1
|
de Lima RMS, Leão LKR, Martins LC, Passos ADCF, Batista EDJO, Herculano AM, Oliveira KRHM. Unveiling new perspectives about the onset of neurological and cognitive deficits in cerebral malaria: exploring cellular and neurochemical mechanisms. Front Cell Infect Microbiol 2025; 15:1506282. [PMID: 39981376 PMCID: PMC11839640 DOI: 10.3389/fcimb.2025.1506282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/02/2025] [Indexed: 02/22/2025] Open
Abstract
Cerebral malaria is the most severe and lethal complication caused by Plasmodium falciparum infection, leading to critical neurological impairments and long-term cognitive, behavioral, and neurological sequelae in survivors, particularly affecting children under the age of five. Various hypotheses have been proposed to explain the neurological syndrome associated to cerebral malaria condition, including vascular occlusion and sequestration, cytokine storm or inflammatory response, or a combination of these mechanisms and despite extensive research and a growing range of scientific information, the precise pathophysiological mechanism remains poorly understood. In this sense, this review aims to explore the neurological impairment in cerebral malaria and elucidate novel mechanisms to explain the severity of this disease. Recent evidence implicates glutamate and glutamatergic pathways in the onset of cerebral malaria, alongside the impairments in the metabolic activity of other molecules such as dopamine and kynurenic acid. These neurotransmitters pathways may play a crucial role in the pathogenesis of cerebral malaria, potentially interacting with other molecular players. By enhancing our understanding in the pathophysiology of cerebral malaria, this article seeks to explore new hypotheses regarding the involvement of neurotransmitters and their interactions with other molecular targets, thereby contributing to the overall pathology of cerebral malaria.
Collapse
Affiliation(s)
- Renato M. S. de Lima
- Laboratory of Experimental Neuropharmacology, Biological Science Institute, Federal University of Pará, Belém, Brazil
| | - Luana K. R. Leão
- Laboratory of Experimental Neuropharmacology, Biological Science Institute, Federal University of Pará, Belém, Brazil
| | - Luana C. Martins
- Laboratory of Experimental Neuropharmacology, Biological Science Institute, Federal University of Pará, Belém, Brazil
| | - Adelaide da C. Fonseca Passos
- Laboratory of Experimental Neuropharmacology, Biological Science Institute, Federal University of Pará, Belém, Brazil
| | | | - Anderson M. Herculano
- Laboratory of Experimental Neuropharmacology, Biological Science Institute, Federal University of Pará, Belém, Brazil
| | - Karen R. H. M. Oliveira
- Laboratory of Experimental Neuropharmacology, Biological Science Institute, Federal University of Pará, Belém, Brazil
| |
Collapse
|
2
|
Pollenus E, Possemiers H, Knoops S, Prenen F, Vandermosten L, Pham TT, Buysrogge L, Matthys P, Van den Steen PE. NK cells contribute to the resolution of experimental malaria-associated acute respiratory distress syndrome after antimalarial treatment. Front Immunol 2024; 15:1433904. [PMID: 39355242 PMCID: PMC11442241 DOI: 10.3389/fimmu.2024.1433904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 08/27/2024] [Indexed: 10/03/2024] Open
Abstract
In both humans and mice, natural killer (NK) cells are important lymphocytes of the innate immune system. They are often considered pro-inflammatory effector cells but may also have a regulatory or pro-resolving function by switching their cytokine profile towards the production of anti-inflammatory cytokines, including interleukin-10 (IL-10) and transforming growth factor-β, and by killing pro-inflammatory immune cells. Here, the role of NK cells in the resolution of malaria lung pathology was studied. Malaria complications, such as malaria-associated acute respiratory distress syndrome (MA-ARDS), are often lethal despite the rapid and efficient killing of Plasmodium parasites with antimalarial drugs. Hence, studying the resolution and healing mechanisms involved in the recovery from these complications could be useful to develop adjunctive treatments. Treatment of Plasmodium berghei NK65-infected C57BL/6 mice with a combination of artesunate and chloroquine starting at the appearance of symptoms was used as a model to study the resolution of MA-ARDS. The role of NK cells was studied using anti-NK1.1 depletion antibodies and NK cell-deficient mice. Using both methods, NK cells were found to be dispensable in the development of MA-ARDS, as shown previously. In contrast, NK cells were crucial in the initiation of resolution upon antimalarial treatment, as survival was significantly decreased in the absence of NK cells. Considerably increased IL-10 expression by NK cells suggested an anti-inflammatory and pro-resolving phenotype. Despite the increase in Il10 expression in the NK cells, inhibition of the IL-10/IL-10R axis using anti-IL10R antibodies had no effect on the resolution for MA-ARDS, suggesting that the pro-resolving effect of NK cells cannot solely be attributed to their IL-10 production. In conclusion, NK cells contribute to the resolution of experimental MA-ARDS.
Collapse
Affiliation(s)
- Emilie Pollenus
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Hendrik Possemiers
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Sofie Knoops
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Fran Prenen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Leen Vandermosten
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Thao-Thy Pham
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
- Clinical Immunology Unit, Department of Clinical Sciences, Institute of Tropical Medicine Antwerp, Antwerp, Belgium
| | - Laura Buysrogge
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Patrick Matthys
- Laboratory of Immunobiology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| | - Philippe E. Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology, Immunology & Transplantation, Rega Institute for Medical Research, KU Leuven, Leuven, Belgium
| |
Collapse
|
3
|
Simião GM, Parreira KS, Klein SG, Ferreira FB, Freitas FDS, Silva EFD, Silva NM, Silva MVD, Lima WR. Involvement of Inflammatory Cytokines, Renal NaPi-IIa Cotransporter, and TRAIL Induced-Apoptosis in Experimental Malaria-Associated Acute Kidney Injury. Pathogens 2024; 13:376. [PMID: 38787228 DOI: 10.3390/pathogens13050376] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/24/2024] [Accepted: 04/26/2024] [Indexed: 05/25/2024] Open
Abstract
The murine model of experimental cerebral malaria (ECM) induced by Plasmodium berghei ANKA was used to investigate the relationship among pro-inflammatory cytokines, alterations in renal function biomarkers, and the induction of the TRAIL apoptosis pathway during malaria-associated acute kidney injury (AKI). Renal function was evaluated through the measurement of plasma creatinine and blood urea nitrogen (BUN). The mRNA expression of several cytokines and NaPi-IIa was quantified. Kidney sections were examined and cytokine levels were assessed using cytometric bead array (CBA) assays. The presence of glomerular IgG deposits and apoptosis-related proteins were investigated using in situ immunofluorescence assays and quantitative real-time PCR, respectively. NaPi-IIa downregulation in the kidneys provided novel insights into the pathogenesis of hypophosphatemia during CM. Histopathological analysis revealed characteristic features of severe malaria-associated nephritis, including glomerular collapse and tubular alterations. Pro-inflammatory cytokines, such as TNF-α, IL-1β, and IL-6, were upregulated. The TRAIL apoptosis pathway was significantly activated, implicating its role in renal apoptosis. The observed alterations in renal biomarkers and the downregulation of NaPi-IIa shed light on potential mechanisms contributing to renal dysfunction in ECM. The intricate balance between pro- and anti-inflammatory cytokines, along with the activation of the TRAIL apoptosis pathway, highlights the complexity of malaria-associated AKI and provides new therapeutic targets.
Collapse
Affiliation(s)
- Gustavo Martins Simião
- Faculty of Health Sciences, Federal University of Rondonopolis, Rondonópolis 78736-900, MT, Brazil
| | | | - Sandra Gabriela Klein
- Laboratory of Biotechnology in Experimental Models, Federal University of Uberlandia, Uberlândia 38410-337, MG, Brazil
| | - Flávia Batista Ferreira
- Laboratory of Biotechnology in Experimental Models, Federal University of Uberlandia, Uberlândia 38410-337, MG, Brazil
- Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlândia 38405-318, MG, Brazil
| | | | | | - Neide Maria Silva
- Institute of Biomedical Sciences, Federal University of Uberlandia, Uberlândia 38405-318, MG, Brazil
| | - Murilo Vieira da Silva
- Laboratory of Biotechnology in Experimental Models, Federal University of Uberlandia, Uberlândia 38410-337, MG, Brazil
| | - Wânia Rezende Lima
- Faculty of Health Sciences, Federal University of Rondonopolis, Rondonópolis 78736-900, MT, Brazil
- Institute of Biotechnology, Federal University of Catalao, Catalão 75706-881, GO, Brazil
- Laboratory of Biotechnology in Experimental Models, Federal University of Uberlandia, Uberlândia 38410-337, MG, Brazil
| |
Collapse
|
4
|
Kim MJ, Chu KB, Yoon KW, Kang HJ, Lee DH, Moon EK, Quan FS. Virus-like particles expressing microneme-associated antigen of Plasmodium berghei confer better protection than those expressing apical membrane antigen 1. PARASITES, HOSTS AND DISEASES 2024; 62:193-204. [PMID: 38835260 PMCID: PMC11150920 DOI: 10.3347/phd.24017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/18/2024] [Indexed: 06/06/2024]
Abstract
Malaria is a global disease affecting a large portion of the world's population. Although vaccines have recently become available, their efficacies are suboptimal. We generated virus-like particles (VLPs) that expressed either apical membrane antigen 1 (AMA1) or microneme-associated antigen (MIC) of Plasmodium berghei and compared their efficacy in BALB/c mice. We found that immune sera acquired from AMA1 VLP- or MIC VLP-immunized mice specifically interacted with the antigen of choice and the whole P. berghei lysate antigen, indicating that the antibodies were highly parasite-specific. Both VLP vaccines significantly enhanced germinal center B cell frequencies in the inguinal lymph nodes of mice compared with the control, but only the mice that received MIC VLPs showed significantly enhanced CD4+ T cell responses in the blood following P. berghei challenge infection. AMA1 and MIC VLPs significantly suppressed TNF-α and interleukin-10 production but had a negligible effect on interferon-γ. Both VLPs prevented excessive parasitemia buildup in immunized mice, although parasite burden reduction induced by MIC VLPs was slightly more effective than that induced by AMA1. Both VLPs were equally effective at preventing body weight loss. Our findings demonstrated that the MIC VLP was an effective inducer of protection against murine experimental malaria and should be the focus of further development.
Collapse
Affiliation(s)
- Min-Ju Kim
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447,
Korea
| | - Ki Back Chu
- Department of Parasitology, Inje University College of Medicine, Busan 47392,
Korea
- Department of Infectious Disease and Malaria, Paik Institute of Clinical Research, Inje University, Busan 47392,
Korea
| | - Keon-Woong Yoon
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447,
Korea
| | - Hae-Ji Kang
- Department of Microbiology, Dongguk University College of Medicine, Gyeongju 38066,
Korea
| | - Dong-Hun Lee
- Department of Biomedical Science, Graduate School, Kyung Hee University, Seoul 02447,
Korea
| | - Eun-Kyung Moon
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul 02447,
Korea
| | - Fu-Shi Quan
- Department of Medical Zoology, Kyung Hee University School of Medicine, Seoul 02447,
Korea
- Medical Research Center for Bioreaction to Reactive Oxygen Species and Biomedical Science Institute, School of Medicine, Graduate School, Kyung Hee University, Seoul 02447,
Korea
| |
Collapse
|
5
|
Carpinter BA, Renhe DC, Bellei JCB, Vieira CD, Rodolphi CM, Ferreira MVR, de Freitas CS, Neto AFDS, Coelho EAF, Mietto BDS, Gomes FLR, Rocha VN, Scopel KKG. DHA-rich fish oil plays a protective role against experimental cerebral malaria by controlling inflammatory and mechanical events from infection. J Nutr Biochem 2024; 123:109492. [PMID: 37866427 DOI: 10.1016/j.jnutbio.2023.109492] [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: 02/04/2023] [Revised: 05/17/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Every year, thousands of children, particularly those under 5 years old, die because of cerebral malaria (CM). Following conventional treatment, approximately 25% of surviving individuals have lifelong severe neurocognitive sequelae. Therefore, improved conventional therapies or effective alternative therapies that prevent the severe infection are crucial. Omega-3 (Ω-3) polyunsaturated fatty acids (PUFAs) are known to have antioxidative and anti-inflammatory effects and protect against diverse neurological disorders, including Alzheimer's and Parkinson's diseases. However, little is known regarding the effects of Ω-3 PUFAs against parasitic infections. In this study, C57BL/6 mice received supplemental treatment of a fish oil rich in the Ω-3 PUFA, docosahexaenoic acid (DHA), which was started 15 days prior to infection with Plasmodium berghei ANKA and was maintained until the end of the study. Animals treated with the highest doses of DHA, 3.0 and 6.0 g/kg body weight, had 60 and 80% chance of survival, respectively, while all nontreated mice died by the 7th day postinfection due to CM. Furthermore, the parasite load during the critical period for CM development (5th to 11th day postinfection) was controlled in treated mice. However, after this period all animals developed high levels of parasitemia until the 20th day of infection. DHA treatment also effectively reduced blood-brain barrier (BBB) damage and brain edema and completely prevented brain hemorrhage and vascular occlusion. A strong anti-inflammatory profile was observed in the brains of DHA-treated mice, as well as, an increased number of neutrophil and reduced number of CD8+ T leukocytes in the spleen. Thus, this is the first study to demonstrate that the prophylactic use of DHA-rich fish oil exerts protective effects against experimental CM, reducing the mechanical and immunological events caused by the P. berghei ANKA infection.
