1
|
Haley MJ, Barroso R, Jasim DA, Haigh M, Green J, Dickie B, Craig AG, Brough D, Couper KN. Lymphatic network drainage resolves cerebral edema and facilitates recovery from experimental cerebral malaria. Cell Rep 2024; 43:114217. [PMID: 38728141 DOI: 10.1016/j.celrep.2024.114217] [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: 11/22/2022] [Revised: 11/29/2023] [Accepted: 04/25/2024] [Indexed: 05/12/2024] Open
Abstract
While brain swelling, associated with fluid accumulation, is a known feature of pediatric cerebral malaria (CM), how fluid and macromolecules are drained from the brain during recovery from CM is unknown. Using the experimental CM (ECM) model, we show that fluid accumulation in the brain during CM is driven by vasogenic edema and not by perivascular cerebrospinal fluid (CSF) influx. We identify that fluid and molecules are removed from the brain extremely quickly in mice with ECM to the deep cervical lymph nodes (dcLNs), predominantly through basal routes and across the cribriform plate and the nasal lymphatics. In agreement, we demonstrate that ligation of the afferent lymphatic vessels draining to the dcLNs significantly impairs fluid drainage from the brain and lowers anti-malarial drug recovery from the ECM syndrome. Collectively, our results provide insight into the pathways that coordinate recovery from CM.
Collapse
Affiliation(s)
- Michael J Haley
- Division of Immunology, Immunity to Infection & Respiratory Medicine, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Ruben Barroso
- Division of Immunology, Immunity to Infection & Respiratory Medicine, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Dhifaf A Jasim
- Nanomedicine Lab, National Graphene Institute and Faculty of Biology, Medicine & Health, The University of Manchester, AV Hill Building, Manchester M13 9PT, UK; Medicines Discovery Catapult (MDC), Alderley Park, Macclesfield SK10 4TG, UK
| | - Megan Haigh
- Division of Immunology, Immunity to Infection & Respiratory Medicine, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M13 9PT, UK
| | - Jack Green
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK; Division of Neuroscience, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M13 9PT, UK
| | - Ben Dickie
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; Division of Informatics, Imaging & Data Sciences, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M13 9PT, UK
| | - Alister G Craig
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - David Brough
- Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK; Division of Neuroscience, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M13 9PT, UK
| | - Kevin N Couper
- Division of Immunology, Immunity to Infection & Respiratory Medicine, Faculty of Biology, Medicine & Health, The University of Manchester, Manchester M13 9PT, UK; Geoffrey Jefferson Brain Research Centre, The Manchester Academic Health Science Centre, Northern Care Alliance NHS Group, University of Manchester, Manchester, UK; The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.
| |
Collapse
|
2
|
Sharma I, Kataria P, Das J. Cerebral malaria pathogenesis: Dissecting the role of CD4 + and CD8 + T-cells as major effectors in disease pathology. Int Rev Immunol 2024; 43:309-325. [PMID: 38618863 DOI: 10.1080/08830185.2024.2336539] [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: 09/25/2023] [Revised: 03/22/2024] [Accepted: 03/24/2024] [Indexed: 04/16/2024]
Abstract
Cerebral malaria (CM) is a severe complication of Plasmodium falciparum (P. falciparum) infection, with complex pathogenesis involving multiple factors, including the host's immunological response. T lymphocytes, specifically CD4+ T helper cells and CD8+ cytotoxic T cells, are crucial in controlling parasite growth and activating cells for parasite clearance via cytokine secretion. Contrary to this, reports also suggest the pathogenic nature of T lymphocytes as they are often involved in disease progression and severity. CD8+ cytotoxic T cells migrate to the host's brain vasculature, disrupting the blood-brain barrier and causing neurological manifestations. CD4+ T helper cells on the other hand play a variety of functions as they differentiate into different subtypes which may function as pro-inflammatory or anti-inflammatory. The excessive pro-inflammatory response in CM can lead to multi-organ failure, necessitating a check mechanism to maintain immune homeostasis. This is achieved by regulatory T cells and their characteristic cytokines, which counterbalance the pro-inflammatory immune response. Maintaining a critical balance between pro and anti-inflammatory responses is crucial for determining disease outcomes in CM. A slight change in this balance may contribute to a disease severity owing to an extreme inflammatory response or unrestricted parasite growth, a potential target for designing immunotherapeutic treatment approaches. The review briefly discusses the pathogenesis of CM and various mechanisms responsible for the disruption of the blood-brain barrier. It also highlights the role of different T cell subsets during infection and emphasizes the importance of balance between pro and anti-inflammatory T cells that ultimately decides the outcome of the disease.
Collapse
Affiliation(s)
- Indu Sharma
- Academy of Scientific and Innovative Research (AcSIR), Noida, India
- Division of Immunology, National Institute of Malaria Research, Dwarka, New Delhi, India
| | - Poonam Kataria
- Academy of Scientific and Innovative Research (AcSIR), Noida, India
- Division of Immunology, National Institute of Malaria Research, Dwarka, New Delhi, India
| | - Jyoti Das
- Academy of Scientific and Innovative Research (AcSIR), Noida, India
- Division of Immunology, National Institute of Malaria Research, Dwarka, New Delhi, India
| |
Collapse
|
3
|
Oelschlegel AM, Bhattacharjee R, Wenk P, Harit K, Rothkötter HJ, Koch SP, Boehm-Sturm P, Matuschewski K, Budinger E, Schlüter D, Goldschmidt J, Nishanth G. Beyond the microcirculation: sequestration of infected red blood cells and reduced flow in large draining veins in experimental cerebral malaria. Nat Commun 2024; 15:2396. [PMID: 38493187 PMCID: PMC10944460 DOI: 10.1038/s41467-024-46617-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 02/29/2024] [Indexed: 03/18/2024] Open
Abstract
Sequestration of infected red blood cells (iRBCs) in the microcirculation is a hallmark of cerebral malaria (CM) in post-mortem human brains. It remains controversial how this might be linked to the different disease manifestations, in particular brain swelling leading to brain herniation and death. The main hypotheses focus on iRBC-triggered inflammation and mechanical obstruction of blood flow. Here, we test these hypotheses using murine models of experimental CM (ECM), SPECT-imaging of radiolabeled iRBCs and cerebral perfusion, MR-angiography, q-PCR, and immunohistochemistry. We show that iRBC accumulation and reduced flow precede inflammation. Unexpectedly, we find that iRBCs accumulate not only in the microcirculation but also in large draining veins and sinuses, particularly at the rostral confluence. We identify two parallel venous streams from the superior sagittal sinus that open into the rostral rhinal veins and are partially connected to infected skull bone marrow. The flow in these vessels is reduced early, and the spatial patterns of pathology correspond to venous drainage territories. Our data suggest that venous efflux reductions downstream of the microcirculation are causally linked to ECM pathology, and that the different spatiotemporal patterns of edema development in mice and humans could be related to anatomical differences in venous anatomy.
Collapse
Affiliation(s)
- A M Oelschlegel
- Combinatorial NeuroImaging Core Facility, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
- Research group Neuroplasticity, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - R Bhattacharjee
- Combinatorial NeuroImaging Core Facility, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625, Hannover, Germany
| | - P Wenk
- Combinatorial NeuroImaging Core Facility, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
| | - K Harit
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625, Hannover, Germany
| | - H-J Rothkötter
- Institute of Anatomy, Medical Faculty, Otto-von-Guericke-University Magdeburg, Leipziger Strasse 44, 39120, Magdeburg, Germany
| | - S P Koch
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Experimental Neurology and Center for Stroke Research, Charitéplatz 1, 10117, Berlin, Germany
- Charité-Universitätsmedizin Berlin, NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, 10117, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Charité 3R | Replace, Reduce, Refine, Charitéplatz 1, 10117, Berlin, Germany
| | - P Boehm-Sturm
- Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Experimental Neurology and Center for Stroke Research, Charitéplatz 1, 10117, Berlin, Germany
- Charité-Universitätsmedizin Berlin, NeuroCure Cluster of Excellence and Charité Core Facility 7T Experimental MRIs, 10117, Berlin, Germany
- Charité-Universitätsmedizin Berlin, Charité 3R | Replace, Reduce, Refine, Charitéplatz 1, 10117, Berlin, Germany
| | - K Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt University, 10115, Berlin, Germany
| | - E Budinger
- Combinatorial NeuroImaging Core Facility, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany
- Center of Behavioural Brain Sciences, Universitätsplatz 2, 39106, Magdeburg, Germany
| | - D Schlüter
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625, Hannover, Germany
| | - J Goldschmidt
- Combinatorial NeuroImaging Core Facility, Leibniz Institute for Neurobiology, 39118, Magdeburg, Germany.
- Center of Behavioural Brain Sciences, Universitätsplatz 2, 39106, Magdeburg, Germany.
| | - G Nishanth
- Institute of Medical Microbiology and Hospital Epidemiology, Hannover Medical School, 30625, Hannover, Germany.
| |
Collapse
|
4
|
Hellani F, Leleu I, Saidi N, Martin N, Lecoeur C, Werkmeister E, Koffi D, Trottein F, Yapo-Etté H, Das B, Abbadie C, Pied S. Role of astrocyte senescence regulated by the non- canonical autophagy in the neuroinflammation associated to cerebral malaria. Brain Behav Immun 2024; 117:20-35. [PMID: 38157948 DOI: 10.1016/j.bbi.2023.12.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 12/12/2023] [Accepted: 12/21/2023] [Indexed: 01/03/2024] Open
Abstract
BACKGROUND Cerebral malaria (CM) is a fatal neuroinflammatory syndrome caused (in humans) by the protozoa Plasmodium (P.) falciparum. Glial cell activation is one of the mechanisms that contributes to neuroinflammation in CM. RESULT By studying a mouse model of CM (caused by P. berghei ANKA), we describe that the induction of autophagy promoted p21-dependent senescence in astrocytes and that CXCL-10 was part of the senescence-associated secretory phenotype. Furthermore, p21 expression was observed in post-mortem brain and peripheral blood samples from patients with CM. Lastly, we found that the depletion of senescent astrocytes with senolytic drugs abrogated inflammation and protected mice from CM. CONCLUSION Our data provide evidence for a novel mechanism through which astrocytes could be involved in the neuropathophysiology of CM. p21 gene expression in blood cell and an elevated plasma CXCL-10 concentration could be valuable biomarkers of CM in humans. In the end, we believe senolytic drugs shall open up new avenues to develop newer treatment options.
Collapse
Affiliation(s)
- Fatima Hellani
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France
| | - Inès Leleu
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France
| | - Nasreddine Saidi
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France
| | - Nathalie Martin
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies F-59000 Lille, France
| | - Cécile Lecoeur
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France
| | - Elisabeth Werkmeister
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, US 41 - UMS 2014 - PLBS F-59000 Lille, France
| | - David Koffi
- Parasitology and Mycology Department, Institut Pasteur de Côte d'Ivoire, Ivory Coast
| | - François Trottein
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France
| | - Hélène Yapo-Etté
- Institute of Forensic Medicine-Faculty of Health, University Félix Houphouët-Boigny of Abidjan, Ivory Coast
| | - Bidyut Das
- SCB Medical College, Cuttack, Orissa, India
| | - Corinne Abbadie
- Univ. Lille, CNRS, INSERM, CHU Lille, Institut Pasteur de Lille, UMR9020-U1277 - CANTHER - Cancer Heterogeneity, Plasticity and Resistance to Therapies F-59000 Lille, France
| | - Sylviane Pied
- Univ. Lille, CNRS UMR 9017-INSERM U1019, Center for Infection and Immunity of Lille-CIIL, Institut Pasteur de Lille F-59019 Lille, France.
| |
Collapse
|
5
|
Silva AF, Torres MDT, Silva LS, Alves FL, Miranda A, Oliveira VX, de la Fuente-Nunez C, Pinheiro AAS. Synthetic angiotensin II peptide derivatives confer protection against cerebral and severe non-cerebral malaria in murine models. Sci Rep 2024; 14:4682. [PMID: 38409185 PMCID: PMC10897374 DOI: 10.1038/s41598-024-51267-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 01/03/2024] [Indexed: 02/28/2024] Open
Abstract
Malaria can have severe long-term effects. Even after treatment with antimalarial drugs eliminates the parasite, survivors of cerebral malaria may suffer from irreversible brain damage, leading to cognitive deficits. Angiotensin II, a natural human peptide hormone that regulates blood pressure, has been shown to be active against Plasmodium spp., the etiologic agent of malaria. Here, we tested two Ang II derivatives that do not elicit vasoconstriction in mice: VIPF, a linear tetrapeptide, which constitutes part of the hydrophobic portion of Ang II; and Ang II-SS, a disulfide-bridged derivative. The antiplasmodial potential of both peptides was evaluated with two mouse models: an experimental cerebral malaria model and a mouse model of non-cerebral malaria. The latter consisted of BALB/c mice infected with Plasmodium berghei ANKA. The peptides had no effect on mean blood pressure and significantly reduced parasitemia in both mouse models. Both peptides reduced the SHIRPA score, an assay used to assess murine health and behavior. However, only the constrained derivative (Ang II-SS), which was also resistant to proteolytic degradation, significantly increased mouse survival. Here, we show that synthetic peptides derived from Ang II are capable of conferring protection against severe manifestations of malaria in mouse models while overcoming the vasoconstrictive side effects of the parent peptide.
