45 results on '"Claser C"'
Search Results
2. Immunodominance: a new hypothesis to explain parasite escape and host/parasite equilibrium leading to the chronic phase of Chagas' disease?
- Author
-
Rodrigues, M.M., primary, Alencar, B.C.G. de, additional, Claser, C., additional, and Tzelepis, F., additional
- Published
- 2009
- Full Text
- View/download PDF
3. Immunodominance: a new hypothesis to explain parasite escape and host/parasite equilibrium leading to the chronic phase of Chagas' disease?
- Author
-
Rodrigues, M. M., Bruna de Alencar, Claser, C., and Tzelepis, F.
- Subjects
lcsh:R5-920 ,Trypanosoma cruzi ,Major histocompatibility complex ,Ocean Engineering ,Host-Parasite Interactions ,Chagas' disease ,lcsh:Biology (General) ,Immunodominance ,Chronic Disease ,parasitic diseases ,Immune Tolerance ,Animals ,Humans ,Chagas Disease ,lcsh:Medicine (General) ,lcsh:QH301-705.5 - Abstract
Intense immune responses are observed during human or experimental infection with the digenetic protozoan parasite Trypanosoma cruzi. The reasons why such immune responses are unable to completely eliminate the parasites are unknown. The survival of the parasite leads to a parasite-host equilibrium found during the chronic phase of chagasic infection in most individuals. Parasite persistence is recognized as the most likely cause of the chagasic chronic pathologies. Therefore, a key question in Chagas' disease is to understand how this equilibrium is established and maintained for a long period. Understanding the basis for this equilibrium may lead to new approaches to interventions that could help millions of individuals at risk for infection or who are already infected with T. cruzi. Here, we propose that the phenomenon of immunodominance may be significant in terms of regulating the host-parasite equilibrium observed in Chagas' disease. T. cruzi infection restricts the repertoire of specific T cells generating, in some cases, an intense immunodominant phenotype and in others causing a dramatic interference in the response to distinct epitopes. This immune response is sufficiently strong to maintain the host alive during the acute phase carrying them to the chronic phase where transmission usually occurs. At the same time, immunodominance interferes with the development of a higher and broader immune response that could be able to completely eliminate the parasite. Based on this, we discuss how we can interfere with or take advantage of immunodominance in order to provide an immunotherapeutic alternative for chagasic individuals.
4. Infection with Trypanosoma cruzi restricts the repertoire of parasite-specific CD8+ T cells leading to immunodominance
- Author
-
Tzelepis, F., Alencar, B. C. G., Penido, M. L. O., Claser, C., Machado, A. V., OSCAR BRUNA-ROMERO, Gazzinelli, R. T., and Rodrigues, M. M.
5. Silencing cytokeratin 18 gene inhibits intracellular replication of Trypanosoma cruzi in HeLa cells but not binding and invasion of trypanosomes
- Author
-
de Mello Samanta M, Curcio Marli, Claser Carla, Silveira Eduardo V, Monteiro Hugo P, and Rodrigues Mauricio M
- Subjects
Cytology ,QH573-671 - Abstract
Abstract Background As an obligatory intracellular parasite, Trypanosoma cruzi, the etiological agent of Chagas' disease, must invade and multiply within mammalian cells. Cytokeratin 18 (CK18) is among the host molecules that have been suggested as a mediator of important events during T. cruzi-host cell interaction. Based on that possibility, we addressed whether RNA interference (RNAi)-mediated down regulation of the CK18 gene could interfere with the parasite life cycle in vitro. HeLa cells transiently transfected with CK18-RNAi had negligible levels of CK18 transcripts, and significantly reduced levels of CK18 protein expression as determined by immunoblotting or immunofluorescence. Results CK18 negative or positive HeLa cells were invaded equally as well by trypomastigotes of different T. cruzi strains. Also, in CK18 negative or positive cells, parasites recruited host cells lysosomes and escaped from the parasitophorous vacuole equally as well. After that, the growth of amastigotes of the Y or CL-Brener strains, was drastically arrested in CK18 RNAi-treated cells. After 48 hours, the number of amastigotes was several times lower in CK18 RNAi-treated cells when compared to control cells. Simultaneous staining of parasites and CK18 showed that in HeLa cells infected with the Y strain both co-localize. Although the amastigote surface protein-2 contains the domain VTVXNVFLYNR previously described to bind to CK18, in several attempts, we failed to detect binding of a recombinant protein to CK-18. Conclusion The study demonstrates that silencing CK18 by transient RNAi, inhibits intracellular multiplication of the Y and CL strain of T. cruzi in HeLa cells, but not trypanosome binding and invasion.
- Published
- 2008
- Full Text
- View/download PDF
6. Unraveling the complex interplay: immunopathology and immune evasion strategies of alphaviruses with emphasis on neurological implications.
- Author
-
de Oliveira Souza R, Duarte Júnior JWB, Della Casa VS, Santoro Rosa D, Renia L, and Claser C
- Subjects
- Humans, Animals, Nervous System Diseases immunology, Nervous System Diseases virology, Immune Evasion, Alphavirus pathogenicity, Alphavirus immunology, Alphavirus Infections immunology, Alphavirus Infections virology, Host-Pathogen Interactions immunology, Blood-Brain Barrier immunology
- Abstract
Arthritogenic alphaviruses pose a significant public health concern due to their ability to cause joint inflammation, with emerging evidence of potential neurological consequences. In this review, we examine the immunopathology and immune evasion strategies employed by these viruses, highlighting their complex mechanisms of pathogenesis and neurological implications. We delve into how these viruses manipulate host immune responses, modulate inflammatory pathways, and potentially establish persistent infections. Further, we explore their ability to breach the blood-brain barrier, triggering neurological complications, and how co-infections exacerbate neurological outcomes. This review synthesizes current research to provide a comprehensive overview of the immunopathological mechanisms driving arthritogenic alphavirus infections and their impact on neurological health. By highlighting knowledge gaps, it underscores the need for research to unravel the complexities of virus-host interactions. This deeper understanding is crucial for developing targeted therapies to address both joint and neurological manifestations of these infections., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2024 de Oliveira Souza, Duarte Júnior, Della Casa, Santoro Rosa, Renia and Claser.)
- Published
- 2024
- Full Text
- View/download PDF
7. DUSP4 modulates RIG-I- and STING-mediated IRF3-type I IFN response.
- Author
-
Jiao H, James SJ, Png CW, Cui C, Li H, Li L, Chia WN, Min N, Li W, Claser C, Rénia L, Wang H, Chen MI, Chu JJH, Tan KSW, Deng Y, and Zhang Y
- Subjects
- Animals, Mice, Immunity, Innate, Protein Serine-Threonine Kinases genetics, Protein Serine-Threonine Kinases metabolism, Signal Transduction, Interferon Type I metabolism, Virus Diseases immunology, Virus Diseases metabolism, Membrane Proteins metabolism, Roundabout Proteins metabolism, Protein Tyrosine Phosphatases metabolism, Receptors, Cell Surface metabolism
- Abstract
Detection of cytosolic nucleic acids by pattern recognition receptors, including STING and RIG-I, leads to the activation of multiple signalling pathways that culminate in the production of type I interferons (IFNs) which are vital for host survival during virus infection. In addition to protective immune modulatory functions, type I IFNs are also associated with autoimmune diseases. Hence, it is important to elucidate the mechanisms that govern their expression. In this study, we identified a critical regulatory function of the DUSP4 phosphatase in innate immune signalling. We found that DUSP4 regulates the activation of TBK1 and ERK1/2 in a signalling complex containing DUSP4, TBK1, ERK1/2 and IRF3 to regulate the production of type I IFNs. Mice deficient in DUSP4 were more resistant to infections by both RNA and DNA viruses but more susceptible to malaria parasites. Therefore, our study establishes DUSP4 as a regulator of nucleic acid sensor signalling and sheds light on an important facet of the type I IFN regulatory system., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
8. CCR6 expression reduces mouse survival upon malarial challenge with Plasmodium berghei NK65 strain.
- Author
-
Silveira ELVD, Rai U, Bonezi V, Zárate-Bladés CR, and Claser C
- Subjects
- Animals, DNA, Complementary, Mice, Mice, Inbred C57BL, Parasitemia parasitology, Receptors, CCR6, Malaria parasitology, Plasmodium berghei
- Abstract
Background: It has been demonstrated that proteins expressed by liver-stage Plasmodium parasites can inhibit the translocation of transcription factors to the nucleus of different cells. This process would hinder the expression of immune genes, such as the CCL20 chemokine., Objective: Since CCR6 is the only cognate receptor for CCL20, we investigated the importance of this chemokine-receptor axis against rodent malaria., Methods: CCR6-deficient (KO) and wild-type (WT) C57BL/6 mice were challenged with Plasmodium berghei (Pb) NK65 sporozoites or infected red blood cells (iRBCs). Liver parasitic cDNA, parasitemia and serum cytokine concentrations were respectively evaluated through reverse transcription-polymerase chain reaction (RT-PCR), staining thin-blood smears with Giemsa solution, and enzyme-linked immunosorbent assay (ELISA)., Findings: Although the sporozoite challenges yielded similar liver parasitic cDNA and parasitemia, KO mice presented a prolonged survival than WT mice. After iRBC challenges, KO mice kept displaying higher survival rates as well as a decreased IL-12 p70 concentration in the serum than WT mice., Conclusion: Our data suggest that malaria triggered by PbNK65 liver- or blood-stage forms elicit a pro-inflammatory environment that culminates with a decreased survival of infected C57BL/6 mice.
- Published
- 2022
- Full Text
- View/download PDF
9. Experimental Models to Study the Pathogenesis of Malaria-Associated Acute Respiratory Distress Syndrome.
- Author
-
Nguee SYT, Júnior JWBD, Epiphanio S, Rénia L, and Claser C
- Subjects
- Animals, Disease Models, Animal, Humans, Lung pathology, Mice, Mice, Inbred C57BL, Plasmodium berghei physiology, Malaria pathology, Respiratory Distress Syndrome etiology
- Abstract
Malaria-associated acute respiratory distress syndrome (MA-ARDS) is increasingly gaining recognition as a severe malaria complication because of poor prognostic outcomes, high lethality rate, and limited therapeutic interventions. Unfortunately, invasive clinical studies are challenging to conduct and yields insufficient mechanistic insights. These limitations have led to the development of suitable MA-ARDS experimental mouse models. In patients and mice, MA-ARDS is characterized by edematous lung, along with marked infiltration of inflammatory cells and damage of the alveolar-capillary barriers. Although, the pathogenic pathways have yet to be fully understood, the use of different experimental mouse models is fundamental in the identification of mediators of pulmonary vascular damage. In this review, we discuss the current knowledge on endothelial activation, leukocyte recruitment, leukocyte induced-endothelial dysfunction, and other important findings, to better understand the pathogenesis pathways leading to endothelial pulmonary barrier lesions and increased vascular permeability. We also discuss how the advances in imaging techniques can contribute to a better understanding of the lung lesions induced during MA-ARDS, and how it could aid to monitor MA-ARDS severity., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest., (Copyright © 2022 Nguee, Júnior, Epiphanio, Rénia and Claser.)
