (rSj CP1412) via IPTG induction. The RNase activity of Sj CP1412 was predicted by bioinformatic analysis and confirmed by digesting the yeast tRNA with rSj CP1412.C57BL/6j mice were immunized with rSj CP1412, and its immune regulatory effects in vivo and in vitro were investigated. Meanwhile, the relationship between the RNase activity of Sj CP1412 and its immune regulation was observed. Results Sj CP1412 was confirmed as a novel RNase T2 family protein with RNase activity. Immunoblotting and RT-PCR analyses demonstrated Sj CP1412 as a protein exclusively secreted/excreted from eggs, but not cercariae and adult worms. Stimulating RAW264.7 macrophages with rSj CP1412 raised the expression of CD206, Arg-1 and IL-10, which are related to M2 type macrophage differentiation. Stimulating dendritic cells (DCs) with rSjCP1412 failed to induce their maturation, and the recombinant protein also inhibited LPS-stimulated DC maturation. Depletion of Sj CP1412 from soluble egg antigen (SEA) impaired the ability of SEA to induce M2 type polarization of RAW264.7 macrophages. Immunizing mice with rSj CP1412 induced high antibody titers, increased serum IL-4 and TGF-β levels and splenic CD4 + CD25 + Foxp3 + T cells, downregulated serum IFN-γ levels and alleviated the egg granuloma pathology of schistosome infection. In vitro stimulation by rSj CP1412 significantly increased CD4 + CD25 + Foxp3 + T cell numbers in splenocytes of healthy mice. The rSj CP1412 protein with RNase activity inactivated by DEPC failed to induce M2 surface marker CD206 expression in RAW264.7 macrophages. Conclusions The Sj CP1412 protein expressed specifically in S. japonicum eggs is a novel member of the RNase T2 family. Similar to Omega-1 of Schistosoma mansoni, the Sj CP1412 protein drives polarization of the host Th2 immune response, which is dependent on its RNase activity. These data provide new evidence towards understanding the immune regulatory role of RNase T2 family proteins during schistosome infection. Electronic supplementary material The online version of this article (doi:10.1186/s13071-016-1962-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Xue-Dan Ke
- Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health; Jiangsu Provincial Key Laboratory on Technology for Parasite and Vector Control, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Shuang Shen
- Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health; Jiangsu Provincial Key Laboratory on Technology for Parasite and Vector Control, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China.,Medical College, Jiangnan University, Wuxi, 214122, China.,Public Health Research Center, Jiangnan University, Wuxi, 214122, People's Republic of China.,Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, People's Republic of China
| | - Li-Jun Song
- Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health; Jiangsu Provincial Key Laboratory on Technology for Parasite and Vector Control, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Chuan-Xin Yu
- Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health; Jiangsu Provincial Key Laboratory on Technology for Parasite and Vector Control, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China. .,Medical College, Jiangnan University, Wuxi, 214122, China. .,Public Health Research Center, Jiangnan University, Wuxi, 214122, People's Republic of China.
| | - Mihoko Kikuchi
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Kenji Hirayama
- Department of Immunogenetics, Institute of Tropical Medicine (NEKKEN), Nagasaki University, 1-12-4 Sakamoto, Nagasaki, 852-8523, Japan
| | - Hong Gao
- Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health; Jiangsu Provincial Key Laboratory on Technology for Parasite and Vector Control, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China.,Department of Pathology, Nanjing Drum Tower Hospital, Nanjing, 210003, People's Republic of China
| | - Jie Wang
- Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health; Jiangsu Provincial Key Laboratory on Technology for Parasite and Vector Control, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Xuren Yin
- Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health; Jiangsu Provincial Key Laboratory on Technology for Parasite and Vector Control, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Yuan Yao
- Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health; Jiangsu Provincial Key Laboratory on Technology for Parasite and Vector Control, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Qian Liu
- Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health; Jiangsu Provincial Key Laboratory on Technology for Parasite and Vector Control, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| | - Wei Zhou
- Key Laboratory on Technology for Parasitic Disease Prevention and Control, Ministry of Health; Jiangsu Provincial Key Laboratory on Technology for Parasite and Vector Control, Jiangsu Institute of Parasitic Diseases, Wuxi, 214064, People's Republic of China