Collapse
Affiliation(s)
- Bárbara Albuquerque Carpinter
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Daniela Chaves Renhe
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Jéssica Correa Bezerra Bellei
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Carolina David Vieira
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Cinthia Magalhães Rodolphi
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Camila Simões de Freitas
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adolfo Firmino da Silva Neto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eduardo Antônio Ferraz Coelho
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno de Siqueira Mietto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Vinicius Novaes Rocha
- Department of Veterinary Medicine, Research Centre of Pathology and Veterinary Histology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Kézia Katiani Gorza Scopel
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil.
| |
Collapse
|
6
|
Plirat W, Chaniad P, Phuwajaroanpong A, Konyanee A, Viriyavejakul P, Septama AW, Punsawad C. Efficacy of artesunate combined with Atractylodes lancea or Prabchompoothaweep remedy extracts as adjunctive therapy for the treatment of cerebral malaria. BMC Complement Med Ther 2023; 23:332. [PMID: 37730604 PMCID: PMC10510250 DOI: 10.1186/s12906-023-04150-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/04/2023] [Indexed: 09/22/2023] Open
Abstract
BACKGROUND Cerebral malaria is one of the most serious complications of Plasmodium infection and causes behavioral changes. However, current antimalarial drugs have shown poor outcomes. Therefore, new antimalarials with neuroprotective effects are urgently needed. This study aimed to evaluate the effects of selected extracts as monotherapy or adjunctive therapy with artesunate on antimalarial, anti-inflammatory, antioxidant, and neuroprotective properties in experimental cerebral malaria (ECM). METHODS ECM was induced in male C57BL/6 mice by infection with Plasmodium berghei ANKA (PbA). Ethanolic extracts of Atractylodes lancea (a dose of 400 mg/kg) and Prabchompoothaweep remedy (a dose of 600 mg/kg) were evaluated as monotherapy and adjunctive therapy combined with artesunate at the onset of signs of cerebral malaria and continued for 7 consecutive days. Parasitemia, clinical scores, and body weight were recorded throughout the study. At day 13 post-infection, mouse brains were dissected and processed for the study of the inflammatory response, oxidative stress, blood-brain barrier (BBB) integrity, histopathological changes, and neurocognitive impairments. RESULTS Ethanolic extracts of A. lancea and Prabchompoothaweep remedy alone improved cerebral malaria outcome in ECM, whereas artesunate combined with extracts of A. lancea or Prabchompoothaweep remedy significantly improved the outcome of artesunate and crude extracts alone. Using real-time PCR, PbA-infected mice that had received the combination treatment showed significantly reduced gene expression of inflammatory cytokines (TNF-α, IL-1β, IL-6, and IL-10), chemokines (CXCL4 and CXCL10), and adhesion molecules (ICAM-1, VCAM1, and CD36). The PbA-infected mice that received the combination treatment showed a significantly decreased malondialdehyde level compared to the untreated group. Similarly, the Evans blue dye assay revealed significantly less dye extravasation in the brains of infected mice administered the combination treatment, indicating improved BBB integrity. Combination treatment improved survival and reduced pathology in the PbA-infected group. Additionally, combination treatment resulted in a significantly reduced level of cognitive impairment, which was analyzed using a novel object recognition test. CONCLUSIONS This study demonstrated that artesunate combined with A. lancea or Prabchompoothaweep remedy extracts as adjunctive therapy reduced mortality, neuroinflammation, oxidative stress, BBB integrity protection, and neurocognitive impairment in the ECM.
Collapse
Affiliation(s)
- Walaiporn Plirat
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Prapaporn Chaniad
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Arisara Phuwajaroanpong
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | - Atthaphon Konyanee
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand
| | | | - Abdi Wira Septama
- Research Center for Pharmaceutical Ingredient and Traditional Medicine, National Research and Innovation Agency (BRIN), Cibinong Science Center, Cibinong, West Java, 16915, Indonesia
| | - Chuchard Punsawad
- Department of Medical Sciences, School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand.
- Research Center in Tropical Pathobiology, Walailak University, Nakhon Si Thammarat, 80160, Thailand.
| |
Collapse
|
7
|
de Roquetaillade C, Laouenan C, Mira JP, Roy C, Thuong M, Azoulay É, Gruson D, Jacobs F, Chommeloux J, Raffi F, Hocqueloux L, Imbert P, Jeantils V, Delassus JL, Matheron S, Fitting C, Timsit JF, Bruneel F. Cytokine profiles in adults with imported malaria. Sci Rep 2023; 13:10347. [PMID: 37365194 DOI: 10.1038/s41598-023-36212-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 05/31/2023] [Indexed: 06/28/2023] Open
Abstract
The increase in worldwide travel is making imported malaria a growing health concern in non-endemic countries. Most data on the pathophysiology of malaria come from endemic areas. Little is known about cytokine profiles during imported malaria. This study aimed at deciphering the relationship between cytokine host response and malaria severity among imported cases in France. This study reports cytokine profiles in adults with Plasmodium falciparum malaria included in the PALUREA prospective study conducted between 2006 and 2010. The patients were classified as having uncomplicated malaria (UM) or severe malaria (SM), with this last further categorized as very severe malaria (VSM) or less severe malaria (LSM). At hospital admission, eight blood cytokines were assayed in duplicate using Luminex® technology: interleukin (IL)-1α, IL-1β, IL-2, IL-4, IL-10, tumor necrosis factor (TNF)α, interferon (IFN)γ, and macrophage migration inhibitory factor (MIF). These assays were repeated on days 1 and 2 in the SM group. Of the 278 patients, 134 had UM and 144 SM. At hospital admission, over half the patients had undetectable levels of IL-1α, IL-1β, IL-2, IL-4, IFNγ, and TNFα, while IL-10 and MIF were significantly higher in the SM vs. the UM group. Higher IL-10 was significantly associated with higher parasitemia (R = 0.32 [0.16-0.46]; P = 0.0001). In the SM group, IL-10 elevation persisting from admission to day 2 was significantly associated with subsequent nosocomial infection. Of eight tested cytokines, only MIF and IL-10 were associated with disease severity in adults with imported P. falciparum malaria. At admission, many patients had undetectable cytokine levels, suggesting that circulating cytokine assays may not be helpful as part of the routine evaluation of adults with imported malaria. Persisting high IL-10 concentration was associated with subsequent nosocomial infection, suggesting its possible interest in immune monitoring of most severe patients.
Collapse
Affiliation(s)
- Charles de Roquetaillade
- Department of Anesthesiology and Critical Care, Hôpital Lariboisière, FHU PROMICE, DMU Parabol, AP-HP Nord, Paris, France.
- INSERM UMR 942 MASCOT, Université de Paris-Cité, Paris, France.
| | - Cédric Laouenan
- Département Epidémiologie Biostatistiques et Recherche Clinique, AP-HP, INSERM, Centre d'Investigation Clinique-Epidémiologie Clinique 1425, Hôpital Bichat, Paris, France
- UMR 1137, Université de Paris-Cité, INSERM, IAME, Hôpital Bichat, AP-HP, Paris, France
| | - Jean-Paul Mira
- Service de medecine intensive-reanimation, Hôpital Cochin, AP-HP Centre, Paris, France
- Université Paris Cité, Paris, France
- Institut Cochin, INSERM U1016, CNRS UMR 8104, Université Paris Cité, AP-HP, Paris, France
| | - Carine Roy
- Département Epidémiologie Biostatistiques et Recherche Clinique, AP-HP, INSERM, Centre d'Investigation Clinique-Epidémiologie Clinique 1425, Hôpital Bichat, Paris, France
- UMR 1137, Université de Paris-Cité, INSERM, IAME, Hôpital Bichat, AP-HP, Paris, France
| | - Marie Thuong
- Service de medecine intensive-reanimation, Centre Hospitalier René Dubos, Pontoise, France
| | - Élie Azoulay
- Service de medecine intensive-reanimation, Hôpital Saint-Louis, Hôpitaux Universitaires Paris-Nord, AP-HP, Paris, France
| | - Didier Gruson
- Service de medecine intensive-reanimation, Hôpital Pellegrin-Tripode, Bordeaux, France
- Centre de Recherche Cardio-Thoracique, CHU Bordeaux, Bordeaux, France
| | - Frédéric Jacobs
- Service de medecine intensive-reanimation, Hôpital Antoine Béclère, Université Paris-Saclay, AP-HP, Clamart, France
| | - Juliette Chommeloux
- Service de medecine intensive reanimation, Institut de Cardiologie, ICAN, Groupe Hospitalier Pitié-Salpêtrière, Sorbonne Université, AP-HP, Paris, France
| | | | - Laurent Hocqueloux
- Service de maladies infectieuses, Centre Hospitalier Régional d'Orléans, Orléans, France
| | - Patrick Imbert
- Centre de vaccinations internationales, Hôpital d'instruction des armees Bégin, Saint-Mandé, France
| | - Vincent Jeantils
- Service de maladies infectieuses, Hôpital Jean Verdier, AP-HP, Bondy, France
| | - Jean-Luc Delassus
- Service de medecine interne et de maladies infectieuses, Centre hospitalier intercommunal Robert-Ballanger, Aulnay-sous-Bois, France
| | - Sophie Matheron
- Service de maladies infectieuses et tropicales, Hôpital Bichat, GHU Paris Nord, AP-HP, Paris, France
| | | | - Jean-François Timsit
- Service de medecine intensive et reanimation (MI2), Hôpital Bichat, Paris, France
- IAME, Université de Paris, INSERM U1137, AP-HP, Paris, France
| | - Fabrice Bruneel
- Service de reanimation, Centre Hospitalier de Versailles, Hôpital André Mignot, Le Chesnay, France
| |
Collapse
|
8
|
Rai S, Girdhar M, Siraj F, Sharma S, Kumar M, Katyal A. Mechanistic insights into immunopathogenesis of murine cerebral malaria: Cues from "young" C57BL/6J and BALB/c mice. Immunol Lett 2023; 256-257:9-19. [PMID: 36931472 DOI: 10.1016/j.imlet.2023.03.004] [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: 01/11/2023] [Revised: 03/04/2023] [Accepted: 03/13/2023] [Indexed: 03/17/2023]
Abstract
Cerebral malaria (CM), a major cause of mortality in children <5 years, presents disparity in pathophysiological features and poor prognosis compared to adults. Adult C57BL/6J mice infected with Plasmodium berghei ANKA (PbA) are widely used to understand CM pathogenesis compared to relatively less prone BALB/c mice; however, age and immune status of the host also influence disease sequelae and cerebral manifestations. Murine models of CM known so far do not project complete disease spectrum of pediatric CM. The present study was designed to dissect and differentiate CM immunopathogenesis in "young" BALB/c and C57BL/6J mice infected with PbA, in search of a competent mouse model mimicking pediatric CM. Multipronged approach including the analysis of blood-brain barrier (BBB) permeability and parasite infiltration, histopathology, nitric oxide levels, and pro/anti-inflammatory (TNF-α, IFN-γ, IL-4, and IL-10) cytokine expression were compared in the cortices of both young BALB/c and C57BL/6J mice. The results illustrate severe course of infection and typical CM like histopathological alterations including monocytic plugging in PbA-infected "young" BALB/c compared to C57BL/6J mice. The decreased expression of tight junction proteins (ZO-1 and Claudin-3) and Evan's blue extravasation was also more evident in BALB/c mice indicating a more permeable BBB. The increased cortical expression of TNF-α, IFN-γ, IL-4, IL-10, iNOS, eNOS, nNOS, and associated activation of brain resident cells in cortices of BALB/c with progressive parasitaemia depicts the cumulative involvement of host immune responses and parasite accumulation in progression of CM. Thus, the incongruity of cytokine balance resulted in worsening of disease manifestation in "young" BALB/c similar to pediatric CM.
Collapse
Affiliation(s)
- Shweta Rai
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North campus, New Delhi 110007, India
| | - Meetali Girdhar
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North campus, New Delhi 110007, India
| | - Fouzia Siraj
- Department of Pathology, National Institute of Pathology, ICMR, Safdarjung Hospital, New Delhi, India
| | - Sheetal Sharma
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North campus, New Delhi 110007, India
| | - Mukesh Kumar
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North campus, New Delhi 110007, India
| | - Anju Katyal
- Dr. B.R. Ambedkar Center for Biomedical Research, University of Delhi, North campus, New Delhi 110007, India.
| |
Collapse
|
9
|
Deng S, Graham ML, Chen XM. The Complexity of Interferon Signaling in Host Defense against Protozoan Parasite Infection. Pathogens 2023; 12:319. [PMID: 36839591 PMCID: PMC9962834 DOI: 10.3390/pathogens12020319] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 02/11/2023] [Accepted: 02/13/2023] [Indexed: 02/17/2023] Open
Abstract
Protozoan parasites, such as Plasmodium, Leishmania, Toxoplasma, Cryptosporidium, and Trypanosoma, are causative agents of health-threatening diseases in both humans and animals, leading to significant health risks and socioeconomic losses globally. The development of effective therapeutic and prevention strategies for protozoan-caused diseases requires a full understanding of the pathogenesis and protective events occurring in infected hosts. Interferons (IFNs) are a family of cytokines with diverse biological effects in host antimicrobial defense and disease pathogenesis, including protozoan parasite infection. Type II IFN (IFN-γ) has been widely recognized as the essential defense cytokine in intracellular protozoan parasite infection, whereas recent studies also revealed the production and distinct function of type I and III IFNs in host defense against these parasites. Decoding the complex network of the IFN family in host-parasite interaction is critical for exploring potential new therapeutic strategies against intracellular protozoan parasite infection. Here, we review the complex effects of IFNs on the host defense against intracellular protozoan parasites and the crosstalk between distinct types of IFN signaling during infections.
Collapse
Affiliation(s)
- Silu Deng
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
- Department of Medical Microbiology and Immunology, Creighton University School of Medicine, Omaha, NE 68178, USA
| | - Marion L. Graham
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| | - Xian-Ming Chen
- Department of Microbial Pathogens and Immunity, Rush University Medical Center, Chicago, IL 60612, USA
| |
Collapse
|
10
|
Ibraheem Y, Bayarsaikhan G, Inoue SI. Host immunity to Plasmodium infection: Contribution of Plasmodium berghei to our understanding of T cell-related immune response to blood-stage malaria. Parasitol Int 2022; 92:102646. [PMID: 35998816 DOI: 10.1016/j.parint.2022.102646] [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: 04/06/2022] [Revised: 08/09/2022] [Accepted: 08/12/2022] [Indexed: 10/15/2022]
Abstract
Malaria is a life-threatening disease caused by infection with Plasmodium parasites. The goal of developing an effective malaria vaccine is yet to be reached despite decades of massive research efforts. CD4+ helper T cells, CD8+ cytotoxic T cells, and γδ T cells are associated with immune responses to both liver-stage and blood-stage Plasmodium infection. The immune responses of T cell-lineages to Plasmodium infection are associated with both protection and immunopathology. Studies with mouse model of malaria contribute to our understanding of host immune response. In this paper, we focus primarily on mouse malaria model with blood-stage Plasmodium berghei infection and review our knowledge of T cell immune responses against Plasmodium infection. Moreover, we also discuss findings of experimental human studies. Uncovering the precise mechanisms of T cell-mediated immunity to Plasmodium infection can be accomplished through further investigations using mouse models of malaria with rodent Plasmodium parasites. Those findings would be invaluable to advance the efforts for development of an effective malaria vaccine.
Collapse
Affiliation(s)
- Yarob Ibraheem
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki-city, Nagasaki 852-8523, Japan
| | - Ganchimeg Bayarsaikhan
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki-city, Nagasaki 852-8523, Japan
| | - Shin-Ichi Inoue
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki-city, Nagasaki 852-8523, Japan.
| |
Collapse
|
11
|
Surette FA, Butler NS. Temporally Evolving and Context-Dependent Functions of Cytokines That Regulate Murine Anti-Plasmodium Humoral Immunity. Pathogens 2022; 11:pathogens11050523. [PMID: 35631044 PMCID: PMC9144513 DOI: 10.3390/pathogens11050523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 04/21/2022] [Accepted: 04/26/2022] [Indexed: 02/01/2023] Open
Abstract
Protective immunity against blood-stage Plasmodium infection and the disease malaria depends on antibodies secreted from high-affinity B cells selected during the germinal center (GC) response. The induction and stability of the GC response require the activation and direct cell–cell communication between parasite-specific CD4 helper T cells and B cells. However, cytokines secreted by helper T cells, B cells, and multiple other innate and adaptive immune cells also contribute to regulating the magnitude and protective functions of GC-dependent humoral immune responses. Here, we briefly review emerging data supporting the finding that specific cytokines can exhibit temporally distinct and context-dependent influences on the induction and maintenance of antimalarial humoral immunity.