- Published
- 2022
- Full Text
- View/download PDF
10. PET Imaging of Translocator Protein as a Marker of Malaria-Associated Lung Inflammation.
- Author
-
Goggi JL, Claser C, Hartimath SV, Hor PX, Tan PW, Ramasamy B, Abdul Rahman H, Cheng P, Chang ZW, Nguee SYT, Tang JR, Robins EG, and Renia L
- Subjects
- Animals, Disease Models, Animal, Leukocytes metabolism, Macrophages metabolism, Male, Mice, Mice, Inbred C57BL, Monocytes metabolism, Plasmodium berghei pathogenicity, Positron-Emission Tomography methods, Respiratory Distress Syndrome metabolism, Biomarkers metabolism, Lung metabolism, Malaria metabolism, Pneumonia metabolism
- Abstract
Malaria-associated acute respiratory distress syndrome (MA-ARDS) is a severe complication of malaria that occurs despite effective antimalarial treatment. Currently, noninvasive imaging procedures such as chest X-rays are used to assess edema in established MA-ARDS, but earlier detection methods are needed to reduce morbidity and mortality. The early stages of MA-ARDS are characterized by the infiltration of leukocytes, in particular monocytes/macrophages; thus, monitoring of immune infiltrates may provide a useful indicator of early pathology. In this study, Plasmodium berghei ANKA-infected C57BL/6 mice, a rodent model of MA-ARDS, were longitudinally imaged using the 18-kDa translocator protein (TSPO) imaging agent [
18 F]FEPPA as a marker of macrophage accumulation during the development of pathology and in response to combined artesunate and chloroquine diphosphate (ART+CQ) therapy. [18 F]FEPPA uptake was compared to blood parasitemia levels and to levels of pulmonary immune cell infiltrates by using flow cytometry. Infected animals showed rapid increases in lung retention of [18 F]FEPPA, correlating well with increases in blood parasitemia and pulmonary accumulation of interstitial inflammatory macrophages and major histocompatibility complex class II (MHC-II)-positive alveolar macrophages. Treatment with ART+CQ abrogated this increase in parasitemia and significantly reduced both lung uptake of [18 F]FEPPA and levels of macrophage infiltrates. We conclude that retention of [18 F]FEPPA in the lungs is well correlated with changes in blood parasitemia and levels of lung-associated macrophages during disease progression and in response to ART+CQ therapy. With further development, TSPO biomarkers may have the potential to accurately assess the early onset of MA-ARDS.- Published
- 2021
- Full Text
- View/download PDF
11. Author Correction: Lung endothelial cell antigen cross-presentation to CD8 + T cells drives malaria-associated lung injury.
- Author
-
Claser C, Nguee SYT, Balachander A, Howland SW, Becht E, Gunasegaran B, Hartimath SV, Lee AWQ, Ho JTT, Ong CB, Newell EW, Goggi J, Ng LG, and Renia L
- Abstract
An amendment to this paper has been published and can be accessed via a link at the top of the paper.
- Published
- 2019
- Full Text
- View/download PDF
12. Lung endothelial cell antigen cross-presentation to CD8 + T cells drives malaria-associated lung injury.
- Author
-
Claser C, Nguee SYT, Balachander A, Wu Howland S, Becht E, Gunasegaran B, Hartimath SV, Lee AWQ, Theng Theng Ho J, Bing Ong C, Newell EW, Goggi J, Guan Ng L, and Renia L
- Subjects
- Acute Lung Injury immunology, Acute Lung Injury parasitology, Animals, Disease Models, Animal, Endothelial Cells immunology, Female, Lung parasitology, Lung pathology, Malaria drug therapy, Malaria parasitology, Male, Mice, Mice, Inbred C57BL, Mice, Knockout, Plasmodium berghei immunology, Pulmonary Edema parasitology, Pulmonary Edema pathology, Respiratory Distress Syndrome immunology, Respiratory Distress Syndrome parasitology, Acute Lung Injury pathology, CD8-Positive T-Lymphocytes immunology, Cross-Priming immunology, Interferon-gamma immunology, Malaria immunology, Respiratory Distress Syndrome pathology
- Abstract
Malaria-associated acute respiratory distress syndrome (ARDS) and acute lung injury (ALI) are life-threatening manifestations of severe malaria infections. The pathogenic mechanisms that lead to respiratory complications, such as vascular leakage, remain unclear. Here, we confirm that depleting CD8
+ T cells with anti-CD8β antibodies in C57BL/6 mice infected with P. berghei ANKA (PbA) prevent pulmonary vascular leakage. When we transfer activated parasite-specific CD8+ T cells into PbA-infected TCRβ-/- mice (devoid of all T-cell populations), pulmonary vascular leakage recapitulates. Additionally, we demonstrate that PbA-infected erythrocyte accumulation leads to lung endothelial cell cross-presentation of parasite antigen to CD8+ T cells in an IFNγ-dependent manner. In conclusion, pulmonary vascular damage in ALI is a consequence of IFNγ-activated lung endothelial cells capturing, processing, and cross-presenting malaria parasite antigen to specific CD8+ T cells induced during infection. The mechanistic understanding of the immunopathogenesis in malaria-associated ARDS and ALI provide the basis for development of adjunct treatments.- Published
- 2019
- Full Text
- View/download PDF
13. Plasmodium co-infection protects against chikungunya virus-induced pathologies.
- Author
-
Teo TH, Lum FM, Ghaffar K, Chan YH, Amrun SN, Tan JJL, Lee CYP, Chua TK, Carissimo G, Lee WWL, Claser C, Rajarethinam R, Rénia L, and Ng LFP
- Subjects
- Animals, Apoptosis immunology, Arthritis genetics, Arthritis immunology, Arthritis metabolism, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes metabolism, Chikungunya Fever virology, Chikungunya virus physiology, Coinfection parasitology, Coinfection virology, Female, Humans, Interferon-gamma genetics, Interferon-gamma immunology, Interferon-gamma metabolism, Malaria metabolism, Malaria parasitology, Male, Mice, Inbred C57BL, Mice, Knockout, Plasmodium physiology, Viral Load immunology, Viremia immunology, Viremia virology, Chikungunya Fever immunology, Chikungunya virus immunology, Coinfection immunology, Malaria immunology, Plasmodium immunology
- Abstract
Co-infection with Plasmodium and chikungunya virus (CHIKV) has been reported in humans, but the impact of co-infection on pathogenesis remains unclear. Here, we show that prior exposure to Plasmodium suppresses CHIKV-associated pathologies in mice. Mechanistically, Plasmodium infection induces IFNγ, which reduces viraemia of a subsequent CHIKV infection and suppresses tissue viral load and joint inflammation. Conversely, concomitant infection with both pathogens limits the peak of joint inflammation with no effect on CHIKV viraemia. Reduced peak joint inflammation is regulated by elevated apoptosis of CD4
+ T-cells in the lymph nodes and disrupted CXCR3-mediated CD4+ T-cell migration that abolishes their infiltration into the joints. Virus clearance from tissues is delayed in both infection scenarios, and is associated with a disruption of B cell affinity-maturation in the spleen that reduces CHIKV-neutralizing antibody production.- Published
- 2018
- Full Text
- View/download PDF
14. Interferon regulatory factor 1 is essential for pathogenic CD8+ T cell migration and retention in the brain during experimental cerebral malaria.
- Author
-
Gun SY, Claser C, Teo TH, Howland SW, Poh CM, Chye RRY, Ng LFP, and Rénia L
- Subjects
- Animals, Brain microbiology, Brain pathology, CD8-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes microbiology, Cell Movement genetics, Disease Models, Animal, Gene Expression Regulation, Humans, Lymphocyte Activation genetics, Lymphocyte Activation immunology, Malaria, Cerebral immunology, Malaria, Cerebral microbiology, Mice, Mice, Knockout, Interferon Regulatory Factor-1 genetics, Malaria, Cerebral genetics, Plasmodium berghei pathogenicity, Receptors, CXCR3 genetics
- Abstract
Host immune response has a key role in controlling the progression of malaria infection. In the well-established murine model of experimental cerebral malaria (ECM) with Plasmodium berghei ANKA infection, proinflammatory Th1 and CD8+ T cell response are essential for disease development. Interferon regulatory factor 1 (IRF1) is a transcription factor that promotes Th1 responses, and its absence was previously shown to protect from ECM death. Yet the exact mechanism of protection remains unknown. Here we demonstrated that IRF1-deficient mice (IRF1 knockout) were protected from ECM death despite displaying early neurological signs. Resistance to ECM death was a result of reduced parasite sequestration and pathogenic CD8+ T cells in the brain. Further analysis revealed that IRF1 deficiency suppress interferon-γ production and delayed CD8+ T cell proliferation. CXCR3 expression was found to be decreased in pathogenic CD8+ T cells, which limited their migration to the brain. In addition, reduced expression of adhesion molecules by brain endothelial cells hampered leucocyte retention in the brain. Taken together, these factors limited sequestration of pathogenic CD8+ T cells and consequently its ability to induce extensive damage to the blood-brain barrier., (© 2017 John Wiley & Sons Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
15. Dual modal ultra-bright nanodots with aggregation-induced emission and gadolinium-chelation for vascular integrity and leakage detection.