| |
Collapse
|
8
|
Stempin CC, Rojas Marquez JD, Ana Y, Cerban FM. GRAIL and Otubain-1 are Related to T Cell Hyporesponsiveness during Trypanosoma cruzi Infection. PLoS Negl Trop Dis 2017; 11:e0005307. [PMID: 28114324 PMCID: PMC5289611 DOI: 10.1371/journal.pntd.0005307] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Revised: 02/02/2017] [Accepted: 01/06/2017] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Trypanosoma cruzi infection is associated with severe T cell unresponsiveness to antigens and mitogens and is characterized by decreased IL-2 synthesis. In addition, the acquisition of the anergic phenotype is correlated with upregulation of "gene related to anergy in lymphocytes" (GRAIL) protein in CD4 T cells. We therefore sought to examine the role of GRAIL in CD4 T cell proliferation during T. cruzi infection. METHODOLOGY/PRINCIPAL FINDINGS Balb/c mice were infected intraperitoneally with 500 blood-derived trypomastigotes of Tulahuen strain, and spleen cells from control non-infected or infected animals were obtained. CD4 T cell proliferation was assessed by CFSE staining, and the expression of GRAIL in splenic T cells was measured by real-time PCR, flow cytometry and Western blot. We found increased GRAIL expression at the early stages of infection, coinciding with the peak of parasitemia, with these findings correlating with impaired proliferation and poor IL-2 and IFN-γ secretion in response to plate-bound antibodies. In addition, we showed that the expression of GRAIL E3-ubiquitin ligase in CD4 T cells during the acute phase of infection was complemented by a high expression of inhibitory receptors such as PD-1 and CTLA-4. We demonstrated that GRAIL expression during infection was modulated by the mammalian target of the rapamycin (mTOR) pathway, since addition of IL-2 or CTLA-4 blockade in splenocytes from mice 21 days post infection led to a reduction in GRAIL expression. Furthermore, addition of IL-2 was able to activate the mTOR pathway, inducing Otubain-1 expression, which mediated GRAIL degradation and improved T cell proliferation. CONCLUSIONS We hypothesize that GRAIL expression induced by the parasite may be maintained by the increased expression of inhibitory molecules, which blocked mTOR activation and IL-2 secretion. Consequently, the GRAIL regulator Otubain-1 was not expressed and GRAIL maintained the brake on T cell proliferation. Our findings reveal a novel association between increased GRAIL expression and impaired CD4 T cell proliferation during Trypanosoma cruzi infection.
Collapse
Affiliation(s)
- Cinthia C. Stempin
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Medina Allende y Haya de la Torre, Ciudad Universitaria, Córdoba, Argentina
| | - Jorge D. Rojas Marquez
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Medina Allende y Haya de la Torre, Ciudad Universitaria, Córdoba, Argentina
| | - Yamile Ana
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Medina Allende y Haya de la Torre, Ciudad Universitaria, Córdoba, Argentina
| | - Fabio M. Cerban
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), CONICET, Departamento de Bioquímica Clínica, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Medina Allende y Haya de la Torre, Ciudad Universitaria, Córdoba, Argentina
- * E-mail:
| |
Collapse
|
9
|
Cabanelas E, López CM, Díaz P, Pérez-Creo A, Morrondo P, Díez-Baños P, Panadero R. Modulatory effects of interferon-γ and interleukin-4 on cellular immune responses against Hypoderma lineatum antigens. MEDICAL AND VETERINARY ENTOMOLOGY 2016; 30:439-443. [PMID: 27611850 DOI: 10.1111/mve.12190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2016] [Revised: 05/21/2016] [Accepted: 05/23/2016] [Indexed: 06/06/2023]
Abstract
This study investigates the in vitro modulatory effects of interferon-γ (IFN-γ) and interleukin-4 (IL-4) on both proliferative bovine T cell responses and IL-10 production induced by different antigens [crude larval extract and the purified fractions hypodermin A, B and C (HyA, HyB, HyC)] obtained from first instars of Hypoderma lineatum (Diptera: Oestridae), alone or in the presence of the mitogen concanavalin A. Incubation with the different parasitic antigens resulted in significant inhibition of T cell proliferation and IL-10 production, which, in general, did not revert after the addition of IFN-γ and IL-4. In the absence of antigens, IL-4 induced significant inhibition of mitogen-induced T cell responses. Exogenous IFN-γ exhibited an inhibitory effect on cell proliferation in the presence of the purified fractions HyB and HyC. These in vitro data suggest that far from neutralizing the effects of larval antigens, the addition of IFN-γ potentiates their anti-proliferative activity; by contrast, IL-4 had no consistent effects on proliferative responses to Hypoderma. IL-4 provoked an increment of IL-10 levels in supernatants of HyB-stimulated cells. In conclusion, exogenous IFN-γ and IL-4 were unable to counteract the suppressor effects of H. lineatum antigens.