Collapse
Affiliation(s)
- Adriana F Silva
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, SP, Brazil
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Marcelo D T Torres
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA
| | - Leandro S Silva
- Division of Infectious Diseases and Immunology, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Flavio L Alves
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
- Faculdade de Ciências Médicas da Santa Casa de São Paulo, São Paulo, SP, Brazil
| | - Antonio Miranda
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Vani X Oliveira
- Centro de Ciências Naturais e Humanas, Universidade Federal do ABC, Santo André, SP, Brazil
- Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, SP, Brazil
| | - Cesar de la Fuente-Nunez
- Machine Biology Group, Departments of Psychiatry and Microbiology, Institute for Biomedical Informatics, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Departments of Bioengineering and Chemical and Biomolecular Engineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA.
- Penn Institute for Computational Science, University of Pennsylvania, Philadelphia, PA, USA.
| | - Ana Acacia S Pinheiro
- Instituto de Biofísica Carlos Chagas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
6
|
Fain CE, Zheng J, Jin F, Ayasoufi K, Wu Y, Lilley MT, Dropik AR, Wolf DM, Rodriguez RC, Aibaidula A, Tritz ZP, Bouchal SM, Pewe LL, Urban SL, Chen Y, Chang SY, Hansen MJ, Kachergus JM, Shi J, Thompson EA, Jensen HE, Harty JT, Parney IF, Sun J, Wu LJ, Johnson AJ. Discrete class I molecules on brain endothelium differentially regulate neuropathology in experimental cerebral malaria. Brain 2024; 147:566-589. [PMID: 37776513 DOI: 10.1093/brain/awad319] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 08/15/2023] [Accepted: 08/31/2023] [Indexed: 10/02/2023] Open
Abstract
Cerebral malaria is the deadliest complication that can arise from Plasmodium infection. CD8 T-cell engagement of brain vasculature is a putative mechanism of neuropathology in cerebral malaria. To define contributions of brain endothelial cell major histocompatibility complex (MHC) class I antigen-presentation to CD8 T cells in establishing cerebral malaria pathology, we developed novel H-2Kb LoxP and H-2Db LoxP mice crossed with Cdh5-Cre mice to achieve targeted deletion of discrete class I molecules, specifically from brain endothelium. This strategy allowed us to avoid off-target effects on iron homeostasis and class I-like molecules, which are known to perturb Plasmodium infection. This is the first endothelial-specific ablation of individual class-I molecules enabling us to interrogate these molecular interactions. In these studies, we interrogated human and mouse transcriptomics data to compare antigen presentation capacity during cerebral malaria. Using the Plasmodium berghei ANKA model of experimental cerebral malaria (ECM), we observed that H-2Kb and H-2Db class I molecules regulate distinct patterns of disease onset, CD8 T-cell infiltration, targeted cell death and regional blood-brain barrier disruption. Strikingly, ablation of either molecule from brain endothelial cells resulted in reduced CD8 T-cell activation, attenuated T-cell interaction with brain vasculature, lessened targeted cell death, preserved blood-brain barrier integrity and prevention of ECM and the death of the animal. We were able to show that these events were brain-specific through the use of parabiosis and created the novel technique of dual small animal MRI to simultaneously scan conjoined parabionts during infection. These data demonstrate that interactions of CD8 T cells with discrete MHC class I molecules on brain endothelium differentially regulate development of ECM neuropathology. Therefore, targeting MHC class I interactions therapeutically may hold potential for treatment of cases of severe malaria.
Collapse
Affiliation(s)
- Cori E Fain
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Jiaying Zheng
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905USA
| | - Fang Jin
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
| | | | - Yue Wu
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Meredith T Lilley
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Abigail R Dropik
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Delaney M Wolf
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
| | | | - Abudumijiti Aibaidula
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905USA
| | - Zachariah P Tritz
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Samantha M Bouchal
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Lecia L Pewe
- Department of Pathology, University of Iowa, Iowa City, IA 52242USA
| | - Stina L Urban
- Department of Pathology, University of Iowa, Iowa City, IA 52242USA
| | - Yin Chen
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Graduate School of Biomedical Sciences, Mayo Clinic, Rochester, MN 55905USA
| | - Su-Youne Chang
- Department of Neurosurgery, Mayo Clinic, Rochester, MN 55905USA
| | | | | | - Ji Shi
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224USA
| | - E Aubrey Thompson
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL 32224USA
| | - Hadley E Jensen
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
| | - John T Harty
- Department of Pathology, University of Iowa, Iowa City, IA 52242USA
| | - Ian F Parney
- Department of Neurosurgery, Mayo Clinic, Rochester, MN 55905USA
| | - Jie Sun
- Department of Medicine, University of Virginia, Charlottesville, VA 22903USA
| | - Long-Jun Wu
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905USA
| | - Aaron J Johnson
- Department of Immunology, Mayo Clinic, Rochester, MN 55905USA
- Department of Molecular Medicine, Mayo Clinic, Rochester, MN 55905USA
- Department of Neurology, Mayo Clinic, Rochester, MN 55905USA
| |
Collapse
|
7
|
Zafar I, Taniguchi T, Baghdadi HB, Kondoh D, Rizk MA, Galon EM, Ji S, El-Sayed SAES, Do T, Li H, Amer MM, Zhuowei M, Yihong M, Zhou J, Inoue N, Xuan X. Babesia microti alleviates disease manifestations caused by Plasmodium berghei ANKA in murine co-infection model of complicated malaria. Front Cell Infect Microbiol 2023; 13:1226088. [PMID: 37492527 PMCID: PMC10364126 DOI: 10.3389/fcimb.2023.1226088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 06/20/2023] [Indexed: 07/27/2023] Open
Abstract
Malaria remains one of the most significant health issues worldwide, accounting for 2.6% of the total global disease burden, and efforts to eliminate this threat continue. The key focus is to develop an efficient and long-term immunity to this disease via vaccination or therapeutic approach, and innovative strategies would enable us to achieve this target. Previously, using a mouse co-infection disease model, cross-protection was illustrated between Babesia microti and Plasmodium chabaudi. Hence, this study was planned to elucidate the impact of acute B. microti Peabody mjr and Plasmodium berghei ANKA co-infection on the consequence of complicated malaria in the C57BL/6J mouse model of malaria. Furthermore, immune response and pathological features were analyzed, and the course of the disease was compared among experimental groups. Our study established that acute B. microti infection activated immunity which was otherwise suppressed by P. berghei. The immunosuppressive tissue microenvironment was counteracted as evidenced by the enhanced immune cell population in co-infected mice, in contrast to P. berghei-infected control mice. Parasite sequestration in the brain, liver, lung, and spleen of co-infected mice was significantly decreased and tissue injury was ameliorated. Meanwhile, the serum levels of IFN-γ, TNF-α, and IL-12p70 were reduced while the secretion of IL-10 was promoted in co-infected mice. Eventually, co-infected mice showed an extended rate of survival. Hereby, the principal cytokines associated with the severity of malaria by P. berghei infection were TNF-α, IFN-γ, and IL-12p70. Moreover, it was evident from our flow cytometry results that innate immunity is crucial and macrophages are at the frontline of immunity against P. berghei infection. Our study recommended further investigations to shed light on the effects of babesiosis in suppressing malaria with the goal of developing Babesia-based therapy against malaria.
Collapse
Affiliation(s)
- Iqra Zafar
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
- Livestock and Dairy Development Department, Veterinary Research Institute, Lahore, Punjab, Pakistan
| | - Tomoyo Taniguchi
- Department of Immunology and Parasitology, Graduate School of Medicine University of the Ryukyus, Nishihara Cho, Japan
| | - Hanadi B. Baghdadi
- Biology Department, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
- Basic and Applied Scientific Research Center (BASRC), Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Daisuke Kondoh
- Department of Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Mohamed Abdo Rizk
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Eloiza May Galon
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
- College of Veterinary Medicine and Biomedical Sciences, Cavite State University, Indang, Cavite, Philippines
| | - Shengwei Ji
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Shimaa Abd El-Salam El-Sayed
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Thom Do
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Hang Li
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Moaz M. Amer
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Ma Zhuowei
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Ma Yihong
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Jinlin Zhou
- Shanghai Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Shanghai, China
| | - Noboru Inoue
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Xuenan Xuan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| |
Collapse
|
8
|
Anand A, Chandana M, Ghosh S, Das R, Singh N, Vaishalli PM, Gantasala NP, Padmanaban G, Nagaraj VA. Significance of Plasmodium berghei Amino Acid Transporter 1 in Food Vacuole Functionality and Its Association with Cerebral Pathogenesis. Microbiol Spectr 2023; 11:e0494322. [PMID: 36976018 PMCID: PMC10101031 DOI: 10.1128/spectrum.04943-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Accepted: 03/07/2023] [Indexed: 03/29/2023] Open
Abstract
The food vacuole plays a central role in the blood stage of parasite development by digesting host hemoglobin acquired from red blood cells and detoxifying the host heme released during hemoglobin digestion into hemozoin. Blood-stage parasites undergo periodic schizont bursts, releasing food vacuoles containing hemozoin. Clinical studies in malaria-infected patients and in vivo animal studies have shown the association of hemozoin with disease pathogenesis and abnormal host immune responses in malaria. Here, we perform a detailed in vivo characterization of putative Plasmodium berghei amino acid transporter 1 localized in the food vacuole to understand its significance in the malaria parasite. We show that the targeted deletion of amino acid transporter 1 in Plasmodium berghei leads to a swollen food vacuole phenotype with the accumulation of host hemoglobin-derived peptides. Plasmodium berghei amino acid transporter 1-knockout parasites produce less hemozoin, and the hemozoin crystals display a thin morphology compared with wild-type parasites. The knockout parasites show reduced sensitivity to chloroquine and amodiaquine by showing recrudescence. More importantly, mice infected with the knockout parasites are protected from cerebral malaria and display reduced neuronal inflammation and cerebral complications. Genetic complementation of the knockout parasites restores the food vacuole morphology with hemozoin levels similar to that of wild-type parasites, causing cerebral malaria in the infected mice. The knockout parasites also show a significant delay in male gametocyte exflagellation. Our findings highlight the significance of amino acid transporter 1 in food vacuole functionality and its association with malaria pathogenesis and gametocyte development. IMPORTANCE Food vacuoles of the malaria parasite are involved in the degradation of red blood cell hemoglobin. The amino acids derived from hemoglobin degradation support parasite growth, and the heme released is detoxified into hemozoin. Antimalarials such as quinolines target hemozoin formation in the food vacuole. Food vacuole transporters transport hemoglobin-derived amino acids and peptides from the food vacuole to the parasite cytosol. Such transporters are also associated with drug resistance. Here, we show that the deletion of amino acid transporter 1 in Plasmodium berghei leads to swollen food vacuoles with the accumulation of hemoglobin-derived peptides. The transporter-deleted parasites generate less hemozoin with thin crystal morphology and show reduced sensitivity to quinolines. Mice infected with transporter-deleted parasites are protected from cerebral malaria. There is also a delay in male gametocyte exflagellation, affecting transmission. Our findings uncover the functional significance of amino acid transporter 1 in the life cycle of the malaria parasite.
Collapse
Affiliation(s)
- Aditya Anand
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Manjunatha Chandana
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, Odisha, India
| | - Sourav Ghosh
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Rahul Das
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | - Nalini Singh
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
| | - Pradeep Mini Vaishalli
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, Odisha, India
- Regional Centre for Biotechnology, Faridabad, Haryana, India
| | | | | | | |
Collapse
|
9
|
Das A, Sahu W, Ojha DK, Reddy KS, Suar M. Comparative Analysis of Host Metabolic Alterations in Murine Malaria Models with Uncomplicated or Severe Malaria. J Proteome Res 2022; 21:2261-2276. [PMID: 36169658 DOI: 10.1021/acs.jproteome.2c00123] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Malaria varies in severity, with complications ranging from uncomplicated to severe malaria. Severe malaria could be attributed to peripheral hyperparasitemia or cerebral malaria. The metabolic interactions between the host and Plasmodium species are yet to be understood during these infections of varied pathology and severity. An untargeted metabolomics approach utilizing the liquid chromatography-mass spectrometry platform has been used to identify the affected host metabolic pathways and associated metabolites in the serum of murine malaria models with uncomplicated malaria, hyperparasitemia, and experimental cerebral malaria. We report that mice with malaria share similar metabolic attributes like higher levels of bile acids, bile pigments, and steroid hormones that have been reported for human malaria infections. Moreover, in severe malaria, upregulated levels of metabolites like phenylalanine, histidine, valine, pipecolate, ornithine, and pantothenate, with decreased levels of arginine and hippurate, were observed. Metabolites of sphingolipid metabolism were upregulated in experimental cerebral malaria. Higher levels of 20-hydroxy-leukotriene B4 and epoxyoctadecamonoenoic acids were found in uncomplicated malaria, with lower levels observed for experimental cerebral malaria. Our study provides insights into host biology during different pathological stages of malaria disease and would be useful for the selection of animal models for evaluating diagnostic and therapeutic interventions against malaria. The raw data files are available via MetaboLights with the identifier MTBLS4387.