Collapse
|
12
|
Stijlemans B, Schoovaerts M, De Baetselier P, Magez S, De Trez C. The Role of MIF and IL-10 as Molecular Yin-Yang in the Modulation of the Host Immune Microenvironment During Infections: African Trypanosome Infections as a Paradigm. Front Immunol 2022; 13:865395. [PMID: 35464430 PMCID: PMC9022210 DOI: 10.3389/fimmu.2022.865395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
African trypanosomes are extracellular flagellated unicellular protozoan parasites transmitted by tsetse flies and causing Sleeping Sickness disease in humans and Nagana disease in cattle and other livestock. These diseases are usually characterized by the development of a fatal chronic inflammatory disease if left untreated. During African trypanosome infection and many other infectious diseases, the immune response is mediating a see-saw balance between effective/protective immunity and excessive infection-induced inflammation that can cause collateral tissue damage. African trypanosomes are known to trigger a strong type I pro-inflammatory response, which contributes to peak parasitaemia control, but this can culminate into the development of immunopathologies, such as anaemia and liver injury, if not tightly controlled. In this context, the macrophage migration inhibitory factor (MIF) and the interleukin-10 (IL-10) cytokines may operate as a molecular “Yin-Yang” in the modulation of the host immune microenvironment during African trypanosome infection, and possibly other infectious diseases. MIF is a pleiotropic pro-inflammatory cytokine and critical upstream mediator of immune and inflammatory responses, associated with exaggerated inflammation and immunopathology. For example, it plays a crucial role in the pro-inflammatory response against African trypanosomes and other pathogens, thereby promoting the development of immunopathologies. On the other hand, IL-10 is an anti-inflammatory cytokine, acting as a master regulator of inflammation during both African trypanosomiasis and other diseases. IL-10 is crucial to counteract the strong MIF-induced pro-inflammatory response, leading to pathology control. Hence, novel strategies capable of blocking MIF and/or promoting IL-10 receptor signaling pathways, could potentially be used as therapy to counteract immunopathology development during African trypanosome infection, as well as during other infectious conditions. Together, this review aims at summarizing the current knowledge on the opposite immunopathological molecular “Yin-Yang” switch roles of MIF and IL-10 in the modulation of the host immune microenvironment during infection, and more particularly during African trypanosomiasis as a paradigm.
Collapse
Affiliation(s)
- Benoit Stijlemans
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Myeloid Cell Immunology Laboratory, Vlaams Instituut voor Biotechnologie (VIB) Centre for Inflammation Research, Brussels, Belgium
| | - Maxime Schoovaerts
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| | - Patrick De Baetselier
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Myeloid Cell Immunology Laboratory, Vlaams Instituut voor Biotechnologie (VIB) Centre for Inflammation Research, Brussels, Belgium
| | - Stefan Magez
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium.,Laboratory of Biomedical Research, Ghent University Global Campus, Incheon, South Korea
| | - Carl De Trez
- Laboratory of Cellular and Molecular Immunology, Vrije Universiteit Brussel (VUB), Brussels, Belgium
| |
Collapse
|
13
|
IL-10 Producing Regulatory B Cells Mediated Protection against Murine Malaria Pathogenesis. BIOLOGY 2022; 11:biology11050669. [PMID: 35625397 PMCID: PMC9138363 DOI: 10.3390/biology11050669] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/05/2022] [Accepted: 03/06/2022] [Indexed: 02/06/2023]
Abstract
Simple Summary The immunomodulatory role of B cell subset called regulatory B cells was evaluated during Plasmodium infection to study their role in susceptibility or resistance during infection. The expansion of regulatory B cells during Plasmodium infection indicated their important role in regulating the immune response. Adoptive transfer of regulatory B cells following infection with a lethal parasite resulted in enhanced survival of mice and inhibited growth of the Plasmodium parasite. Moreover, by inhibiting the production of the pro-inflammatory cytokine, IFN-γ, and stimulating anti-inflammatory IL-10 production, regulatory B cells may serve as an important contributor to protective immune response. Abstract Various immune cells are known to participate in combating infection. Regulatory B cells represent a subset of B cells that take part in immunomodulation and control inflammation. The immunoregulatory function of regulatory B cells has been shown in various murine models of several disorders. In this study, a comparable IL-10 competent B-10 cell subset (regulatory B cells) was characterized during lethal and non-lethal infection with malaria parasites using the mouse model. We observed that infection of Balb/c mice with P. yoelii I 7XL was lethal, and a rapid increase in dynamics of IL-10 producing B220+CD5+CD1d+ regulatory B cells over the course of infection was observed. However, animals infected with a less virulent strain of the parasite P. yoelii I7XNL attained complete resistance. It was observed that there is an increase in the population of regulatory B cells with an increase of parasitemia; however, a sudden drop in the frequency of these cells was observed with parasite clearance. Adoptive transfer of regulatory B cells to naïve mice followed by infection results in slow parasite growth and enhancement of survival in P. yoelii 17XL (lethal) infected animals. Adoptively transferred regulatory B cells also resulted in decreased production of pro-inflammatory cytokine (IFN-γ) and enhanced production of anti-inflammatory cytokine (IL-10). It infers that these regulatory B cells may contribute in immune protection by preventing the inflammation associated with disease and inhibiting the parasite growth.
Collapse
|
14
|
Kalkal M, Kalkal A, Dhanda SK, Das E, Pande V, Das J. A comprehensive study of epitopes and immune reactivity among Plasmodium species. BMC Microbiol 2022; 22:74. [PMID: 35277125 PMCID: PMC8913861 DOI: 10.1186/s12866-022-02480-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2021] [Accepted: 02/21/2022] [Indexed: 02/04/2023] Open
Abstract
Background Malaria is a life-threatening disease caused by protozoan parasite of genus Plasmodium. Various antigenic proteins of Plasmodium are considered as the major targets for the development of an effective vaccine. The aim of the current study was a comprehensive analysis of the experimentally validated epitopes of Plasmodium obtained from various immunoassays. Methods Plasmodium species epitopes were prefetched from Immune Epitope Database (IEDB). Species specific classification of available epitopes was done for both human and murine malaria parasites. Further, these T cell and B cell epitopes along with MHC I/II binders of different Plasmodium species were examined to find out their capability to induce IFN-γ and IL-10 using IFNepitope and IL-10 Pred, respectively. Results The species-specific classification of 6874 unique epitopes resulted in the selection of predominant human and murine Plasmodium species. Further, the attempt was made to analyse the immune reactivity of these epitopes for their ability to induce cytokines namely IFN-γ and IL-10. Total, 2775 epitopes were predicted to possess IFN-γ inducing ability, whereas 1275 epitopes were found to be involved in the induction of IL-10. Conclusions This study facilitates the assessment of Plasmodium epitopes and associated proteins as a potential approach to design and develop an epitope-based vaccine. Moreover, the results highlight the epitope-based immunization in malaria to induce a protective immune response. Supplementary Information The online version contains supplementary material available at 10.1186/s12866-022-02480-7.
Collapse
|
15
|
Mukherjee S, Ray G, Saha B, Kar SK. Oral Therapy Using a Combination of Nanotized Antimalarials and Immunomodulatory Molecules Reduces Inflammation and Prevents Parasite Induced Pathology in the Brain and Spleen of P. berghei ANKA Infected C57BL/6 Mice. Front Immunol 2022; 12:819469. [PMID: 35095923 PMCID: PMC8793777 DOI: 10.3389/fimmu.2021.819469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 12/22/2021] [Indexed: 11/13/2022] Open
Abstract
In malaria, anti-parasite immune response of the host may lead to dysregulated inflammation causing severe neuropathology arising from extensive damage to the Blood Brain Barrier (BBB). Use of anti-malarial drugs alone can control parasitemia and reduce inflammation but it cannot reduce pathology if chronic inflammation has already set in. In the present study, we have tested the efficacy of a new oral artemsinin based combination therapy (ACT) regimen using a combination of anti-malarial compounds like nanoartemisinin and nanoallylated-chalcone9 [{1-(4-Chlorophenyl)-3-[3-methoxy-4-(prop-2-en-1-yloxy) phenyl]-prop-2-en-1-one}]given together with anti-inflammatory-cum- anti-malarial compounds like nanoandrographolide and nanocurcumin to C57BL/6 mice infected with P. berghei ANKA. Untreated infected mice developed Experimental Cerebral Malaria (ECM) and died between 10 to 12 days after infection from severe BBB damage. We observed that oral treatments with nanoartemisinin or nano allylated chalcone 9 or nanoandrographolide alone, for 4 days after the onset of ECM, delayed the development of severe neurolopathology but could not prevent it. Nanocurcumin treatment for 4 days on the other hand, prevented damage to the BBB but the mice died because of hyperparasitemia. A single time oral administration of our ACT controlled blood parasitemia and prevented damage to the BBB, but recrudescence occurred due to persistence of parasites in the spleen. However the recrudescent parasites failed to induce ECM and BBB damage, leading to prolonged survival of the animals. A second time treatment at the start of recrudescence led to complete parasite clearance and survival of mice without pathology or parasitemia for 90 days. FACS analysis of spleen cells and gene expression profile in brain and spleen as well as quantitation of serum cytokine by ELISA showed that P. berghei ANKA infection in C57Bl/6 mice leads to a Th1-skewed immune response that result in severe inflammation and early death from ECM. Oral treatment with our ACT prevented a heightened pro-inflammatory response by modulating the Th1, Th2 and Treg immune responses and prevented ECM and death.
Collapse
Affiliation(s)
- Sitabja Mukherjee
- School of Biotechnology, KIIT deemed to be University, Bhubaneswar, India
| | - Gopesh Ray
- Nano Herb Research Laboratory, Kalinga Institute of Industrial Technology (KIIT) Technology Business Incubator, KIIT deemed to be University, Bhubaneswar, India
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune, India
| | - Santosh K. Kar
- Nano Herb Research Laboratory, Kalinga Institute of Industrial Technology (KIIT) Technology Business Incubator, KIIT deemed to be University, Bhubaneswar, India,*Correspondence: Santosh K. Kar,
| |
Collapse
|
16
|
Wang Q, Tang Y, Pan Z, Yuan Y, Zou Y, Zhang H, Guo X, Guo W, Huang X, Wu Z, Li C, Xu Q, Song J, Deng C. RNA-seq-based transcriptome analysis of the anti-inflammatory effect of artesunate in the early treatment of the mouse cerebral malaria model. Mol Omics 2022; 18:716-730. [DOI: 10.1039/d1mo00491c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The present study provides new insights into the molecular mechanisms by which artesunate improves prognosis in cerebral malaria, in particular inhibition of host cytokine storm.
Collapse
Affiliation(s)
- Qi Wang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
- Institute of Pulmonary Disease, Guangzhou Chest Hospital, Guangzhou, Guangdong, P. R. China
| | - Yexiao Tang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
- Guangzhou Eighth People’s Hospital, Guangzhou Medical University, Guangzhou, P. R. China
| | - Ziyi Pan
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Yueming Yuan
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
- Institute of Science and Technology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Yuanyuan Zou
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Hongying Zhang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
- Institute of Science and Technology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Xueying Guo
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Wenfeng Guo
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Xinan Huang
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Zhibin Wu
- Institute of Science and Technology, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Changqing Li
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Qin Xu
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Jianping Song
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| | - Changsheng Deng
- Artemisinin Research Center, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, P. R. China
| |
Collapse
|
17
|
Raacke M, Kerr A, Dörpinghaus M, Brehmer J, Wu Y, Lorenzen S, Fink C, Jacobs T, Roeder T, Sellau J, Bachmann A, Metwally NG, Bruchhaus I. Altered Cytokine Response of Human Brain Endothelial Cells after Stimulation with Malaria Patient Plasma. Cells 2021; 10:cells10071656. [PMID: 34359826 PMCID: PMC8303479 DOI: 10.3390/cells10071656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022] Open
Abstract
Infections with the deadliest malaria parasite, Plasmodium falciparum, are accompanied by a strong immunological response of the human host. To date, more than 30 cytokines have been detected in elevated levels in plasma of malaria patients compared to healthy controls. Endothelial cells (ECs) are a potential source of these cytokines, but so far it is not known if their cytokine secretion depends on the direct contact of the P. falciparum-infected erythrocytes (IEs) with ECs in terms of cytoadhesion. Culturing ECs with plasma from malaria patients (27 returning travellers) resulted in significantly increased secretion of IL-11, CXCL5, CXCL8, CXCL10, vascular endothelial growth factor (VEGF) and angiopoietin-like protein 4 (ANGPTL4) if compared to matching controls (22 healthy individuals). The accompanying transcriptome study of the ECs identified 43 genes that were significantly increased in expression (≥1.7 fold) after co-incubation with malaria patient plasma, including cxcl5 and angptl4. Further bioinformatic analyses revealed that biological processes such as cell migration, cell proliferation and tube development were particularly affected in these ECs. It can thus be postulated that not only the cytoadhesion of IEs, but also molecules in the plasma of malaria patients exerts an influence on ECs, and that not only the immunological response but also other processes, such as angiogenesis, are altered.
Collapse
Affiliation(s)
- Michaela Raacke
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; (M.R.); (A.K.); (M.D.); (J.B.); (Y.W.); (S.L.); (T.J.); (J.S.); (A.B.); (N.G.M.)
| | - Amy Kerr
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; (M.R.); (A.K.); (M.D.); (J.B.); (Y.W.); (S.L.); (T.J.); (J.S.); (A.B.); (N.G.M.)
| | - Michael Dörpinghaus
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; (M.R.); (A.K.); (M.D.); (J.B.); (Y.W.); (S.L.); (T.J.); (J.S.); (A.B.); (N.G.M.)
| | - Jana Brehmer
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; (M.R.); (A.K.); (M.D.); (J.B.); (Y.W.); (S.L.); (T.J.); (J.S.); (A.B.); (N.G.M.)
| | - Yifan Wu
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; (M.R.); (A.K.); (M.D.); (J.B.); (Y.W.); (S.L.); (T.J.); (J.S.); (A.B.); (N.G.M.)
| | - Stephan Lorenzen
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; (M.R.); (A.K.); (M.D.); (J.B.); (Y.W.); (S.L.); (T.J.); (J.S.); (A.B.); (N.G.M.)
| | - Christine Fink
- Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany; (C.F.); (T.R.)
| | - Thomas Jacobs
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; (M.R.); (A.K.); (M.D.); (J.B.); (Y.W.); (S.L.); (T.J.); (J.S.); (A.B.); (N.G.M.)
| | - Thomas Roeder
- Department of Molecular Physiology, Kiel University, 24118 Kiel, Germany; (C.F.); (T.R.)