Collapse
Affiliation(s)
- E Cabanelas
- Department of Animal Pathology, Sanidad Animal (INVESAGA Group), Faculty of Veterinary Science, University of Santiago de Compostela, Lugo, Spain
| | - C M López
- Department of Animal Pathology, Sanidad Animal (INVESAGA Group), Faculty of Veterinary Science, University of Santiago de Compostela, Lugo, Spain
| | - P Díaz
- Department of Animal Pathology, Sanidad Animal (INVESAGA Group), Faculty of Veterinary Science, University of Santiago de Compostela, Lugo, Spain
| | - A Pérez-Creo
- Department of Animal Pathology, Sanidad Animal (INVESAGA Group), Faculty of Veterinary Science, University of Santiago de Compostela, Lugo, Spain
| | - P Morrondo
- Department of Animal Pathology, Sanidad Animal (INVESAGA Group), Faculty of Veterinary Science, University of Santiago de Compostela, Lugo, Spain
| | - P Díez-Baños
- Department of Animal Pathology, Sanidad Animal (INVESAGA Group), Faculty of Veterinary Science, University of Santiago de Compostela, Lugo, Spain
| | - R Panadero
- Department of Animal Pathology, Sanidad Animal (INVESAGA Group), Faculty of Veterinary Science, University of Santiago de Compostela, Lugo, Spain.
| |
Collapse
|
10
|
Sodji Q, Patil V, Jain S, Kornacki JR, Mrksich M, Tekwani BL, Oyelere AK. The antileishmanial activity of isoforms 6- and 8-selective histone deacetylase inhibitors. Bioorg Med Chem Lett 2014; 24:4826-30. [PMID: 25240614 PMCID: PMC4225773 DOI: 10.1016/j.bmcl.2014.08.060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Accepted: 08/27/2014] [Indexed: 11/23/2022]
Abstract
Histone deacetylase inhibitors (HDACi) pleiotropy is largely due to their nonselective inhibition of various cellular HDAC isoforms. Connecting inhibition of a specific isoform to biological responses and/or phenotypes is essential toward deconvoluting HDACi pleiotropy. The contribution of classes I and II HDACs to the antileishmanial activity of HDACi was investigated using the amastigote and promastigote forms of Leishmania donovani. We observed that the antileishmanial activities of HDACi are largely due to the inhibition of HDAC6-like activity. This observation could facilitate the development of HDACi as antileishmanial agents.
Collapse
Affiliation(s)
- Quaovi Sodji
- School of Chemistry and Biochemistry, Parker H. Petit for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Vishal Patil
- School of Chemistry and Biochemistry, Parker H. Petit for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA
| | - Surendra Jain
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS 38677-1848, USA
| | - James R Kornacki
- Department of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Rd, Evanston, IL 60208-3113, USA
| | - Milan Mrksich
- Department of Chemistry and Biomedical Engineering, Northwestern University, 2145 Sheridan Rd, Evanston, IL 60208-3113, USA
| | - Babu L Tekwani
- National Center for Natural Products Research, School of Pharmacy, University of Mississippi, University, MS 38677-1848, USA.
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry, Parker H. Petit for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332-0400, USA.
| |
Collapse
|
11
|
Dzitko K, Dziadek B, Gatkowska J, Długońska H. Toxoplasma gondii binds sheep prolactin. Exp Parasitol 2013; 134:216-9. [PMID: 23499881 DOI: 10.1016/j.exppara.2013.02.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2012] [Revised: 10/05/2012] [Accepted: 02/19/2013] [Indexed: 01/08/2023]
Abstract
Taking into account the literature reports on the involvement of prolactin (PRL) in the regulation of immunity against Toxoplasma gondii, we decided to check whether this parasite has the ability to bind the lactotrophic hormone. We examined T. gondii binding of sheep fluoresceine- and biotine-labeled prolactin isolated from pituitary (shPRL). In this work we announced for the first time that shPRL was bound to live tachyzoites of RH (type I) and ME49 (type II) strains. Furthermore, by use of competitive inhibition analysis, we confirmed that this binding was specific for both tested T. gondii strains.