Collapse
Affiliation(s)
- Aleena Das
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India.,Technology Business Incubator, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India
| | - Welka Sahu
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India
| | - Deepak Kumar Ojha
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India
| | - K Sony Reddy
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India
| | - Mrutyunjay Suar
- School of Biotechnology, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India.,Technology Business Incubator, Kalinga Institute of Industrial Technology (Deemed University), Bhubaneswar751024, India
| |
Collapse
|
10
|
Olatunde AC, Cornwall DH, Roedel M, Lamb TJ. Mouse Models for Unravelling Immunology of Blood Stage Malaria. Vaccines (Basel) 2022; 10:1525. [PMID: 36146602 PMCID: PMC9501382 DOI: 10.3390/vaccines10091525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 11/16/2022] Open
Abstract
Malaria comprises a spectrum of disease syndromes and the immune system is a major participant in malarial disease. This is particularly true in relation to the immune responses elicited against blood stages of Plasmodium-parasites that are responsible for the pathogenesis of infection. Mouse models of malaria are commonly used to dissect the immune mechanisms underlying disease. While no single mouse model of Plasmodium infection completely recapitulates all the features of malaria in humans, collectively the existing models are invaluable for defining the events that lead to the immunopathogenesis of malaria. Here we review the different mouse models of Plasmodium infection that are available, and highlight some of the main contributions these models have made with regards to identifying immune mechanisms of parasite control and the immunopathogenesis of malaria.
Collapse
Affiliation(s)
| | | | | | - Tracey J. Lamb
- Department of Pathology, University of Utah, Emma Eccles Jones Medical Research Building, 15 N Medical Drive E, Room 1420A, Salt Lake City, UT 84112, USA
| |
Collapse
|
11
|
Chandana M, Anand A, Ghosh S, Das R, Beura S, Jena S, Suryawanshi AR, Padmanaban G, Nagaraj VA. Malaria parasite heme biosynthesis promotes and griseofulvin protects against cerebral malaria in mice. Nat Commun 2022; 13:4028. [PMID: 35821013 PMCID: PMC9276668 DOI: 10.1038/s41467-022-31431-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 06/16/2022] [Indexed: 11/08/2022] Open
Abstract
Heme-biosynthetic pathway of malaria parasite is dispensable for asexual stages, but essential for mosquito and liver stages. Despite having backup mechanisms to acquire hemoglobin-heme, pathway intermediates and/or enzymes from the host, asexual parasites express heme pathway enzymes and synthesize heme. Here we show heme synthesized in asexual stages promotes cerebral pathogenesis by enhancing hemozoin formation. Hemozoin is a parasite molecule associated with inflammation, aberrant host-immune responses, disease severity and cerebral pathogenesis. The heme pathway knockout parasites synthesize less hemozoin, and mice infected with knockout parasites are protected from cerebral malaria and death due to anemia is delayed. Biosynthetic heme regulates food vacuole integrity and the food vacuoles from knockout parasites are compromised in pH, lipid unsaturation and proteins, essential for hemozoin formation. Targeting parasite heme synthesis by griseofulvin-a FDA-approved antifungal drug, prevents cerebral malaria in mice and provides an adjunct therapeutic option for cerebral and severe malaria.
Collapse
Affiliation(s)
- Manjunatha Chandana
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, Odisha, India
| | - Aditya Anand
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Sourav Ghosh
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Rahul Das
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Subhashree Beura
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
| | - Sarita Jena
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
| | | | - Govindarajan Padmanaban
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | | |
Collapse
|
12
|
Wai CH, Jin J, Cyrklaff M, Genoud C, Funaya C, Sattler J, Maceski A, Meier S, Heiland S, Lanzer M, Frischknecht F, Kuhle J, Bendszus M, Hoffmann A. Neurofilament light chain plasma levels are associated with area of brain damage in experimental cerebral malaria. Sci Rep 2022; 12:10726. [PMID: 35750882 PMCID: PMC9232608 DOI: 10.1038/s41598-022-14291-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 06/03/2022] [Indexed: 11/23/2022] Open
Abstract
Neurofilament light chain (NfL), released during central nervous injury, has evolved as a powerful serum marker of disease severity in many neurological disorders, including infectious diseases. So far NfL has not been assessed in cerebral malaria in human or its rodent model experimental cerebral malaria (ECM), a disease that can lead to fatal brain edema or reversible brain edema. In this study we assessed if NfL serum levels can also grade disease severity in an ECM mouse model with reversible (n = 11) and irreversible edema (n = 10). Blood–brain-barrier disruption and brain volume were determined by magnetic resonance imaging. Neurofilament density volume as well as structural integrity were examined by electron microscopy in regions of most severe brain damage (olfactory bulb (OB), cortex and brainstem). NfL plasma levels in mice with irreversible edema (317.0 ± 45.01 pg/ml) or reversible edema (528.3 ± 125.4 pg/ml) were significantly increased compared to controls (103.4 ± 25.78 pg/ml) by three to five fold, but did not differ significantly in mice with reversible or irreversible edema. In both reversible and irreversible edema, the brain region most affected was the OB with highest level of blood–brain-barrier disruption and most pronounced decrease in neurofilament density volume, which correlated with NfL plasma levels (r = − 0.68, p = 0.045). In cortical and brainstem regions neurofilament density was only decreased in mice with irreversible edema and strongest in the brainstem. In reversible edema NfL plasma levels, MRI findings and neurofilament volume density normalized at 3 months’ follow-up. In conclusion, NfL plasma levels are elevated during ECM confirming brain damage. However, NfL plasma levels fail short on reliably indicating on the final outcomes in the acute disease stage that could be either fatal or reversible. Increased levels of plasma NfL during the acute disease stage are thus likely driven by the anatomical location of brain damage, the olfactory bulb, a region that serves as cerebral draining pathway into the nasal lymphatics.
Collapse
Affiliation(s)
- Chi Ho Wai
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Jessica Jin
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Marek Cyrklaff
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Christel Genoud
- Electron Microscopy Facility, Faculty of Biology and Medicine, University of Lausanne, Lausanne, Switzerland
| | - Charlotta Funaya
- Electron Microscopy Core Facility, Heidelberg University, Heidelberg, Germany
| | - Julia Sattler
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Aleksandra Maceski
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Stephanie Meier
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Sabine Heiland
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Lanzer
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Friedrich Frischknecht
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Jens Kuhle
- Neurologic Clinic and Policlinic, MS Center and Research Center for Clinical Neuroimmunology and Neuroscience Basel (RC2NB), University Hospital Basel, University of Basel, Basel, Switzerland
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Angelika Hoffmann
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany. .,Department of Neuroradiology, University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Freiburgstrasse, 3010, Bern, Switzerland.
| |
Collapse
|
13
|
Chaudhary A, Kataria P, Surela N, Das J. Pathophysiology of Cerebral Malaria: Implications of MSCs as A Regenerative Medicinal Tool. Bioengineering (Basel) 2022; 9:bioengineering9060263. [PMID: 35735506 PMCID: PMC9219920 DOI: 10.3390/bioengineering9060263] [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: 04/10/2022] [Revised: 05/02/2022] [Accepted: 05/03/2022] [Indexed: 11/16/2022] Open
Abstract
The severe form of malaria, i.e., cerebral malaria caused by Plasmodium falciparum, is a complex neurological syndrome. Surviving persons have a risk of behavioral difficulties, cognitive disorders, and epilepsy. Cerebral malaria is associated with multiple organ dysfunctions. The adhesion and accumulation of infected RBCs, platelets, and leucocytes (macrophages, CD4+ and CD8+ T cells, and monocytes) in the brain microvessels play an essential role in disease progression. Micro-vascular hindrance by coagulation and endothelial dysfunction contributes to neurological damage and the severity of the disease. Recent studies in human cerebral malaria and the murine model of cerebral malaria indicate that different pathogens as well as host-derived factors are involved in brain microvessel adhesion and coagulation that induces changes in vascular permeability and impairment of the blood-brain barrier. Efforts to alleviate blood-brain barrier dysfunction and de-sequestering of RBCs could serve as adjunct therapies. In this review, we briefly summarize the current understanding of the pathogenesis of cerebral malaria, the role of some factors (NK cells, platelet, ANG-2/ANG-1 ratio, and PfEMP1) in disease progression and various functions of Mesenchymal stem cells. This review also highlighted the implications of MSCs as a regenerative medicine.
Collapse
Affiliation(s)
- Amrendra Chaudhary
- Parasite-Host Biology, National Institute of Malaria Research, New Delhi 110077, India; (A.C.); (P.K.); (N.S.)
| | - Poonam Kataria
- Parasite-Host Biology, National Institute of Malaria Research, New Delhi 110077, India; (A.C.); (P.K.); (N.S.)
| | - Neha Surela
- Parasite-Host Biology, National Institute of Malaria Research, New Delhi 110077, India; (A.C.); (P.K.); (N.S.)
| | - Jyoti Das
- Parasite-Host Biology, National Institute of Malaria Research, New Delhi 110077, India; (A.C.); (P.K.); (N.S.)
- AcSIR, Ghaziabad 201002, India
- Correspondence: or ; Tel.: +91-25307203; Fax: +91-25307177
| |
Collapse
|
14
|
dos Santos EC, Silva LS, Pinheiro AS, Teixeira DE, Peruchetti DB, Silva-Aguiar RP, Wendt CHC, Miranda KR, Coelho-de-Souza AN, Leal-Cardoso JH, Caruso-Neves C, Pinheiro AAS. The monoterpene 1,8-cineole prevents cerebral edema in a murine model of severe malaria. PLoS One 2022; 17:e0268347. [PMID: 35550638 PMCID: PMC9098050 DOI: 10.1371/journal.pone.0268347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Accepted: 04/27/2022] [Indexed: 11/18/2022] Open
Abstract
1,8-Cineole is a naturally occurring compound found in essential oils of different plants and has well-known anti-inflammatory and antimicrobial activities. In the present work, we aimed to investigate its potential antimalarial effect, using the following experimental models: (1) the erythrocytic cycle of Plasmodium falciparum; (2) an adhesion assay using brain microvascular endothelial cells; and (3) an experimental cerebral malaria animal model induced by Plasmodium berghei ANKA infection in susceptible mice. Using the erythrocytic cycle of Plasmodium falciparum, we characterized the schizonticidal effect of 1,8-cineole. This compound decreased parasitemia in a dose-dependent manner with a half maximal inhibitory concentration of 1045.53 ± 63.30 μM. The inhibitory effect of 972 μM 1,8-cineole was irreversible and independent of parasitemia. Moreover, 1,8-cineole reduced the progression of intracellular development of the parasite over 2 cycles, inducing important morphological changes. Ultrastructure analysis revealed a massive loss of integrity of endomembranes and hemozoin crystals in infected erythrocytes treated with 1,8-cineole. The monoterpene reduced the adhesion index of infected erythrocytes to brain microvascular endothelial cells by 60%. Using the experimental cerebral malaria model, treatment of infected mice for 6 consecutive days with 100 mg/kg/day 1,8-cineole reduced cerebral edema with a 50% reduction in parasitemia. Our data suggest a potential antimalarial effect of 1,8-cineole with an impact on the parasite erythrocytic cycle and severe disease.
Collapse
Affiliation(s)
- Edgleyson C. dos Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, Brazil
| | - Leandro S. Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro S. Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas E. Teixeira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo B. Peruchetti
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo P. Silva-Aguiar
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Camila H. C. Wendt
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kildare R. Miranda
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, Conselho Nacional de Desenvolvimento Científico e Tecnológico, Rio de Janeiro, Brazil
| | | | | | - Celso Caruso-Neves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, Conselho Nacional de Desenvolvimento Científico e Tecnológico, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health, Rio de Janeiro, Brazil
| | - Ana Acacia S. Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
- Rio de Janeiro Innovation Network in Nanosystems for Health, Rio de Janeiro, Brazil
- * E-mail:
| |
Collapse
|
15
|
Imai T, Ngo-Thanh H, Suzue K, Shimo A, Nakamura A, Horiuchi Y, Hisaeda H, Murakami T. Live Vaccination with Blood-Stage Plasmodium yoelii 17XNL Prevents the Development of Experimental Cerebral Malaria. Vaccines (Basel) 2022; 10:vaccines10050762. [PMID: 35632518 PMCID: PMC9145751 DOI: 10.3390/vaccines10050762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 05/09/2022] [Accepted: 05/09/2022] [Indexed: 01/27/2023] Open
Abstract
In our work, we aim to develop a malaria vaccine with cross-strain (-species) protection. C57BL/6 mice infected with the P. berghei ANKA strain (PbA) develop experimental cerebral malaria (ECM). In contrast, ECM development is inhibited in infected mice depleted of T cells. The clinical applications of immune-cell depletion are limited due to the benefits of host defense against infectious diseases. Therefore, in the present study we attempted to develop a new method for preventing ECM without immune cell depletion. We demonstrated that mice inoculated with a heterologous live-vaccine of P. yoelii 17XNL were able to prevent both ECM and lung pathology and survived longer than control mice when challenged with PbA. Live vaccination protected blood–organ barriers from PbA infection. Meanwhile, live vaccination conferred sterile protection against homologous challenge with the P. yoelii 17XL virulent strain for the long-term. Analysis of the immune response induced by live vaccination showed that cross-reactive antibodies against PbA antigens were generated. IL-10, which has an immunosuppressive effect, was strongly induced in mice challenged with PbA, unlike the pro-inflammatory cytokine IFNγ. These results suggest that the protective effect of heterologous live vaccination against ECM development results from IL-10-mediated host protection.