- Author
-
Feng G, Li JLY, Claser C, Balachander A, Tan Y, Goh CC, Kwok IWH, Rénia L, Tang BZ, Ng LG, and Liu B
- Subjects
- Animals, Brain blood supply, Chelating Agents chemistry, Female, Fluorescent Dyes chemistry, Light, Malaria, Cerebral diagnostic imaging, Mice, Inbred C57BL, Particle Size, Skin blood supply, Skin diagnostic imaging, Surface Properties, Tissue Distribution, Blood-Brain Barrier metabolism, Brain diagnostic imaging, Coordination Complexes chemistry, Gadolinium chemistry, Metal Nanoparticles chemistry, Optical Imaging methods
- Abstract
The study of blood brain barrier (BBB) functions is important for neurological disorder research. However, the lack of suitable tools and methods has hampered the progress of this field. Herein, we present a hybrid nanodot strategy, termed AIE-Gd dots, comprising of a fluorogen with aggregation-induced emission (AIE) characteristics as the core to provide bright and stable fluorescence for optical imaging, and gadolinium (Gd) for accurate quantification of vascular leakage via inductively-coupled plasma mass spectrometry (ICP-MS). In this report, we demonstrate that AIE-Gd dots enable direct visualization of brain vascular networks under resting condition, and that they form localized punctate aggregates and accumulate in the brain tissue during experimental cerebral malaria, indicative of hemorrhage and BBB malfunction. With its superior detection sensitivity and multimodality, we hereby propose that AIE-Gd dots can serve as a better alternative to Evans blue for visualization and quantification of changes in brain barrier functions., (Copyright © 2017. Published by Elsevier Ltd.)
- Published
- 2018
- Full Text
- View/download PDF
16. Co-infection with Chikungunya virus alters trafficking of pathogenic CD8 + T cells into the brain and prevents Plasmodium -induced neuropathology.
- Author
-
Teo TH, Howland SW, Claser C, Gun SY, Poh CM, Lee WW, Lum FM, Ng LF, and Rénia L
- Subjects
- Animals, Brain parasitology, Brain virology, CD8-Positive T-Lymphocytes parasitology, CD8-Positive T-Lymphocytes virology, Cell Movement, Chikungunya Fever parasitology, Chikungunya Fever virology, Coinfection parasitology, Coinfection virology, Female, Malaria, Cerebral parasitology, Malaria, Cerebral virology, Male, Mice, Mice, Inbred C57BL, Neuropathology, Protective Factors, Brain pathology, CD8-Positive T-Lymphocytes pathology, Chikungunya Fever pathology, Chikungunya virus physiology, Coinfection pathology, Malaria, Cerebral pathology, Plasmodium berghei physiology
- Abstract
Arboviral diseases have risen significantly over the last 40 years, increasing the risk of co-infection with other endemic disease such as malaria. However, nothing is known about the impact arboviruses have on the host response toward heterologous pathogens during co-infection. Here, we investigate the effects of Chikungunya virus (CHIKV) co-infection on the susceptibility and severity of malaria infection. Using the Plasmodium berghei ANKA (PbA) experimental cerebral malaria (ECM) model, we show that concurrent co-infection induced the most prominent changes in ECM manifestation. Concurrent co-infection protected mice from ECM mortality without affecting parasite development in the blood. This protection was mediated by the alteration of parasite-specific CD8
+ T-cell trafficking through an IFNγ-mediated mechanism. Co-infection with CHIKV induced higher splenic IFNγ levels that lead to high local levels of CXCL9 and CXCL10. This induced retention of CXCR3-expressing pathogenic CD8+ T cells in the spleen and prevented their migration to the brain. This then averts all downstream pathogenic events such as parasite sequestration in the brain and disruption of blood-brain barrier that prevents ECM-induced mortality in co-infected mice., (© 2017 Agency for Science, Technology and Research (A*STAR). Published under the terms of the CC BY 4.0 license.)- Published
- 2018
- Full Text
- View/download PDF
17. Adaptive immunity is essential in preventing recrudescence of Plasmodium yoelii malaria parasites after artesunate treatment.
- Author
-
Claser C, Chang ZW, Russell B, and Rénia L
- Subjects
- Adaptive Immunity immunology, Animals, Artesunate, Disease Models, Animal, Drug Resistance, Female, Malaria immunology, Malaria parasitology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Knockout, Recurrence, Antimalarials therapeutic use, Artemisinins therapeutic use, B-Lymphocytes immunology, CD4-Positive T-Lymphocytes immunology, Malaria drug therapy, Plasmodium yoelii drug effects, Plasmodium yoelii immunology
- Abstract
Artemisinin-based antimalarials, such as artesunate (ART), alone or in combination, are the mainstay of the therapy against malaria caused by Plasmodium falciparum. However, the emergence and spread of artemisinin resistance threatens the future success of its global malaria eradication. Although much of the reported artemisinin resistance can be attributed to mutations intrinsic to the parasite, a significant proportion of treatment failures are thought to be due to other factors such as the host's immune system. Exactly how the immune system participates in the clearance and elimination of malaria parasites during ART treatment is unknown. Here, we show that a developing primary immune response, involving both B and CD4
+ T cells, is necessary for the complete elimination but not initial clearance, of Plasmodium yoelii YM parasites in mice treated with ART. Our study uncovers a dynamic interplay between ART and host adaptive immunity in Plasmodium sp. elimination., (© 2017 John Wiley & Sons Ltd.)- Published
- 2017
- Full Text
- View/download PDF
18. Host Resistance to Plasmodium-Induced Acute Immune Pathology Is Regulated by Interleukin-10 Receptor Signaling.
- Author
-
Claser C, De Souza JB, Thorburn SG, Grau GE, Riley EM, Rénia L, and Hafalla JCR
- Subjects
- Animals, Antibodies, Blocking administration & dosage, Antibodies, Neutralizing administration & dosage, Brain pathology, CD8-Positive T-Lymphocytes drug effects, Erythrocytes drug effects, Erythrocytes parasitology, Female, Liver pathology, Lymphocyte Activation immunology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Parasitemia immunology, Receptors, Interleukin-10 antagonists & inhibitors, Signal Transduction, CD8-Positive T-Lymphocytes immunology, Interferon-gamma blood, Malaria, Cerebral immunology, Plasmodium berghei immunology, Receptors, Interleukin-10 immunology
- Abstract
The resolution of malaria infection is dependent on a balance between proinflammatory and regulatory immune responses. While early effector T cell responses are required for limiting parasitemia, these responses need to be switched off by regulatory mechanisms in a timely manner to avoid immune-mediated tissue damage. Interleukin-10 receptor (IL-10R) signaling is considered to be a vital component of regulatory responses, although its role in host resistance to severe immune pathology during acute malaria infections is not fully understood. In this study, we have determined the contribution of IL-10R signaling to the regulation of immune responses during Plasmodium berghei ANKA-induced experimental cerebral malaria (ECM). We show that antibody-mediated blockade of the IL-10R during P. berghei ANKA infection in ECM-resistant BALB/c mice leads to amplified T cell activation, higher serum gamma interferon (IFN-γ) concentrations, enhanced intravascular accumulation of both parasitized red blood cells and CD8
+ T cells to the brain, and an increased incidence of ECM. Importantly, the pathogenic effects of IL-10R blockade during P. berghei ANKA infection were reversible by depletion of T cells and neutralization of IFN-γ. Our findings underscore the importance of IL-10R signaling in preventing T-cell- and cytokine-mediated pathology during potentially lethal malaria infections., (Copyright © 2017 Claser et al.)- Published
- 2017
- Full Text
- View/download PDF
19. Ex Vivo Maturation Assay for Testing Antimalarial Sensitivity of Rodent Malaria Parasites.
- Author
-
Chang ZW, Malleret B, Russell B, Rénia L, and Claser C
- Subjects
- Animals, Disease Models, Animal, Female, Flow Cytometry methods, Malaria drug therapy, Male, Mice, Inbred C57BL, Microbial Sensitivity Tests, Plasmodium pathogenicity, Plasmodium berghei drug effects, Plasmodium berghei pathogenicity, Plasmodium berghei physiology, Antimalarials pharmacology, Drug Evaluation, Preclinical methods, Malaria parasitology, Plasmodium drug effects, Plasmodium physiology
- Abstract
Ex vivo assay systems provide a powerful approach to studying human malaria parasite biology and to testing antimalarials. For rodent malaria parasites, short-term in vitro culture and ex vivo antimalarial susceptibility assays are relatively cumbersome, relying on in vivo passage for synchronization, since ring-stage parasites are an essential starting material. Here, we describe a new approach based on the enrichment of ring-stage Plasmodium berghei, P. yoelii, and P. vinckei vinckei using a single-step Percoll gradient. Importantly, we demonstrate that the enriched ring-stage parasites develop synchronously regardless of the parasite strain or species used. Using a flow cytometry assay with Hoechst and ethidium or MitoTracker dye, we show that parasite development is easily and rapidly monitored. Finally, we demonstrate that this approach can be used to screen antimalarial drugs., (Copyright © 2016, American Society for Microbiology. All Rights Reserved.)
- Published
- 2016
- Full Text
- View/download PDF
20. Tissue-Resident CD169(+) Macrophages Form a Crucial Front Line against Plasmodium Infection.
- Author
-
Gupta P, Lai SM, Sheng J, Tetlak P, Balachander A, Claser C, Renia L, Karjalainen K, and Ruedl C
- Subjects
- Animals, Erythrocytes parasitology, Hemeproteins metabolism, Macrophages pathology, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Macrophages parasitology, Malaria immunology, Plasmodium berghei parasitology, Sialic Acid Binding Ig-like Lectin 1 genetics
- Abstract
Tissue macrophages exhibit diverse functions, ranging from the maintenance of tissue homeostasis, including clearance of senescent erythrocytes and cell debris, to modulation of inflammation and immunity. Their contribution to the control of blood-stage malaria remains unclear. Here, we show that in the absence of tissue-resident CD169(+) macrophages, Plasmodium berghei ANKA (PbA) infection results in significantly increased parasite sequestration, leading to vascular occlusion and leakage and augmented tissue deposition of the malarial pigment hemozoin. This leads to widespread tissue damage culminating in multiple organ inflammation. Thus, the capacity of CD169(+) macrophages to contain the parasite burden and its sequestration into different tissues and to limit infection-induced inflammation is crucial to mitigating Plasmodium infection and pathogenesis., (Copyright © 2016 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
21. Measuring antigen presentation in mouse brain endothelial cells ex vivo and in vitro.
- Author
-
Howland SW, Gun SY, Claser C, Poh CM, and Rénia L
- Subjects
- Animals, Brain blood supply, Brain cytology, Cell Culture Techniques, Cell Line, Cells, Cultured, Disease Models, Animal, Endothelial Cells cytology, Major Histocompatibility Complex, Malaria immunology, Mice, Mice, Inbred C57BL, Microvessels cytology, Plasmodium berghei immunology, Receptors, Antigen, T-Cell immunology, Antigen Presentation, Brain immunology, Endothelial Cells immunology, Microvessels immunology
- Abstract
We have recently demonstrated that brain endothelial cells cross-present parasite antigen during mouse experimental cerebral malaria (ECM). Here we describe a 2-d protocol to detect cross-presentation by isolating the brain microvessels and incubating them with a reporter cell line that expresses lacZ upon detection of the relevant peptide-major histocompatibility complex. After X-gal staining, a typical positive result consists of hundreds of blue spots, compared with fewer than 20 spots from a naive brain. The assay is generalizable to other disease contexts by using reporter cells that express appropriate specific T cell receptors. Also described is the protocol for culturing endothelial cells from brain microvessels isolated from naive mice. After 7-10 d, an in vitro cross-presentation assay can be performed by adding interferon-γ, antigen (e.g., Plasmodium berghei-infected red blood cells) and reporter cells in sequence over 3 d. This is useful for comparing different antigen forms or for probing the effects of various interventions.
