1
|
McManus CM, Maizels RM. Regulatory T cells in parasite infections: susceptibility, specificity and specialisation. Trends Parasitol 2023; 39:547-562. [PMID: 37225557 DOI: 10.1016/j.pt.2023.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 04/06/2023] [Accepted: 04/06/2023] [Indexed: 05/26/2023]
Abstract
Regulatory T cells (Tregs) are essential to control immune system responses to innocuous self-specificities, intestinal and environmental antigens. However, they may also interfere with immunity to parasites, particularly in chronic infection. Susceptibility to many parasite infections is, to a greater or lesser extent, controlled by Tregs, but often they play a more prominent role in moderating the immunopathological consequences of parasitism, and dampening bystander reactions in an antigen-nonspecific manner. More recently, Treg subtypes have been defined which may preferentially act in different contexts; we also discuss the degree to which this specialisation is now being mapped onto how Tregs maintain the delicate balance between tolerance, immunity, and pathology in infection.
Collapse
Affiliation(s)
- Caitlin M McManus
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK
| | - Rick M Maizels
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, 120 University Place, Glasgow G12 8TA, UK.
| |
Collapse
|
2
|
Chen G, Du JW, Nie Q, Du YT, Liu SC, Liu DH, Zhang HM, Wang FF. Plasmodium yoelii 17XL infection modified maturation and function of dendritic cells by skewing Tregs and amplificating Th17. BMC Infect Dis 2020; 20:266. [PMID: 32252652 PMCID: PMC7132900 DOI: 10.1186/s12879-020-04990-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2019] [Accepted: 03/25/2020] [Indexed: 11/25/2022] Open
Abstract
Background Emerging data has suggested that Tregs, Th17, Th1 and Th2 are correlated with early immune mechanisms by controlling Plasmodium infection. Plasmodium infection appeared to impair the antigen presentation and maturation of DCs, leading to attenuation of specific cellular immune response ultimately. Hence, in this study, we aim to evaluate the relevance between DCs and Tregs/Th17 populations in the process and outcomes of infection with Plasmodium yoelii 17XL (P.y17XL). Methods DCs detection/analysis dynamically was performed by Tregs depletion or Th17 neutralization in P.y17XL infected BALB/c mice via flow cytometry. Then the levels of cytokines production were detected using enzyme-linked mmunosorbent assay (ELISA). Results Our results indicated that Tregs depletion or Th17 neutralization in BALB/c mice infected with P.y17XL significantly up-regulated the percentages of mDC and pDC, increased the expressions of major histocompatibility complex (MHC) class II, CD80, CD86 on DCs and the levels of IL-10/IL-12 secreted by DCs, indicating that abnormal amplification of Tregs or Th17 may damage the maturation and function of DCs during the early stage of malaria infection. Interestingly, we also found that the abnormal amplification of Th17, as well as Tregs, could inhibit the maturation of DCs. Conclusions Tregs skewing or Th17 amplification during the early stage of malaria infection may inhibit the maturation and function of DCs by modifying the subsets of DCs, expressions of surface molecules on DCs and secretion mode of cytokines.
Collapse
Affiliation(s)
- Guang Chen
- Department of Basic Medical Sciences, Taizhou University Hospital, Taizhou University, No 1139 Shifu Road, Jiaojiang District, Taizhou, 318000, China.
| | - Ji-Wei Du
- Nursing Department, Xiang'An Hospital, Xiamen University, No 2000, Xian'an East Road, Xiang'an District, Xiamen, 361005, China
| | - Qing Nie
- Weifang Centers for Disease Control and Prevention, No 4801 Huixian Road, Gaoxin District, Shandong Province, Weifang, 261061, China
| | - Yun-Ting Du
- Department of Laboratory Medicine, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, No 44, Xiaoheyan Road, Dadong District, Shenyang, 110042, China
| | - Shuang-Chun Liu
- Municipal Hospital Affiliated to Medical School of Taizhou University, No 381, Zhongshan East Road, Jiaojiang District, Taizhou, 318000, China
| | - De-Hui Liu
- Weifang Centers for Disease Control and Prevention, No 4801 Huixian Road, Gaoxin District, Shandong Province, Weifang, 261061, China
| | - Hui-Ming Zhang
- College of Basic Medical Sciences, Jiamusi University, No 148 Xuefu Street, Jiamusi, 154007, China
| | - Fang-Fang Wang
- College of Basic Medical Sciences, Jiamusi University, No 148 Xuefu Street, Jiamusi, 154007, China
| |
Collapse
|
3
|
Haeberlein S, Chevalley-Maurel S, Ozir-Fazalalikhan A, Koppejan H, Winkel BMF, Ramesar J, Khan SM, Sauerwein RW, Roestenberg M, Janse CJ, Smits HH, Franke-Fayard B. Protective immunity differs between routes of administration of attenuated malaria parasites independent of parasite liver load. Sci Rep 2017; 7:10372. [PMID: 28871201 PMCID: PMC5583236 DOI: 10.1038/s41598-017-10480-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Accepted: 08/10/2017] [Indexed: 12/27/2022] Open
Abstract
In humans and murine models of malaria, intradermal immunization (ID-I) with genetically attenuated sporozoites that arrest in liver induces lower protective immunity than intravenous immunization (IV-I). It is unclear whether this difference is caused by fewer sporozoites migrating into the liver or by suboptimal hepatic and injection site-dependent immune responses. We therefore developed a Plasmodium yoelii immunization/boost/challenge model to examine parasite liver loads as well as hepatic and lymph node immune responses in protected and unprotected ID-I and IV-I animals. Despite introducing the same numbers of genetically attenuated parasites in the liver, ID-I resulted in lower sterile protection (53-68%) than IV-I (93-95%). Unprotected mice developed less sporozoite-specific CD8+ and CD4+ effector T-cell responses than protected mice. After immunization, ID-I mice showed more interleukin-10-producing B and T cells in livers and skin-draining lymph nodes, but fewer hepatic CD8 memory T cells and CD8+ dendritic cells compared to IV-I mice. Our results indicate that the lower protection efficacy obtained by intradermal sporozoite administration is not linked to low hepatic parasite numbers as presumed before, but correlates with a shift towards regulatory immune responses. Overcoming these immune suppressive responses is important not only for live-attenuated malaria vaccines but also for other live vaccines administered in the skin.