Collapse
Affiliation(s)
- K Dzitko
- Department of Immunoparasitology, Faculty of Biology and Environmental Protection, University of Łódź, ul. Banacha 12/16, 90-237 Łódź, Poland.
| | | | | | | |
Collapse
|
12
|
Trypanosoma cruzi SSP4 Amastigote Protein Induces Expression of Immunoregulatory and Immunosuppressive Molecules in Peripheral Blood Mononuclear Cells. J Trop Med 2012; 2012:829139. [PMID: 23209478 PMCID: PMC3503440 DOI: 10.1155/2012/829139] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2012] [Revised: 09/25/2012] [Accepted: 10/02/2012] [Indexed: 12/20/2022] Open
Abstract
The acute phase of Chagas' disease in mice and human is marked by states of immunosuppression, in which Trypanosoma cruzi replicates extensively and releases immunomodulatory molecules that delay parasite-specific responses mediated by effector T cells. This mechanism of evasion allows the parasite to spread in the host. Parasite molecules that regulate the host immune response during Chagas' disease have not been fully identified, particularly proteins of the amastigote stage. In this work, we evaluated the role of the GPI anchored SSP4 protein of T. cruzi as an immunomodulatory molecule in peripheral blood mononuclear cells (PBMCs). rMBP::SSP4 protein was able to stimulate nitric oxide (NO) production. Likewise, rMBP::SSP4 induced the expression of genes and production of molecules involved in the inflammatory process, such as, cytokines, chemokines, and adhesion molecules (CAMs) as determined by RT-PCR and ELISA. These results suggest that the amastigote SSP4 molecule could play a key role in the immunoregulatory and/or immunosuppressive process observed in the acute phase of infection with T. cruzi.
Collapse
|
13
|
Adalid-Peralta L, Fragoso G, Fleury A, Sciutto E. Mechanisms underlying the induction of regulatory T cells and its relevance in the adaptive immune response in parasitic infections. Int J Biol Sci 2011; 7:1412-26. [PMID: 22110392 PMCID: PMC3221948 DOI: 10.7150/ijbs.7.1412] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2011] [Accepted: 10/01/2011] [Indexed: 12/22/2022] Open
Abstract
To fulfill its function, the immune system must detect and interpret a wide variety of signals and adjust the magnitude, duration, and specific traits of each response during the complex host-parasite relationships in parasitic infections. Inflammation must be tightly regulated since uncontrolled inflammation may be as destructive as the triggering stimulus and leads to immune-mediated tissue injury. During recent years, increasing evidence points to regulatory T cells (Tregs) as key anti-inflammatory cells, critically involved in limiting the inflammatory response. Herein, we review the published information on the induction of Tregs and summarize the most recent findings on Treg generation in parasitic diseases.
Collapse
|
14
|
Ribechini E, Greifenberg V, Sandwick S, Lutz MB. Subsets, expansion and activation of myeloid-derived suppressor cells. Med Microbiol Immunol 2010; 199:273-81. [PMID: 20376485 DOI: 10.1007/s00430-010-0151-4] [Citation(s) in RCA: 126] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2010] [Indexed: 12/20/2022]
Abstract
Tumor cells and microorganisms manipulate the immune system to minimize any counter response in order to survive. Myeloid-derived suppressor cells (MDSC) in the mouse represent activated Gr-1(+) CD11b(+) myeloid precursor cells. Activation may occur through endogenous or exogenous factors leading to the suppression of immune responses. Under steady state conditions the same precursors differentiate into dendritic cells, macrophages and neutrophils. Their linkage to tumor progression and several suppression mechanisms employing the arginine metabolism are well documented, but knowledge of their role in chronic infections, autoimmune diseases and graft-versus-host reactions is just emerging. Several factors have been described to promote MDSC expansion and activation in bone marrow, spleen and tumor sites. New evidence suggests that the Gr-1 antibody itself may differentially trigger myelopoiesis under steady state conditions or induce apoptosis in inflammatory situations after binding to a common epitope expressed on Ly-6C and Ly-6G molecules, respectively. Moreover, two subsets of neutrophil- and monocyte-related MDSC have been described in tumor-bearing and healthy mice. In the present review, we summarize some early work leading to recent findings on these two MDSC subsets, the factors supporting MDSC expansion and activation, as well as novel insights on Gr-1 antibody functions.
Collapse
Affiliation(s)
- Eliana Ribechini
- Institute of Virology and Immunobiology, University of Würzburg, Versbacherstrasse 7, Würzburg, Germany
| | | | | | | |
Collapse
|