- Published
- 2015
- Full Text
- View/download PDF
22. Immunization with the MAEBL M2 Domain Protects against Lethal Plasmodium yoelii Infection.
- Author
-
Leite JA, Bargieri DY, Carvalho BO, Albrecht L, Lopes SC, Kayano AC, Farias AS, Chia WN, Claser C, Malleret B, Russell B, Castiñeiras C, Santos LM, Brocchi M, Wunderlich G, Soares IS, Rodrigues MM, Rénia L, and Costa FT
- Subjects
- Animals, Antibodies, Protozoan immunology, Erythrocytes parasitology, Female, Humans, Immunization, Malaria immunology, Malaria mortality, Malaria parasitology, Malaria Vaccines administration & dosage, Malaria Vaccines chemistry, Malaria Vaccines genetics, Male, Merozoites chemistry, Merozoites growth & development, Merozoites immunology, Mice, Mice, Inbred C57BL, Plasmodium yoelii chemistry, Plasmodium yoelii genetics, Plasmodium yoelii growth & development, Protein Structure, Tertiary, Protozoan Proteins administration & dosage, Protozoan Proteins genetics, Sporozoites chemistry, Sporozoites growth & development, Sporozoites immunology, Malaria prevention & control, Malaria Vaccines immunology, Plasmodium yoelii immunology, Protozoan Proteins chemistry, Protozoan Proteins immunology
- Abstract
Malaria remains a world-threatening disease largely because of the lack of a long-lasting and fully effective vaccine. MAEBL is a type 1 transmembrane molecule with a chimeric cysteine-rich ectodomain homologous to regions of the Duffy binding-like erythrocyte binding protein and apical membrane antigen 1 (AMA1) antigens. Although MAEBL does not appear to be essential for the survival of blood-stage forms, ectodomains M1 and M2, homologous to AMA1, seem to be involved in parasite attachment to erythrocytes, especially M2. MAEBL is necessary for sporozoite infection of mosquito salivary glands and is expressed in liver stages. Here, the Plasmodium yoelii MAEBL-M2 domain was expressed in a prokaryotic vector. C57BL/6J mice were immunized with doses of P. yoelii recombinant protein rPyM2-MAEBL. High levels of antibodies, with balanced IgG1 and IgG2c subclasses, were achieved. rPyM2-MAEBL antisera were capable of recognizing the native antigen. Anti-MAEBL antibodies recognized different MAEBL fragments expressed in CHO cells, showing stronger IgM and IgG responses to the M2 domain and repeat region, respectively. After a challenge with P. yoelii YM (lethal strain)-infected erythrocytes (IE), up to 90% of the immunized animals survived and a reduction of parasitemia was observed. Moreover, splenocytes harvested from immunized animals proliferated in a dose-dependent manner in the presence of rPyM2-MAEBL. Protection was highly dependent on CD4(+), but not CD8(+), T cells toward Th1. rPyM2-MAEBL antisera were also able to significantly inhibit parasite development, as observed in ex vivo P. yoelii erythrocyte invasion assays. Collectively, these findings support the use of MAEBL as a vaccine candidate and open perspectives to understand the mechanisms involved in protection., (Copyright © 2015, American Society for Microbiology. All Rights Reserved.)
- Published
- 2015
- Full Text
- View/download PDF
23. Pathogenic CD8+ T cells in experimental cerebral malaria.
- Author
-
Howland SW, Claser C, Poh CM, Gun SY, and Rénia L
- Subjects
- Animals, CD8-Positive T-Lymphocytes metabolism, Cell Movement, Cytotoxicity, Immunologic, Disease Models, Animal, Epitopes, T-Lymphocyte chemistry, Epitopes, T-Lymphocyte immunology, Extracellular Matrix immunology, Humans, Immunomodulation, Malaria, Cerebral parasitology, Phenotype, Plasmodium immunology, CD8-Positive T-Lymphocytes immunology, Host-Parasite Interactions immunology, Malaria, Cerebral immunology
- Abstract
Cerebral malaria (CM) is one the major complications occurring during malaria infection. The mechanisms leading to this syndrome are still not completely understood. Although it is clear that parasite sequestration is the key initiation factor, the downstream pathological processes are still highly debated. The experimental cerebral malaria (ECM) model, in which susceptible mice are infected with Plasmodium berghei ANKA, has led to the identification of CD8(+) T cells as the major mediator of ECM death. In this review, we discuss the recent advances and future developments in the understanding of the role of CD8(+) T cells in CM.
- Published
- 2015
- Full Text
- View/download PDF
24. Plasmodium vivax: restricted tropism and rapid remodeling of CD71-positive reticulocytes.
- Author
-
Malleret B, Li A, Zhang R, Tan KS, Suwanarusk R, Claser C, Cho JS, Koh EG, Chu CS, Pukrittayakamee S, Ng ML, Ginhoux F, Ng LG, Lim CT, Nosten F, Snounou G, Rénia L, and Russell B
- Subjects
- Biomechanical Phenomena, Cells, Cultured, Erythrocyte Deformability, Humans, Malaria, Vivax blood, Malaria, Vivax parasitology, Reticulocytes metabolism, Antigens, CD metabolism, Plasmodium vivax growth & development, Receptors, Transferrin metabolism, Reticulocytes parasitology, Reticulocytes physiology, Tropism physiology
- Abstract
Plasmodium vivax merozoites only invade reticulocytes, a minor though heterogeneous population of red blood cell precursors that can be graded by levels of transferrin receptor (CD71) expression. The development of a protocol that allows sorting reticulocytes into defined developmental stages and a robust ex vivo P vivax invasion assay has made it possible for the first time to investigate the fine-scale invasion preference of P vivax merozoites. Surprisingly, it was the immature reticulocytes (CD71(+)) that are generally restricted to the bone marrow that were preferentially invaded, whereas older reticulocytes (CD71(-)), principally found in the peripheral blood, were rarely invaded. Invasion assays based on the CD71(+) reticulocyte fraction revealed substantial postinvasion modification. Thus, 3 to 6 hours after invasion, the initially biomechanically rigid CD71(+) reticulocytes convert into a highly deformable CD71(-) infected red blood cell devoid of host reticular matter, a process that normally spans 24 hours for uninfected reticulocytes. Concurrent with these changes, clathrin pits disappear by 3 hours postinvasion, replaced by distinctive caveolae nanostructures. These 2 hitherto unsuspected features of P vivax invasion, a narrow preference for immature reticulocytes and a rapid remodeling of the host cell, provide important insights pertinent to the pathobiology of the P vivax infection., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
25. Spatiotemporal requirements for IRF7 in mediating type I IFN-dependent susceptibility to blood-stage Plasmodium infection.
- Author
-
Edwards CL, Best SE, Gun SY, Claser C, James KR, de Oca MM, Sebina I, Rivera Fde L, Amante FH, Hertzog PJ, Engwerda CR, Renia L, and Haque A
- Subjects
- Animals, Antibodies, Monoclonal pharmacology, Brain drug effects, Brain parasitology, Disease Susceptibility, Erythrocytes parasitology, Female, Gene Expression Regulation, Host-Parasite Interactions, Interferon Regulatory Factor-7 genetics, Malaria, Cerebral parasitology, Mice, Mice, Inbred C57BL, Plasmodium berghei immunology, Plasmodium chabaudi immunology, Receptor, Interferon alpha-beta antagonists & inhibitors, Receptor, Interferon alpha-beta genetics, Signal Transduction, Spleen drug effects, Spleen parasitology, Th1 Cells parasitology, Time Factors, Brain immunology, Interferon Regulatory Factor-7 immunology, Malaria, Cerebral immunology, Receptor, Interferon alpha-beta immunology, Spleen immunology, Th1 Cells immunology
- Abstract
Type I IFN signaling suppresses splenic T helper 1 (Th1) responses during blood-stage Plasmodium berghei ANKA (PbA) infection in mice, and is crucial for mediating tissue accumulation of parasites and fatal cerebral symptoms via mechanisms that remain to be fully characterized. Interferon regulatory factor 7 (IRF7) is considered to be a master regulator of type I IFN responses. Here, we assessed IRF7 for its roles during lethal PbA infection and nonlethal Plasmodium chabaudi chabaudi AS (PcAS) infection as two distinct models of blood-stage malaria. We found that IRF7 was not essential for tissue accumulation of parasites, cerebral symptoms, or brain pathology. Using timed administration of anti-IFNAR1 mAb, we show that late IFNAR1 signaling promotes fatal disease via IRF7-independent mechanisms. Despite this, IRF7 significantly impaired early splenic Th1 responses and limited control of parasitemia during PbA infection. Finally, IRF7 also suppressed antiparasitic immunity and Th1 responses during nonlethal PcAS infection. Together, our data support a model in which IRF7 suppresses antiparasitic immunity in the spleen, while IFNAR1-mediated, but IRF7-independent, signaling contributes to pathology in the brain during experimental blood-stage malaria., (© 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2015
- Full Text
- View/download PDF
26. CD41 is a reliable identification and activation marker for murine basophils in the steady state and during helminth and malarial infections.