Collapse
Affiliation(s)
- Simone Haeberlein
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.,Institute of Parasitology, Justus-Liebig-University Giessen, Schubertstrasse 81, 35392, Giessen, Germany
| | - Séverine Chevalley-Maurel
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Arifa Ozir-Fazalalikhan
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Hester Koppejan
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Beatrice M F Winkel
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Jai Ramesar
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Shahid M Khan
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Robert W Sauerwein
- Department of Medical Microbiology, Radboud University Medical Center, Geert-Grooteplein 28, 6525 GA, Nijmegen, The Netherlands
| | - Meta Roestenberg
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.,Department of Infectious Diseases, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Chris J Janse
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Hermelijn H Smits
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Blandine Franke-Fayard
- Department of Parasitology, Leiden University Medical Center, Albinusdreef 2, 2333 ZA, Leiden, The Netherlands.
| |
Collapse
|
4
|
Kaewraemruaen C, Sermswan RW, Wongratanacheewin S. Induction of regulatory T cells by Opisthorchis viverrini. Parasite Immunol 2016; 38:688-697. [PMID: 27552546 DOI: 10.1111/pim.12358] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2015] [Accepted: 08/19/2016] [Indexed: 01/04/2023]
Abstract
Opisthorchis viverrini causes public health problems in South-East Asia. Recently, TGF-β and IL-10 have been reported to increase in O. viverrini-infected hamsters but the sources of these cytokines are still unknown. In this study, the CD4+ T cells in infected hamsters were investigated. It was demonstrated that IL-4+ CD4+ T cells were significantly increased in hamster spleens and mesenteric lymph nodes (MLNs) during chronic infection. Interestingly, IL-10+ CD4+ T cells were also discovered at a significant level while Treg (T regulatory)-like TGF- β+ CD4+ T cells were in MLNs of infected hamsters. Moreover, the CD4+ CD25+ Foxp3+ Treg cell response was significantly found both in spleens and MLNs in infected hamsters. The findings were then confirmed by development of T-cell clones against crude somatic antigens (CSAg) in immunized BALB/c mice. Five clones named TCC21, TCC23, TCC35, TCC41 and TCC108 were established. The TCC21 was found to be the TGF-β+ CD4+ while TCC35, TCC41 and TCC108 were IL-4+ CD4+ and TCC23 was IFN-γ+ CD4+ . This TGF-β+ CD4+ T clone showed an inhibitory function in vitro in mononuclear cell proliferation via TGF-β-mediated mechanisms. This study indicated that O. viverrini-infected hamsters could induce TGF-β+ CD4+ Treg-like cells. The CSAg-specific Tregs secreted high TGF-β, and limited immune cell proliferation.
Collapse
Affiliation(s)
- C Kaewraemruaen
- Department of Microbiology, Khon Kaen University, Khon Kaen, Thailand.,Melioidosis Research Center, Khon Kaen University, Khon Kaen, Thailand
| | - R W Sermswan
- Melioidosis Research Center, Khon Kaen University, Khon Kaen, Thailand.,Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - S Wongratanacheewin
- Department of Microbiology, Khon Kaen University, Khon Kaen, Thailand. .,Melioidosis Research Center, Khon Kaen University, Khon Kaen, Thailand.
| |
Collapse
|
5
|
TLR4 and TLR9 signals stimulate protective immunity against blood-stage Plasmodium yoelii infection in mice. Exp Parasitol 2016; 170:73-81. [PMID: 27646627 DOI: 10.1016/j.exppara.2016.09.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Revised: 08/01/2016] [Accepted: 09/15/2016] [Indexed: 01/07/2023]
Abstract
The mechanisms regulating the induction of protective immunity against blood-stage malaria remain unclear. Resistant DBA/2 mouse develops a higher Th1 response compared with a susceptible BALB/c strain during Plasmodium yoelii (Py) infection. It is known that the T helper cell response is initiated and polarized by dendritic cells (DCs) of the innate immune system, during which TLR4 and TLR9 are important receptors for the innate recognition of the malaria parasite and its products. We hypothesized that TLR4/9 may play critical roles in the induction of protective immunity against Py infection. We used TLR4/9 antagonists and agonists to study their effects on mouse resistance to Py infection. We found that the administration of an antagonist prior to infection aggravated disease outcomes, impaired DC functions and suppressed the pro-inflammatory response to Py infection in resistant DBA/2 mice. Treatment with the TLR4 agonist lipopolysaccharide (LPS) but not TLR9 agonist significantly improved the survival rate of susceptible Py-infected BALB/c mice. LPS administration promoted the activation and expansion of DCs and drove a Th1-biased response. Our data demonstrate the important roles of TLR4/9 signals in inducing resistance to malaria parasites and provide evidence for the rational use of TLR agonists to potentiate protective immunity against Plasmodium infection.
Collapse
|
6
|
Pang W, Sun X, Feng H, Wang J, Cui L, Cao Y. The role of regulatory T cells during Plasmodium chabaudi chabaudi AS infection in BALB/c mice. Parasite Immunol 2016; 38:439-50. [PMID: 27139002 DOI: 10.1111/pim.12333] [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: 07/09/2015] [Accepted: 04/26/2016] [Indexed: 11/30/2022]
Abstract
An inappropriate immune response to parasite infection is one of the primary drivers of malaria pathogenesis. Regulatory T cells (Tregs), an important subset of CD4(+) T cells, can maintain self-tolerance and prevent autoimmune diseases. However, there is little consensus about their role in malaria pathogenesis. In this study, we transiently depleted Tregs (CD25(+) T cells) using an anti-CD25 mAb (7D4 clone) at different time points following Plasmodium chabaudi chabaudi AS infection in BALB/c mice and investigated the effect of depletion of Tregs in this model. In control mice, Tregs proliferated significantly and their suppressive function was enhanced after infection. IL-10 was increased drastically during infection. Depletion of Tregs at various time points can lead to divergent outcomes. When Tregs were depleted prior to or during the early phase of infection, most mice survived and had a robust Th1 immune response. In contrast, when Tregs were depleted close to peak parasitemia, all mice died as a result of inflammation. Taken together, these data suggest that in P. c. chabaudi AS-infected BALB/c mice, Tregs inhibit the Th1 response and macrophage activation, leading to increased parasite load; however, they also control inflammation-mediated pathology by secreting high levels of IL-10.