- Airway Research Center North (ARCN), Member of the German Center for Lung Research (DZL), 24118 Kiel, Germany
| | - Julie Sellau
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; (M.R.); (A.K.); (M.D.); (J.B.); (Y.W.); (S.L.); (T.J.); (J.S.); (A.B.); (N.G.M.)
| | - Anna Bachmann
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; (M.R.); (A.K.); (M.D.); (J.B.); (Y.W.); (S.L.); (T.J.); (J.S.); (A.B.); (N.G.M.)
| | - Nahla Galal Metwally
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; (M.R.); (A.K.); (M.D.); (J.B.); (Y.W.); (S.L.); (T.J.); (J.S.); (A.B.); (N.G.M.)
| | - Iris Bruchhaus
- Bernhard Nocht Institute for Tropical Medicine, 20359 Hamburg, Germany; (M.R.); (A.K.); (M.D.); (J.B.); (Y.W.); (S.L.); (T.J.); (J.S.); (A.B.); (N.G.M.)
- Department of Biology, University of Hamburg, 20148 Hamburg, Germany
- Correspondence: ; Tel.: +49-404-281-8472
| |
Collapse
|
18
|
Mohan M, Bhattacharya D. Host-directed Therapy: A New Arsenal to Come. Comb Chem High Throughput Screen 2021; 24:59-70. [PMID: 32723230 DOI: 10.2174/1386207323999200728115857] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Revised: 06/04/2020] [Accepted: 06/15/2020] [Indexed: 11/22/2022]
Abstract
The emergence of drug-resistant strains among the variety of pathogens worsens the situation in today's scenario. In such a situation, a very heavy demand for developing the new antibiotics has arisen, but unfortunately, very limited success has been achieved in this arena till now. Infectious diseases usually make their impression in the form of severe pathology. Intracellular pathogens use the host's cell machinery for their survival. They alter the gene expression of several host's pathways and endorse to shut down the cell's innate defense pathway like apoptosis and autophagy. Intracellular pathogens are co-evolved with hosts and have a striking ability to manipulate the host's factors. They also mimic the host molecules and secrete them to prevent the host's proper immune response against them for their survival. Intracellular pathogens in chronic diseases create excessive inflammation. This excessive inflammation manifests in pathology. Host directed therapy could be alternative medicine in this situation; it targets the host factors, and abrogates the replication and persistence of pathogens inside the cell. It also provokes the anti-microbial immune response against the pathogen and reduces the exacerbation by enhancing the healing process to the site of pathology. HDT targets the host's factor involved in a certain pathway that ultimately targets the pathogen life cycle and helps in eradication of the pathogen. In such a scenario, HDT could also play a significant role in the treatment of drugsensitive as well with drug resistance strains because it targets the host's factors, which favors the pathogen survival inside the cell.
Collapse
Affiliation(s)
- Mradul Mohan
- National Institute of Malaria Research, New Delhi, India
| | - Debapriya Bhattacharya
- Center for Biotechnology, School of Pharmaceutical Sciences, SOA Deemed University, Bhubaneswar, Odisha, India
| |
Collapse
|
19
|
Patel H, Dunican C, Cunnington AJ. Predictors of outcome in childhood Plasmodium falciparum malaria. Virulence 2020; 11:199-221. [PMID: 32063099 PMCID: PMC7051137 DOI: 10.1080/21505594.2020.1726570] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022] Open
Abstract
Plasmodium falciparum malaria is classified as either uncomplicated or severe, determining clinical management and providing a framework for understanding pathogenesis. Severe malaria in children is defined by the presence of one or more features associated with adverse outcome, but there is wide variation in the predictive value of these features. Here we review the evidence for the usefulness of these features, alone and in combination, to predict death and other adverse outcomes, and we consider the role that molecular biomarkers may play in augmenting this prediction. We also examine whether a more personalized approach to predicting outcome for specific presenting syndromes of severe malaria, particularly cerebral malaria, has the potential to be more accurate. We note a general need for better external validation in studies of outcome predictors and for the demonstration that predictors can be used to guide clinical management in a way that improves survival and long-term health.
Collapse
Affiliation(s)
- Harsita Patel
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| | - Claire Dunican
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| | - Aubrey J. Cunnington
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| |
Collapse
|
20
|
Kuehlwein JM, Borsche M, Korir PJ, Risch F, Mueller A, Hübner MP, Hildner K, Hoerauf A, Dunay IR, Schumak B. Protection of Batf3-deficient mice from experimental cerebral malaria correlates with impaired cytotoxic T-cell responses and immune regulation. Immunology 2020; 159:193-204. [PMID: 31631339 PMCID: PMC6954726 DOI: 10.1111/imm.13137] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2019] [Revised: 09/30/2019] [Accepted: 10/14/2019] [Indexed: 12/28/2022] Open
Abstract
Excessive inflammatory immune responses during infections with Plasmodium parasites are responsible for severe complications such as cerebral malaria (CM) that can be studied experimentally in mice. Dendritic cells (DCs) activate cytotoxic CD8+ T-cells and initiate immune responses against the parasites. Batf3-/- mice lack a DC subset, which efficiently induces strong CD8 T-cell responses by cross-presentation of exogenous antigens. Here we show that Batf3-/- mice infected with Plasmodium berghei ANKA (PbA) were protected from experimental CM (ECM), characterized by a stable blood-brain barrier (BBB) and significantly less infiltrated peripheral immune cells in the brain. Importantly, the absence of ECM in Batf3-/- mice correlated with attenuated responses of cytotoxic T-cells, as their parasite-specific lytic activity as well as the production of interferon gamma and granzyme B were significantly decreased. Remarkably, spleens of ECM-protected Batf3-/- mice had elevated levels of regulatory immune cells and interleukin 10. Thus, protection from ECM in PbA-infected Batf3-/- mice was associated with the absence of strong CD8+ T-cell activity and induction of immunoregulatory mediators and cells.
Collapse
MESH Headings
- Animals
- Basic-Leucine Zipper Transcription Factors/deficiency
- Basic-Leucine Zipper Transcription Factors/genetics
- Blood-Brain Barrier/immunology
- Blood-Brain Barrier/parasitology
- Brain/immunology
- Brain/metabolism
- Brain/parasitology
- Cells, Cultured
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/parasitology
- Disease Models, Animal
- Female
- Granzymes/immunology
- Granzymes/metabolism
- Host-Parasite Interactions
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Interleukin-10/immunology
- Interleukin-10/metabolism
- Malaria, Cerebral/immunology
- Malaria, Cerebral/metabolism
- Malaria, Cerebral/parasitology
- Malaria, Cerebral/prevention & control
- Mice, Inbred C57BL
- Mice, Knockout
- Plasmodium berghei/immunology
- Plasmodium berghei/pathogenicity
- Repressor Proteins/deficiency
- Repressor Proteins/genetics
- Spleen/immunology
- Spleen/metabolism
- Spleen/parasitology
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/parasitology
Collapse
Affiliation(s)
- Janina M. Kuehlwein
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Max Borsche
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Patricia J. Korir
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Frederic Risch
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Ann‐Kristin Mueller
- Parasitology UnitCentre for Infectious DiseasesHeidelberg University HospitalHeidelbergGermany
- DZIF German Center for Infection ResearchPartner Site HeidelbergHeidelbergGermany
| | - Marc P. Hübner
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| | - Kai Hildner
- Medical Department 1University Hospital ErlangenErlangenGermany
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
- DZIF German Center for Infection ResearchPartner Site Bonn‐CologneBonnGermany
| | - Ildiko Rita Dunay
- Institute of Inflammation and NeurodegenerationUniversity of MagdeburgMagdeburgGermany
| | - Beatrix Schumak
- Institute of Medical Microbiology, Immunology and ParasitologyUniversity Hospital BonnBonnGermany
| |
Collapse
|
21
|
Zhang Y, Jiang N, Zhang T, Chen R, Feng Y, Sang X, Yang N, Chen Q. Tim-3 signaling blockade with α-lactose induces compensatory TIGIT expression in Plasmodium berghei ANKA-infected mice. Parasit Vectors 2019; 12:534. [PMID: 31711531 PMCID: PMC6849286 DOI: 10.1186/s13071-019-3788-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/04/2019] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Malaria, one of the largest health burdens worldwide, is caused by Plasmodium spp. infection. Upon infection, the host's immune system begins to clear the parasites. However, Plasmodium species have evolved to escape the host's immune clearance. T-cell immunoglobulin and mucin domain 3 (Tim-3), a surface molecule on most immune cells, is often referred to as an exhaustion marker. Galectin (Gal)-9 is a Tim-3 ligand and the T helper (Th) 1 cell response is inhibited when Gal-9 binds to Tim-3. In the present study, dynamic expression of Tim-3 on key populations of lymphocytes during infection periods of Plasmodium berghei and its significance in disease resistance and pathogenesis were explored. METHODS Tim-3 expression on critical lymphocyte populations and the proportion of these cells, as well as the levels of cytokines in the sera of infected mice, were detected by flow cytometry. Further, in vitro anti-Tim-3 assay using an anti-Tim-3 antibody and in vivo Tim-3-Gal-9 signaling blockade assays using α-lactose (an antagonist of Gal-9) were conducted. An Annexin V Apoptosis Detection Kit with propidium iodide was used to detect apoptosis. In addition, proteins associated with apoptosis in lung and spleen tissues were confirmed by Western blotting assays. RESULTS Increased Tim-3 expression on splenic CD8+ and splenic CD4+, and circulatory CD4+ T cells was associated with a reduction in the proportion of these cells. Furthermore, the levels of interleukin (IL)-2, IL-4, IL-6, IL-22, and interferon (IFN)-γ, but not that of tumor necrosis factor alpha (TNF-α), IL-10, and IL-9, increased to their highest levels at day 4 post-infection and decreased thereafter. Blocking Tim-3 signaling in vitro inhibited lymphocyte apoptosis. Tim-3-Gal-9 signaling blockade in vivo did not protect the mice, but induced the expression of the immunosuppressive molecule, T cell immunoreceptor with Ig and ITIM domains (TIGIT), in Plasmodium berghei ANKA-infected mice. CONCLUSIONS Tim-3 on lymphocytes negatively regulates cell-mediated immunity against Plasmodium infection, and blocking Tim-3-galectin 9 signaling using α-lactose did not significantly protect the mice; however, it induced the compensatory expression of TIGIT. Further investigations are required to identify whether combined blockade of Tim-3 and TIGIT signaling could achieve a better protective effect.
Collapse
Affiliation(s)
- Yiwei Zhang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang, 110866, China
| | - Ning Jiang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang, 110866, China
| | - Ting Zhang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China
| | - Ran Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang, 110866, China
| | - Ying Feng
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang, 110866, China
| | - Xiaoyu Sang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang, 110866, China
| | - Na Yang
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China.,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang, 110866, China
| | - Qijun Chen
- Key Laboratory of Livestock Infectious Diseases in Northeast China, Ministry of Education, College of Animal Science and Veterinary Medicine, Shenyang Agricultural University, Shenyang, 110866, China. .,The Research Unit for Pathogenic Mechanisms of Zoonotic Parasites, Chinese Academy of Medical Sciences, 120 Dongling Road, Shenyang, 110866, China.
| |
Collapse
|
22
|
Sato Y, Ries S, Stenzel W, Fillatreau S, Matuschewski K. The Liver-Stage Plasmodium Infection Is a Critical Checkpoint for Development of Experimental Cerebral Malaria. Front Immunol 2019; 10:2554. [PMID: 31736970 PMCID: PMC6837997 DOI: 10.3389/fimmu.2019.02554] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2019] [Accepted: 10/15/2019] [Indexed: 12/20/2022] Open
Abstract
Cerebral malaria is a life-threatening complication of malaria in humans, and the underlying pathogenic mechanisms are widely analyzed in a murine model of experimental cerebral malaria (ECM). Here, we show abrogation of ECM by hemocoel sporozoite-induced infection of a transgenic Plasmodium berghei line that overexpresses profilin, whereas these parasites remain fully virulent in transfusion-mediated blood infection. We, thus, demonstrate the importance of the clinically silent liver-stage infection for modulating the onset of ECM. Even though both parasites triggered comparable splenic immune cell expansion and accumulation of antigen-experienced CD8+ T cells in the brain, infection with transgenic sporozoites did not lead to cerebral vascular damages and suppressed the recruitment of overall lymphocyte populations. Strikingly, infection with the transgenic strain led to maintenance of CD115+Ly6C+ monocytes, which disappear in infected animals prone to ECM. An early induction of IL-10, IL-12p70, IL-6, and TNF at the time when parasites emerge from the liver might lead to a diminished induction of hepatic immunity. Collectively, our study reveals the essential role of early host interactions in the liver that may dampen the subsequent pro-inflammatory immune responses and influence the occurrence of ECM, highlighting a novel checkpoint in this fatal pathology.
Collapse
Affiliation(s)
- Yuko Sato
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Stefanie Ries
- Immune Regulation Research Group, Deutsches Rheuma-Forschungszentrum, Berlin, Germany
| | - Werner Stenzel
- Department of Neuropathology, Charité - Universitätmedizin, Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany
| | - Simon Fillatreau
- Immune Regulation Research Group, Deutsches Rheuma-Forschungszentrum, Berlin, Germany.,Department of Immunology, Infectiology and Haematology (I2H), Institut Necker-Enfants Malades, INSERM U1151-CNRS UMR 8253, Paris, France.,Faculté de Médecine, Université Paris Descartes, Sorbonne Paris Cité, Paris, France.,AP-HP, Hôpital Necker Enfants Malades, Paris, France
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany.,Department of Molecular Parasitology, Institute of Biology, Humboldt University, Berlin, Germany
| |
Collapse
|
23
|
Hortle E, Starrs L, Brown FC, Jane SM, Curtis DJ, McMorran BJ, Foote SJ, Burgio G. KCC1 Activation protects Mice from the Development of Experimental Cerebral Malaria. Sci Rep 2019; 9:6356. [PMID: 31015511 PMCID: PMC6478876 DOI: 10.1038/s41598-019-42782-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Accepted: 04/08/2019] [Indexed: 11/15/2022] Open
Abstract
Plasmodium falciparum malaria causes half a million deaths per year, with up to 9% of this mortality caused by cerebral malaria (CM). One of the major processes contributing to the development of CM is an excess of host inflammatory cytokines. Recently K+ signaling has emerged as an important mediator of the inflammatory response to infection; we therefore investigated whether mice carrying an ENU induced activation of the electroneutral K+ channel KCC1 had an altered response to Plasmodium berghei. Here we show that Kcc1M935K/M935K mice are protected from the development of experimental cerebral malaria, and that this protection is associated with an increased CD4+ and TNFa response. This is the first description of a K+ channel affecting the development of experimental cerebral malaria.