Collapse
Affiliation(s)
- Takashi Imai
- Department of Infectious Diseases and Host Defense, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan; (H.N.-T.); (K.S.)
- Department of Microbiology, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; (A.S.); (A.N.); (Y.H.); (T.M.)
- Correspondence: ; Tel.: +81-49-276-1166
| | - Ha Ngo-Thanh
- Department of Infectious Diseases and Host Defense, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan; (H.N.-T.); (K.S.)
- National Hospital for Tropical Disease, 78 Giai Phong, Dong Da, Hanoi 10000, Vietnam
| | - Kazutomo Suzue
- Department of Infectious Diseases and Host Defense, Graduate School of Medicine, Gunma University, Maebashi 371-8511, Japan; (H.N.-T.); (K.S.)
| | - Aoi Shimo
- Department of Microbiology, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; (A.S.); (A.N.); (Y.H.); (T.M.)
| | - Akihiro Nakamura
- Department of Microbiology, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; (A.S.); (A.N.); (Y.H.); (T.M.)
| | - Yutaka Horiuchi
- Department of Microbiology, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; (A.S.); (A.N.); (Y.H.); (T.M.)
| | - Hajime Hisaeda
- Department of Parasitology, National Institute of Infectious Diseases, Tokyo 162-0052, Japan;
| | - Takashi Murakami
- Department of Microbiology, Saitama Medical University, Moroyama-machi, Iruma-gun, Saitama 350-0495, Japan; (A.S.); (A.N.); (Y.H.); (T.M.)
| |
Collapse
|
16
|
Albrecht-Schgoer K, Lackner P, Schmutzhard E, Baier G. Cerebral Malaria: Current Clinical and Immunological Aspects. Front Immunol 2022; 13:863568. [PMID: 35514965 PMCID: PMC9067128 DOI: 10.3389/fimmu.2022.863568] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/21/2022] [Indexed: 11/23/2022] Open
Abstract
This review focuses on current clinical and immunological aspects of cerebral malaria induced by Plasmodium falciparum infection. Albeit many issues concerning the inflammatory responses remain unresolved and need further investigations, current knowledge of the underlying molecular mechanisms is highlighted. Furthermore, and in the light of significant limitations in preventative diagnosis and treatment of cerebral malaria, this review mainly discusses our understanding of immune mechanisms in the light of the most recent research findings. Remarkably, the newly proposed CD8+ T cell-driven pathophysiological aspects within the central nervous system are summarized, giving first rational insights into encouraging studies with immune-modulating adjunctive therapies that protect from symptomatic cerebral participation of Plasmodium falciparum infection.
Collapse
Affiliation(s)
- Karin Albrecht-Schgoer
- Division of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| | - Peter Lackner
- Department of Neurology, Klinik Floridsdorf, Wien, Austria
| | - Erich Schmutzhard
- Department of Neurology, Medical University of Innsbruck, Innsbruck, Austria
| | - Gottfried Baier
- Division of Translational Cell Genetics, Medical University of Innsbruck, Innsbruck, Austria
| |
Collapse
|
17
|
Leleu I, Alloo J, Cazenave PA, Roland J, Pied S. Autophagy Pathways in the Genesis of Plasmodium-Derived Microvesicles: A Double-Edged Sword? Life (Basel) 2022; 12:life12030415. [PMID: 35330166 PMCID: PMC8955828 DOI: 10.3390/life12030415] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 03/02/2022] [Accepted: 03/08/2022] [Indexed: 11/16/2022] Open
Abstract
Malaria, caused by Plasmodium species (spp.), is a deadly parasitic disease that results in approximately 400,000 deaths per year globally. Autophagy pathways play a fundamental role in the developmental stages of the parasite within the mammalian host. They are also involved in the production of Plasmodium-derived extracellular vesicles (EVs), which play an important role in the infection process, either by providing nutrients for parasite growth or by contributing to the immunopathophysiology of the disease. For example, during the hepatic stage, Plasmodium-derived EVs contribute to parasite virulence by modulating the host immune response. EVs help in evading the different autophagy mechanisms deployed by the host for parasite clearance. During cerebral malaria, on the other hand, parasite-derived EVs promote an astrocyte-mediated inflammatory response, through the induction of a non-conventional host autophagy pathway. In this review, we will discuss the cross-talk between Plasmodium-derived microvesicles and autophagy, and how it influences the outcome of infection.
Collapse
|
18
|
Jin J, Ba MA, Wai CH, Mohanty S, Sahu PK, Pattnaik R, Pirpamer L, Fischer M, Heiland S, Lanzer M, Frischknecht F, Mueller AK, Pfeil J, Majhi M, Cyrklaff M, Wassmer SC, Bendszus M, Hoffmann A. Transcellular blood-brain barrier disruption in malaria-induced reversible brain edema. Life Sci Alliance 2022; 5:5/6/e202201402. [PMID: 35260473 PMCID: PMC8905774 DOI: 10.26508/lsa.202201402] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2022] [Revised: 02/19/2022] [Accepted: 02/22/2022] [Indexed: 12/12/2022] Open
Abstract
We present how reversible edema can reliably be induced in experimental cerebral malaria and show that it is associated with transcellular blood–brain barrier disruption and delayed microhemorrhages. Brain swelling occurs in cerebral malaria (CM) and may either reverse or result in fatal outcome. It is currently unknown how brain swelling in CM reverses, as brain swelling at the acute stage is difficult to study in humans and animal models with reliable induction of reversible edema are not known. In this study, we show that reversible brain swelling in experimental murine CM can be induced reliably after single vaccination with radiation-attenuated sporozoites as proven by in vivo high-field magnetic resonance imaging. Our results provide evidence that brain swelling results from transcellular blood–brain barrier disruption (BBBD), as revealed by electron microscopy. This mechanism enables reversal of brain swelling but does not prevent persistent focal brain damage, evidenced by microhemorrhages, in areas of most severe BBBD. In adult CM patients magnetic resonance imaging demonstrate microhemorrhages in more than one third of patients with reversible edema, emphasizing similarities of the experimental model and human disease. Our data suggest that targeting transcellular BBBD may represent a promising adjunct therapeutic approach to reduce edema and may improve neurological outcome.
Collapse
Affiliation(s)
- Jessica Jin
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Mame Aida Ba
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Chi Ho Wai
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Sanjib Mohanty
- Center for the Study of Complex Malaria in India, Ispat General Hospital, Rourkela, India
| | - Praveen K Sahu
- Center for the Study of Complex Malaria in India, Ispat General Hospital, Rourkela, India
| | | | - Lukas Pirpamer
- Department of Neurology, Division of Neurogeriatrics, Medical University of Graz, Graz, Austria
| | - Manuel Fischer
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Division of Experimental Radiology, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Sabine Heiland
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Division of Experimental Radiology, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Michael Lanzer
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Friedrich Frischknecht
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Ann-Kristin Mueller
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany
| | - Johannes Pfeil
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infection Research (DZIF), Heidelberg, Germany.,Center for Childhood and Adolescent Medicine, General Pediatrics, University Hospital, Heidelberg, Germany
| | - Megharay Majhi
- Department of Radiology, Ispat General Hospital, Rourkela, India
| | - Marek Cyrklaff
- Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Samuel C Wassmer
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, UK
| | - Martin Bendszus
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany
| | - Angelika Hoffmann
- Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany .,Division of Experimental Radiology, Department of Neuroradiology, Heidelberg University Hospital, Heidelberg, Germany.,Centre for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany.,University Institute of Diagnostic and Interventional Neuroradiology, University Hospital Bern, Inselspital, University of Bern, Bern, Switzerland
| |
Collapse
|
19
|
Simwela NV, Waters AP. Current status of experimental models for the study of malaria. Parasitology 2022; 149:1-22. [PMID: 35357277 PMCID: PMC9378029 DOI: 10.1017/s0031182021002134] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Infection by malaria parasites (Plasmodium spp.) remains one of the leading causes of morbidity and mortality, especially in tropical regions of the world. Despite the availability of malaria control tools such as integrated vector management and effective therapeutics, these measures have been continuously undermined by the emergence of vector resistance to insecticides or parasite resistance to frontline antimalarial drugs. Whilst the recent pilot implementation of the RTS,S malaria vaccine is indeed a remarkable feat, highly effective vaccines against malaria remain elusive. The barriers to effective vaccines result from the complexity of both the malaria parasite lifecycle and the parasite as an organism itself with consequent major gaps in our understanding of their biology. Historically and due to the practical and ethical difficulties of working with human malaria infections, research into malaria parasite biology has been extensively facilitated by animal models. Animals have been used to study disease pathogenesis, host immune responses and their (dys)regulation and further disease processes such as transmission. Moreover, animal models remain at the forefront of pre-clinical evaluations of antimalarial drugs (drug efficacy, mode of action, mode of resistance) and vaccines. In this review, we discuss commonly used animal models of malaria, the parasite species used and their advantages and limitations which hinder their extrapolation to actual human disease. We also place into this context the most recent developments such as organoid technologies and humanized mice.
Collapse
Affiliation(s)
- Nelson V. Simwela
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Andrew P. Waters
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| |
Collapse
|
20
|
Heme oxygenase-1, carbon monoxide, and malaria – The interplay of chemistry and biology. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2021.214285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
21
|
Ghazanfari N, Gregory JL, Devi S, Fernandez-Ruiz D, Beattie L, Mueller SN, Heath WR. CD8 + and CD4 + T Cells Infiltrate into the Brain during Plasmodium berghei ANKA Infection and Form Long-Term Resident Memory. THE JOURNAL OF IMMUNOLOGY 2021; 207:1578-1590. [PMID: 34400523 DOI: 10.4049/jimmunol.2000773] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 07/18/2021] [Indexed: 12/31/2022]
Abstract
In the Plasmodium berghei ANKA mouse model of malaria, accumulation of CD8+ T cells and infected RBCs in the brain promotes the development of experimental cerebral malaria (ECM). In this study, we used malaria-specific transgenic CD4+ and CD8+ T cells to track evolution of T cell immunity during the acute and memory phases of P. berghei ANKA infection. Using a combination of techniques, including intravital multiphoton and confocal microscopy and flow cytometric analysis, we showed that, shortly before onset of ECM, both CD4+ and CD8+ T cell populations exit the spleen and begin infiltrating the brain blood vessels. Although dominated by CD8+ T cells, a proportion of both T cell subsets enter the brain parenchyma, where they are largely associated with blood vessels. Intravital imaging shows these cells moving freely within the brain parenchyma. Near the onset of ECM, leakage of RBCs into areas of the brain can be seen, implicating severe damage. If mice are cured before ECM onset, brain infiltration by T cells still occurs, but ECM is prevented, allowing development of long-term resident memory T cell populations within the brain. This study shows that infiltration of malaria-specific T cells into the brain parenchyma is associated with cerebral immunopathology and the formation of brain-resident memory T cells. The consequences of these resident memory populations is unclear but raises concerns about pathology upon secondary infection.
Collapse
Affiliation(s)
- Nazanin Ghazanfari
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Julia L Gregory
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Sapna Devi
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Daniel Fernandez-Ruiz
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Lynette Beattie
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Scott N Mueller
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and.,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - William R Heath
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Victoria, Australia; and .,The Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
22
|
Shaw TN, Haley MJ, Dookie RS, Godfrey JJ, Cheeseman AJ, Strangward P, Zeef LAH, Villegas-Mendez A, Couper KN. Memory CD8 + T cells exhibit tissue imprinting and non-stable exposure-dependent reactivation characteristics following blood-stage Plasmodium berghei ANKA infections. Immunology 2021; 164:737-753. [PMID: 34407221 PMCID: PMC8561116 DOI: 10.1111/imm.13405] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Revised: 08/09/2021] [Accepted: 08/13/2021] [Indexed: 02/06/2023] Open
Abstract
Experimental cerebral malaria (ECM) is a severe complication of Plasmodium berghei ANKA (PbA) infection in mice, characterized by CD8+ T‐cell accumulation within the brain. Whilst the dynamics of CD8+ T‐cell activation and migration during extant primary PbA infection have been extensively researched, the fate of the parasite‐specific CD8+ T cells upon resolution of ECM is not understood. In this study, we show that memory OT‐I cells persist systemically within the spleen, lung and brain following recovery from ECM after primary PbA‐OVA infection. Whereas memory OT‐I cells within the spleen and lung exhibited canonical central memory (Tcm) and effector memory (Tem) phenotypes, respectively, memory OT‐I cells within the brain post‐PbA‐OVA infection displayed an enriched CD69+CD103− profile and expressed low levels of T‐bet. OT‐I cells within the brain were excluded from short‐term intravascular antibody labelling but were targeted effectively by longer‐term systemically administered antibodies. Thus, the memory OT‐I cells were extravascular within the brain post‐ECM but were potentially not resident memory cells. Importantly, whilst memory OT‐I cells exhibited strong reactivation during secondary PbA‐OVA infection, preventing activation of new primary effector T cells, they had dampened reactivation during a fourth PbA‐OVA infection. Overall, our results demonstrate that memory CD8+ T cells are systemically distributed but exhibit a unique phenotype within the brain post‐ECM, and that their reactivation characteristics are shaped by infection history. Our results raise important questions regarding the role of distinct memory CD8+ T‐cell populations within the brain and other tissues during repeat Plasmodium infections.