Collapse
Affiliation(s)
- W Pang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - X Sun
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - H Feng
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - J Wang
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | - L Cui
- Department of Entomology, Pennsylvania State University, University Park, PA, USA
| | - Y Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
7
|
Cao DH, Wang JC, Liu J, Du YT, Cui LW, Cao YM. Bacillus Calmette-Guérin-inoculation at different time points influences the outcome of C57BL/6 mice infected with Plasmodium chabaudi chabaudi AS. Folia Parasitol (Praha) 2016; 63. [PMID: 27188912 DOI: 10.14411/fp.2016.010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Accepted: 03/03/2016] [Indexed: 11/19/2022]
Abstract
Bacillus Calmette-Guérin (BCG) is an attenuated Mycobacterium tuberculosis vaccine. We performed a series of co-infection experiments with BCG-Plasmodium chabaudi chabaudi Landau, 1965 AS using C57BL/6 mice to analyse whether BCG can affect the development of protective immunity to infection with Plasmodium spp. and the mechanism of this protection. We divided mice into four groups: BCG-inoculation 4 weeks prior to P. c. chabaudi AS infection (B-4w-Pc); simultaneous BCG-inoculation and P. c. chabaudi AS infection (Pc+B); BCG-inoculation 3 days post P. c. chabaudi AS (Pc-3-B) infection; and mono-P. c. chabaudi AS infection as control (Pc). The parasitemia level in the B-4w-Pc group was noticeably higher than control group at 6-19 days post infection (dpi). Compared with the control group, the proportion of CD4(+)CD69(+) T cells was significantly reduced 5, 8 and 12 dpi, but the proportion of CD4(+)CD25(+)Foxp3(+) Tregs was significantly increased in the B-4w-Pc group on 5 and 8 dpi. The B-4w-Pc group also demonstrated reduced levels of IFN-γ and TNF-α on 5 and 8 dpi and significantly elevated level of IL-10 on 12 dpi. There were significantly fewer mDCs (CD11c(+)CD11b(+)) and pDCs (CD11c(+)B220(+)) in the B-4w-Pc group than the control group at all the time points post infection and the expression of MHC II was noticeably reduced on day 8 pi. Our findings confirmed that BCG inoculation prior to Plasmodium infection resulted in excessive activation and proliferation of Tregs and upregulation of anti-inflammatory mediators, which inhibited establishment of a Th1-dominant immune response during the early stages of Plasmodium infection by inhibiting dendritive cells response. BCG inoculation prior to P. c. chabaudi AS infection may contribute to overgrowth of parasites as well as mortality in mice.
Collapse
Affiliation(s)
- Dong-Hua Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China.,Aristogenesis Center, Hospital of People's Liberation Army, Shenyang, China
| | - Ji-Chun Wang
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Jun Liu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Yun-Ting Du
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| | - Li-Wang Cui
- Department of Entomology, The Pennsylvania State University, University Park, Pennsylvania, USA
| | - Ya-Ming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, China
| |
Collapse
|
8
|
Abstract
Naturally acquired immunity to the blood-stage of the malaria parasite develops slowly in areas of high endemicity, but is not sterilizing. It manifests as a reduction in parasite density and clinical symptoms. Immunity as a result of blood-stage vaccination has not yet been achieved in humans, although there are many animal models where vaccination has been successful. The development of a blood-stage vaccine has been complicated by a number of factors including limited knowledge of human-parasite interactions and which antigens and immune responses are critical for protection. Opinion is divided as to whether this vaccine should aim to accelerate the acquisition of responses acquired following natural exposure, or whether it should induce a different response. Animal and experimental human models suggest that cell-mediated immune responses can control parasite growth, but these responses can also contribute to significant immunopathology if unregulated. They are largely ignored in most blood-stage malaria vaccine development strategies. Here, we discuss key observations relating to cell-mediated immune responses in the context of experimental human systems and field studies involving naturally exposed individuals and how this may inform the development of a blood-stage malaria vaccine.
Collapse
|
9
|
Deroost K, Pham TT, Opdenakker G, Van den Steen PE. The immunological balance between host and parasite in malaria. FEMS Microbiol Rev 2015; 40:208-57. [PMID: 26657789 DOI: 10.1093/femsre/fuv046] [Citation(s) in RCA: 81] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/04/2015] [Indexed: 12/16/2022] Open
Abstract
Coevolution of humans and malaria parasites has generated an intricate balance between the immune system of the host and virulence factors of the parasite, equilibrating maximal parasite transmission with limited host damage. Focusing on the blood stage of the disease, we discuss how the balance between anti-parasite immunity versus immunomodulatory and evasion mechanisms of the parasite may result in parasite clearance or chronic infection without major symptoms, whereas imbalances characterized by excessive parasite growth, exaggerated immune reactions or a combination of both cause severe pathology and death, which is detrimental for both parasite and host. A thorough understanding of the immunological balance of malaria and its relation to other physiological balances in the body is of crucial importance for developing effective interventions to reduce malaria-related morbidity and to diminish fatal outcomes due to severe complications. Therefore, we discuss in this review the detailed mechanisms of anti-malarial immunity, parasite virulence factors including immune evasion mechanisms and pathogenesis. Furthermore, we propose a comprehensive classification of malaria complications according to the different types of imbalances.