Collapse
Affiliation(s)
- Elinor Hortle
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Australian Capital Territory, Australia
| | - Lora Starrs
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Australian Capital Territory, Australia
| | - Fiona C Brown
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia
| | - Stephen M Jane
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia.,The Alfred Hospital, Melbourne, Australia.,Department of Medicine, Central Clinical School, Monash University, Melbourne, Australia
| | - David J Curtis
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Australia.,The Alfred Hospital, Melbourne, Australia
| | - Brendan J McMorran
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Australian Capital Territory, Australia
| | - Simon J Foote
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Australian Capital Territory, Australia
| | - Gaetan Burgio
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research, Australian National University, Australian Capital Territory, Australia.
| |
Collapse
|
24
|
Kumar R, Ng S, Engwerda C. The Role of IL-10 in Malaria: A Double Edged Sword. Front Immunol 2019; 10:229. [PMID: 30809232 PMCID: PMC6379449 DOI: 10.3389/fimmu.2019.00229] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 01/28/2019] [Indexed: 12/11/2022] Open
Abstract
IL-10 produced by CD4+ T cells suppresses inflammation by inhibiting T cell functions and the upstream activities of antigen presenting cells (APCs). IL-10 was first identified in Th2 cells, but has since been described in IFNγ-producing Tbet+ Th1, FoxP3+ CD4+ regulatory T (Treg) and IL-17-producing CD4+ T (Th17) cells, as well as many innate and innate-like immune cell populations. IL-10 production by Th1 cells has emerged as an important mechanism to dampen inflammation in the face of intractable infection, including in African children with malaria. However, although these type I regulatory T (Tr1) cells protect tissue from inflammation, they may also promote disease by suppressing Th1 cell-mediated immunity, thereby allowing infection to persist. IL-10 produced by other immune cells during malaria can also influence disease outcome, but the full impact of this IL-10 production is still unclear. Together, the actions of this potent anti-inflammatory cytokine along with other immunoregulatory mechanisms that emerge following Plasmodium infection represent a potential hurdle for the development of immunity against malaria, whether naturally acquired or vaccine-induced. Recent advances in understanding how IL-10 production is initiated and regulated have revealed new opportunities for manipulating IL-10 for therapeutic advantage. In this review, we will summarize our current knowledge about IL-10 production during malaria and discuss its impact on disease outcome. We will highlight recent advances in our understanding about how IL-10 production by specific immune cell subsets is regulated and consider how this knowledge may be used in drug delivery and vaccination strategies to help eliminate malaria.
Collapse
Affiliation(s)
- Rajiv Kumar
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, India.,Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Susanna Ng
- Immunology and Infection Lab, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Christian Engwerda
- Immunology and Infection Lab, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
25
|
T-lymphocytes response persists following Plasmodium berghei strain Anka infection resolution and may contribute to later experimental cerebral malaria outcomes. J Neuroimmunol 2019; 330:5-11. [PMID: 30763800 DOI: 10.1016/j.jneuroim.2019.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 02/04/2019] [Accepted: 02/07/2019] [Indexed: 01/20/2023]
Abstract
Several studies have proposed cerebral malaria (CM) as a CD4+ and CD8+ T lymphocyte-mediated disease. However, there are no data regarding the recruitment and/or persistence of these cells in the CNS following the phase of infection resolution. Glutamate-mediate excitotoxicity has also been implicated in CM. Blockade of glutamate NMDA receptors by its noncompetitive antagonist MK801 modulates cytokine and neurotrophic factors expression preventing cognitive and depressive-like behavior in experimental CM. Herein, we aim to investigate the role of T lymphocytes in later outcomes in CM, and whether the protective role of MK801 is associated with T lymphocytes response.
Collapse
|
26
|
Nakamae S, Kimura D, Miyakoda M, Sukhbaatar O, Inoue SI, Yui K. Role of IL-10 in inhibiting protective immune responses against infection with heterologous Plasmodium parasites. Parasitol Int 2019; 70:5-15. [PMID: 30639137 DOI: 10.1016/j.parint.2019.01.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 01/04/2019] [Accepted: 01/07/2019] [Indexed: 11/25/2022]
Abstract
Malaria is induced by infection with Plasmodium parasites, which are genetically diverse, and the immune response to Plasmodium infection has both allele-specific and cross-reactive components. To determine the role of the cross-reactive immune response in the protection and disease manifestation in heterologous Plasmodium infection, we used infection models of P. chabaudi chabaudi (Pcc) and P. berghei ANKA (PbA). CD4+ T cells primed with Pcc infection exhibited strong cross-reactivity to PbA antigens. We infected C57BL/6 mice with Pcc and subsequently treated them with an anti-Plasmodium drug. The Pcc-primed mice exhibited reduced parasitemia and showed no signs of experimental cerebral malaria after infection with PbA. CD4+ T cells from the Pcc-primed mice produced high levels of IFN-γ and IL-10 in response to PbA early after PbA infection. The blockade of IL-10 signaling with anti-IL-10 receptor antibody increased the proportion of activated CD4+ and γδ T cells and the IFN-γ production by CD4+ T cells in response to PbA antigens, while markedly reducing the levels of parasitemia. In contrast, IL-10 blockade did not have a significant effect on parasitemia levels in unprimed mice after PbA infection. These data suggest a potent regulatory role of IL-10 in the cross-reactive memory response to the infection with heterologous Plasmodium parasites leading to the inhibition of the protective immunity and pathogenesis.
Collapse
Affiliation(s)
- Sayuri Nakamae
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Infectious Diseases, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Daisuke Kimura
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Immunology, School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Health, Sports, and Nutrition, Faculty of Health and Welfare, Kobe Women's University, 4-7-2 Minatojima-nakamachi, Chuo-ku, Kobe 650-0046, Japan
| | - Mana Miyakoda
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Immunology, School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Research and Education Center for Drug Fostering and Evolution, School of Pharmaceutical Sciences, Nagasaki University, 1-14 Bunkyomachi, Nagasaki 852-8521, Japan
| | - Odsuren Sukhbaatar
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Infectious Diseases, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Shin-Ichi Inoue
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Immunology, School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan
| | - Katsuyuki Yui
- Division of Immunology, Department of Molecular Microbiology and Immunology, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Program for Nurturing Global Leaders in Tropical and Emerging Infectious Diseases, Graduate School of Biomedical Sciences, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Department of Immunology, School of Medicine, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan; Graduate School of Tropical Medicine and Global Health, Nagasaki University, 1-12-4 Sakamoto, Nagasaki 852-8523, Japan.
| |
Collapse
|
27
|
Abstract
Nontyphoidal salmonellae (NTS) are a major cause of invasive (iNTS) disease in sub-Saharan Africa, manifesting as bacteremia and meningitis. Available epidemiological data indicate that iNTS disease is endemic in much of the region. Antimicrobial resistance is common and case fatality rates are high. There are well-characterized clinical associations with iNTS disease, including young age, HIV infection, malaria, malnutrition, anemia, and sickle cell disease. However, the clinical presentation of iNTS disease is often with fever alone, so clinical diagnosis is impossible without blood culture confirmation. No vaccine is currently available, making this a priority area for global health research. Over the past ten years, it has emerged that iNTS disease in Africa is caused by distinct pathovars of Salmonella Typhimurium, belonging to sequence type ST313, and Salmonella Enteritidis. These are characterized by genome degradation and appear to be adapting to an invasive lifestyle. Investigation of rare patients with primary immunodeficiencies has suggested a key role for interferon gamma-mediated immunity in host defense against NTS. This concept has been supported by recent population-based host genetic studies in African children. In contrast, immunoepidemiological studies from Africa indicate an important role for antibody for protective immunity, supporting the development of antibody-inducing vaccines against iNTS disease. With candidate O-antigen-based vaccines due to enter clinical trials in the near future, research efforts should focus on understanding the relative contributions of antibody and cell-mediated immunity to protection against iNTS disease in humans.
Collapse
Affiliation(s)
| | - Calman A MacLennan
- Jenner Institute, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| |
Collapse
|
28
|
Cassin-Sackett L, Callicrate TE, Fleischer RC. Parallel evolution of gene classes, but not genes: Evidence from Hawai'ian honeycreeper populations exposed to avian malaria. Mol Ecol 2018; 28:568-583. [PMID: 30298567 DOI: 10.1111/mec.14891] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2017] [Revised: 10/14/2018] [Accepted: 10/19/2018] [Indexed: 12/29/2022]
Abstract
Adaptation in nature is ubiquitous, yet characterizing its genomic basis is difficult because population demographics cause correlations with nonadaptive loci. Introduction events provide opportunities to observe adaptation over known spatial and temporal scales, facilitating the identification of genes involved in adaptation. The pathogen causing avian malaria, Plasmodium relictum, was introduced to Hawai'i in the 1930s and elicited extinctions and precipitous population declines in native honeycreepers. After a sharp initial population decline, the Hawai'i 'amakihi (Chlorodrepanis virens) has evolved tolerance to the parasite at low elevations where P. relictum exists, and can sustain infection without major fitness consequences. High-elevation, unexposed populations of 'amakihi display little to no tolerance. To explore the genomic basis of adaptation to P. relictum in low-elevation 'amakihi, we genotyped 125 'amakihi from the island of Hawai'i via hybridization capture to 40,000 oligonucleotide baits containing SNPs and used the reference 'amakihi genome to identify genes potentially under selection from malaria. We tested for outlier loci between low- and high-elevation population pairs and identified loci with signatures of selection within low-elevation populations. In some cases, genes commonly involved in the immune response (e.g., major histocompatibility complex) were associated with malaria presence in the population. We also detected several novel candidate loci that may be implicated in surviving malaria infection (e.g., beta-defensin, glycoproteins and interleukin-related genes). Our results suggest that rapid adaptation to pathogens may occur through changes in different immune genes, but in the same classes of genes, across populations.
Collapse
Affiliation(s)
- Loren Cassin-Sackett
- Center for Conservation Genomics, Smithsonian Conservation Biology Institute, National Zoological Park, Washington, District of Columbia.,Department of Integrative Biology, University of South Florida, Tampa, Florida
| | - Taylor E Callicrate
- Center for Conservation Genomics, Smithsonian Conservation Biology Institute, National Zoological Park, Washington, District of Columbia.,Species Conservation Toolkit Initiative, Department of Conservation Science, Chicago Zoological Society, Brookfield, Illinois
| | - Robert C Fleischer
- Center for Conservation Genomics, Smithsonian Conservation Biology Institute, National Zoological Park, Washington, District of Columbia
| |
Collapse
|
29
|
Burrack KS, Huggins MA, Taras E, Dougherty P, Henzler CM, Yang R, Alter S, Jeng EK, Wong HC, Felices M, Cichocki F, Miller JS, Hart GT, Johnson AJ, Jameson SC, Hamilton SE. Interleukin-15 Complex Treatment Protects Mice from Cerebral Malaria by Inducing Interleukin-10-Producing Natural Killer Cells. Immunity 2018; 48:760-772.e4. [PMID: 29625893 DOI: 10.1016/j.immuni.2018.03.012] [Citation(s) in RCA: 55] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 11/27/2017] [Accepted: 03/05/2018] [Indexed: 12/21/2022]
Abstract
Cerebral malaria is a deadly complication of Plasmodium infection and involves blood brain barrier (BBB) disruption following infiltration of white blood cells. During experimental cerebral malaria (ECM), mice inoculated with Plasmodium berghei ANKA-infected red blood cells develop a fatal CM-like disease caused by CD8+ T cell-mediated pathology. We found that treatment with interleukin-15 complex (IL-15C) prevented ECM, whereas IL-2C treatment had no effect. IL-15C-expanded natural killer (NK) cells were necessary and sufficient for protection against ECM. IL-15C treatment also decreased CD8+ T cell activation in the brain and prevented BBB breakdown without influencing parasite load. IL-15C induced NK cells to express IL-10, which was required for IL-15C-mediated protection against ECM. Finally, we show that ALT-803, a modified human IL-15C, mediates similar induction of IL-10 in NK cells and protection against ECM. These data identify a regulatory role for cytokine-stimulated NK cells in the prevention of a pathogenic immune response.
Collapse
Affiliation(s)
- Kristina S Burrack
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Matthew A Huggins
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA; Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Emily Taras
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Philip Dougherty
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Christine M Henzler
- Supercomputing Institute for Advanced Computational Research, University of Minnesota, Minneapolis, MN 55414, USA
| | - Rendong Yang
- Supercomputing Institute for Advanced Computational Research, University of Minnesota, Minneapolis, MN 55414, USA
| | - Sarah Alter
- Altor BioScience Corporation, Miramar, FL 33025, USA
| | - Emily K Jeng
- Altor BioScience Corporation, Miramar, FL 33025, USA
| | - Hing C Wong
- Altor BioScience Corporation, Miramar, FL 33025, USA
| | - Martin Felices
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Frank Cichocki
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Jeffrey S Miller
- Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Geoffrey T Hart
- Center for Immunology, Department of Medicine, University of Minnesota, Minneapolis, MN 55414, USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN 55905, USA
| | - Stephen C Jameson
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA
| | - Sara E Hamilton
- Center for Immunology, Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN 55414, USA.
| |
Collapse
|
30
|
Perillyl alcohol exhibits in vitro inhibitory activity against Plasmodium falciparum and protects against experimental cerebral malaria. Int J Antimicrob Agents 2018; 51:370-377. [DOI: 10.1016/j.ijantimicag.2017.08.025] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 08/08/2017] [Accepted: 08/15/2017] [Indexed: 11/20/2022]
|
31
|
Schmidt KE, Kuepper JM, Schumak B, Alferink J, Hofmann A, Howland SW, Rénia L, Limmer A, Specht S, Hoerauf A. Doxycycline inhibits experimental cerebral malaria by reducing inflammatory immune reactions and tissue-degrading mediators. PLoS One 2018; 13:e0192717. [PMID: 29438386 PMCID: PMC5811026 DOI: 10.1371/journal.pone.0192717] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 01/29/2018] [Indexed: 12/22/2022] Open
Abstract
Malaria ranks among the most important infectious diseases worldwide and affects mostly people living in tropical countries. Mechanisms involved in disease progression are still not fully understood and specific treatments that might interfere with cerebral malaria (CM) are limited. Here we show that administration of doxycycline (DOX) prevented experimental CM (ECM) in Plasmodium berghei ANKA (PbA)-infected C57BL/6 wildtype (WT) mice in an IL-10-independent manner. DOX-treated mice showed an intact blood-brain barrier (BBB) and attenuated brain inflammation. Importantly, if WT mice were infected with a 20-fold increased parasite load, they could be still protected from ECM if they received DOX from day 4-6 post infection, despite similar parasitemia compared to control-infected mice that did not receive DOX and developed ECM. Infiltration of T cells and cytotoxic responses were reduced in brains of DOX-treated mice. Analysis of brain tissue by RNA-array revealed reduced expression of chemokines and tumour necrosis factor (TNF) in brains of DOX-treated mice. Furthermore, DOX-administration resulted in brains of the mice in reduced expression of matrix metalloproteinase 2 (MMP2) and granzyme B, which are both factors associated with ECM pathology. Systemic interferon gamma production was reduced and activated peripheral T cells accumulated in the spleen in DOX-treated mice. Our results suggest that DOX targeted inflammatory processes in the central nervous system (CNS) and prevented ECM by impaired brain access of effector T cells in addition to its anti-parasitic effect, thereby expanding the understanding of molecular events that underlie DOX-mediated therapeutic interventions.