- Author
-
Bakocevic N, Claser C, Yoshikawa S, Jones LA, Chew S, Goh CC, Malleret B, Larbi A, Ginhoux F, de Lafaille MC, Karasuyama H, Renia L, and Ng LG
- Subjects
- Animals, Antibodies, Helminth immunology, Basophils pathology, Female, Immunoglobulin E immunology, Interleukin-3 immunology, Interleukin-4 immunology, Malaria pathology, Membrane Glycoproteins immunology, Mice, Mice, Inbred BALB C, Mice, Knockout, Strongylida Infections pathology, Basophils immunology, Malaria immunology, Nippostrongylus immunology, Plasmodium yoelii immunology, Platelet Membrane Glycoprotein IIb immunology, Strongylida Infections immunology
- Abstract
Basophils, a rare leukocyte population in peripheral circulation, are conventionally identified as CD45(int) CD49b(+) FcεRI(+) cells. Here, we show that basophils from blood and several organs of naïve wild-type mice express CD41, the α subunit of α(IIb)β₃ integrin. CD41 expression on basophils is upregulated after in vivo IL-3 treatment and during infection with Nippostrongylus brasiliensis (Nb). Moreover, CD41 can be used as a reliable marker for basophils, circumventing technical difficulties associated with FcεRI for basophil identification in a Nb infection model. In vitro anti-IgE cross-linking and IL-3 basophil stimulation showed that CD41 upregulation positively correlates with augmented surface expression of CD200R and increased production of IL-4/IL-13, indicating that CD41 is a basophil activation marker. Furthermore, we found that infection with Plasmodium yoelii 17X (Py17x) induced a profound basophilia and using Mcpt8(DTR) reporter mice as a basophil-specific depletion model, we verified that CD41 can be used as a marker to track basophils in the steady state and during infection. During malarial infection, CD41 expression on basophils is negatively regulated by IFN-γ and positively correlates with increased basophil IL-4 production. In conclusion, we provide evidence that CD41 can be used as both an identification and activation marker for basophils during homeostasis and immune challenge., (© 2014 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim.)
- Published
- 2014
- Full Text
- View/download PDF
27. Rodent Plasmodium-infected red blood cells: imaging their fates and interactions within their hosts.
- Author
-
Claser C, Malleret B, Peng K, Bakocevic N, Gun SY, Russell B, Ng LG, and Rénia L
- Subjects
- Animals, Plasmodium physiology, Rodentia, Luminescent Measurements methods, Magnetic Resonance Imaging methods, Microscopy methods, Plasmodium cytology, Positron-Emission Tomography methods
- Abstract
Malaria, a disease caused by the Plasmodium parasite, remains one of the most deadly infectious diseases known to mankind. The parasite has a complex life cycle, of which only the erythrocytic stage is responsible for the diverse pathologies induced during infection. To date, the disease mechanisms that underlie these pathologies are still poorly understood. In the case of infections caused by Plasmodium falciparum, the species responsible for most malaria related deaths, pathogenesis is thought to be due to the sequestration of infected red blood cells (IRBCs) in deep tissues. Other human and rodent malaria parasite species are also known to exhibit sequestration. Here, we review the different techniques that allow researchers to study how rodent malaria parasites modify their host cells, the distribution of IRBCs in vivo as well as the interactions between IRBCs and host tissues., (© 2013. Published by Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2014
- Full Text
- View/download PDF
28. Interferons and interferon regulatory factors in malaria.
- Author
-
Gun SY, Claser C, Tan KS, and Rénia L
- Subjects
- Animals, Humans, Interferon Regulatory Factors metabolism, Interferons metabolism, Malaria metabolism
- Abstract
Malaria is one of the most serious infectious diseases in humans and responsible for approximately 500 million clinical cases and 500 thousand deaths annually. Acquired adaptive immune responses control parasite replication and infection-induced pathologies. Most infections are clinically silent which reflects on the ability of adaptive immune mechanisms to prevent the disease. However, a minority of these can become severe and life-threatening, manifesting a range of overlapping syndromes of complex origins which could be induced by uncontrolled immune responses. Major players of the innate and adaptive responses are interferons. Here, we review their roles and the signaling pathways involved in their production and protection against infection and induced immunopathologies.
- Published
- 2014
- Full Text
- View/download PDF
29. Brain microvessel cross-presentation is a hallmark of experimental cerebral malaria.
- Author
-
Howland SW, Poh CM, Gun SY, Claser C, Malleret B, Shastri N, Ginhoux F, Grotenbreg GM, and Rénia L
- Subjects
- Amino Acid Sequence, Animals, Antigens, Protozoan analysis, Antimalarials therapeutic use, Blood-Brain Barrier immunology, Blood-Brain Barrier parasitology, Blood-Brain Barrier pathology, Brain immunology, Brain pathology, CD8-Positive T-Lymphocytes chemistry, CD8-Positive T-Lymphocytes parasitology, Female, Humans, Malaria, Cerebral drug therapy, Malaria, Cerebral parasitology, Malaria, Cerebral pathology, Mice, Mice, Inbred C57BL, Microvessels immunology, Microvessels parasitology, Microvessels pathology, Molecular Sequence Data, Parasite Load, Plasmodium berghei drug effects, Receptors, Antigen, T-Cell, alpha-beta chemistry, Receptors, Antigen, T-Cell, alpha-beta immunology, Antigens, Protozoan immunology, Brain blood supply, Brain parasitology, CD8-Positive T-Lymphocytes immunology, Cross-Priming drug effects, Malaria, Cerebral immunology, Plasmodium berghei immunology
- Abstract
Cerebral malaria is a devastating complication of Plasmodium falciparum infection. Its pathogenesis is complex, involving both parasite- and immune-mediated events. CD8(+) T cells play an effector role in murine experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA (PbA) infection. We have identified a highly immunogenic CD8 epitope in glideosome-associated protein 50 that is conserved across rodent malaria species. Epitope-specific CD8(+) T cells are induced during PbA infection, migrating to the brain just before neurological signs manifest. They are functional, cytotoxic and can damage the blood-brain barrier in vivo. Such CD8(+) T cells are also found in the brain during infection with parasite strains/species that do not induce neuropathology. We demonstrate here that PbA infection causes brain microvessels to cross-present parasite antigen, while non-ECM-causing parasites do not. Further, treatment with fast-acting anti-malarial drugs before the onset of ECM reduces parasite load and thus antigen presentation in the brain, preventing ECM death. Thus our data suggest that combined therapies targeting both the parasite and host antigen-presenting cells may improve the outcome of CM patients., (© 2013 The Authors. Published by John Wiley and Sons, Ltd on behalf of EMBO.)
- Published
- 2013
- Full Text
- View/download PDF
30. Inflammatory Flt3l is essential to mobilize dendritic cells and for T cell responses during Plasmodium infection.
- Author
-
Guermonprez P, Helft J, Claser C, Deroubaix S, Karanje H, Gazumyan A, Darasse-Jèze G, Telerman SB, Breton G, Schreiber HA, Frias-Staheli N, Billerbeck E, Dorner M, Rice CM, Ploss A, Klein F, Swiecki M, Colonna M, Kamphorst AO, Meredith M, Niec R, Takacs C, Mikhail F, Hari A, Bosque D, Eisenreich T, Merad M, Shi Y, Ginhoux F, Rénia L, Urban BC, and Nussenzweig MC
- Subjects
- Animals, CD8 Antigens analysis, Cell Movement, Female, Humans, Interferon Type I physiology, Male, Mast Cells physiology, Mice, Mice, Inbred C57BL, Toll-Like Receptors physiology, Uric Acid metabolism, Uric Acid pharmacology, Dendritic Cells physiology, Malaria immunology, Membrane Proteins physiology, T-Lymphocytes immunology
- Abstract
Innate sensing mechanisms trigger a variety of humoral and cellular events that are essential to adaptive immune responses. Here we describe an innate sensing pathway triggered by Plasmodium infection that regulates dendritic cell homeostasis and adaptive immunity through Flt3 ligand (Flt3l) release. Plasmodium-induced Flt3l release in mice requires Toll-like receptor (TLR) activation and type I interferon (IFN) production. We found that type I IFN supports the upregulation of xanthine dehydrogenase, which metabolizes the xanthine accumulating in infected erythrocytes to uric acid. Uric acid crystals trigger mast cells to release soluble Flt3l from a pre-synthesized membrane-associated precursor. During infection, Flt3l preferentially stimulates expansion of the CD8-α(+) dendritic cell subset or its BDCA3(+) human dendritic cell equivalent and has a substantial impact on the magnitude of T cell activation, mostly in the CD8(+) compartment. Our findings highlight a new mechanism that regulates dendritic cell homeostasis and T cell responses to infection.
- Published
- 2013
- Full Text
- View/download PDF
31. A pathogenic role for CD4+ T cells during Chikungunya virus infection in mice.
- Author
-
Teo TH, Lum FM, Claser C, Lulla V, Lulla A, Merits A, Rénia L, and Ng LF
- Subjects
- Adaptive Immunity genetics, Alphavirus Infections genetics, Animals, Arthritis, Experimental genetics, Arthritis, Experimental immunology, Arthritis, Experimental virology, CD4 Antigens genetics, CD4-Positive T-Lymphocytes pathology, Cell Movement genetics, Cell Movement immunology, Chikungunya Fever, DNA-Binding Proteins deficiency, DNA-Binding Proteins genetics, Female, Interferon-gamma deficiency, Interferon-gamma genetics, Lymphocyte Depletion, Mice, Mice, Inbred C57BL, Mice, Knockout, Signal Transduction genetics, Signal Transduction immunology, Vero Cells, Alphavirus Infections immunology, Alphavirus Infections pathology, CD4-Positive T-Lymphocytes immunology, CD4-Positive T-Lymphocytes virology, Chikungunya virus immunology
- Abstract
Chikungunya virus (CHIKV) is an alphavirus that causes chronic and incapacitating arthralgia in humans. Injury to the joint is believed to occur because of viral and host immune-mediated effects. However, the exact involvement of the different immune mediators in CHIKV-induced pathogenesis is unknown. In this study, we assessed the roles of T cells in primary CHIKV infection, virus replication and dissemination, and virus persistence, as well as in the mediation of disease severity in adult RAG2(-/-), CD4(-/-), CD8(-/-), and wild-type CHIKV C57BL/6J mice and in wild-type mice depleted of CD4(+) or CD8(+) T cells after Ab treatment. CHIKV-specific T cells in the spleen and footpad were investigated using IFN-γ ELISPOT. Interestingly, our results indicated that CHIKV-specific CD4(+), but not CD8(+), T cells are essential for the development of joint swelling without any effect on virus replication and dissemination. Infection in IFN-γ(-/-) mice demonstrated that pathogenic CD4(+) T cells do not mediate inflammation via an IFN-γ-mediated pathway. Taken together, these observations strongly indicate that mechanisms of joint pathology induced by CHIKV in mice resemble those in humans and differ from infections caused by other arthritogenic viruses, such as Ross River virus.