Collapse
Affiliation(s)
- Katrien Deroost
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven - University of Leuven, 3000 Leuven, Belgium The Francis Crick Institute, Mill Hill Laboratory, London, NW71AA, UK
| | - Thao-Thy Pham
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven - University of Leuven, 3000 Leuven, Belgium
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven - University of Leuven, 3000 Leuven, Belgium
| | - Philippe E Van den Steen
- Laboratory of Immunobiology, Rega Institute for Medical Research, KU Leuven - University of Leuven, 3000 Leuven, Belgium
| |
Collapse
|
10
|
Xu H, Feng Y, Chen G, Zhu X, Pang W, Du Y, Wang Q, Qi Z, Cao Y. L-arginine exacerbates experimental cerebral malaria by enhancing pro-inflammatory responses. TOHOKU J EXP MED 2015; 236:21-31. [PMID: 25925198 DOI: 10.1620/tjem.236.21] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
L-Arginine (L-Arg), the substrate for nitric oxide (NO) synthase, has been used to treat malaria to reverse endothelial dysfunction in adults. However, the safety and efficacy of L-Arg remains unknown in malaria patients under the age of five, who are at the greatest risk of developing cerebral malaria (CM), a severe malaria complication. Here, we tested effects of L-Arg treatment on the outcomes of CM using a mouse model. Experimental cerebral malaria (ECM) was induced in female C57BL/6 mice infected with Plasmodium berghei ANKA, and L-Arg was administrated either prophylactically or after parasite infection. Surprisingly, both types of L-Arg administration caused a decline in survival time and raised CM clinical scores. L-Arg treatment increased the population of CD4(+)T-bet(+)IFN-γ(+) Th1 cells and the activated macrophages (F4/80(+)CD36(+)) in the spleen. The levels of pro-inflammatory cytokines, IFN-γ and TNF-α, in splenocyte cultures were also increased by L-Arg treatment. The above changes were accompanied with a rise in the number of dendritic cells (DCs) and an increase in their maturation. However, L-Arg did not affect the population of regulatory T cells or the level of IL-10 in the spleen. Taken together, these data suggest that L-Arg may enhance the Th1 immune response, which is essential for a protective response in uncomplicated malaria but could be lethal in CM patients. Therefore, the prophylactic use of L-Arg to treat CM, based on the assumption that restoring the bioavailability of endothelial NO improves the outcome of CM, may need to be reconsidered especially for children.
Collapse
Affiliation(s)
- Hongbin Xu
- Department of Immunology, College of Basic Medical Sciences, China Medical University
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Keswani T, Bhattacharyya A. Differential role of T regulatory and Th17 in Swiss mice infected with Plasmodium berghei ANKA and Plasmodium yoelii. Exp Parasitol 2014; 141:82-92. [PMID: 24675415 DOI: 10.1016/j.exppara.2014.03.003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2013] [Revised: 01/21/2014] [Accepted: 03/04/2014] [Indexed: 12/31/2022]
Abstract
The outcome of malaria infection is determined, in part, by the balance of pro-inflammatory and regulatory immune responses. Host immune responses in disease including malaria are finely regulated by the opposing effects of Th17 and T regulatory (Treg) cells. Here we have examined the role of Treg cells and Th17 cells during malaria infection and find that low levels of Treg cells possibly influence the outcome of infections with the lethal strain of Plasmodium berghei ANKA (PbA). In contrast, high level of Treg cells may influence the outcome of nonlethal Plasmodium yoelii NXL (P. yoelii) infections. We observed decreased expressions of key regulators of Treg inductions-TGF-β, CD4IL-2 and IL-10 during PbA infection, whereas their expression remains high during P. yoelii infection. On the other hand TNF-α, IL-6, IFN-γ and IL-23 expression is high during PbA infection and lower during P. yoelii infection. Thus, results from this study suggest that the differential expression of Treg and Th17 might have a key role on host pathogenesis during malaria infection. The high level of IL-6 and low level of TGF-β may composite of the advantaged local microenvironment for the production of Th17 cells in the spleen of the PBA infected mice and vice verse during nonlethal P. yoelii.
Collapse
Affiliation(s)
- Tarun Keswani
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, 35, Ballygunge Circular Road, Kolkata 700019, West Bengal, India.
| |
Collapse
|
12
|
Azcárate IG, Marín-García P, Kamali AN, Pérez-Benavente S, Puyet A, Diez A, Bautista JM. Differential immune response associated to malaria outcome is detectable in peripheral blood following Plasmodium yoelii infection in mice. PLoS One 2014; 9:e85664. [PMID: 24465641 PMCID: PMC3900426 DOI: 10.1371/journal.pone.0085664] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2013] [Accepted: 12/01/2013] [Indexed: 11/18/2022] Open
Abstract
Malaria infection in humans elicits a wide range of immune responses that can be detected in peripheral blood, but we lack detailed long-term follow-up data on the primary and subsequent infections that lead to naturally acquired immunity. Studies on antimalarial immune responses in mice have been based on models yielding homogenous infection profiles. Here, we present a mouse model in which a heterogeneous course of Plasmodium yoelii lethal malaria infection is produced in a non-congenic ICR strain to allow comparison among different immunological and clinical outcomes. Three different disease courses were observed ranging from a fatal outcome, either early or late, to a self-resolved infection that conferred long-term immunity against re-infection. Qualitative and quantitative changes produced in leukocyte subpopulations and cytokine profiles detected in peripheral blood during the first week of infection revealed that monocytes, dendritic cells and immature B cells were the main cell subsets present in highly-parasitized mice dying in the first week after infection. Besides, CD4(+)CD25(high) T cells expanded at an earlier time point in early deceased mice than in surviving mice and expressed higher levels of intracellular Foxp3 protein. In contrast, survivors showed a limited increase of cytokines release and stable circulating innate cells. From the second week of infection, mice that would die or survive showed similar immune profiles, although CD4(+)CD25(high) T cells number increased earlier in mice with the worst prognosis. In surviving mice the expansion of activated circulating T cell and switched-class B cells with a long-term protective humoral response from the second infection week is remarkable. Our results demonstrate that the follow-up studies of immunological blood parameters during a malaria infection can offer information about the course of the pathological process and the immune response.