Collapse
Affiliation(s)
- Kim E. Schmidt
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Janina M. Kuepper
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Beatrix Schumak
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
- * E-mail:
| | - Judith Alferink
- Department of Psychiatry and Psychotherapy, University Hospital Muenster, Muenster, Germany
| | - Andrea Hofmann
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Shanshan W. Howland
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Laurent Rénia
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Andreas Limmer
- Clinic for Anaesthesiology and Intensive Care, University Hospital Essen, Essen, Germany
- Institutes of Molecular Medicine and Experimental Immunology, University Hospital Bonn, Bonn, Germany
| | - Sabine Specht
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| | - Achim Hoerauf
- Institute of Medical Microbiology, Immunology and Parasitology, University Hospital Bonn, Bonn, Germany
| |
Collapse
|
32
|
Crowley VM, Ayi K, Lu Z, Liby KT, Sporn M, Kain KC. Synthetic oleanane triterpenoids enhance blood brain barrier integrity and improve survival in experimental cerebral malaria. Malar J 2017; 16:463. [PMID: 29137631 PMCID: PMC5686938 DOI: 10.1186/s12936-017-2109-0] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2017] [Accepted: 11/04/2017] [Indexed: 12/31/2022] Open
Abstract
Background Cerebral malaria (CM) is a severe complication of Plasmodium falciparum infection associated with high mortality and neurocognitive impairment in survivors. New anti-malarials and host-based adjunctive therapy may improve clinical outcome in CM. Synthetic oleanane triterpenoid (SO) compounds have shown efficacy in the treatment of diseases where inflammation and oxidative stress contribute to pathogenesis. Methods A derivative of the SO 2-cyano-3,12-dioxooleana-1,9-dien-28-oic acid (CDDO), CDDO-ethyl amide (CDDO-EA) was investigated for the treatment of severe malaria in a pre-clinical model. CDDO-EA was evaluated in vivo as a monotherapy as well as adjunctive therapy with parenteral artesunate in the Plasmodium berghei strain ANKA experimental cerebral malaria (ECM) model. Results CDDO-EA alone improved outcome in ECM and, given as adjunctive therapy in combination with artesunate, it significantly improved outcome over artesunate alone (p = 0.009). Improved survival was associated with reduced inflammation, enhanced endothelial stability and blood–brain barrier integrity. Survival was improved even when administered late in the disease course after the onset of neurological symptoms. Conclusions These results indicate that SO are a new class of immunomodulatory drugs and support further studies investigating this class of agents as potential adjunctive therapy for severe malaria.
Collapse
Affiliation(s)
- Valerie M Crowley
- S. A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, Canada
| | - Kodjo Ayi
- S. A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, Canada
| | - Ziyue Lu
- S. A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, Canada
| | - Karen T Liby
- Department of Pharmacology, Dartmouth Medical School, Hanover, NH, USA
| | - Michael Sporn
- Department of Pharmacology, Dartmouth Medical School, Hanover, NH, USA
| | - Kevin C Kain
- S. A. Rotman Laboratories, Sandra Rotman Centre for Global Health, University Health Network-Toronto General Hospital, Toronto, Canada. .,Department of Medicine, University of Toronto, Toronto, ON, Canada. .,Tropical Diseases Unit, Division of Infectious Diseases, Department of Medicine, UHN-Toronto General Hospital, Toronto, ON, Canada.
| |
Collapse
|
33
|
Keita Alassane S, Nicolau-Travers ML, Menard S, Andreoletti O, Cambus JP, Gaudre N, Wlodarczyk M, Blanchard N, Berry A, Abbes S, Colongo D, Faye B, Augereau JM, Lacroux C, Iriart X, Benoit-Vical F. Young Sprague Dawley rats infected by Plasmodium berghei: A relevant experimental model to study cerebral malaria. PLoS One 2017; 12:e0181300. [PMID: 28742109 PMCID: PMC5524346 DOI: 10.1371/journal.pone.0181300] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2016] [Accepted: 06/14/2017] [Indexed: 02/06/2023] Open
Abstract
Cerebral malaria (CM) is the most severe manifestation of human malaria yet is still poorly understood. Mouse models have been developed to address the subject. However, their relevance to mimic human pathogenesis is largely debated. Here we study an alternative cerebral malaria model with an experimental Plasmodium berghei Keyberg 173 (K173) infection in Sprague Dawley rats. As in Human, not all infected subjects showed cerebral malaria, with 45% of the rats exhibiting Experimental Cerebral Malaria (ECM) symptoms while the majority (55%) of the remaining rats developed severe anemia and hyperparasitemia (NoECM). These results allow, within the same population, a comparison of the noxious effects of the infection between ECM and severe malaria without ECM. Among the ECM rats, 77.8% died between day 5 and day 12 post-infection, while the remaining rats were spontaneously cured of neurological signs within 24-48 hours. The clinical ECM signs observed were paresis quickly evolving to limb paralysis, global paralysis associated with respiratory distress, and coma. The red blood cell (RBC) count remained normal but a drastic decrease of platelet count and an increase of white blood cell numbers were noted. ECM rats also showed a decrease of glucose and total CO2 levels and an increase of creatinine levels compared to control rats or rats with no ECM. Assessment of the blood-brain barrier revealed loss of integrity, and interestingly histopathological analysis highlighted cyto-adherence and sequestration of infected RBCs in brain vessels from ECM rats only. Overall, this ECM rat model showed numerous clinical and histopathological features similar to Human CM and appears to be a promising model to achieve further understanding the CM pathophysiology in Humans and to evaluate the activity of specific antimalarial drugs in avoiding/limiting cerebral damages from malaria.
Collapse
Affiliation(s)
- Sokhna Keita Alassane
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, Toulouse, France
- Université de Toulouse, UPS, INPT, Toulouse, France
- UFR Sciences de la Santé, Université Gaston Berger, St Louis, Sénégal
| | - Marie-Laure Nicolau-Travers
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, Toulouse, France
- Université de Toulouse, UPS, INPT, Toulouse, France
| | - Sandie Menard
- CPTP (Centre de Physiopathologie de Toulouse Purpan), INSERM U1043, CNRS UMR5282, Université de Toulouse III, Toulouse, France
| | - Olivier Andreoletti
- UMR INRA ENVT 1225, Interactions Hôte Agent Pathogène, Ecole Nationale Vétérinaire de Toulouse, 23 Chemin des Capelles, Toulouse, France
| | - Jean-Pierre Cambus
- Laboratoire Hématologie, Centre Hospitalier Universitaire, Toulouse, France
| | - Noémie Gaudre
- CPTP (Centre de Physiopathologie de Toulouse Purpan), INSERM U1043, CNRS UMR5282, Université de Toulouse III, Toulouse, France
| | - Myriam Wlodarczyk
- CPTP (Centre de Physiopathologie de Toulouse Purpan), INSERM U1043, CNRS UMR5282, Université de Toulouse III, Toulouse, France
| | - Nicolas Blanchard
- CPTP (Centre de Physiopathologie de Toulouse Purpan), INSERM U1043, CNRS UMR5282, Université de Toulouse III, Toulouse, France
| | - Antoine Berry
- Service de Parasitologie-Mycologie, Centre Hospitalier Universitaire, Toulouse, France
| | - Sarah Abbes
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, Toulouse, France
- Université de Toulouse, UPS, INPT, Toulouse, France
| | | | - Babacar Faye
- UFR Sciences de la Santé, Université Gaston Berger, St Louis, Sénégal
| | - Jean-Michel Augereau
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, Toulouse, France
- Université de Toulouse, UPS, INPT, Toulouse, France
| | - Caroline Lacroux
- UMR INRA ENVT 1225, Interactions Hôte Agent Pathogène, Ecole Nationale Vétérinaire de Toulouse, 23 Chemin des Capelles, Toulouse, France
| | - Xavier Iriart
- CPTP (Centre de Physiopathologie de Toulouse Purpan), INSERM U1043, CNRS UMR5282, Université de Toulouse III, Toulouse, France
- Service de Parasitologie-Mycologie, Centre Hospitalier Universitaire, Toulouse, France
| | - Françoise Benoit-Vical
- CNRS, LCC (Laboratoire de Chimie de Coordination), 205 route de Narbonne, Toulouse, France
- Université de Toulouse, UPS, INPT, Toulouse, France
| |
Collapse
|
34
|
Van Den Ham KM, Smith LK, Richer MJ, Olivier M. Protein Tyrosine Phosphatase Inhibition Prevents Experimental Cerebral Malaria by Precluding CXCR3 Expression on T Cells. Sci Rep 2017; 7:5478. [PMID: 28710387 PMCID: PMC5511231 DOI: 10.1038/s41598-017-05609-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 05/31/2017] [Indexed: 11/25/2022] Open
Abstract
Cerebral malaria induced by Plasmodium berghei ANKA infection is dependent on the sequestration of cytotoxic T cells within the brain and augmentation of the inflammatory response. Herein, we demonstrate that inhibition of protein tyrosine phosphatase (PTP) activity significantly attenuates T cell sequestration within the brain and prevents the development of neuropathology. Mechanistically, the initial upregulation of CXCR3 on splenic T cells upon T cell receptor stimulation was critically decreased through the reduction of T cell-intrinsic PTP activity. Furthermore, PTP inhibition markedly increased IL-10 production by splenic CD4+ T cells by enhancing the frequency of LAG3+CD49b+ type 1 regulatory cells. Overall, these findings demonstrate that modulation of PTP activity could possibly be utilized in the treatment of cerebral malaria and other CXCR3-mediated diseases.
Collapse
Affiliation(s)
- Kristin M Van Den Ham
- Department of Microbiology and Immunology, McGill University, Montréal, QC, H3A 0G4, Canada.,Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, H4A 3J1, Canada
| | - Logan K Smith
- Department of Microbiology and Immunology, McGill University, Montréal, QC, H3A 0G4, Canada.,Microbiome and Disease Tolerance Centre and Associate Member, Goodman Cancer Research Centre, McGill University, Montréal, QC, H3A 2B4, Canada
| | - Martin J Richer
- Department of Microbiology and Immunology, McGill University, Montréal, QC, H3A 0G4, Canada. .,Microbiome and Disease Tolerance Centre and Associate Member, Goodman Cancer Research Centre, McGill University, Montréal, QC, H3A 2B4, Canada.
| | - Martin Olivier
- Department of Microbiology and Immunology, McGill University, Montréal, QC, H3A 0G4, Canada. .,Infectious Diseases and Immunity in Global Health Program, Research Institute of the McGill University Health Centre, Montréal, QC, H4A 3J1, Canada.
| |
Collapse
|
35
|
Loevenich K, Ueffing K, Abel S, Hose M, Matuschewski K, Westendorf AM, Buer J, Hansen W. DC-Derived IL-10 Modulates Pro-inflammatory Cytokine Production and Promotes Induction of CD4 +IL-10 + Regulatory T Cells during Plasmodium yoelii Infection. Front Immunol 2017; 8:152. [PMID: 28293237 PMCID: PMC5328999 DOI: 10.3389/fimmu.2017.00152] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Accepted: 01/30/2017] [Indexed: 02/02/2023] Open
Abstract
The cytokine IL-10 plays a crucial role during malaria infection by counteracting the pro-inflammatory immune response. We and others demonstrated that Plasmodium yoelii infection results in enhanced IL-10 production in CD4+ T cells accompanied by the induction of an immunosuppressive phenotype. However, it is unclear whether this is a direct effect caused by the parasite or an indirect consequence due to T cell activation by IL-10-producing antigen-presenting cells. Here, we demonstrate that CD11c+CD11b+CD8− dendritic cells (DCs) produce elevated levels of IL-10 after P. yoelii infection of BALB/c mice. DC-specific ablation of IL-10 in P. yoelii-infected IL-10flox/flox/CD11c-cre mice resulted in increased IFN-γ and TNF-α production with no effect on MHC-II, CD80, or CD86 expression in CD11c+ DCs. Accordingly, DC-specific ablation of IL-10 exacerbated systemic IFN-γ and IL-12 production without altering P. yoelii blood stage progression. Strikingly, DC-specific inactivation of IL-10 in P. yoelii-infected mice interfered with the induction of IL-10-producing CD4+ T cells while raising the frequency of IFN-γ-secreting CD4+ T cells. These results suggest that P. yoelii infection promotes IL-10 production in DCs, which in turn dampens secretion of pro-inflammatory cytokines and supports the induction of CD4+IL-10+ T cells.
Collapse
Affiliation(s)
- Katharina Loevenich
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Kristina Ueffing
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Simone Abel
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Matthias Hose
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Kai Matuschewski
- Institute of Biology, Humboldt University, Berlin, Germany; Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
| | - Astrid M Westendorf
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Jan Buer
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| | - Wiebke Hansen
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen , Essen , Germany
| |
Collapse
|
36
|
Roussilhon C, Bang G, Bastaert F, Solhonne B, Garcia-Verdugo I, Peronet R, Druilhe P, Sakuntabhai A, Mecheri S, Sallenave JM. The antimicrobial molecule trappin-2/elafin has anti-parasitic properties and is protective in vivo in a murine model of cerebral malaria. Sci Rep 2017; 7:42243. [PMID: 28181563 PMCID: PMC5299836 DOI: 10.1038/srep42243] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 01/05/2017] [Indexed: 11/18/2022] Open
Abstract
According to the WHO, and despite reduction in mortality rates, there were an estimated 438 000 malaria deaths in 2015. Therefore new antimalarials capable of limiting organ damage are still required. We show that systemic and lung adenovirus (Ad)-mediated over-expression of trappin-2 (T-2) an antibacterial molecule with anti-inflammatory activity, increased mice survival following infection with the cerebral malaria-inducing Plasmodium berghei ANKA (PbANKA) strain. Systemically, T-2 reduced PbANKA sequestration in spleen, lung, liver and brain, associated with a decrease in pro-inflammatory cytokines (eg TNF-α in spleen and lung) and an increase in IL-10 production in the lung. Similarly, local lung instillation of Ad-T-2 resulted in a reduced organ parasite sequestration and a shift towards an anti-inflammatory/repair response, potentially implicating monocytes in the protective phenotype. Relatedly, we demonstrated in vitro that human monocytes incubated with Plasmodium falciparum-infected red blood cells (Pf-iRBCs) and IgGs from hyper-immune African human sera produced T-2 and that the latter colocalized with merozoites and inhibited Pf multiplication. This array of data argues for the first time for the potential therapeutic usefulness of this host defense peptide in human malaria patients, with the aim to limit acute lung injury and respiratory distress syndrom often observed during malaria episodes.