Collapse
Affiliation(s)
- Tovah N Shaw
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK.,School of Biological Sciences, Institute of Immunology and Infection, University of Edinburgh, Edinburgh, UK
| | - Michael J Haley
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Rebecca S Dookie
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Jenna J Godfrey
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Antonn J Cheeseman
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Patrick Strangward
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Leo A H Zeef
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Ana Villegas-Mendez
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| | - Kevin N Couper
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, UK
| |
Collapse
|
23
|
Villegas-Mendez A, Stafford N, Haley MJ, Pravitasari NE, Baudoin F, Ali A, Asih PBS, Siregar JE, Baena E, Syafruddin D, Couper KN, Oceandy D. The plasma membrane calcium ATPase 4 does not influence parasite levels but partially promotes experimental cerebral malaria during murine blood stage malaria. Malar J 2021; 20:297. [PMID: 34215257 PMCID: PMC8252299 DOI: 10.1186/s12936-021-03832-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 06/24/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND Recent genome wide analysis studies have identified a strong association between single nucleotide variations within the human ATP2B4 gene and susceptibility to severe malaria. The ATP2B4 gene encodes the plasma membrane calcium ATPase 4 (PMCA4), which is responsible for controlling the physiological level of intracellular calcium in many cell types, including red blood cells (RBCs). It is, therefore, postulated that genetic differences in the activity or expression level of PMCA4 alters intracellular Ca2+ levels and affects RBC hydration, modulating the invasion and growth of the Plasmodium parasite within its target host cell. METHODS In this study the course of three different Plasmodium spp. infections were examined in mice with systemic knockout of Pmca4 expression. RESULTS Ablation of PMCA4 reduced the size of RBCs and their haemoglobin content but did not affect RBC maturation and reticulocyte count. Surprisingly, knockout of PMCA4 did not significantly alter peripheral parasite burdens or the dynamics of blood stage Plasmodium chabaudi infection or reticulocyte-restricted Plasmodium yoelii infection. Interestingly, although ablation of PMCA4 did not affect peripheral parasite levels during Plasmodium berghei infection, it did promote slight protection against experimental cerebral malaria, associated with a minor reduction in antigen-experienced T cell accumulation in the brain. CONCLUSIONS The finding suggests that PMCA4 may play a minor role in the development of severe malarial complications, but that this appears independent of direct effects on parasite invasion, growth or survival within RBCs.
Collapse
Affiliation(s)
- Ana Villegas-Mendez
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | - Nicholas Stafford
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Michael J Haley
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK
| | | | - Florence Baudoin
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK
| | - Adnan Ali
- Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Manchester, SK10 4TG, UK
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
| | | | | | - Esther Baena
- Cancer Research UK Manchester Institute, The University of Manchester, Alderley Park, Manchester, SK10 4TG, UK
| | - Din Syafruddin
- Eijkman Institute for Molecular Biology, Jakarta, 10430, Indonesia
| | - Kevin N Couper
- The Lydia Becker Institute of Immunology and Inflammation, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT, UK.
| | - Delvac Oceandy
- Division of Cardiovascular Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, M13 9PT, UK.
| |
Collapse
|
24
|
Whole blood transfusion improves vascular integrity and increases survival in artemether-treated experimental cerebral malaria. Sci Rep 2021; 11:12077. [PMID: 34103601 PMCID: PMC8187502 DOI: 10.1038/s41598-021-91499-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 05/25/2021] [Indexed: 01/28/2023] Open
Abstract
Pathological features observed in both human and experimental cerebral malaria (ECM) are endothelial dysfunction and changes in blood components. Blood transfusion has been routinely used in patients with severe malarial anemia and can also benefit comatose and acidotic malaria patients. In the present study Plasmodium berghei-infected mice were transfused intraperitoneally with 200 μL of whole blood along with 20 mg/kg of artemether. ECM mice showed severe thrombocytopenia and decreases in hematocrit. Artemether treatment markedly aggravated anemia within 24 h. Whole blood administration significantly prevented further drop in hematocrit and partially restored the platelet count. Increased levels of plasma angiopoietin-2 (Ang-2) remained high 24 h after artemether treatment but returned to normal levels 24 h after blood transfusion, indicating reversal to quiescence. Ang-1 was depleted in ECM mice and levels were not restored by any treatment. Blood transfusion prevented the aggravation of the breakdown of blood brain barrier after artemether treatment and decreased spleen congestion without affecting splenic lymphocyte populations. Critically, blood transfusion resulted in markedly improved survival of mice with ECM (75.9% compared to 50.9% receiving artemether only). These findings indicate that whole blood transfusion can be an effective adjuvant therapy for cerebral malaria.
Collapse
|
25
|
Galán-Salinas A, Corral-Ruíz G, Pérez-Vega MJ, Fabila-Castillo L, Silva-García R, Marquina-Castillo B, León-Contreras JC, Barrios-Payán J, Francisco-Cruz A, Montecillo-Aguado M, Huerta-Yepez S, Calderón-Amador J, Flores-Romo L, Hernández-Pando R, Sánchez-Torres LE. Monocyte Locomotion Inhibitory Factor confers neuroprotection and prevents the development of murine cerebral malaria. Int Immunopharmacol 2021; 97:107674. [PMID: 34044183 DOI: 10.1016/j.intimp.2021.107674] [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: 02/10/2021] [Revised: 03/30/2021] [Accepted: 04/10/2021] [Indexed: 10/21/2022]
Abstract
Cerebral malaria (CM) is a neurological complication derived from the Plasmodium falciparum infection in humans. The mechanisms involved in the disease progression are still not fully understood, but both the sequestration of infected red blood cells (iRBC) and leukocytes and an exacerbated host inflammatory immune response are significant factors. In this study, we investigated the effect of Monocyte Locomotion Inhibitory Factor (MLIF), an anti-inflammatory peptide, in a well-characterized murine model of CM. Our data showed that the administration of MLIF increased the survival and avoided the neurological signs of CM in Plasmodium berghei ANKA (PbA) infected C57BL/6 mice. MLIF administration down-regulated systemic inflammatory mediators such as IFN-γ, TNF-α, IL-6, CXCL2, and CCL2, as well as the in situ expression of TNF-α in the brain. In the same way, MLIF reduced the expression of CD31, CD36, CD54, and CD106 in the cerebral endothelium of infected animals and prevented the sequestration of iRBC and leucocytes in the brain microvasculature. Furthermore, MLIF inhibited the activation of astrocytes and microglia and preserved the integrity of the blood-brain barrier (BBB). In conclusion, our results demonstrated that the administration of MLIF increased survival and conferred neuroprotection by decreasing neuroinflammation in murine CM.
Collapse
Affiliation(s)
- A Galán-Salinas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico; Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico
| | - G Corral-Ruíz
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico; Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico
| | - M J Pérez-Vega
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico; Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico
| | - L Fabila-Castillo
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico; Departamento de Farmacia, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico
| | - R Silva-García
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, CMN-Siglo XXI, IMSS, México City, Mexico
| | - B Marquina-Castillo
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - J C León-Contreras
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - J Barrios-Payán
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico
| | - A Francisco-Cruz
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Montecillo-Aguado
- Unidad de Investigación en Enfermedades Oncológicas, Hospital Infantil de México, Federico Gómez, México City, Mexico
| | - S Huerta-Yepez
- Unidad de Investigación en Enfermedades Oncológicas, Hospital Infantil de México, Federico Gómez, México City, Mexico
| | - J Calderón-Amador
- Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico; Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados, Instituto Politécnico Nacional, México City, Mexico
| | - L Flores-Romo
- Departamento de Biología Celular, Centro de Investigación y Estudios Avanzados, Instituto Politécnico Nacional, México City, Mexico
| | - R Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, México City, Mexico.
| | - L E Sánchez-Torres
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México City, Mexico.
| |
Collapse
|
26
|
Hai L, Shi X, Wang Q. Attenuated T Cell Responses Are Associated With the Blockade of Cerebral Malaria Development by YOP1-Deficient Plasmodium berghei ANKA. Front Immunol 2021; 12:642585. [PMID: 34025654 PMCID: PMC8134684 DOI: 10.3389/fimmu.2021.642585] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Accepted: 04/21/2021] [Indexed: 11/13/2022] Open
Abstract
Reticulon and the REEP family of proteins stabilize the high curvature of endoplasmic reticulum tubules. The REEP5 homolog in Plasmodium, Plasmodium berghei YOP1 (PbYOP1), plays an important role in the erythrocytic cycle of the P. berghei ANKA and the pathogenesis of experimental cerebral malaria (ECM), but the mechanisms are largely unknown. Here, we show that protection from ECM in Pbyop1Δ-infected mice is associated with reduced intracerebral Th1 accumulation, decreased expression of pro-inflammatory cytokines and chemokines, and attenuated pathologies in the brainstem, though the total number of CD4+ and CD8+ T cells sequestered in the brain are not reduced. Expression of adhesive molecules on brain endothelial cells, including ICAM-1, VCAM-1, and CD36, are decreased, particularly in the brainstem, where fatal pathology is always induced during ECM. Subsequently, CD8+ T cell-mediated cell apoptosis in the brain is compromised. These findings suggest that Pbyop1Δ parasites can be a useful tool for mechanistic investigation of cerebral malaria pathogenesis.
Collapse
Affiliation(s)
- Lei Hai
- Department of Immunology, School of Basic Medical Sciences, and Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Xiaoyu Shi
- Department of Immunology, School of Basic Medical Sciences, and Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Qian Wang
- Department of Immunology, School of Basic Medical Sciences, and Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China.,National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
27
|
Perillyl alcohol reduces parasite sequestration and cerebrovascular dysfunction during experimental cerebral malaria. Antimicrob Agents Chemother 2021; 65:AAC.00004-21. [PMID: 33649109 PMCID: PMC8092904 DOI: 10.1128/aac.00004-21] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cerebral malaria (CM) is a severe immunovasculopathy which presents high mortality rate (15-20%), despite the availability of artemisinin-based therapy. More effective immunomodulatory and/or antiparasitic therapies are urgently needed. Experimental Cerebral Malaria (ECM) in mice is used to elucidate aspects involved in this pathology since manifests many of the neurological features of CM. In the present study, we evaluated the potential mechanisms involved in the protection afforded by perillyl alcohol (POH) in mouse strains susceptible to CM caused by Plasmodium berghei ANKA (PbA) infection through intranasal preventive treatment. Additionally, to evaluate the interaction of POH with the cerebral endothelium using an in vitro model of human brain endothelial cells (HBEC). Pharmacokinetic approaches demonstrated constant and prolonged levels of POH in the plasma and brain after a single intranasal dose. Treatment with POH effectively prevented vascular dysfunction. Furthermore, treatment with POH reduced the endothelial cell permeability and PbA s in the brain and spleen. Finally, POH treatment decreased the accumulation of macrophages and T and B cells in the spleen and downregulated the expression of endothelial adhesion molecules (ICAM-1, VCAM-1, and CD36) in the brain. POH is a potent monoterpene that prevents cerebrovascular dysfunction in vivo and in vitro, decreases parasite sequestration, and modulates different processes related to the activation, permeability, and integrity of the blood brain barrier (BBB), thereby preventing cerebral oedema and inflammatory infiltrates.
Collapse
|
28
|
Pontes KAO, Silva LS, Santos EC, Pinheiro AS, Teixeira DE, Peruchetti DB, Silva-Aguiar RP, Wendt CHC, Miranda KR, Coelho-de-Souza AN, Leal-Cardoso JH, Caruso-Neves C, Pinheiro AAS. Eugenol disrupts Plasmodium falciparum intracellular development during the erythrocytic cycle and protects against cerebral malaria. Biochim Biophys Acta Gen Subj 2020; 1865:129813. [PMID: 33321150 DOI: 10.1016/j.bbagen.2020.129813] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 10/22/2022]
Abstract
BACKGROUND Malaria is a parasitic disease that compromises the human host. Currently, control of the Plasmodium falciparum burden is centered on artemisinin-based combination therapies. However, decreased sensitivity to artemisinin and derivatives has been reported, therefore it is important to identify new therapeutic strategies. METHOD We used human erythrocytes infected with P. falciparum and experimental cerebral malaria (ECM) animal model to assess the potential antimalarial effect of eugenol, a component of clove bud essential oil. RESULTS Plasmodium falciparum cultures treated with increasing concentrations of eugenol reduced parasitemia in a dose-dependent manner, with IC50 of 532.42 ± 29.55 μM. This effect seems to be irreversible and maintained even in the presence of high parasitemia. The prominent effect of eugenol was detected in the evolution from schizont to ring forms, inducing important morphological changes, indicating a disruption in the development of the erythrocytic cycle. Aberrant structural modification was observed by electron microscopy, showing the separation of the two nuclear membrane leaflets as well as other subcellular membranes, such as from the digestive vacuole. Importantly, in vivo studies using ECM revealed a reduction in blood parasitemia and cerebral edema when mice were treated for 6 consecutive days upon infection. CONCLUSIONS These data suggest a potential effect of eugenol against Plasmodium sp. with an impact on cerebral malaria. GENERAL SIGNIFICANCE Our results provide a rational basis for the use of eugenol in therapeutic strategies to the treatment of malaria.