Collapse
Affiliation(s)
- Isabel G. Azcárate
- Department of Biochemistry and Molecular Biology IV, Universidad Complutense de Madrid, Facultad de Veterinaria, Ciudad Universitaria, Madrid, Spain
- Research Institute Hospital 12 de Octubre, University Hospital 12 de Octubre, Madrid, Spain,
| | - Patricia Marín-García
- Department of Biochemistry and Molecular Biology IV, Universidad Complutense de Madrid, Facultad de Veterinaria, Ciudad Universitaria, Madrid, Spain
| | - Alí N. Kamali
- Department of Biochemistry and Molecular Biology IV, Universidad Complutense de Madrid, Facultad de Veterinaria, Ciudad Universitaria, Madrid, Spain
| | - Susana Pérez-Benavente
- Department of Biochemistry and Molecular Biology IV, Universidad Complutense de Madrid, Facultad de Veterinaria, Ciudad Universitaria, Madrid, Spain
| | - Antonio Puyet
- Department of Biochemistry and Molecular Biology IV, Universidad Complutense de Madrid, Facultad de Veterinaria, Ciudad Universitaria, Madrid, Spain
- Research Institute Hospital 12 de Octubre, University Hospital 12 de Octubre, Madrid, Spain,
| | - Amalia Diez
- Department of Biochemistry and Molecular Biology IV, Universidad Complutense de Madrid, Facultad de Veterinaria, Ciudad Universitaria, Madrid, Spain
- Research Institute Hospital 12 de Octubre, University Hospital 12 de Octubre, Madrid, Spain,
| | - José M. Bautista
- Department of Biochemistry and Molecular Biology IV, Universidad Complutense de Madrid, Facultad de Veterinaria, Ciudad Universitaria, Madrid, Spain
- Research Institute Hospital 12 de Octubre, University Hospital 12 de Octubre, Madrid, Spain,
- * E-mail:
| |
Collapse
|
13
|
Erythropoietin protects against murine cerebral malaria through actions on host cellular immunity. Infect Immun 2013; 82:165-73. [PMID: 24126529 DOI: 10.1128/iai.00929-13] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Cerebral malaria (CM) is associated with excessive host proinflammatory responses and endothelial activation. The hematopoietic hormone erythropoietin (EPO) possesses neuroprotective functions in animal models of ischemic-hypoxic, traumatic, and inflammatory injuries. In the Plasmodium berghei ANKA model of experimental CM (ECM), recombinant human EPO (rhEPO) has shown evident protection against ECM. To elucidate the mechanism of EPO in this ECM model, we investigated the effect of rhEPO on host cellular immune responses. We demonstrated that improved survival of mice with ECM after rhEPO treatment was associated with reduced endothelial activation and improved integrity of the blood-brain barrier. Our results revealed that rhEPO downregulated the inflammatory responses by directly inhibiting the levels and functions of splenic dendritic cells. Conversely, rhEPO treatment led to significant expansion of regulatory T cells and increased expression of the receptor cytotoxic T lymphocyte antigen 4 (CTLA-4). The data presented here provide evidence of the direct effect of rhEPO on host cellular immunity during ECM.
Collapse
|
14
|
Wang ML, Cao YM, Luo EJ, Zhang Y, Guo YJ. Pre-existing Schistosoma japonicum infection alters the immune response to Plasmodium berghei infection in C57BL/6 mice. Malar J 2013; 12:322. [PMID: 24034228 PMCID: PMC3848616 DOI: 10.1186/1475-2875-12-322] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 08/18/2013] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Since helminths and malaria parasites are often co-endemic, it is important to clarify the immunoregulatory mechanism that occurs during the process of co-infection. A previous study confirmed that dendritic cells (DCs) are involved in the establishment and regulation of the T-cell-mediated immune response to malaria infection. In the current study, distinct response profiles for splenic DCs and regulatory T cell (Treg) responses were assessed to evaluate the effects of a pre-existing Schistosoma japonicum infection on malaria infection. METHODS Malaria parasitaemia, survival rate, brain histopathology and clinical experimental cerebral malaria (ECM) were assessed in both Plasmodium berghei ANKA-mono-infected and S. japonicum-P. berghei ANKA-co-infected mice. Cell surface/intracellular staining and flow cytometry were used to analyse the level of splenic DC subpopulations, toll-like receptors (TLRs), DC surface molecules, Tregs (CD4⁺CD25⁺Foxp3⁺), IFN-γ/IL-10-secreting Tregs, and IFN-γ⁺/IL-10⁺-Foxp3⁻CD4⁺ T cells. IFN-γ, IL-4, IL-5, IL-10 and IL-13 levels were determined in splenocyte supernatants using enzyme-linked immunosorbent assay (ELISA). RESULTS The co-infected mice had significantly higher malaria parasitaemia, compared with the mono-infected mice, on days 2, 3, 7 and 8 after P. berghei ANKA infection. Mono-infected mice had a slightly lower survival rate, while clinical ECM symptoms, and brain pathology, were significantly more severe during the period of susceptibility to ECM. On days 5 and 8 post P. berghei ANKA infection, co-infected mice had significantly lower levels of CD11c⁺CD11b⁺, CD11c⁺CD45R/B220⁺, CD11c⁺TLR4⁺, CD11c⁺TLR9⁺, CD11c⁺MHCII⁺, CD11c⁺CD86⁺, IFN-γ-secreting Tregs, and IFN-γ⁺Foxp3⁻CD4⁺ T cells in single-cell suspensions of splenocytes when compared with P. berghei ANKA-mono-infected mice. Co-infected mice also had significantly lower levels of IFN-γ and higher levels of IL-4, IL-5, and IL-13 in splenocyte supernatants compared to mono-infected mice. There were no differences in the levels of IL-10-secreting Tregs or IL-10⁺Foxp3⁻CD4⁺ T cells between co-infected and mono-infected mice. CONCLUSIONS A Tregs-associated Th2 response plays an important role in protecting against ECM pathology. Pre-existing S. japonicum infection suppressed TLR ligand-induced DC maturation and had an anti-inflammatory effect during malaria infection not only by virtue of its ability to induce Th2 responses, but also by directly suppressing the ability of DC to produce pro-inflammatory mediators.