Collapse
Affiliation(s)
- Christian Roussilhon
- Unité de génétique fonctionnelle des maladies infectieuses and CNRS Unité de recherche associée 3012; Paris, 75015, France
| | - Gilles Bang
- Unité de génétique fonctionnelle des maladies infectieuses and CNRS Unité de recherche associée 3012; Paris, 75015, France
| | - Fabien Bastaert
- Unité de Défense Innée et Inflammation, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
- INSERM U874, Institut Pasteur
- INSERM U1152, Faculté de Médicine site Bichat, Université Paris Diderot, Université Sorbonne Paris-Cité, 16, rue Henri Huchard, Paris, 75018, France
| | - Brigitte Solhonne
- Unité de Défense Innée et Inflammation, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
- INSERM U874, Institut Pasteur
- INSERM U1152, Faculté de Médicine site Bichat, Université Paris Diderot, Université Sorbonne Paris-Cité, 16, rue Henri Huchard, Paris, 75018, France
| | - Ignacio Garcia-Verdugo
- Unité de Défense Innée et Inflammation, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
- INSERM U874, Institut Pasteur
- INSERM U1152, Faculté de Médicine site Bichat, Université Paris Diderot, Université Sorbonne Paris-Cité, 16, rue Henri Huchard, Paris, 75018, France
| | - Roger Peronet
- Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, CNRS ERL9195 and INSERM U1201, Paris F-75015, France
- CNRS ERL9195 and INSERM U1201, Paris F-75015, France
- INSERM U1201, Paris F-75015, France
| | | | - Anavaj Sakuntabhai
- Unité de génétique fonctionnelle des maladies infectieuses and CNRS Unité de recherche associée 3012; Paris, 75015, France
| | - Salaheddine Mecheri
- Institut Pasteur, Unité de Biologie des Interactions Hôte Parasites, CNRS ERL9195 and INSERM U1201, Paris F-75015, France
- CNRS ERL9195 and INSERM U1201, Paris F-75015, France
- INSERM U1201, Paris F-75015, France
| | - Jean-Michel Sallenave
- Unité de Défense Innée et Inflammation, Institut Pasteur, 25 rue du Dr Roux, Paris, 75015, France
- INSERM U874, Institut Pasteur
- INSERM U1152, Faculté de Médicine site Bichat, Université Paris Diderot, Université Sorbonne Paris-Cité, 16, rue Henri Huchard, Paris, 75018, France
| |
Collapse
|
37
|
de Miranda AS, Brant F, Vieira LB, Rocha NP, Vieira ÉLM, Rezende GHS, de Oliveira Pimentel PM, Moraes MFD, Ribeiro FM, Ransohoff RM, Teixeira MM, Machado FS, Rachid MA, Teixeira AL. A Neuroprotective Effect of the Glutamate Receptor Antagonist MK801 on Long-Term Cognitive and Behavioral Outcomes Secondary to Experimental Cerebral Malaria. Mol Neurobiol 2016; 54:7063-7082. [PMID: 27796746 DOI: 10.1007/s12035-016-0226-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 10/13/2016] [Indexed: 02/06/2023]
Abstract
Cerebral malaria (CM) is a life-threatening complication of Plasmodium falciparum infection, which can result in long-term cognitive and behavioral deficits despite successful anti-malarial therapy. Due to the substantial social and economic burden of CM, the development of adjuvant therapies is a scientific goal of highest priority. Apart from vascular and immune responses, changes in glutamate system have been reported in CM pathogenesis suggesting a potential therapeutic target. Based on that, we hypothesized that interventions in the glutamatergic system induced by blockage of N-methyl-D-aspartate (NMDA) receptors could attenuate experimental CM long-term cognitive and behavioral outcomes. Before the development of evident CM signs, susceptible mice infected with Plasmodium berghei ANKA (PbA) strain were initiated on treatment with dizocilpine maleate (MK801, 0.5 mg/kg), a noncompetitive NMDA receptor antagonist. On day 5 post-infection, mice were treated orally with a 10-day course chloroquine (CQ, 30 mg/kg). Control mice also received saline, CQ or MK801 + CQ therapy. After 10 days of cessation of CQ treatment, magnetic resonance images (MRI), behavioral and immunological assays were performed. Indeed, MK801 combined with CQ prevented long-term memory impairment and depressive-like behavior following successful PbA infection resolution. In addition, MK801 also modulated the immune system by promoting a balance of TH1/TH2 response and upregulating neurotrophic factors levels in the frontal cortex and hippocampus. Moreover, hippocampus abnormalities observed by MRI were partially prevented by MK801 treatment. Our results indicate that NMDA receptor antagonists can be neuroprotective in CM and could be a valuable adjuvant strategy for the management of the long-term impairment observed in CM.
Collapse
Affiliation(s)
- Aline Silva de Miranda
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Laboratory of Neurobiology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Departamento de Morfologia, ICB, UFMG, Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901, Brazil.
| | - Fátima Brant
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciene Bruno Vieira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Natália Pessoa Rocha
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Érica Leandro Marciano Vieira
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gustavo Henrique Souza Rezende
- Department of Physiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Marcio F D Moraes
- Department of Physiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabíola Mara Ribeiro
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Mauro Martins Teixeira
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabiana Simão Machado
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Milene Alvarenga Rachid
- Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antônio Lúcio Teixeira
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
38
|
|
39
|
Alferink J, Specht S, Arends H, Schumak B, Schmidt K, Ruland C, Lundt R, Kemter A, Dlugos A, Kuepper JM, Poppensieker K, Findeiss M, Albayram Ö, Otte DM, Marazzi J, Gertsch J, Förster I, Maier W, Scheu S, Hoerauf A, Zimmer A. Cannabinoid Receptor 2 Modulates Susceptibility to Experimental Cerebral Malaria through a CCL17-dependent Mechanism. J Biol Chem 2016; 291:19517-31. [PMID: 27474745 DOI: 10.1074/jbc.m116.746594] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Indexed: 11/06/2022] Open
Abstract
Cerebral malaria is a severe and often fatal complication of Plasmodium falciparum infection. It is characterized by parasite sequestration, a breakdown of the blood-brain barrier, and a strong inflammation in the brain. We investigated the role of the cannabinoid receptor 2 (CB2), an important modulator of neuroinflammatory responses, in experimental cerebral malaria (ECM). Strikingly, mice with a deletion of the CB2-encoding gene (Cnr2(-/-)) inoculated with Plasmodium berghei ANKA erythrocytes exhibited enhanced survival and a diminished blood-brain barrier disruption. Therapeutic application of a specific CB2 antagonist also conferred increased ECM resistance in wild type mice. Hematopoietic derived immune cells were responsible for the enhanced protection in bone marrow (BM) chimeric Cnr2(-/-) mice. Mixed BM chimeras further revealed that CB2-expressing cells contributed to ECM development. A heterogeneous CD11b(+) cell population, containing macrophages and neutrophils, expanded in the Cnr2(-/-) spleen after infection and expressed macrophage mannose receptors, arginase-1 activity, and IL-10. Also in the Cnr2(-/-) brain, CD11b(+) cells that expressed selected anti-inflammatory markers accumulated, and expression of inflammatory mediators IFN-γ and TNF-α was reduced. Finally, the M2 macrophage chemokine CCL17 was identified as an essential factor for enhanced survival in the absence of CB2, because CCL17 × Cnr2 double-deficient mice were fully susceptible to ECM. Thus, targeting CB2 may be promising for the development of alternative treatment regimes of ECM.
Collapse
Affiliation(s)
- Judith Alferink
- From the Institute of Molecular Psychiatry, Medical Faculty, and the Department of Psychiatry, University of Münster, 48149 Münster, Germany, the Cluster of Excellence EXC 1003, Cells in Motion, 48149 Münster, Germany,
| | - Sabine Specht
- the Institute of Medical Microbiology, Immunology, and Parasitology and
| | - Hannah Arends
- From the Institute of Molecular Psychiatry, Medical Faculty, and
| | - Beatrix Schumak
- the Institute of Medical Microbiology, Immunology, and Parasitology and
| | - Kim Schmidt
- the Institute of Medical Microbiology, Immunology, and Parasitology and
| | - Christina Ruland
- the Department of Psychiatry, University of Münster, 48149 Münster, Germany
| | - Ramona Lundt
- From the Institute of Molecular Psychiatry, Medical Faculty, and
| | - Andrea Kemter
- From the Institute of Molecular Psychiatry, Medical Faculty, and
| | - Andrea Dlugos
- the Department of Psychiatry, University of Münster, 48149 Münster, Germany
| | - Janina M Kuepper
- the Institute of Medical Microbiology, Immunology, and Parasitology and
| | | | | | - Önder Albayram
- From the Institute of Molecular Psychiatry, Medical Faculty, and
| | - David-M Otte
- From the Institute of Molecular Psychiatry, Medical Faculty, and
| | - Janine Marazzi
- the Institute of Biochemistry and Molecular Medicine, University of Bern, 3012 Bern, Switzerland, and
| | - Jürg Gertsch
- the Institute of Biochemistry and Molecular Medicine, University of Bern, 3012 Bern, Switzerland, and
| | - Irmgard Förster
- the Department of Immunology and Environment, Life and Medical Sciences Institute (LIMES), University of Bonn, 53127 Bonn, Germany
| | - Wolfgang Maier
- the Department of Psychiatry, University Hospital Bonn, 53105 Bonn, Germany
| | - Stefanie Scheu
- the Institute of Medical Microbiology and Hospital Hygiene, University of Düsseldorf, 40225 Düsseldorf, Germany
| | - Achim Hoerauf
- the Institute of Medical Microbiology, Immunology, and Parasitology and
| | - Andreas Zimmer
- From the Institute of Molecular Psychiatry, Medical Faculty, and
| |
Collapse
|
40
|
Gupta P, Lai SM, Sheng J, Tetlak P, Balachander A, Claser C, Renia L, Karjalainen K, Ruedl C. Tissue-Resident CD169(+) Macrophages Form a Crucial Front Line against Plasmodium Infection. Cell Rep 2016; 16:1749-1761. [PMID: 27477286 DOI: 10.1016/j.celrep.2016.07.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 05/23/2016] [Accepted: 07/01/2016] [Indexed: 12/31/2022] Open
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.
Collapse
Affiliation(s)
- Pravesh Gupta
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Si Min Lai
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Singapore; Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Singapore 138648, Singapore
| | - Jianpeng Sheng
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Piotr Tetlak
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Akhila Balachander
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Singapore 138648, Singapore
| | - Carla Claser
- Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Singapore 138648, Singapore
| | - Laurent Renia
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Singapore; Singapore Immunology Network, Agency for Science, Technology and Research (A(∗)STAR), 8A Biomedical Grove, Singapore 138648, Singapore
| | - Klaus Karjalainen
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Singapore
| | - Christiane Ruedl
- Nanyang Technological University, School of Biological Sciences, 60 Nanyang Drive, Singapore 637551, Singapore.
| |
Collapse
|
41
|
Dalko E, Tchitchek N, Pays L, Herbert F, Cazenave PA, Ravindran B, Sharma S, Nataf S, Das B, Pied S. Erythropoietin Levels Increase during Cerebral Malaria and Correlate with Heme, Interleukin-10 and Tumor Necrosis Factor-Alpha in India. PLoS One 2016; 11:e0158420. [PMID: 27441662 PMCID: PMC4956275 DOI: 10.1371/journal.pone.0158420] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Accepted: 06/15/2016] [Indexed: 12/12/2022] Open
Abstract
Cerebral malaria (CM) caused by Plasmodium falciparum parasites often leads to the death of infected patients or to persisting neurological sequelae despite anti-parasitic treatments. Erythropoietin (EPO) was recently suggested as a potential adjunctive treatment for CM. However diverging results were obtained in patients from Sub-Saharan countries infected with P. falciparum. In this study, we measured EPO levels in the plasma of well-defined groups of P. falciparum-infected patients, from the state of Odisha in India, with mild malaria (MM), CM, or severe non-CM (NCM). EPO levels were then correlated with biological parameters, including parasite biomass, heme, tumor necrosis factor (TNF)-α, interleukin (IL)-10, interferon gamma-induced protein (IP)-10, and monocyte chemoattractant protein (MCP)-1 plasma concentrations by Spearman’s rank and multiple correlation analyses. We found a significant increase in EPO levels with malaria severity degree, and more specifically during fatal CM. In addition, EPO levels were also found correlated positively with heme, TNF-α, IL-10, IP-10 and MCP-1 during CM. We also found a significant multivariate correlation between EPO, TNF-α, IL-10, IP-10 MCP-1 and heme, suggesting an association of EPO with a network of immune factors in CM patients. The contradictory levels of circulating EPO reported in CM patients in India when compared to Africa highlights the need for the optimization of adjunctive treatments according to the targeted population.
Collapse
Affiliation(s)
- Esther Dalko
- Centre for Infection and Immunity of Lille, INSERM U1019, CNRS UMR 8204, Université Lille Nord de France, Institut Pasteur de Lille, Lille 59019, France
| | - Nicolas Tchitchek
- CEA, DSV/iMETI, Immunology of viral infections and autoimmune diseases research unit, UMR1184, IDMIT infrastructure, Fontenay-aux-Roses, France
| | - Laurent Pays
- Lyon 1 University, CarMeN Laboratory, INSERM U-1060, INRA USC-1235, 69921, Oullins, France; Banque de Tissus et de Cellules des Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| | - Fabien Herbert
- Centre for Infection and Immunity of Lille, INSERM U1019, CNRS UMR 8204, Université Lille Nord de France, Institut Pasteur de Lille, Lille 59019, France
| | - Pierre-André Cazenave
- Centre for Infection and Immunity of Lille, INSERM U1019, CNRS UMR 8204, Université Lille Nord de France, Institut Pasteur de Lille, Lille 59019, France
| | | | - Shobhona Sharma
- Department of Biological Sciences, Tata Institute of Fundamental Research, Mumbai, Maharashtra 400005, India
| | - Serge Nataf
- Lyon 1 University, CarMeN Laboratory, INSERM U-1060, INRA USC-1235, 69921, Oullins, France; Banque de Tissus et de Cellules des Hospices Civils de Lyon, Hôpital Edouard Herriot, Lyon, France
| | - Bidyut Das
- SCB Medical College, Cuttack, Odisha 753007, India
| | - Sylviane Pied
- Centre for Infection and Immunity of Lille, INSERM U1019, CNRS UMR 8204, Université Lille Nord de France, Institut Pasteur de Lille, Lille 59019, France
- * E-mail:
| |
Collapse
|
42
|
Van Braeckel-Budimir N, Kurup SP, Harty JT. Regulatory issues in immunity to liver and blood-stage malaria. Curr Opin Immunol 2016; 42:91-97. [PMID: 27351448 DOI: 10.1016/j.coi.2016.06.008] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Revised: 06/10/2016] [Accepted: 06/15/2016] [Indexed: 02/01/2023]
Abstract
T cells play a major role in control of both blood and liver stage of plasmodium infection. While immunization with certain attenuated whole-parasite vaccines that are attenuated at the liver stage of the infection induces protective T cell responses, even multiple exposures to natural infection in endemic areas do not lead to stable T cell memory or humoral immunity and sterilizing protection. One of the key differences between vaccination and natural exposure is the absence of blood stage during vaccination. Here we will discuss possible immunoregulatory strategies employed by blood stage of malaria leading to generation of severely compromised T cell and humoral immune responses and subsequent lack of sterilizing immunity.