- Published
- 2013
- Full Text
- View/download PDF
32. Cutting edge: Clec9A+ dendritic cells mediate the development of experimental cerebral malaria.
- Author
-
Piva L, Tetlak P, Claser C, Karjalainen K, Renia L, and Ruedl C
- Subjects
- Animals, CD11c Antigen biosynthesis, Cell Death immunology, Clone Cells, Dendritic Cells parasitology, Diphtheria Toxin administration & dosage, Diphtheria Toxin toxicity, Disease Models, Animal, Disease Resistance genetics, Disease Resistance immunology, Female, Humans, Lectins, C-Type biosynthesis, Malaria, Cerebral genetics, Male, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Mice, Transgenic, Plasmodium berghei immunology, Receptors, Immunologic biosynthesis, Dendritic Cells immunology, Dendritic Cells metabolism, Lectins, C-Type physiology, Malaria, Cerebral immunology, Malaria, Cerebral pathology, Receptors, Immunologic physiology
- Abstract
Plasmodium infections trigger strong innate and acquired immune responses, which can lead to severe complications, including the most feared and often fatal cerebral malaria (CM). To begin to dissect the roles of different dendritic cell (DC) subsets in Plasmodium-induced pathology, we have generated a transgenic strain, Clec9A-diphtheria toxin receptor that allows us to ablate in vivo Clec9A(+) DCs. Specifically, we have analyzed the in vivo contribution of this DC subset in an experimental CM model using Plasmodium berghei, and we provide strong evidence that the absence of this DC subset resulted in complete resistance to experimental CM. This was accompanied with dramatic reduction of brain CD8(+) T cells, and those few cerebral CD8(+) T cells present had a less activated phenotype, unlike their wildtype counterparts that expressed IFN-γ and especially granzyme B. This almost complete absence of local cellular responses was also associated with reduced parasite load in the brain.
- Published
- 2012
- Full Text
- View/download PDF
33. Cerebral malaria: mysteries at the blood-brain barrier.
- Author
-
Rénia L, Howland SW, Claser C, Charlotte Gruner A, Suwanarusk R, Hui Teo T, Russell B, and Ng LF
- Subjects
- Animals, Disease Models, Animal, Humans, Mice, Blood-Brain Barrier immunology, Blood-Brain Barrier parasitology, Host-Pathogen Interactions, Malaria, Cerebral parasitology, Malaria, Cerebral pathology, Plasmodium falciparum immunology, Plasmodium falciparum pathogenicity
- Abstract
Cerebral malaria is the most severe pathology caused by the malaria parasite, Plasmodium falciparum. The pathogenic mechanisms leading to cerebral malaria are still poorly defined as studies have been hampered by limited accessibility to human tissues. Nevertheless, histopathology of post-mortem human tissues and mouse models of cerebral malaria have indicated involvement of the blood-brain barrier in cerebral malaria. In contrast to viruses and bacteria, malaria parasites do not infiltrate and infect the brain parenchyma. Instead, rupture of the blood-brain barrier occurs and may lead to hemorrhages resulting in neurological alterations. Here, we review the most recent findings from human studies and mouse models on the interactions of malaria parasites and the blood-brain barrier, shedding light on the pathogenesis of cerebral malaria, which may provide directions for possible interventions.
- Published
- 2012
- Full Text
- View/download PDF
34. The CTLA-4 and PD-1/PD-L1 inhibitory pathways independently regulate host resistance to Plasmodium-induced acute immune pathology.
- Author
-
Hafalla JC, Claser C, Couper KN, Grau GE, Renia L, de Souza JB, and Riley EM
- Subjects
- Animals, Antigens, Differentiation metabolism, B7-H1 Antigen metabolism, CD8-Positive T-Lymphocytes metabolism, CTLA-4 Antigen metabolism, Erythrocytes parasitology, Interferon-gamma immunology, Lymphocyte Activation immunology, Malaria, Cerebral immunology, Malaria, Cerebral microbiology, Malaria, Cerebral pathology, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Plasmodium berghei immunology, Programmed Cell Death 1 Receptor, Antigens, Differentiation immunology, B7-H1 Antigen immunology, CD8-Positive T-Lymphocytes immunology, CTLA-4 Antigen immunology, Plasmodium berghei pathogenicity
- Abstract
The balance between pro-inflammatory and regulatory immune responses in determining optimal T cell activation is vital for the successful resolution of microbial infections. This balance is maintained in part by the negative regulators of T cell activation, CTLA-4 and PD-1/PD-L, which dampen effector responses during chronic infections. However, their role in acute infections, such as malaria, remains less clear. In this study, we determined the contribution of CTLA-4 and PD-1/PD-L to the regulation of T cell responses during Plasmodium berghei ANKA (PbA)-induced experimental cerebral malaria (ECM) in susceptible (C57BL/6) and resistant (BALB/c) mice. We found that the expression of CTLA-4 and PD-1 on T cells correlates with the extent of pro-inflammatory responses induced during PbA infection, being higher in C57BL/6 than in BALB/c mice. Thus, ECM develops despite high levels of expression of these inhibitory receptors. However, antibody-mediated blockade of either the CTLA-4 or PD-1/PD-L1, but not the PD-1/PD-L2, pathways during PbA-infection in ECM-resistant BALB/c mice resulted in higher levels of T cell activation, enhanced IFN-γ production, increased intravascular arrest of both parasitised erythrocytes and CD8(+) T cells to the brain, and augmented incidence of ECM. Thus, in ECM-resistant BALB/c mice, CTLA-4 and PD-1/PD-L1 represent essential, independent and non-redundant pathways for maintaining T cell homeostasis during a virulent malaria infection. Moreover, neutralisation of IFN-γ or depletion of CD8(+) T cells during PbA infection was shown to reverse the pathologic effects of regulatory pathway blockade, highlighting that the aetiology of ECM in the BALB/c mice is similar to that in C57BL/6 mice. In summary, our results underscore the differential and complex regulation that governs immune responses to malaria parasites.
- Published
- 2012
- Full Text
- View/download PDF
35. CD8+ T cells and IFN-γ mediate the time-dependent accumulation of infected red blood cells in deep organs during experimental cerebral malaria.
- Author
-
Claser C, Malleret B, Gun SY, Wong AY, Chang ZW, Teo P, See PC, Howland SW, Ginhoux F, and Rénia L
- Subjects
- Animals, Biomass, Brain immunology, Brain parasitology, Brain pathology, DNA-Binding Proteins metabolism, Erythrocytes immunology, Female, Lymphocyte Depletion, Male, Mice, Mice, Inbred C57BL, Myeloid Cells immunology, Plasmodium berghei physiology, Time Factors, CD8-Positive T-Lymphocytes immunology, Erythrocytes parasitology, Interferon-gamma metabolism, Malaria, Cerebral immunology, Malaria, Cerebral parasitology, Organ Specificity immunology
- Abstract
Background: Infection with Plasmodium berghei ANKA (PbA) in susceptible mice induces a syndrome called experimental cerebral malaria (ECM) with severe pathologies occurring in various mouse organs. Immune mediators such as T cells or cytokines have been implicated in the pathogenesis of ECM. Red blood cells infected with PbA parasites have been shown to accumulate in the brain and other tissues during infection. This accumulation is thought to be involved in PbA-induced pathologies, which mechanisms are poorly understood., Methods and Findings: Using transgenic PbA parasites expressing the luciferase protein, we have assessed by real-time in vivo imaging the dynamic and temporal contribution of different immune factors in infected red blood cell (IRBC) accumulation and distribution in different organs during PbA infection. Using deficient mice or depleting antibodies, we observed that CD8(+) T cells and IFN-γ drive the rapid increase in total parasite biomass and accumulation of IRBC in the brain and in different organs 6-12 days post-infection, at a time when mice develop ECM. Other cells types like CD4(+) T cells, monocytes or neutrophils or cytokines such as IL-12 and TNF-α did not influence the early increase of total parasite biomass and IRBC accumulation in different organs., Conclusions: CD8(+) T cells and IFN-γ are the major immune mediators controlling the time-dependent accumulation of P. berghei-infected red blood cells in tissues.
- Published
- 2011
- Full Text
- View/download PDF
36. A rapid and robust tri-color flow cytometry assay for monitoring malaria parasite development.
- Author
-
Malleret B, Claser C, Ong AS, Suwanarusk R, Sriprawat K, Howland SW, Russell B, Nosten F, and Rénia L
- Subjects
- Animals, Antimalarials pharmacology, Drug Evaluation, Preclinical, Fluorescent Dyes, Green Fluorescent Proteins genetics, Humans, Malaria blood, Malaria diagnosis, Malaria drug therapy, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Parasitemia parasitology, Plasmodium genetics, Plasmodium isolation & purification, Plasmodium berghei growth & development, Plasmodium vivax growth & development, Plasmodium yoelii growth & development, Staining and Labeling, Flow Cytometry methods, Malaria parasitology, Plasmodium growth & development
- Abstract
Microscopic examination of Giemsa-stained thin blood smears remains the gold standard method used to quantify and stage malaria parasites. However, this technique is tedious, and requires trained microscopists. We have developed a fast and simple flow cytometry method to quantify and stage, various malaria parasites in red blood cells in whole blood or in vitro cultured Plasmodium falciparum. The parasites were stained with dihydroethidium and Hoechst 33342 or SYBR Green I and leukocytes were identified with an antibody against CD45. Depending on the DNA stains used, samples were analyzed using different models of flow cytometers. This protocol, which does not require any washing steps, allows infected red blood cells to be distinguished from leukocytes, as well as allowing non-infected reticulocytes and normocytes to be identified. It also allows assessing the proportion of parasites at different developmental stages. Lastly, we demonstrate how this technique can be applied to antimalarial drug testing.
- Published
- 2011
- Full Text
- View/download PDF
37. Adaptive immunity against Leishmania nucleoside hydrolase maps its c-terminal domain as the target of the CD4+ T cell-driven protective response.