Collapse
Affiliation(s)
- Kesley A O Pontes
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto de Ciências Biomédicas, Universidade Estadual do Ceará, Fortaleza, Brazil
| | - Leandro S Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Edgleyson C Santos
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Alessandro S Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Douglas E Teixeira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Diogo B Peruchetti
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rodrigo P Silva-Aguiar
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Camila H C Wendt
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Kildare R Miranda
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem /CENABIO, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Biologia Estrutural e Bioimagem, INBEB, Conselho Nacional de Desenvolvimento Científico e Tecnológico/MCTIC, Rio de Janeiro, Brazil
| | | | | | - Celso Caruso-Neves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Instituto Nacional de Ciência e Tecnologia em Medicina Regenerativa, INCT-Regenera, Conselho Nacional de Desenvolvimento Científico e Tecnológico/MCTIC, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health - NanoSAUDE/FAPERJ, Rio de Janeiro, Brazil
| | - Ana Acacia S Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health - NanoSAUDE/FAPERJ, Rio de Janeiro, Brazil.
| |
Collapse
|
29
|
Jiang X, Chen L, Zheng Z, Chen Y, Weng X, Guo Y, Li K, Yang T, Qu S, Liu H, Li Y, Zhu X. Synergistic Effect of Combined Artesunate and Tetramethylpyrazine in Experimental Cerebral Malaria. ACS Infect Dis 2020; 6:2400-2409. [PMID: 32786270 DOI: 10.1021/acsinfecdis.0c00124] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Intravenous artesunate is effective against cerebral malaria (CM), but high mortality and neurological sequelae in survivors are inevitable. We investigated the effect of combined artesunate and tetramethylpyrazine using mouse models of experimental cerebral malaria (ECM). Artesunate + tetramethylpyrazine reduced microvascular blockage and improved neurological function, including the rapid murine coma and behavior scale (RMCBS), leading to improved survival and reduced pathology in ECM. This combination downregulated the expression of adhesion molecules and sequestration of parasitized red blood cells (pRBCs), increased cerebral blood flow, nerve growth factor (b-NGF), vascular endothelial growth factor A (VEGF-A), and neurotrophin (brain-derived neurotrophic factor (BDNF), neurotrophic factor-3 (NT-3)) levels, and alleviated hippocampal neuronal damage and astrocyte activation. Down- (n = 128) and upregulated (n = 64) proteins were identified in the artesunate group, while up- (n = 217) and downregulated (n = 177) proteins were identified in the artesunate + tetramethylpyrazine group, presenting a significantly altered proteome profile. KEGG analysis showed that 166 differentially expressed proteins were enriched in the Art group and 234, in the artesunate + tetramethylpyrazine group. The neuroprotective effects of artesunate + tetramethylpyrazine were mainly related to proteins involved in axon development and transportation between blood and brain. These results suggested that artesunate + tetramethylpyrazine could be a potential adjuvant therapy against CM, but this will have to be confirmed in future studies and trials.
Collapse
Affiliation(s)
- Xiaohui Jiang
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| | - Lina Chen
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| | - Zhongyuan Zheng
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| | - Ying Chen
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| | - Xiaogang Weng
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| | - Yuan Guo
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| | - Kai Li
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| | - Ting Yang
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| | - Shuiqing Qu
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| | - Hui Liu
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| | - Yujie Li
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| | - Xiaoxin Zhu
- Artemisinin Research Center, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
- Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, No. 16 Dongzhimen Nei Avenue, Beijing 100700, China
| |
Collapse
|
30
|
Akkaya M, Bansal A, Sheehan PW, Pena M, Cimperman CK, Qi CF, Yazew T, Otto TD, Billker O, Miller LH, Pierce SK. Testing the impact of a single nucleotide polymorphism in a Plasmodium berghei ApiAP2 transcription factor on experimental cerebral malaria in mice. Sci Rep 2020; 10:13630. [PMID: 32788672 PMCID: PMC7424516 DOI: 10.1038/s41598-020-70617-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/28/2020] [Indexed: 12/15/2022] Open
Abstract
Cerebral malaria (CM) is the deadliest form of severe Plasmodium infections. Currently, we have limited understanding of the mechanisms by which Plasmodium parasites induce CM. The mouse model of CM, experimental CM (ECM), induced by infection with the rodent parasite, Plasmodium berghei ANKA (PbANKA) has been extensively used to study the pathophysiology of CM. Recent genomic analyses revealed that the coding regions of PbANKA and the closely related Plasmodium berghei NK65 (PbNK65), that does not cause ECM, differ in only 21 single nucleotide polymorphysims (SNPs). Thus, the SNP-containing genes might contribute to the pathogenesis of ECM. Although the majority of these SNPs are located in genes of unknown function, one SNP is located in the DNA binding site of a member of the Plasmodium ApiAP2 transcription factor family, that we recently showed functions as a virulence factor alternating the host’s immune response to the parasite. Here, we investigated the impact of this SNP on the development of ECM. Our results using CRISPR-Cas9 engineered parasites indicate that despite its immune modulatory function, the SNP is neither necessary nor sufficient to induce ECM and thus cannot account for parasite strain-specific differences in ECM phenotypes.
Collapse
Affiliation(s)
- Munir Akkaya
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Lane, Room 4S04, Rockville, MD, USA.
| | - Abhisheka Bansal
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA.,School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Patrick W Sheehan
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Lane, Room 4S04, Rockville, MD, USA.,Department of Neurology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Mirna Pena
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Lane, Room 4S04, Rockville, MD, USA
| | - Clare K Cimperman
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Lane, Room 4S04, Rockville, MD, USA.,Department of Microbiology, University of Pennsylvania, Philadelphia, PA, USA
| | - Chen Feng Qi
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Lane, Room 4S04, Rockville, MD, USA
| | - Takele Yazew
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Lane, Room 4S04, Rockville, MD, USA.,Department of Veterinary Medicine, College of Agriculture and Natural Resources, University of Maryland, College Park, MD, USA
| | - Thomas D Otto
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| | - Oliver Billker
- Laboratory for Molecular Infection Medicine Sweden and Molecular Biology Department, Umea University, Umea, Sweden
| | - Louis H Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Lane, Room 4S04, Rockville, MD, USA.
| |
Collapse
|
31
|
Shi X, Hai L, Govindasamy K, Gao J, Coppens I, Hu J, Wang Q, Bhanot P. A Plasmodium homolog of ER tubule-forming proteins is required for parasite virulence. Mol Microbiol 2020; 114:454-467. [PMID: 32432369 DOI: 10.1111/mmi.14526] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/04/2020] [Accepted: 05/05/2020] [Indexed: 01/27/2023]
Abstract
Reticulon and REEP family of proteins stabilize the high curvature of endoplasmic reticulum (ER) tubules. Plasmodium berghei Yop1 (PbYop1) is a REEP5 homolog in Plasmodium. Here, we characterize its function using a gene-knockout (Pbyop1∆). Pbyop1∆ asexual stage parasites display abnormal ER architecture and an enlarged digestive vacuole. The erythrocytic cycle of Pbyop1∆ parasites is severely attenuated and the incidence of experimental cerebral malaria is significantly decreased in Pbyop1∆-infected mice. Pbyop1∆ sporozoites have reduced speed, are slower to invade host cells but give rise to equal numbers of infected HepG2 cells, as WT sporozoites. We propose that PbYOP1's disruption may lead to defects in trafficking and secretion of a subset of proteins required for parasite development and invasion of erythrocytes. Furthermore, the maintenance of ER morphology in different parasite stages is likely to depend on different proteins.
Collapse
Affiliation(s)
- Xiaoyu Shi
- Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Lei Hai
- Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Kavitha Govindasamy
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| | - Jian Gao
- Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD, USA
| | - Junjie Hu
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Qian Wang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin Medical University, Tianjin, China.,National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Purnima Bhanot
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers New Jersey Medical School, Newark, NJ, USA
| |
Collapse
|
32
|
Georgiadou A, Cunnington AJ. Shedding of the Vascular Endothelial Glycocalyx: A Common Pathway to Severe Malaria? Clin Infect Dis 2020; 69:1721-1723. [PMID: 30698670 DOI: 10.1093/cid/ciz043] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2018] [Accepted: 01/14/2019] [Indexed: 12/28/2022] Open
Affiliation(s)
- Athina Georgiadou
- Section of Pediatrics, Department of Medicine, Imperial College London, United Kingdom
| | - Aubrey J Cunnington
- Section of Pediatrics, Department of Medicine, Imperial College London, United Kingdom
| |
Collapse
|
33
|
Meehan GR, Scales HE, Osii R, De Niz M, Lawton JC, Marti M, Garside P, Craig A, Brewer JM. Developing a xenograft model of human vasculature in the mouse ear pinna. Sci Rep 2020; 10:2058. [PMID: 32029768 PMCID: PMC7004987 DOI: 10.1038/s41598-020-58650-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Accepted: 01/14/2020] [Indexed: 12/31/2022] Open
Abstract
Humanised xenograft models allow for the analysis of human tissue within a physiological environment in vivo. However, current models often rely on the angiogenesis and ingrowth of recipient vasculature to perfuse tissues, preventing analysis of biological processes and diseases involving human blood vessels. This limits the effectiveness of xenografts in replicating human physiology and may lead to issues with translating findings into human research. We have designed a xenograft model of human vasculature to address this issue. Human subcutaneous fat was cultured in vitro to promote blood vessel outgrowth prior to implantation into immunocompromised mice. We demonstrate that implants survived, retained human vasculature and anastomosed with the circulatory system of the recipient mouse. Significantly, by performing transplants into the ear pinna, this system enabled intravital observation of xenografts by multiphoton microscopy, allowing us to visualise the steps leading to vascular cytoadherence of erythrocytes infected with the human parasite Plasmodium falciparum. This model represents a useful tool for imaging the interactions that occur within human tissues in vivo and permits visualization of blood flow and cellular recruitment in a system which is amenable to intervention for various studies in basic biology together with drug evaluation and mechanism of action studies.
Collapse
Affiliation(s)
- Gavin R Meehan
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Hannah E Scales
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Rowland Osii
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Mariana De Niz
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
- Instituto de Medicina Molecular, University of Lisbon, Lisbon, Portugal
| | - Jennifer C Lawton
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Matthias Marti
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Paul Garside
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK
| | - Alister Craig
- Liverpool School of Tropical Medicine, Liverpool, UK
| | - James M Brewer
- Institute of Infection, Immunity & Inflammation, University of Glasgow, Scotland, UK.
| |
Collapse
|
34
|
Riggle BA, Miller LH, Pierce SK. Desperately Seeking Therapies for Cerebral Malaria. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:327-334. [PMID: 31907275 PMCID: PMC6951433 DOI: 10.4049/jimmunol.1900829] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/10/2019] [Indexed: 02/07/2023]
Abstract
Malaria is a deadly infectious disease caused by parasites of the Plasmodium spp. that takes an estimated 435,000 lives each year, primarily among young African children. For most children, malaria is a febrile illness that resolves with time, but in ∼1% of cases, for reasons we do not understand, malaria becomes severe and life threatening. Cerebral malaria (CM) is the most common form of severe malaria, accounting for the vast majority of childhood deaths from malaria despite highly effective antiparasite chemotherapy. Thus, CM is one of the most prevalent lethal brain diseases, and one for which we have no effective therapy. CM is, in part, an immune-mediated disease, and to fully understand CM, it is essential to appreciate the complex relationship between the malarial parasite and the human immune system. In this study, we provide a primer on malaria for immunologists and, in this context, review progress identifying targets for therapeutic intervention.
Collapse
Affiliation(s)
- Brittany A Riggle
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852; and
| | - Louis H Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852
| | - Susan K Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852; and
| |
Collapse
|
35
|
Darling TK, Mimche PN, Bray C, Umaru B, Brady LM, Stone C, Eboumbou Moukoko CE, Lane TE, Ayong LS, Lamb TJ. EphA2 contributes to disruption of the blood-brain barrier in cerebral malaria. PLoS Pathog 2020; 16:e1008261. [PMID: 31999807 PMCID: PMC6991964 DOI: 10.1371/journal.ppat.1008261] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 12/07/2019] [Indexed: 01/01/2023] Open
Abstract
Disruption of blood-brain barrier (BBB) function is a key feature of cerebral malaria. Increased barrier permeability occurs due to disassembly of tight and adherens junctions between endothelial cells, yet the mechanisms governing junction disassembly and vascular permeability during cerebral malaria remain poorly characterized. We found that EphA2 is a principal receptor tyrosine kinase mediating BBB breakdown during Plasmodium infection. Upregulated on brain microvascular endothelial cells in response to inflammatory cytokines, EphA2 is required for the loss of junction proteins on mouse and human brain microvascular endothelial cells. Furthermore, EphA2 is necessary for CD8+ T cell brain infiltration and subsequent BBB breakdown in a mouse model of cerebral malaria. Blocking EphA2 protects against BBB breakdown highlighting EphA2 as a potential therapeutic target for cerebral malaria.