Collapse
Affiliation(s)
- Mei-lian Wang
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, No. 92 Beier Road, Heping District, Shenyang 110001, China
| | - Ya-ming Cao
- Department of Immunology, College of Basic Medical Sciences, China Medical University, No. 92 Beier Road, Heping District, Shenyang 110001, China
| | - En-jie Luo
- Department of Microbiology and Parasitology, College of Basic Medical Sciences, China Medical University, No. 92 Beier Road, Heping District, Shenyang 110001, China
| | - Ying Zhang
- Department of Sonography, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, China
| | - Ya-jun Guo
- Department of Sonography, Shengjing Hospital of China Medical University, No. 36 Sanhao Street, Heping District, Shenyang 110004, China
| |
Collapse
|
15
|
Shan Y, Liu J, Pan YY, Jiang YJ, Shang H, Cao YM. Age-related CD4(+)CD25(+)Foxp3(+) regulatory T-cell responses during Plasmodium berghei ANKA infection in mice susceptible or resistant to cerebral malaria. THE KOREAN JOURNAL OF PARASITOLOGY 2013; 51:289-95. [PMID: 23864739 PMCID: PMC3712102 DOI: 10.3347/kjp.2013.51.3.289] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 01/19/2013] [Accepted: 02/26/2013] [Indexed: 11/23/2022]
Abstract
Different functions have been attributed to CD4+CD25+Foxp3+ regulatory T-cells (Tregs) during malaria infection. Herein, we describe the disparity in Treg response and pro- and anti-inflammatory cytokines during infection with Plasmodium berghei ANKA between young (3-week-old) and middle-aged (8-month-old) C57BL/6 mice. Young mice were susceptible to cerebral malaria (CM), while the middle-aged mice were resistant to CM and succumbed to hyperparasitemia and severe anemia. The levels of pro-inflammatory cytokines, such as TNF-α, in young CM-susceptible mice were markedly higher than in middle-aged CM-resistant mice. An increased absolute number of Tregs 3-5 days post-inoculation, co-occurring with elevated IL-10 levels, was observed in middle-aged CM-resistant mice but not in young CM-susceptible mice. Our findings suggest that Treg proliferation might be associated with the suppression of excessive pro-inflammatory Th1 response during early malaria infection, leading to resistance to CM in the middle-aged mice, possibly in an IL-10-dependent manner.
Collapse
Affiliation(s)
- Ying Shan
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang 110001, China
| | | | | | | | | | | |
Collapse
|
16
|
Shan Y, Liu J, Jiang YJ, Shang H, Jiang D, Cao YM. Age-related susceptibility and resistance to nonlethal Plasmodium yoelii infection in C57BL/6 mice. Folia Parasitol (Praha) 2012; 59:153-61. [DOI: 10.14411/fp.2012.021] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
17
|
Wang GG, Chen G, Feng H, Liu J, Jiang YJ, Shang H, Cao YM. Plasmodium chabaudi AS: distinct CD4(+)CD25(+)Foxp3(+) regulatory T cell responses during infection in DBA/2 and BALB/c mice. Parasitol Int 2012; 62:24-31. [PMID: 22971347 DOI: 10.1016/j.parint.2012.08.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2012] [Revised: 08/11/2012] [Accepted: 08/19/2012] [Indexed: 11/26/2022]
Abstract
Malaria infections display variation patterns of clinical course and outcome. Although CD4(+)CD25(+)Foxp3(+) regulatory T (Treg) cells play an essential role in immune homeostasis, the immune regulatory roles involved in malaria infection remains to be elucidated. Herein, we compared the disparity in Treg cells response during the course of blood stage Plasmodium chabaudi chabaudi AS (P. c chabaudi AS) infection in DBA/2 and BALB/c mice. BALB/c mice initiated a Th1/Th2 profile respond to P. c chabaudi AS infection, but DBA/2 mice failed to control P. c chabaudi AS infection and almost of them died post-peak parasitemia. At the peak parasitemia, we found that higher proportion of Treg cells with elevated Foxp3 expression in DBA/2 than in BALB/c mice. We used anti-CD25 mAb to deplete Treg cells and found that the survival time and rate were prolonged in DBA/2 mice treated with anti-CD25 mAb. Treatment with anti-CD25 mAb in vivo led to enhanced pro-inflammation responses and Foxp3 expression decline on Treg cells. In contrast, after DBA/2 was treatment with anti-IL-10R mAb, IL-10R blockade in vivo caused excessive pro-inflammation responses and Foxp3 expression loss on CD4(+)CD25(+) T cells. Earlier death was found in all of DBA/2 mice with anti-IL-10R mAb. It suggested that IL-2 and IL-10 signal involved in maintaining Foxp3 expression on Treg cells. In all, the moderate suppressive activity of Treg cells may facilitate resistance to P. c chabaudi AS infection.
Collapse
Affiliation(s)
- Ge-Ge Wang
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Shenyang, Liaoning, China
| | | | | | | | | | | | | |
Collapse
|
18
|
Freitas do Rosario AP, Langhorne J. T cell-derived IL-10 and its impact on the regulation of host responses during malaria. Int J Parasitol 2012; 42:549-55. [PMID: 22549022 DOI: 10.1016/j.ijpara.2012.03.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Revised: 03/02/2012] [Accepted: 03/24/2012] [Indexed: 02/04/2023]
Abstract
Despite intense research, malaria still is the one of the most devastating diseases killing more people than any other parasitic infection. In an attempt to control the infection, the host immune system produces a potent pro-inflammatory response. However, this response is also associated with complications, such as severe anaemia, hypoglycaemia and cerebral malaria. This pronounced production of pro-inflammatory cytokines response is a common feature of malaria caused by parasites infecting humans as well as rodents and primates. A balance between pro- and anti-inflammatory responses may be fundamental to the elimination of the parasite without inducing excessive host pathology. IL-10 is a key cytokine that has been shown to have an important regulatory function in establishing this balance in malaria. Here we discuss which cells can produce IL-10 during infection, and present an overview of the evidence showing that T-cell derived IL-10 plays an important role in regulating malaria pathology. Many different subsets of T cells can produce IL-10, however, evidence is accumulating that it is effector Th1 CD4(+) T cells which provide the crucial source that down-regulates inflammatory pathology during blood-stage malaria infections.