- Author
-
Nico D, Claser C, Borja-Cabrera GP, Travassos LR, Palatnik M, Soares IS, Rodrigues MM, and Palatnik-de-Sousa CB
- Subjects
- Amino Acid Sequence, Animals, CD4-Positive T-Lymphocytes parasitology, Epitope Mapping, Female, Humans, Interferon-gamma immunology, Interleukin-10 immunology, Leishmania donovani chemistry, Leishmania donovani immunology, Leishmaniasis, Visceral parasitology, Mice, Mice, Inbred BALB C, Molecular Sequence Data, N-Glycosyl Hydrolases genetics, Protein Structure, Tertiary, Protozoan Proteins genetics, Adaptive Immunity, CD4-Positive T-Lymphocytes immunology, Leishmania donovani enzymology, Leishmaniasis, Visceral immunology, N-Glycosyl Hydrolases chemistry, N-Glycosyl Hydrolases immunology, Protozoan Proteins chemistry, Protozoan Proteins immunology
- Abstract
Nucleoside hydrolases (NHs) show homology among parasite protozoa, fungi and bacteria. They are vital protagonists in the establishment of early infection and, therefore, are excellent candidates for the pathogen recognition by adaptive immune responses. Immune protection against NHs would prevent disease at the early infection of several pathogens. We have identified the domain of the NH of L. donovani (NH36) responsible for its immunogenicity and protective efficacy against murine visceral leishmaniasis (VL). Using recombinant generated peptides covering the whole NH36 sequence and saponin we demonstrate that protection against L. chagasi is related to its C-terminal domain (amino-acids 199-314) and is mediated mainly by a CD4+ T cell driven response with a lower contribution of CD8+ T cells. Immunization with this peptide exceeds in 36.73±12.33% the protective response induced by the cognate NH36 protein. Increases in IgM, IgG2a, IgG1 and IgG2b antibodies, CD4+ T cell proportions, IFN-γ secretion, ratios of IFN-γ/IL-10 producing CD4+ and CD8+ T cells and percents of antibody binding inhibition by synthetic predicted epitopes were detected in F3 vaccinated mice. The increases in DTH and in ratios of TNFα/IL-10 CD4+ producing cells were however the strong correlates of protection which was confirmed by in vivo depletion with monoclonal antibodies, algorithm predicted CD4 and CD8 epitopes and a pronounced decrease in parasite load (90.5-88.23%; p = 0.011) that was long-lasting. No decrease in parasite load was detected after vaccination with the N-domain of NH36, in spite of the induction of IFN-γ/IL-10 expression by CD4+ T cells after challenge. Both peptides reduced the size of footpad lesions, but only the C-domain reduced the parasite load of mice challenged with L. amazonensis. The identification of the target of the immune response to NH36 represents a basis for the rationale development of a bivalent vaccine against leishmaniasis and for multivalent vaccines against NHs-dependent pathogens.
- Published
- 2010
- Full Text
- View/download PDF
38. Strain-specific protective immunity following vaccination against experimental Trypanosoma cruzi infection.
- Author
-
Haolla FA, Claser C, de Alencar BC, Tzelepis F, de Vasconcelos JR, de Oliveira G, Silvério JC, Machado AV, Lannes-Vieira J, Bruna-Romero O, Gazzinelli RT, dos Santos RR, Soares MB, and Rodrigues MM
- Subjects
- Animals, Antibodies, Protozoan blood, Base Sequence, Cross Reactions, Epitopes, T-Lymphocyte immunology, Female, Humans, Mice, Molecular Sequence Data, Parasitemia prevention & control, Sequence Alignment, Survival Analysis, T-Lymphocytes immunology, Chagas Disease prevention & control, Glycoproteins immunology, Neuraminidase immunology, Protozoan Vaccines immunology, Trypanosoma cruzi immunology, Vaccines, DNA immunology
- Abstract
Immunisation with Amastigote Surface Protein 2 (asp-2) and trans-sialidase (ts) genes induces protective immunity in highly susceptible A/Sn mice, against infection with parasites of the Y strain of Trypanosoma cruzi. Based on immunological and biological strain variations in T. cruzi parasites, our goal was to validate our vaccination results using different parasite strains. Due to the importance of the CD8(+) T cells in protective immunity, we initially determined which strains expressed the immunodominant H-2K(k)-restricted epitope TEWETGQI. We tested eight strains, four of which elicited immune responses to this epitope (Y, G, Colombian and Colombia). We selected the Colombian and Colombia strains for our studies. A/Sn mice were immunised with different regimens using both T. cruzi genes (asp-2 and ts) simultaneously and subsequently challenged with blood trypomastigotes. Immune responses before the challenge were confirmed by the presence of specific antibodies and peptide-specific T cells. Genetic vaccination did not confer protective immunity against acute infection with a lethal dose of the Colombian strain. In contrast, we observed a drastic reduction in parasitemia and a significant increase in survival, following challenge with an otherwise lethal dose of the Colombia strain. In many surviving animals with late-stage chronic infection, we observed alterations in the heart's electrical conductivity, compared to naive mice. In summary, we concluded that immunity against T. cruzi antigens, similar to viruses and bacteria, may be strain-specific and have a negative impact on vaccine development.
- Published
- 2009
- Full Text
- View/download PDF
39. Swimming against the current: genetic vaccination against Trypanosoma cruzi infection in mice.
- Author
-
Rodrigues MM, de Alencar BC, Claser C, Tzelepis F, Silveira EL, Haolla FA, Dominguez MR, and Vasconcelos JR
- Subjects
- Animals, Antigens, Protozoan genetics, Antigens, Protozoan immunology, Chagas Disease prevention & control, Disease Models, Animal, Immunity, Cellular, Mice, Trypanosoma cruzi genetics, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Chagas Disease immunology, Protozoan Vaccines immunology, Trypanosoma cruzi immunology
- Abstract
Vaccines have had an unquestionable impact on public health during the last century. The most likely reason for the success of vaccines is the robust protective properties of specific antibodies. However, antibodies exert a strong selective pressure and many microorganisms, such as the obligatory intracellular parasite Trypanosoma cruzi, have been selected to survive in their presence. Although the host develops a strong immune response to T. cruzi, they do not clear the infection and instead progress to the chronic phase of the disease. Parasite persistence during the chronic phase of infection is now considered the main factor contributing to the chronic symptoms of the disease. Based on this finding, containment of parasite growth and survival may be one method to avoid the immunopathology of the chronic phase. In this context, vaccinologists have looked over the past 20 years for other immune effector mechanisms that could eliminate these antibody-resistant pathogens. We and others have tested the hypothesis that non-antibody-mediated cellular immune responses (CD4+ Th1 and CD8+ Tc1 cells) to specific parasite antigens/genes expressed by T. cruzi could indeed be used for the purpose of vaccination. This hypothesis was confirmed in different mouse models, indicating a possible path for vaccine development.
- Published
- 2009
- Full Text
- View/download PDF
40. Silencing cytokeratin 18 gene inhibits intracellular replication of Trypanosoma cruzi in HeLa cells but not binding and invasion of trypanosomes.
- Author
-
Claser C, Curcio M, de Mello SM, Silveira EV, Monteiro HP, and Rodrigues MM
- Subjects
- Animals, Cell Adhesion genetics, Chagas Disease genetics, DNA Replication genetics, DNA, Protozoan genetics, Down-Regulation, HeLa Cells, Humans, Keratin-18 metabolism, Lysosomes genetics, Phagocytosis genetics, RNA, Small Interfering genetics, Species Specificity, Transfection, Trypanosoma cruzi physiology, Virulence genetics, Chagas Disease metabolism, Chagas Disease parasitology, Host-Pathogen Interactions genetics, Keratin-18 genetics, RNA Interference, Trypanosoma cruzi pathogenicity
- Abstract
Background: As an obligatory intracellular parasite, Trypanosoma cruzi, the etiological agent of Chagas' disease, must invade and multiply within mammalian cells. Cytokeratin 18 (CK18) is among the host molecules that have been suggested as a mediator of important events during T. cruzi-host cell interaction. Based on that possibility, we addressed whether RNA interference (RNAi)-mediated down regulation of the CK18 gene could interfere with the parasite life cycle in vitro. HeLa cells transiently transfected with CK18-RNAi had negligible levels of CK18 transcripts, and significantly reduced levels of CK18 protein expression as determined by immunoblotting or immunofluorescence., Results: CK18 negative or positive HeLa cells were invaded equally as well by trypomastigotes of different T. cruzi strains. Also, in CK18 negative or positive cells, parasites recruited host cells lysosomes and escaped from the parasitophorous vacuole equally as well. After that, the growth of amastigotes of the Y or CL-Brener strains, was drastically arrested in CK18 RNAi-treated cells. After 48 hours, the number of amastigotes was several times lower in CK18 RNAi-treated cells when compared to control cells. Simultaneous staining of parasites and CK18 showed that in HeLa cells infected with the Y strain both co-localize. Although the amastigote surface protein-2 contains the domain VTVXNVFLYNR previously described to bind to CK18, in several attempts, we failed to detect binding of a recombinant protein to CK-18., Conclusion: The study demonstrates that silencing CK18 by transient RNAi, inhibits intracellular multiplication of the Y and CL strain of T. cruzi in HeLa cells, but not trypanosome binding and invasion.
- Published
- 2008
- Full Text
- View/download PDF
41. Novel protective antigens expressed by Trypanosoma cruzi amastigotes provide immunity to mice highly susceptible to Chagas' disease.
- Author
-
Silveira EL, Claser C, Haolla FA, Zanella LG, and Rodrigues MM
- Subjects
- Animals, Antibodies, Protozoan immunology, Antigens, Protozoan administration & dosage, Antigens, Protozoan genetics, Chagas Disease immunology, Chagas Disease mortality, Chagas Disease parasitology, Female, Glycoproteins administration & dosage, Glycoproteins genetics, HeLa Cells, Humans, Mice, Molecular Sequence Data, Neuraminidase administration & dosage, Neuraminidase genetics, Open Reading Frames genetics, Plasmids, Protozoan Vaccines administration & dosage, Protozoan Vaccines genetics, Sequence Analysis, DNA, Vaccination, Vaccines, DNA administration & dosage, Vaccines, DNA immunology, Antibodies, Protozoan blood, Antigens, Protozoan immunology, Chagas Disease prevention & control, Glycoproteins immunology, Neuraminidase immunology, Protozoan Vaccines immunology, Trypanosoma cruzi growth & development, Trypanosoma cruzi immunology
- Abstract
Earlier studies have demonstrated in A/Sn mice highly susceptible to Chagas' disease protective immunity against lethal Trypanosoma cruzi infection elicited by vaccination with an open reading frame (ORF) expressed by amastigotes. In our experiments, we used this mouse model to search for other amastigote-expressed ORFs with a similar property. Fourteen ORFs previously determined to be expressed in this developmental stage were individually inserted into a eukaryotic expression vector containing a nucleotide sequence that encoded a mammalian secretory signal peptide. Immunization with 13 of the 14 ORFs induced specific antibodies which recognized the amastigotes. Three of those immune sera also reacted with trypomastigotes and epimastigotes. After a lethal challenge with Y strain trypomastigotes, the vast majority of plasmid-injected mice succumbed to infection. In some cases, a significant delay in mortality was observed. Only two of these ORFs provided protective immunity against the otherwise lethal infection caused by trypomastigotes of the Y or Colombia strain. These ORFs encode members of the trans-sialidase family of surface antigens related to the previously described protective antigen amastigote surface protein 2 (ASP-2). Nevertheless, at the level of antibody recognition, no cross-reactivity was observed between the ORFs and the previously described ASP-2 from the Y strain. In immunofluorescence analyses, we observed the presence of epitopes related to both proteins expressed by amastigotes of seven different strains. In conclusion, our approach allowed us to successfully identify two novel protective ORFs which we consider interesting for future studies on the immune response to Chagas' disease.