Collapse
Affiliation(s)
| | - Samarchith P Kurup
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA
| | - John T Harty
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA; Department of Pathology, University of Iowa, Iowa City, IA 52242, USA; Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
43
|
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: 92] [Impact Index Per Article: 9.2] [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
|
44
|
Mycobacterium tuberculosis Coinfection Has No Impact on Plasmodium berghei ANKA-Induced Experimental Cerebral Malaria in C57BL/6 Mice. Infect Immun 2015; 84:502-10. [PMID: 26644378 DOI: 10.1128/iai.01290-15] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 11/23/2015] [Indexed: 11/20/2022] Open
Abstract
Cerebral malaria (CM) is the most severe complication of human infection with Plasmodium falciparum. The mechanisms predisposing to CM are still not fully understood. Proinflammatory immune responses are required for the control of blood-stage malaria infection but are also implicated in the pathogenesis of CM. A fine balance between pro- and anti-inflammatory immune responses is required for parasite clearance without the induction of host pathology. The most accepted experimental model to study human CM is Plasmodium berghei ANKA (PbANKA) infection in C57BL/6 mice that leads to the development of a complex neurological syndrome which shares many characteristics with the human disease. We applied this model to study the outcome of PbANKA infection in mice previously infected with Mycobacterium tuberculosis, the causative agent of tuberculosis. Tuberculosis is coendemic with malaria in large regions in the tropics, and mycobacteria have been reported to confer some degree of unspecific protection against rodent Plasmodium parasites in experimental coinfection models. We found that concomitant M. tuberculosis infection did not change the clinical course of PbANKA-induced experimental cerebral malaria (ECM) in C57BL/6 mice. The immunological environments in spleen and brain did not differ between singly infected and coinfected animals; instead, the overall cytokine and T cell responses in coinfected mice were comparable to those in animals solely infected with PbANKA. Our data suggest that M. tuberculosis coinfection is not able to change the outcome of PbANKA-induced disease, most likely because the inflammatory response induced by the parasite rapidly dominates in mice previously infected with M. tuberculosis.
Collapse
|
45
|
Fontana MF, Baccarella A, Kellar D, Oniskey TK, Terinate P, Rosenberg SD, Huang EJ, Herbert DR, Kim CC. Myeloid expression of the AP-1 transcription factor JUNB modulates outcomes of type 1 and type 2 parasitic infections. Parasite Immunol 2015; 37:470-8. [PMID: 26178310 PMCID: PMC4573644 DOI: 10.1111/pim.12215] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 07/03/2015] [Indexed: 12/31/2022]
Abstract
Activation of macrophages is a key step in the initiation of immune responses, but the transcriptional mechanisms governing macrophage activation during infection are not fully understood. It was recently shown that the AP-1 family transcription factor JUNB positively regulates macrophage activation in response to Toll-like receptor agonists that promote classical or M1 polarization, as well as to the cytokine interleukin-4 (IL-4), which elicits an alternatively activated or M2 phenotype. However, a role for JUNB in macrophage activation has never been demonstrated in vivo. Here, to dissect the role of JUNB in macrophage activation in a physiological setting, mice lacking JUNB specifically in myeloid cells were tested in two infection models: experimental cerebral malaria, which elicits a pathological type 1 immune response, and helminth infection, in which type 2 responses are protective. Myeloid-restricted deletion of Junb reduced type 1 immune activation, which was associated with reduced cerebral pathology and improved survival during infection with Plasmodium berghei. Myeloid JUNB deficiency also compromised type 2 activation during infection with the hookworm Nippostrongylus brasiliensis, leading to diminished cytokine production and eosinophil recruitment and increased parasite burden. These results demonstrate that JUNB in myeloid cells shapes host responses and outcomes during type 1 and type 2 infections.
Collapse
Affiliation(s)
- Mary F. Fontana
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alyssa Baccarella
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Danielle Kellar
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Taylor K. Oniskey
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Paul Terinate
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sarah D. Rosenberg
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Eric J. Huang
- Departments of Pathology and Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - De’Broski R. Herbert
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Charles C. Kim
- Division of Experimental Medicine, Department of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
46
|
Disruption of Parasite hmgb2 Gene Attenuates Plasmodium berghei ANKA Pathogenicity. Infect Immun 2015; 83:2771-84. [PMID: 25916985 DOI: 10.1128/iai.03129-14] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 04/19/2015] [Indexed: 12/20/2022] Open
Abstract
Eukaryotic high-mobility-group-box (HMGB) proteins are nuclear factors involved in chromatin remodeling and transcription regulation. When released into the extracellular milieu, HMGB1 acts as a proinflammatory cytokine that plays a central role in the pathogenesis of several immune-mediated inflammatory diseases. We found that the Plasmodium genome encodes two genuine HMGB factors, Plasmodium HMGB1 and HMGB2, that encompass, like their human counterparts, a proinflammatory domain. Given that these proteins are released from parasitized red blood cells, we then hypothesized that Plasmodium HMGB might contribute to the pathogenesis of experimental cerebral malaria (ECM), a lethal neuroinflammatory syndrome that develops in C57BL/6 (susceptible) mice infected with Plasmodium berghei ANKA and that in many aspects resembles human cerebral malaria elicited by P. falciparum infection. The pathogenesis of experimental cerebral malaria was suppressed in C57BL/6 mice infected with P. berghei ANKA lacking the hmgb2 gene (Δhmgb2 ANKA), an effect associated with a reduction of histological brain lesions and with lower expression levels of several proinflammatory genes. The incidence of ECM in pbhmgb2-deficient mice was restored by the administration of recombinant PbHMGB2. Protection from experimental cerebral malaria in Δhmgb2 ANKA-infected mice was associated with reduced sequestration in the brain of CD4(+) and CD8(+) T cells, including CD8(+) granzyme B(+) and CD8(+) IFN-γ(+) cells, and, to some extent, neutrophils. This was consistent with a reduced parasite sequestration in the brain, lungs, and spleen, though to a lesser extent than in wild-type P. berghei ANKA-infected mice. In summary, Plasmodium HMGB2 acts as an alarmin that contributes to the pathogenesis of cerebral malaria.
Collapse
|
47
|
Lewis MD, Behrends J, Sá E Cunha C, Mendes AM, Lasitschka F, Sattler JM, Heiss K, Kooij TWA, Prudêncio M, Bringmann G, Frischknecht F, Mueller AK. Chemical attenuation of Plasmodium in the liver modulates severe malaria disease progression. THE JOURNAL OF IMMUNOLOGY 2015; 194:4860-70. [PMID: 25862814 DOI: 10.4049/jimmunol.1400863] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2014] [Accepted: 03/11/2015] [Indexed: 11/19/2022]
Abstract
Cerebral malaria is one of the most severe complications of malaria disease, attributed to a complicated series of immune reactions in the host. The syndrome is marked by inflammatory immune responses, margination of leukocytes, and parasitized erythrocytes in cerebral vessels leading to breakdown of the blood-brain barrier. We show that chemical attenuation of the parasite at the very early, clinically silent liver stage suppresses parasite development, delays the time until parasites establish blood-stage infection, and provokes an altered host immune response, modifying immunopathogenesis and protecting from cerebral disease. The early response is proinflammatory and cell mediated, with increased T cell activation in the liver and spleen, and greater numbers of effector T cells, cytokine-secreting T cells, and proliferating, proinflammatory cytokine-producing T cells. Dendritic cell numbers, T cell activation, and infiltration of CD8(+) T cells to the brain are decreased later in infection, possibly mediated by the anti-inflammatory cytokine IL-10. Strikingly, protection can be transferred to naive animals by adoptive transfer of lymphocytes from the spleen at very early times of infection. Our data suggest that a subpopulation belonging to CD8(+) T cells as early as day 2 postinfection is responsible for protection. These data indicate that liver stage-directed early immune responses can moderate the overall downstream host immune response and modulate severe malaria outcome.
Collapse
Affiliation(s)
- Matthew D Lewis
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, D 69120 Heidelberg, Germany; German Centre for Infection Research, D 69120 Heidelberg, Germany
| | - Jochen Behrends
- Core Facility Fluorescence Cytometry, Research Center Borstel, D 23845 Borstel, Germany
| | - Cláudia Sá E Cunha
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - António M Mendes
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Felix Lasitschka
- German Centre for Infection Research, D 69120 Heidelberg, Germany; Institute of Pathology, Heidelberg University Hospital, D 69120 Heidelberg, Germany
| | - Julia M Sattler
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, D 69120 Heidelberg, Germany
| | - Kirsten Heiss
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, D 69120 Heidelberg, Germany; MalVa GmbH, D 69121 Heidelberg, Germany
| | - Taco W A Kooij
- Department of Medical Microbiology, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6500 HB Nijmegen, the Netherlands; Centre for Molecular and Biomolecular Informatics, Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, 6500 HB Nijmegen, the Netherlands; and
| | - Miguel Prudêncio
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, 1649-028 Lisbon, Portugal
| | - Gerhard Bringmann
- Institute for Organic Chemistry, University of Würzburg, 97074 Würzburg, Germany
| | - Friedrich Frischknecht
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, D 69120 Heidelberg, Germany
| | - Ann-Kristin Mueller
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, D 69120 Heidelberg, Germany; German Centre for Infection Research, D 69120 Heidelberg, Germany;
| |
Collapse
|
48
|
Howland SW, Claser C, Poh CM, Gun SY, Rénia L. Pathogenic CD8+ T cells in experimental cerebral malaria. Semin Immunopathol 2015; 37:221-31. [PMID: 25772948 DOI: 10.1007/s00281-015-0476-6] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Accepted: 03/01/2015] [Indexed: 11/26/2022]
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.
Collapse
Affiliation(s)
- Shanshan Wu Howland
- Singapore Immunology Network, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | | | | | | | | |
Collapse
|
49
|
Van Den Ham KM, Shio MT, Rainone A, Fournier S, Krawczyk CM, Olivier M. Iron prevents the development of experimental cerebral malaria by attenuating CXCR3-mediated T cell chemotaxis. PLoS One 2015; 10:e0118451. [PMID: 25768944 PMCID: PMC4359107 DOI: 10.1371/journal.pone.0118451] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2014] [Accepted: 01/20/2015] [Indexed: 12/16/2022] Open
Abstract
Cerebral malaria is a severe neurological complication of Plasmodium falciparum infection. Previous studies have suggested that iron overload can suppress the generation of a cytotoxic immune response; however, the effect of iron on experimental cerebral malaria (ECM) is yet unknown. Here we determined that the incidence of ECM was markedly reduced in mice treated with iron dextran. Protection was concomitant with a significant decrease in the sequestration of CD4+ and CD8+ T cells within the brain. CD4+ T cells demonstrated markedly decreased CXCR3 expression and had reduced IFNγ-responsiveness, as indicated by mitigated expression of IFNγR2 and T-bet. Additional analysis of the splenic cell populations indicated that parenteral iron supplementation was also associated with a decrease in NK cells and increase in regulatory T cells. Altogether, these results suggest that iron is able to inhibit ECM pathology by attenuating the capacity of T cells to migrate to the brain.
Collapse
MESH Headings
- Animals
- Brain/drug effects
- Brain/immunology
- Brain/metabolism
- CD4-Positive T-Lymphocytes/drug effects
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/drug effects
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Chemotaxis, Leukocyte/drug effects
- Chemotaxis, Leukocyte/immunology
- Disease Models, Animal
- Female
- Interferon-gamma/immunology
- Interferon-gamma/metabolism
- Iron/immunology
- Iron/pharmacology
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Malaria, Cerebral/etiology
- Malaria, Cerebral/immunology
- Malaria, Cerebral/metabolism
- Malaria, Cerebral/prevention & control
- Malaria, Falciparum/complications
- Malaria, Falciparum/immunology
- Malaria, Falciparum/metabolism
- Mice
- Mice, Inbred C57BL
- Plasmodium falciparum/immunology
- Receptors, CXCR3/immunology
- Receptors, CXCR3/metabolism
- T-Box Domain Proteins/immunology
- T-Box Domain Proteins/metabolism
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
Collapse
Affiliation(s)
- Kristin M. Van Den Ham
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- McGill International TB Centre, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Marina Tiemi Shio
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- McGill International TB Centre, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| | - Anthony Rainone
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | - Sylvie Fournier
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | - Connie M. Krawczyk
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
| | - Martin Olivier
- Department of Microbiology and Immunology, McGill University, Montréal, Québec, Canada
- McGill International TB Centre, Research Institute of the McGill University Health Centre, Montréal, Québec, Canada
| |
Collapse
|
50
|
Fernandes P, Frank R, Lewis MD, Mueller AK. Plasmodium attenuation: connecting the dots between early immune responses and malaria disease severity. Front Microbiol 2014; 5:658. [PMID: 25520710 PMCID: PMC4251431 DOI: 10.3389/fmicb.2014.00658] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/13/2014] [Indexed: 12/21/2022] Open
Abstract
Sterile attenuation of Plasmodium parasites at the liver-stage either by irradiation or genetic modification, or at the blood-stage by chemoprophylaxis, has been shown to induce immune responses that can protect against subsequent wild-type infection. However, following certain interventions, parasite attenuation can be incomplete or non-sterile. Instead parasites are rendered developmentally stunted but still capable of establishing an acute infection. In experiments involving Plasmodium berghei ANKA, a model of experimental cerebral malaria, it has been observed that several forms of attenuated parasites do not induce cerebral pathology. In this perspective we collect evidence from studies on murine malaria in particular, and attempt to “connect the dots” between early immune responses and protection from severe cerebral disease, highlighting potential parallels to human infection.
Collapse
Affiliation(s)
- Priyanka Fernandes
- Parasitology Unit, Centre of Infectious Diseases, University Hospital Heidelberg Heidelberg, Germany
| | - Roland Frank
- Parasitology Unit, Centre of Infectious Diseases, University Hospital Heidelberg Heidelberg, Germany
| | - Matthew D Lewis
- Parasitology Unit, Centre of Infectious Diseases, University Hospital Heidelberg Heidelberg, Germany
| | - Ann-Kristin Mueller
- Parasitology Unit, Centre of Infectious Diseases, University Hospital Heidelberg Heidelberg, Germany
| |
Collapse
|