Collapse
Affiliation(s)
- Thayer K. Darling
- Department of Pathology, University of Utah, Salt Lake City, UT, United States of America
- Department of Pediatric Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Patrice N. Mimche
- Department of Pathology, University of Utah, Salt Lake City, UT, United States of America
| | - Christian Bray
- Department of Pediatric Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Banlanjo Umaru
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroon
| | - Lauren M. Brady
- Department of Pediatric Infectious Diseases, Emory University School of Medicine, Atlanta, GA, United States of America
| | - Colleen Stone
- Department of Pathology, University of Utah, Salt Lake City, UT, United States of America
| | - Carole Else Eboumbou Moukoko
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroon
- Department of Biological Sciences, University of Douala, Douala, Cameroon
| | - Thomas E. Lane
- Department of Pathology, University of Utah, Salt Lake City, UT, United States of America
| | - Lawrence S. Ayong
- Malaria Research Unit, Centre Pasteur du Cameroun, Yaoundé, Cameroon
| | - Tracey J. Lamb
- Department of Pathology, University of Utah, Salt Lake City, UT, United States of America
| |
Collapse
|
36
|
Jensen AR, Adams Y, Hviid L. Cerebral Plasmodium falciparum malaria: The role of PfEMP1 in its pathogenesis and immunity, and PfEMP1-based vaccines to prevent it. Immunol Rev 2020; 293:230-252. [PMID: 31562653 PMCID: PMC6972667 DOI: 10.1111/imr.12807] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Revised: 09/04/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022]
Abstract
Malaria, a mosquito-borne infectious disease caused by parasites of the genus Plasmodium continues to be a major health problem worldwide. The unicellular Plasmodium-parasites have the unique capacity to infect and replicate within host erythrocytes. By expressing variant surface antigens Plasmodium falciparum has evolved to avoid protective immune responses; as a result in endemic areas anti-malaria immunity develops gradually over many years of multiple and repeated infections. We are studying the role of Plasmodium falciparum erythrocyte membrane protein 1 (PfEMP1) expressed by asexual stages of P. falciparum responsible for the pathogenicity of severe malaria. The immunopathology of falciparum malaria has been linked to cyto-adhesion of infected erythrocytes to specific host receptors. A greater appreciation of the PfEMP1 molecules important for the development of protective immunity and immunopathology is a prerequisite for the rational discovery and development of a safe and protective anti-disease malaria vaccine. Here we review the role of ICAM-1 and EPCR receptor adhering falciparum-parasites in the development of severe malaria; we discuss our current research to understand the factors involved in the pathogenesis of cerebral malaria and the feasibility of developing a vaccine targeted specifically to prevent this disease.
Collapse
Affiliation(s)
- Anja Ramstedt Jensen
- Centre for Medical Parasitology at Department of Immunology and MicrobiologyFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Yvonne Adams
- Centre for Medical Parasitology at Department of Immunology and MicrobiologyFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
| | - Lars Hviid
- Centre for Medical Parasitology at Department of Immunology and MicrobiologyFaculty of Health and Medical SciencesUniversity of CopenhagenCopenhagenDenmark
- Department of Infectious DiseasesRigshospitaletCopenhagenDenmark
| |
Collapse
|
37
|
Imaging the dynamic recruitment of monocytes to the blood-brain barrier and specific brain regions during Toxoplasma gondii infection. Proc Natl Acad Sci U S A 2019; 116:24796-24807. [PMID: 31727842 DOI: 10.1073/pnas.1915778116] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Brain infection by the parasite Toxoplasma gondii in mice is thought to generate vulnerability to predation by mechanisms that remain elusive. Monocytes play a key role in host defense and inflammation and are critical for controlling T. gondii However, the dynamic and regional relationship between brain-infiltrating monocytes and parasites is unknown. We report the mobilization of inflammatory (CCR2+Ly6Chi) and patrolling (CX3CR1+Ly6Clo) monocytes into the blood and brain during T. gondii infection of C57BL/6J and CCR2RFP/+CX3CR1GFP/+ mice. Longitudinal analysis of mice using 2-photon intravital imaging of the brain through cranial windows revealed that CCR2-RFP monocytes were recruited to the blood-brain barrier (BBB) within 2 wk of T. gondii infection, exhibited distinct rolling and crawling behavior, and accumulated within the vessel lumen before entering the parenchyma. Optical clearing of intact T. gondii-infected brains using iDISCO+ and light-sheet microscopy enabled global 3D detection of monocytes. Clusters of T. gondii and individual monocytes across the brain were identified using an automated cell segmentation pipeline, and monocytes were found to be significantly correlated with sites of T. gondii clusters. Computational alignment of brains to the Allen annotated reference atlas [E. S. Lein et al., Nature 445:168-176 (2007)] indicated a consistent pattern of monocyte infiltration during T. gondii infection to the olfactory tubercle, in contrast to LPS treatment of mice, which resulted in a diffuse distribution of monocytes across multiple brain regions. These data provide insights into the dynamics of monocyte recruitment to the BBB and the highly regionalized localization of monocytes in the brain during T. gondii CNS infection.
Collapse
|
38
|
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: 17] [Impact Index Per Article: 3.4] [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
|
39
|
Thiam A, Sanka M, Ndiaye Diallo R, Torres M, Mbengue B, Nunez NF, Thiam F, Diop G, Victorero G, Nguyen C, Dieye A, Rihet P. Gene expression profiling in blood from cerebral malaria patients and mild malaria patients living in Senegal. BMC Med Genomics 2019; 12:148. [PMID: 31666081 PMCID: PMC6821028 DOI: 10.1186/s12920-019-0599-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Accepted: 10/09/2019] [Indexed: 01/06/2023] Open
Abstract
Background Plasmodium falciparum malaria remains a major health problem in Africa. The mechanisms of pathogenesis are not fully understood. Transcriptomic studies may provide new insights into molecular pathways involved in the severe form of the disease. Methods Blood transcriptional levels were assessed in patients with cerebral malaria, non-cerebral malaria, or mild malaria by using microarray technology to look for gene expression profiles associated with clinical status. Multi-way ANOVA was used to extract differentially expressed genes. Network and pathways analyses were used to detect enrichment for biological pathways. Results We identified a set of 443 genes that were differentially expressed in the three patient groups after applying a false discovery rate of 10%. Since the cerebral patients displayed a particular transcriptional pattern, we focused our analysis on the differences between cerebral malaria patients and mild malaria patients. We further found 842 differentially expressed genes after applying a false discovery rate of 10%. Unsupervised hierarchical clustering of cerebral malaria-informative genes led to clustering of the cerebral malaria patients. The support vector machine method allowed us to correctly classify five out of six cerebral malaria patients and six of six mild malaria patients. Furthermore, the products of the differentially expressed genes were mapped onto a human protein-protein network. This led to the identification of the proteins with the highest number of interactions, including GSK3B, RELA, and APP. The enrichment analysis of the gene functional annotation indicates that genes involved in immune signalling pathways play a role in the occurrence of cerebral malaria. These include BCR-, TCR-, TLR-, cytokine-, FcεRI-, and FCGR- signalling pathways and natural killer cell cytotoxicity pathways, which are involved in the activation of immune cells. In addition, our results revealed an enrichment of genes involved in Alzheimer’s disease. Conclusions In the present study, we examine a set of genes whose expression differed in cerebral malaria patients and mild malaria patients. Moreover, our results provide new insights into the potential effect of the dysregulation of gene expression in immune pathways. Host genetic variation may partly explain such alteration of gene expression. Further studies are required to investigate this in African populations.
Collapse
Affiliation(s)
- Alassane Thiam
- Unité d'Immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal
| | - Michel Sanka
- Aix Marseille Univ, INSERM, TAGC UMR U1090, 163 Av de Luminy, 13288, Marseille, cedex 9, France
| | - Rokhaya Ndiaye Diallo
- Service de Génétique Humaine, Faculté de Médecine, de Pharmacie et d'Odontostomatologie, UCAD, Dakar, Sénégal
| | - Magali Torres
- Aix Marseille Univ, INSERM, TAGC UMR U1090, 163 Av de Luminy, 13288, Marseille, cedex 9, France
| | - Babacar Mbengue
- Service Immunologie, Faculte de Medecine, Université Cheikh Anta Diop de Dakar, Dakar, Sénégal
| | - Nicolas Fernandez Nunez
- Aix Marseille Univ, INSERM, TAGC UMR U1090, 163 Av de Luminy, 13288, Marseille, cedex 9, France
| | - Fatou Thiam
- Département de Génie chimique et biologie, Ecole Supérieure Polytechnique, Université Cheikh Anta Diop de Dakar, Dakar, Sénégal
| | - Gora Diop
- Unité d'Immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal.,Département de Biologie animale, Faculté des Sciences et Techniques, Université Cheikh Anta Diop de Dakar, Dakar, Sénégal
| | - Geneviève Victorero
- Aix Marseille Univ, INSERM, TAGC UMR U1090, 163 Av de Luminy, 13288, Marseille, cedex 9, France
| | - Catherine Nguyen
- Aix Marseille Univ, INSERM, TAGC UMR U1090, 163 Av de Luminy, 13288, Marseille, cedex 9, France
| | - Alioune Dieye
- Unité d'Immunogénétique, Institut Pasteur de Dakar, Dakar, Sénégal.,Service Immunologie, Faculte de Medecine, Université Cheikh Anta Diop de Dakar, Dakar, Sénégal
| | - Pascal Rihet
- Aix Marseille Univ, INSERM, TAGC UMR U1090, 163 Av de Luminy, 13288, Marseille, cedex 9, France.
| |
Collapse
|
40
|
Barrera V, Haley MJ, Strangward P, Attree E, Kamiza S, Seydel KB, Taylor TE, Milner DA, Craig AG, Couper KN. Comparison of CD8 + T Cell Accumulation in the Brain During Human and Murine Cerebral Malaria. Front Immunol 2019; 10:1747. [PMID: 31396236 PMCID: PMC6668485 DOI: 10.3389/fimmu.2019.01747] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Accepted: 07/10/2019] [Indexed: 01/06/2023] Open
Abstract
CD8+ T cells have been shown to play a critical role in the pathogenesis of experimental cerebral malaria (ECM) in mice, but their role in development of human cerebral malaria (HCM) remains unclear. Thus, in this study we have provided the first direct contrast of the accumulation of CD8+ T cells in the brain during HCM and ECM. HCM cases were from children who died of Plasmodium falciparum cerebral malaria at Queen Elizabeth Central Hospital (Malawi) between 2003 and 2010. ECM was induced by infecting C57BL/6J mice with P. berghei ANKA. We demonstrate similarities in the intracerebral CD8+ T cell responses in ECM and HCM, in particular an apparent shared choroid plexus-meningeal route of CD8+ T cell accumulation in the brain. Nevertheless, we also reveal some potentially important differences in compartmentalization of CD8+ T cells within the cerebrovascular bed in HCM and ECM.
Collapse
Affiliation(s)
- Valentina Barrera
- Department of Eye and Vision Science, University of Liverpool, Liverpool, United Kingdom
| | - Michael J Haley
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Patrick Strangward
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Elizabeth Attree
- Department of Eye and Vision Science, University of Liverpool, Liverpool, United Kingdom
| | - Steve Kamiza
- Department of Histopathology, College of Medicine, University of Malawi, Blantyre, Malawi
| | - Karl B Seydel
- Department of Osteopathic Medical Specialties, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States.,Blantyre Malaria Project, University of Malawi College of Medicine, Blantyre, Malawi
| | - Terrie E Taylor
- Department of Osteopathic Medical Specialties, College of Osteopathic Medicine, Michigan State University, East Lansing, MI, United States.,Blantyre Malaria Project, University of Malawi College of Medicine, Blantyre, Malawi
| | - Danny A Milner
- Center for Global Health, American Society for Clinical Pathology, Chicago, IL, United States
| | - Alister G Craig
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Kevin N Couper
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
41
|
Blood-Brain Barrier in Cerebral Malaria: Pathogenesis and Therapeutic Intervention. Trends Parasitol 2019; 35:516-528. [PMID: 31147271 DOI: 10.1016/j.pt.2019.04.010] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2019] [Revised: 04/25/2019] [Accepted: 04/25/2019] [Indexed: 02/06/2023]
Abstract
Cerebral malaria is a life-threatening complication of malaria caused by the parasite Plasmodium falciparum. The growing problem of drug resistance and the dearth of new antiparasitic drugs are a serious threat to the antimalaria treatment regimes. Studies on humans and the murine model have implicated the disruption of the blood-brain barrier (BBB) in the lethal course of the disease. Therefore, efforts to alleviate the BBB dysfunction could serve as an adjunct therapy. Here, we review the mechanisms associated with the disruption of the BBB. In addition, we discuss the current, still limited, knowledge on the contribution of different cell types, microparticles, and the kynurenine pathway in the regulation of BBB dysfunction, and how these molecules could be used as potential new therapeutic targets.