- Published
- 2008
- Full Text
- View/download PDF
42. Infection with Trypanosoma cruzi restricts the repertoire of parasite-specific CD8+ T cells leading to immunodominance.
- Author
-
Tzelepis F, de Alencar BC, Penido ML, Claser C, Machado AV, Bruna-Romero O, Gazzinelli RT, and Rodrigues MM
- Subjects
- Adenoviridae genetics, Amino Acid Sequence, Animals, Antigen Presentation, Antigen-Presenting Cells immunology, H-2 Antigens chemistry, H-2 Antigens genetics, Heterozygote, Immunization, Mice, Mice, Inbred Strains, Molecular Sequence Data, Antigens, Protozoan immunology, CD8-Positive T-Lymphocytes immunology, Chagas Disease immunology, H-2 Antigens immunology, Immunodominant Epitopes immunology, Trypanosoma cruzi immunology
- Abstract
Interference or competition between CD8(+) T cells restricted by distinct MHC-I molecules can be a powerful means to establish an immunodominant response. However, its importance during infections is still questionable. In this study, we describe that following infection of mice with the human pathogen Trypanosoma cruzi, an immunodominant CD8(+) T cell immune response is developed directed to an H-2K(b)-restricted epitope expressed by members of the trans-sialidase family of surface proteins. To determine whether this immunodominance was exerted over other non-H-2K(b)-restricted epitopes, we measured during infection of heterozygote mice, immune responses to three distinct epitopes, all expressed by members of the trans-sialidase family, recognized by H-2K(b)-, H-2K(k)-, or H-2K(d)-restricted CD8(+) T cells. Infected heterozygote or homozygote mice displayed comparably strong immune responses to the H-2K(b)-restricted immunodominant epitope. In contrast, H-2K(k)- or H-2K(d)-restricted immune responses were significantly impaired in heterozygote infected mice when compared with homozygote ones. This interference was not dependent on the dose of parasite or the timing of infection. Also, it was not seen in heterozygote mice immunized with recombinant adenoviruses expressing T. cruzi Ags. Finally, we observed that the immunodominance was circumvented by concomitant infection with two T. cruzi strains containing distinct immunodominant epitopes, suggesting that the operating mechanism most likely involves competition of T cells for limiting APCs. This type of interference never described during infection with a human parasite may represent a sophisticated strategy to restrict priming of CD8(+) T cells of distinct specificities, avoiding complete pathogen elimination by host effector cells, and thus favoring host parasitism.
- Published
- 2008
- Full Text
- View/download PDF
43. Immunologically relevant strain polymorphism in the Amastigote Surface Protein 2 of Trypanosoma cruzi.
- Author
-
Claser C, Espíndola NM, Sasso G, Vaz AJ, Boscardin SB, and Rodrigues MM
- Subjects
- Amino Acid Sequence, Animals, Antigens, Protozoan genetics, Antigens, Protozoan immunology, Chagas Disease immunology, Chagas Disease parasitology, Chagas Disease prevention & control, Cross Reactions, Epitopes, Female, Genetic Variation, HeLa Cells, Humans, Life Cycle Stages, Mice, Mice, Inbred BALB C, Mice, Inbred C57BL, Molecular Sequence Data, Recombinant Proteins genetics, Recombinant Proteins metabolism, Trypanosoma cruzi genetics, Trypanosoma cruzi growth & development, Vaccination, Isoenzymes chemistry, Isoenzymes genetics, Isoenzymes immunology, Neuraminidase chemistry, Neuraminidase genetics, Neuraminidase immunology, Polymorphism, Genetic, Trypanosoma cruzi classification, Trypanosoma cruzi immunology
- Abstract
Several evidences suggest that the Amastigote Surface Protein-2 (ASP-2) of Trypanosoma cruzi is an important target for immunity during infection. Based on this, we considered it important to evaluate its strain polymorphism. Initially, we observed the presence of conserved cross-reactive epitopes in amastigotes of all parasite strains tested. In addition, the predicted amino acid sequences of the genes isolated from the cDNA of amastigotes of CL-Brener, Tulahuen, Colombian and G strains displayed a high degree of identity (>80%) to the previously described genes of ASP-2. Unexpectedly, Sylvio X10/4 and G strains expressed a new isoform of ASP-2 with limited identity to the previously described genes, but with a high degree of identity when compared to each other. Immunological studies confirmed the presence of cross-reactive epitopes between recombinant proteins representing the different isoforms of ASP-2. However, the genetic vaccination of mice with the new isoform of asp-2 gene expressed by the G strain failed to provide the same degree of protective immunity to a challenge by parasites of the Y strain as did asp-2 genes of Y or CL-Brener strains. In summary, we found that few strains can express different isoforms of ASP-2 which may not share cross-protective epitopes.
- Published
- 2007
- Full Text
- View/download PDF
44. Long-term protective immunity induced against Trypanosoma cruzi infection after vaccination with recombinant adenoviruses encoding amastigote surface protein-2 and trans-sialidase.
- Author
-
Machado AV, Cardoso JE, Claser C, Rodrigues MM, Gazzinelli RT, and Bruna-Romero O
- Subjects
- Amino Acid Sequence, Animals, CD8-Positive T-Lymphocytes immunology, Cell Line, Enzyme-Linked Immunosorbent Assay, Mice, Mice, Inbred Strains, Mice, Knockout, Protozoan Vaccines administration & dosage, Th1 Cells immunology, Trypanosomiasis immunology, Adenoviridae genetics, Glycoproteins genetics, Neuraminidase genetics, Recombination, Genetic, Trypanosoma cruzi immunology, Trypanosomiasis prevention & control
- Abstract
Protection against protozoan parasite Trypanosoma cruzi has been shown to be dependent on the induction of type 1 immune responses. Replication-deficient human type 5 recombinant adenoviruses have an unsurpassed ability to induce type 1 immune responses. Thus, we constructed two type 5 recombinant adenoviruses encoding parasite antigens trans-sialidase (rAdTS) and amastigote surface protein-2 (rAdASP2). Both antigens were genetically engineered to secrete recombinant products in order to induce both optimal antibody and T cell responses. Immunizations of mice with rAdASP2 and rAdTS induced high levels of serum antibodies specific for their recombinant products. In addition, both recombinant viruses were able to elicit a biased helper T cell type 1 (Th1) cellular immune response and a substantial CD8+ T cell-mediated immune response. Moreover, individual immunization with rAdASP2 or rAdTS induced high levels of protection against a challenge with live parasites. CD8+ T cells mediated, at least in part, such protection. Furthermore, when combined in the same inoculum, rAdTS plus rAdASP2 induced complete protection in all animals tested, even when challenges were performed 14 weeks after the last immunization. Taking together, these results show that recombinant adenoviruses expressing TS and ASP-2 antigens of T. cruzi are interesting candidates for the development of a vaccine against Chagas' disease.
- Published
- 2006
- Full Text
- View/download PDF
45. Protective immunity against trypanosoma cruzi infection in a highly susceptible mouse strain after vaccination with genes encoding the amastigote surface protein-2 and trans-sialidase.
- Author
-
Vasconcelos JR, Hiyane MI, Marinho CR, Claser C, Machado AM, Gazzinelli RT, Bruña-Romero O, Alvarez JM, Boscardin SB, and Rodrigues MM
- Subjects
- Animals, Antibodies, Protozoan analysis, Antigens, Surface genetics, CD4-Positive T-Lymphocytes immunology, CD8-Positive T-Lymphocytes immunology, Chagas Disease immunology, Chagas Disease pathology, Glycoproteins, Immunity, Interferon-gamma immunology, Mice, Mice, Inbred Strains, Muscle, Skeletal pathology, Myocardium pathology, Neuraminidase immunology, Parasitemia parasitology, Plasmids administration & dosage, Spleen immunology, Antigens, Protozoan genetics, Chagas Disease prevention & control, Neuraminidase genetics, Protozoan Vaccines, Trypanosoma cruzi immunology
- Abstract
Protective immunity against lethal infection is developed when BALB/c or C57BL/6 mice are immunized with plasmids containing genes from the protozoan parasite Trypanosoma cruzi. However, genetic vaccination of the highly susceptible mouse strain A/Sn promoted limited survival after challenge. This observation questioned whether this type of vaccination would be appropriate for highly susceptible individuals. Here, we compared the protective efficacy and the immune response after individual or combined genetic vaccination of A/Sn mice with genes encoding trans-sialidase (TS) or the amastigote surface protein-2 (ASP-2). After challenge, a significant proportion of A/Sn mice immunized with either the asp-2 gene or simultaneously with asp-2 and ts genes, survived infection. In contrast, the vast majority of mice immunized with the ts gene or the vector alone died. Parasitological and histological studies performed in the surviving mice revealed that these mice harbored parasites; however, minimal inflammatory responses were seen in heart and striated muscle. We used this model to search for an in vitro correlation for protection. We found that protective immunity correlated with a higher secretion of interferon- by spleen cells on in vitro restimulation with ASP-2 and the presence of ASP-2-specific CD8 cells. Depletion of either CD4 or CD8 or both T-cell subpopulations prior to the challenge rendered the mice susceptible to infection demonstrating the critical contribution of both cell types in protective immunity. Our results reinforce the prophylactic potential of genetic vaccination with asp-2 and ts genes by describing protective immunity against lethal T. cruzi infection and chronic tissue pathology in a highly susceptible mouse strain.
- Published
- 2004
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.