Collapse
|
19
|
Zhu X, Pan Y, Zheng L, Cui L, Cao Y. Polysaccharides from the Chinese medicinal herb Achyranthes bidentata enhance anti-malarial immunity during Plasmodium yoelii 17XL infection in mice. Malar J 2012; 11:49. [PMID: 22348301 PMCID: PMC3312874 DOI: 10.1186/1475-2875-11-49] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2011] [Accepted: 02/20/2012] [Indexed: 01/15/2023] Open
Abstract
BACKGROUND Clinical immunity to malaria in human populations is developed after repeated exposure to malaria. Regulation and balance of host immune responses may lead to optimal immunity against malaria parasite infection. Polysaccharides (ABPS) derived from the Chinese herb ox knee Achyranthes bidentata possess immuno-modulatory functions. The aim of this study is to use the rodent malaria model Plasmodium yoelii 17XL (P. y17XL) to examine whether pretreatment with ABPS will modulate host immunity against malaria infection and improve the outcome of the disease. METHODS To determine whether ABPS could modulate immunity against malaria, mice were pretreated with ABPS prior to blood-stage infection by P. y17XL. Host survival and parasitaemia were monitored daily. The effect of pretreatment on host immune responses was studied through the quantitation of cytokines, dendritic cell populations, and natural regulatory T cells (Treg). RESULTS Pretreatment with ABPS prior to infection significantly extended the survival time of mice after P. y17XL infection. At three and five days post-infection, ABPS pretreated mice developed stronger Th1 immune responses against malaria infection with the number of F4/80+CD36+ macrophages and levels of IFN-γ, TNF-α and nitric oxide being significantly higher than in the control group. More importantly, ABPS-treated mice developed more myeloid (CD11c+CD11b+) and plasmacytoid dendritic cells (CD11c+CD45R+/B220+) than control mice. ABPS pretreatment also resulted in modulated expression of MHC-II, CD86, and especially Toll-like receptor 9 by CD11c+ dendritic cells. In comparison, pretreatment with ABPS did not alter the number of natural Treg or the production of the anti-inflammatory cytokine IL-10. CONCLUSION Pretreatment with the immuno-modulatory ABPS selectively enhanced Th1 immune responses to control the proliferation of malaria parasites, and prolonged the survival of mice during subsequent malaria infection.
Collapse
Affiliation(s)
- Xiaotong Zhu
- Department of Immunology, College of Basic Medical Sciences, China Medical University, No,92, Bei'er Road, Heping District, Shenyang, Liaoning 110001, China
| | | | | | | | | |
Collapse
|
20
|
Feng H, Zhu XT, Qi ZM, Wang QH, Wang GG, Pan YY, Li Y, Zheng L, Jiang YJ, Shang H, Cui L, Cao YM. Transient Attenuated Foxp3 Expression on CD4+ T cells Treated with 7D4 mAb Contributes to the Control of Parasite Burden in DBA / 2 Mice Infected with Lethal Plasmodium chabaudi chabaudi AS. Scand J Immunol 2011; 75:46-53. [DOI: 10.1111/j.1365-3083.2011.02622.x] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
21
|
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
|
22
|
Interleukin-10 and immunity against prokaryotic and eukaryotic intracellular pathogens. Infect Immun 2011; 79:2964-73. [PMID: 21576331 DOI: 10.1128/iai.00047-11] [Citation(s) in RCA: 148] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The generation of an effective immune response against an infection while also limiting tissue damage requires a delicate balance between pro- and anti-inflammatory responses. Interleukin-10 (IL-10) has potent immunosuppressive effects and is essential for regulation of immune responses. However, the immunosuppressive properties of IL-10 can also be exploited by pathogens to facilitate their own survival. In this minireview, we discuss the role of IL-10 in modulating intracellular bacterial, fungal, and parasitic infections. Using information from several different infection models, we bring together and highlight some common pathways for IL-10 regulation and function that cannot be fully appreciated by studies of a single pathogen.
Collapse
|
23
|
Sarfo BY, Wilson NO, Bond VC, Stiles JK. Plasmodium berghei ANKA infection increases Foxp3, IL-10 and IL-2 in CXCL-10 deficient C57BL/6 mice. Malar J 2011; 10:69. [PMID: 21439091 PMCID: PMC3078901 DOI: 10.1186/1475-2875-10-69] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2010] [Accepted: 03/28/2011] [Indexed: 12/30/2022] Open
Abstract
Background Cerebral malaria (CM) is a major cause of malaria mortality. Sequestration of infected red blood cells and leukocytes in brain vessels coupled with the production of pro-inflammatory factors contribute to CM. CXCL-10 a chemokine that is chemotactic to T cells has been linked to fatal CM. Mice deficient for CXCL-10 gene are resistant to murine CM, while antibody ablation of CXCL-10 enhanced the production of regulatory T cells (CD4+Cd25+Foxp3+) and IL-10 which regulate the immune system. Interleukin-2 (IL-2), a pro-inflammatory cytokine implicated in malaria pathogenesis has also been shown to be a key regulator of Foxp3. However the role of Foxp3 in resistant murine CM is not well understood. Methods The hypothesis that resistance of CXCL-10-/- mice to murine CM may be due to enhanced expression of Foxp3 in concert with IL-10 and IL-2 was tested. CXCL-10-/- and WT C57BL/6 mice were infected with Plasmodium berghei ANKA and evaluated for CM symptoms. Brain, peripheral blood mononuclear cells (PBMCs) and plasma were harvested from infected and uninfected mice at days 2, 4 and 8. Regulatory T cells (CD4+CD25+) and non-T regs (CD4+CD25-) were isolated from PBMCs and cultured with P. berghei antigens in vitro with dendritic cells as antigen presenting cells. Regulatory T cell transcription and specific factor Foxp3, was evaluated in mouse brain and PBMCs by realtime-PCR and Western blots while IL-10, and IL-2 were evaluated in plasma and cultured supernatants by ELISA. Results Wild type mice exhibited severe murine CM symptoms compared with CXCL-10-/- mice. Foxp3 mRNA and protein in brain and PBMC's of CXCL-10-/- mice was significantly up-regulated (p < 0.05) by day 4 post-infection (p.i) compared with WT. Plasma levels of IL-10 and IL-2 in infected CXCL-10-/- were higher than in WT mice (p < 0.05) at days 2 and 4 p.i. Ex-vivo CD4+CD25+ T cells from CXCL-10-/- re-stimulated with P. berghei antigens produced more IL-10 than WT CD4+CD25+ T cells. Conclusion The results indicate that in the absence of CXCL-10, the resulting up-regulation of Foxp3, IL-10 and IL-2 may be involved in attenuating fatal murine CM.