Collapse
|
42
|
Lv L, Hai L, Wang Q, Shi X. Experimental Cerebral Malaria Alters Blood Lipid Levels during Pathogenesis. J Parasitol 2019. [DOI: 10.1645/18-162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Li Lv
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Lei Hai
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin 3000
| | - Qian Wang
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin 3000
| | - Xiaoyu Shi
- Department of Immunology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin Key Laboratory of Cellular and Molecular Immunology, Key Laboratory of Immune Microenvironment and Diseases of Educational Ministry of China, Tianjin 3000
| |
Collapse
|
43
|
TRPV1 Contributes to Cerebral Malaria Severity and Mortality by Regulating Brain Inflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:9451671. [PMID: 31223430 PMCID: PMC6541938 DOI: 10.1155/2019/9451671] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2018] [Revised: 04/17/2019] [Accepted: 05/05/2019] [Indexed: 02/08/2023]
Abstract
Transient receptor potential vanilloid 1 (TRPV1) is a Ca+2-permeable channel expressed on neuronal and nonneuronal cells, known as an oxidative stress sensor. It plays a protective role in bacterial infection, and recent findings indicate that this receptor modulates monocyte populations in mice with malaria; however, its role in cerebral malaria progression and outcome is unclear. By using TRPV1 wild-type (WT) and knockout (KO) mice, the importance of TRPV1 to this cerebral syndrome was investigated. Infection with Plasmodium berghei ANKA decreased TRPV1 expression in the brain. Mice lacking TRPV1 were protected against Plasmodium-induced mortality and morbidity, a response that was associated with less cerebral swelling, modulation of the brain expression of endothelial tight-junction markers (junctional adhesion molecule A and claudin-5), increased oxidative stress (via inhibition of catalase activity and increased levels of H2O2, nitrotyrosine, and carbonyl residues), and diminished production of cytokines. Plasmodium load was not significantly affected by TRPV1 ablation. Repeated subcutaneous administration of the selective TRPV1 antagonist SB366791 after malaria induction increased TRPV1 expression in the brain tissue and enhanced mouse survival. These data indicate that TRPV1 channels contribute to the development and outcome of cerebral malaria.
Collapse
|
44
|
Sierro F, Grau GER. The Ins and Outs of Cerebral Malaria Pathogenesis: Immunopathology, Extracellular Vesicles, Immunometabolism, and Trained Immunity. Front Immunol 2019; 10:830. [PMID: 31057552 PMCID: PMC6478768 DOI: 10.3389/fimmu.2019.00830] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 03/28/2019] [Indexed: 12/16/2022] Open
Abstract
Complications from malaria parasite infections still cost the lives of close to half a million people every year. The most severe is cerebral malaria (CM). Employing murine models of CM, autopsy results, in vitro experiments, neuroimaging and microscopic techniques, decades of research activity have investigated the development of CM immunopathology in the hope of identifying steps that could be therapeutically targeted. Yet important questions remain. This review summarizes recent findings, primarily mechanistic insights on the essential cellular and molecular players involved gained within the murine experimental cerebral malaria model. It also highlights recent developments in (a) cell-cell communication events mediated through extracellular vesicles (EVs), (b) mounting evidence for innate immune memory, leading to “trained“ increased or tolerised responses, and (c) modulation of immune cell function through metabolism, that could shed light on why some patients develop this life-threatening condition whilst many do not.
Collapse
Affiliation(s)
- Frederic Sierro
- Vascular Immunology Unit, Department of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia.,Human Health, Nuclear Science, Technology, and Landmark Infrastructure, Australian Nuclear Science and Technology Organisation, Sydney, NSW, Australia
| | - Georges E R Grau
- Vascular Immunology Unit, Department of Pathology, Faculty of Medicine and Health, School of Medical Sciences, The University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
45
|
De Niz M, Nacer A, Frischknecht F. Intravital microscopy: Imaging host-parasite interactions in the brain. Cell Microbiol 2019; 21:e13024. [PMID: 30830993 DOI: 10.1111/cmi.13024] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2018] [Revised: 02/14/2019] [Accepted: 02/24/2019] [Indexed: 12/31/2022]
Abstract
Intravital fluorescence microscopy (IVM) is a powerful technique for imaging multiple organs, including the brain of living mice and rats. It enables the direct visualisation of cells in situ providing a real-life view of biological processes that in vitro systems cannot. In addition, to the technological advances in microscopy over the last decade, there have been supporting innovations in data storage and analytical packages that enable the visualisation and analysis of large data sets. Here, we review the advantages and limitations of techniques predominantly used for brain IVM, including thinned skull windows, open skull cortical windows, and a miniaturised optical system based on microendoscopic probes that can be inserted into deep tissues. Further, we explore the relevance of these techniques for the field of parasitology. Several protozoan infections are associated with neurological symptoms including Plasmodium spp., Toxoplasma spp., and Trypanosoma spp. IVM has led to crucial findings on these parasite species, which are discussed in detail in this review.
Collapse
Affiliation(s)
- Mariana De Niz
- Institute of Cell Biology, University of Bern, Bern, Switzerland.,Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasglow, UK
| | - Adéla Nacer
- Division of Bacteriology, National Institute for Biological Standards and Control, Medicines and Healthcare products Regulatory Agency, EN63QG, Potters Bar, UK
| | - Friedrich Frischknecht
- Parasitology-Centre for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| |
Collapse
|
46
|
Abstract
Malaria is a causative factor in about 500.000 deaths each year world-wide. Cerebral malaria is a particularly severe complication of this disease and thus associated with an exceedingly high mortality. Malaria retinopathy is an ocular manifestation often associated with cerebral malaria, and presumably shares a substantial part of its pathophysiology. Here, we describe that indeed murine malaria retinopathy reproduced the main hallmarks of the corresponding human disease. In the living animal, we were able to follow the circulation and cellular localization of malaria parasites transgenically labelled with GFP via non-invasive in vivo retinal imaging. We found that malaria parasites cross the blood-retinal-barrier and infiltrate the neuroretina, concomitant with an extensive, irreversible, and long-lasting retinal neurodegeneration. Furthermore, anti-malarial treatment with dihydroartemisinin strongly diminished the load of circulating parasites but resolved the symptoms of the retinopathy only in part. In summary, we introduce here a novel preclinical model for human cerebral malaria that is much more directly accessible for studies into disease pathophysiology and development of novel treatment approaches. In vivo retinal imaging may furthermore serve as a valuable tool for the early diagnosis of the human disease.
Collapse
|
47
|
Shaw TN, Inkson CA, Villegas-Mendez A, Pattinson DJ, Strangward P, Else KJ, Draper SJ, Zeef LAH, Couper KN. Infection-Induced Resistance to Experimental Cerebral Malaria Is Dependent Upon Secreted Antibody-Mediated Inhibition of Pathogenic CD8 + T Cell Responses. Front Immunol 2019; 10:248. [PMID: 30846985 PMCID: PMC6394254 DOI: 10.3389/fimmu.2019.00248] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Accepted: 01/29/2019] [Indexed: 12/27/2022] Open
Abstract
Cerebral malaria (CM) is one of the most severe complications of Plasmodium falciparum infection. There is evidence that repeated parasite exposure promotes resistance against CM. However, the immunological basis of this infection-induced resistance remains poorly understood. Here, utilizing the Plasmodium berghei ANKA (PbA) model of experimental cerebral malaria (ECM), we show that three rounds of infection and drug-cure protects against the development of ECM during a subsequent fourth (4X) infection. Exposure-induced resistance was associated with specific suppression of CD8+ T cell activation and CTL-related pathways, which corresponded with the development of heterogeneous atypical B cell populations as well as the gradual infection-induced generation and maintenance of high levels of anti-parasite IgG. Mechanistically, transfer of high-titer anti-parasite IgG did not protect 1X infected mice against ECM and depletion of atypical and regulatory B cells during 4X infection failed to abrogate infection-induced resistance to ECM. However, IgMi mice that were unable to produce secreted antibody, or undergo class switching, during the repeated rounds of infection failed to develop resistance against ECM. The failure of infection-induced protection in IgMi mice was associated with impaired development of atypical B cell populations and the inability to suppress pathogenic CD8+ T cell responses. Our results, therefore, suggest the importance of anti-parasite antibody responses, gradually acquired, and maintained through repeated Plasmodium infections, for modulating the B cell compartment and eventually suppressing memory CD8+ T cell reactivation to establish infection-induced resistance to ECM.
Collapse
Affiliation(s)
- Tovah N. Shaw
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
- Manchester Collaborative Centre for Inflammation Research, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Colette A. Inkson
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Ana Villegas-Mendez
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | | | - Patrick Strangward
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Kathryn J. Else
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| | - Simon J. Draper
- The Jenner Institute, University of Oxford, Oxford, United Kingdom
| | - Leo A. H. Zeef
- Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Kevin N. Couper
- Faculty of Biology, Medicine and Health, The Lydia Becker Institute of Immunology and Inflammation, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
48
|
Pais TF, Penha-Gonçalves C. Brain Endothelium: The "Innate Immunity Response Hypothesis" in Cerebral Malaria Pathogenesis. Front Immunol 2019; 9:3100. [PMID: 30761156 PMCID: PMC6361776 DOI: 10.3389/fimmu.2018.03100] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Accepted: 12/14/2018] [Indexed: 01/04/2023] Open
Abstract
Cerebral malaria (CM) is a life-threatening neurological syndrome caused by Plasmodium falciparum infection afflicting mainly children in Africa. Current pathogenesis models implicate parasite and host-derived factors in impairing brain vascular endothelium (BVE) integrity. Sequestration of Plasmodium-infected red blood cells (iRBCs) in brain microvessels is a hallmark of CM pathology. However, the precise mechanisms driving loss of blood-brain barrier (BBB) function with consequent brain injury are still unsettled and it is plausible that distinct pathophysiology trajectories are involved. Studies in humans and in the mouse model of CM indicate that inflammatory reactions intertwined with microcirculatory and coagulation disturbances induce alterations in vascular permeability and impair BBB integrity. Yet, the role of BVE as initiator of immune responses against parasite molecules and iRBCs is largely unexplored. Brain endothelial cells express pattern recognition receptors (PRR) and are privileged sensors of blood-borne infections. Here, we focus on the hypothesis that innate responses initiated by BVE and subsequent interactions with immune cells are critical to trigger local effector immune functions and induce BBB damage. Uncovering mechanisms of BVE involvement in sensing Plasmodium infection, recruiting of immune cells and directing immune effector functions could reveal pharmacological targets to promote BBB protection with potential applications in CM clinical management.
Collapse
|
49
|
Remer I, Pierre-Destine LF, Tay D, Golightly LM, Bilenca A. In vivo noninvasive visualization of retinal perfusion dysfunction in murine cerebral malaria by camera-phone laser speckle imaging. JOURNAL OF BIOPHOTONICS 2019; 12:e201800098. [PMID: 29900690 DOI: 10.1002/jbio.201800098] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 06/08/2018] [Accepted: 06/11/2018] [Indexed: 05/12/2023]
Abstract
Cerebral malaria (CM) is a severe complication of Plasmodium falciparum infection associated with impaired cerebral blood flow. Visualization of the eye vasculature, which is embryologically derived from that of the brain, is used clinically to diagnose the syndrome. Here, we introduce camera-phone laser speckle imaging as a new tool for in vivo, noncontact two-dimensional mapping of blood flow dynamics in the experimental cerebral malaria (ECM) murine model of Plasmodium berghei ANKA. In a longitudinal study, we show that the camera-phone imager can detect an overall decrease in the retinal blood-flow-speed (BFS) as ECM develops in P. berghei ANKA infected mice, with no similar change observed in uninfected control mice or mice infected with a non-ECM inducing strain (P. berghei NK65). Furthermore, by analyzing relative alterations in the BFS of individual retinal vessels during the progression of ECM, we illustrate the strength of our imager in identifying different BFS-change heterogeneities in the retinas of ECM and uninfected mice. The technique creates new possibilities for objective investigations into the diagnosis and pathogenesis of CM noninvasively through the eye. The camera-phone laser speckle imager along with measured spatial blood perfusion maps of the retina of a mouse infected with P. berghei ANKA-a fatal ECM model-on different days during the progression of the infection (top, day 3 after infection; middle, day 5 after infection; and bottom, day 7 after infection).
Collapse
Affiliation(s)
- Itay Remer
- Biomedical Engineering Department, Ben-Gurion University of the Negev, Beersheba, Israel
| | | | - David Tay
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Linnie M Golightly
- Division of Infectious Diseases, Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Alberto Bilenca
- Biomedical Engineering Department, Ben-Gurion University of the Negev, Beersheba, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, Beersheba, Israel
| |
Collapse
|
50
|
Saroa R, Bagai U. Evaluation of T cells infiltration inhibition in brain by immunohistochemistry during experimental cerebral malaria. J Parasit Dis 2018; 42:537-549. [PMID: 30538351 DOI: 10.1007/s12639-018-1030-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Accepted: 09/07/2018] [Indexed: 11/25/2022] Open
Abstract
Plasmodium berghei ANKA is known to be responsible for causing neurological complications in susceptible strain of mice. Despite the decades of research, pathogenesis of cerebral malaria is still unknown. Histopathological and immunofluorescent staining was performed on brain of P. berghei ANKA infected and artesunate-sulphadoxine-pyrimethamine (AS + SP) treated mice to understand the pathogenesis of experimental cerebral malaria in present study. Cerebral vessels were found to be congested with infected/non-infected RBCs and various leukocytes in infected mice. Immunofluorescent staining identified the localisation of CD3+, CD4+ and CD8+ T cells in brain of infected and treated mice. P. berghei infected mice exhibited higher expression of CD3+, CD4+ and CD8+ T cells in brain cortex as compared to mice treated with artesunate-sulphadoxine-pyrimethamine. No sign of injury was observed in brain of treated mice in histopathological analysis. All treated mice survived up to 1 month, whereas, all infected mice died by D15 post infection due to neuro-inflammation.
Collapse
Affiliation(s)
- Ruchika Saroa
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, 160014 India
| | - Upma Bagai
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, 160014 India
| |
Collapse
|