Collapse
Affiliation(s)
- Bismark Y Sarfo
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, 720 Westview Drive, South West, Atlanta Georgia, GA 30310, USA
| | | | | | | |
Collapse
|
24
|
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
|
25
|
In vivo infection by Trypanosoma cruzi: the conserved FLY domain of the gp85/trans-sialidase family potentiates host infection. Parasitology 2010; 138:481-92. [PMID: 21040619 DOI: 10.1017/s0031182010001411] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Trypanosoma cruzi is a protozoan parasite that infects vertebrates, causing in humans a pathological condition known as Chagas' disease. The infection of host cells by T. cruzi involves a vast collection of molecules, including a family of 85 kDa GPI-anchored glycoproteins belonging to the gp85/trans-sialidase superfamily, which contains a conserved cell-binding sequence (VTVXNVFLYNR) known as FLY, for short. Herein, it is shown that BALB/c mice administered with a single dose (1 μg/animal, intraperitoneally) of FLY-synthetic peptide are more susceptible to infection by T. cruzi, with increased systemic parasitaemia (2-fold) and mortality. Higher tissue parasitism was observed in bladder (7·6-fold), heart (3-fold) and small intestine (3·6-fold). Moreover, an intense inflammatory response and increment of CD4+ T cells (1·7-fold) were detected in the heart of FLY-primed and infected animals, with a 5-fold relative increase of CD4+CD25+FoxP3+ T (Treg) cells. Mice treated with anti-CD25 antibodies prior to infection, showed a decrease in parasitaemia in the FLY model employed. In conclusion, the results suggest that FLY facilitates in vivo infection by T. cruzi and concurs with other factors to improve parasite survival to such an extent that might influence the progression of pathology in Chagas' disease.
Collapse
|
26
|
Chi N, Maranchie JK, Appleman LJ, Storkus WJ. Update on vaccine development for renal cell cancer. Res Rep Urol 2010; 2:125-41. [PMID: 24198621 PMCID: PMC3703676 DOI: 10.2147/rru.s7242] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Renal cell carcinoma (RCC) remains a significant health concern that frequently presents as metastatic disease at the time of initial diagnosis. Current first-line therapeutics for the advanced-stage RCC include antiangiogenic drugs that have yielded high rates of objective clinical response; however, these tend to be transient in nature, with many patients becoming refractory to chronic treatment with these agents. Adjuvant immunotherapies remain viable candidates to sustain disease-free and overall patient survival. In particular, vaccines designed to optimize the activation, maintenance, and recruitment of specific immunity within or into the tumor site continue to evolve. Based on the integration of increasingly refined immunomonitoring systems in both translational models and clinical trials, allowing for the improved understanding of treatment mechanism(s) of action, further refined (combinational) vaccine protocols are currently being developed and evaluated. This review provides a brief history of RCC vaccine development, discusses the successes and limitations in such approaches, and provides a rationale for developing combinational vaccine approaches that may provide improved clinical benefits to patients with RCC.
Collapse
Affiliation(s)
- Nina Chi
- Department of immunology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, United States
| | | | | | | |
Collapse
|
27
|
Chen G, Feng H, Liu J, Qi ZM, Wu Y, Guo SY, Li DM, Wang JC, Cao YM. Characterization of immune responses to single or mixed infections with P. yoelii 17XL and P. chabaudi AS in different strains of mice. Parasitol Int 2010; 59:400-6. [PMID: 20609420 DOI: 10.1016/j.parint.2010.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2009] [Revised: 05/03/2010] [Accepted: 05/20/2010] [Indexed: 12/29/2022]
Abstract
The outcome of Plasmodium yoelii 17XL (P.y17XL)-infected BALB/c and DBA/2 mice, ranging from death to spontaneous cure, depends largely on the establishment of effective Th1 and Th2 responses and a successful switch between Th1 and Th2 responses, as well as appropriate functioning of CD4(+)CD25(+)Foxp3(+)regulatory T cells (Tregs). The infection with another malaria-causing parasite, Plasmodium chabaudi AS (P.cAS), leads to a different outcome in BALB/c and DBA/2 mice compared to mice infected with P.y17XL alone. To understand the consequence of co-infection with P.y17XL and P.cAS, we determined the proliferation curve of parasites, pro-inflammatory/anti-inflammatory cytokine profiles, and the dynamic changes of the number of Tregs in DBA/2 and BALB/c mice with single or mixed-species infections. The infective mode in mixed-species infections was the same as single P.y17XL infections. The multiplication of P.y17XL parasites prevailed in BALB/c and DBA/2 mice with early mixed infections, as detected by RTQ-PCR. Subsequently, the multiplication of P.cAS parasites dominated in DBA/2 mice with mixed infections, while BALB/c mice succumbed to infection. In addition, the dynamic changes in IFN-gamma and IL-4 production in mice with mixed infections, used as a measure of Th1 and Th2 responsiveness, were consistent with P.y17XL-infected mice. Treg activation and the IL-10 level were also closely related to susceptibility to infection. Our findings demonstrate that the characteristics of the immune response during infections with mixed species are dependent on the mode of proliferation of different species of Plasmodium. Indeed, different species of Plasmodium can influence each other in the same host.
Collapse
Affiliation(s)
- Guang Chen
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Heping District, Shenyang, China
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Natural regulatory T cells mediate the development of cerebral malaria by modifying the pro-inflammatory response. Parasitol Int 2010; 59:232-41. [DOI: 10.1016/j.parint.2010.02.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2009] [Revised: 01/25/2010] [Accepted: 02/18/2010] [Indexed: 11/17/2022]
|
29
|
Finney OC, Riley EM, Walther M. Regulatory T cells in malaria – friend or foe? Trends Immunol 2010; 31:63-70. [DOI: 10.1016/j.it.2009.12.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2009] [Revised: 12/06/2009] [Accepted: 12/08/2009] [Indexed: 10/20/2022]
|
30
|
Scholzen A, Minigo G, Plebanski M. Heroes or villains? T regulatory cells in malaria infection. Trends Parasitol 2010; 26:16-25. [DOI: 10.1016/j.pt.2009.10.004] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2009] [Revised: 10/09/2009] [Accepted: 10/15/2009] [Indexed: 12/14/2022]
|