1
|
Silveira ELVD, Rai U, Bonezi V, Zárate-Bladés CR, Claser C. CCR6 expression reduces mouse survival upon malarial challenge with Plasmodium berghei NK65 strain. Mem Inst Oswaldo Cruz 2022; 117:e210287. [PMID: 35730803 PMCID: PMC9208320 DOI: 10.1590/0074-02760210287] [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: 08/23/2021] [Accepted: 04/26/2022] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND It has been demonstrated that proteins expressed by liver-stage Plasmodium parasites can inhibit the translocation of transcription factors to the nucleus of different cells. This process would hinder the expression of immune genes, such as the CCL20 chemokine. OBJECTIVE Since CCR6 is the only cognate receptor for CCL20, we investigated the importance of this chemokine-receptor axis against rodent malaria. METHODS CCR6-deficient (KO) and wild-type (WT) C57BL/6 mice were challenged with Plasmodium berghei (Pb) NK65 sporozoites or infected red blood cells (iRBCs). Liver parasitic cDNA, parasitemia and serum cytokine concentrations were respectively evaluated through reverse transcription-polymerase chain reaction (RT-PCR), staining thin-blood smears with Giemsa solution, and enzyme-linked immunosorbent assay (ELISA). FINDINGS Although the sporozoite challenges yielded similar liver parasitic cDNA and parasitemia, KO mice presented a prolonged survival than WT mice. After iRBC challenges, KO mice kept displaying higher survival rates as well as a decreased IL-12 p70 concentration in the serum than WT mice. CONCLUSION Our data suggest that malaria triggered by PbNK65 liver- or blood-stage forms elicit a pro-inflammatory environment that culminates with a decreased survival of infected C57BL/6 mice.
Collapse
Affiliation(s)
- Eduardo Lani Volpe da Silveira
- New York University School of Medicine, Department of Pathology, Michael Heidelberg Division of Immunology, New York, NY, United States of America
| | - Urvashi Rai
- New York University School of Medicine, Department of Pathology, Michael Heidelberg Division of Immunology, New York, NY, United States of America
| | - Vivian Bonezi
- Universidade de São Paulo, Faculdade de Ciências Farmacêuticas, Departamento de Análises Clínicas e Toxicológicas, Laboratório de Imunologia de Células B, São Paulo, SP, Brasil
| | - Carlos Rodrigo Zárate-Bladés
- Universidade Federal de Santa Catarina, Departamento de Microbiologia, Imunologia e Parasitologia, Laboratório de Imunorregulação, Florianópolis, SC, Brasil
| | - Carla Claser
- Universidade de São Paulo, Instituto de Ciências Biomédicas, Departamento de Parasitologia, São Paulo, SP, Brasil
| |
Collapse
|
2
|
Pinto BF, Ribeiro LNB, da Silva GBRF, Freitas CS, Kraemer L, Oliveira FMS, Clímaco MC, Mourão FAG, Santos GSPD, Béla SR, Gurgel ILDS, Leite FDL, de Oliveira AG, Vilela MRSDP, Oliveira-Lima OC, Soriani FM, Fujiwara RT, Birbrair A, Russo RC, Carvalho-Tavares J. Inhalation of dimethyl fumarate-encapsulated solid lipid nanoparticles attenuate clinical signs of experimental autoimmune encephalomyelitis and pulmonary inflammatory dysfunction in mice. Clin Sci (Lond) 2022; 136:81-101. [PMID: 34904644 DOI: 10.1042/cs20210792] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 12/09/2021] [Accepted: 12/14/2021] [Indexed: 11/17/2022]
Abstract
RATIONALE The FDA-approved Dimethyl Fumarate (DMF) as an oral drug for Multiple Sclerosis (MS) treatment based on its immunomodulatory activities. However, it also caused severe adverse effects mainly related to the gastrointestinal system. OBJECTIVE Investigated the potential effects of solid lipid nanoparticles (SLNs) containing DMF, administered by inhalation on the clinical signs, central nervous system (CNS) inflammatory response, and lung function changes in mice with experimental autoimmune encephalomyelitis (EAE). MATERIALS AND METHODS EAE was induced using MOG35-55 peptide in female C57BL/6J mice and the mice were treated via inhalation with DMF-encapsulated SLN (CTRL/SLN/DMF and EAE/SLN/DMF), empty SLN (CTRL/SLN and EAE/SLN), or saline solution (CTRL/saline and EAE/saline), every 72 h during 21 days. RESULTS After 21 days post-induction, EAE mice treated with DMF-loaded SLN, when compared with EAE/saline and EAE/SLN, showed decreased clinical score and weight loss, reduction in brain and spinal cord injury and inflammation, also related to the increased influx of Foxp3+ cells into the spinal cord and lung tissues. Moreover, our data revealed that EAE mice showed signs of respiratory disease, marked by increased vascular permeability, leukocyte influx, production of TNF-α and IL-17, perivascular and peribronchial inflammation, with pulmonary mechanical dysfunction associated with loss of respiratory volumes and elasticity, which DMF-encapsulated reverted in SLN nebulization. CONCLUSION Our study suggests that inhalation of DMF-encapsulated SLN is an effective therapeutic protocol that reduces not only the CNS inflammatory process and disability progression, characteristic of EAE disease, but also protects mice from lung inflammation and pulmonary dysfunction.
Collapse
Affiliation(s)
- Bárbara Fernandes Pinto
- Neuroscience Group, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Lorena Natasha Brito Ribeiro
- Neuroscience Group, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Gisela Bevilacqua Rolfsen Ferreira da Silva
- Nanoneurobiophysics Research Group, Department of Physics, Chemistry and Mathematics, Federal University of São Carlos (UFSCAR), Sorocaba, São Paulo, Brazil
- State of São Paulo University (UNESP), Drugs and Medicines Department, School of Pharmaceutical Sciences, Araraquara, São Paulo, Brazil
| | - Camila Simões Freitas
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Lucas Kraemer
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Fabrício Marcus Silva Oliveira
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Marianna Carvalho Clímaco
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Flávio Afonso Gonçalves Mourão
- Neuroscience Group, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
- Center for Technology and Research in Magneto-Resonance (CTPMAG), Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | | | - Samantha Ribeiro Béla
- Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Isabella Luísa da Silva Gurgel
- Laboratory of Functional Genetics, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Fábio de Lima Leite
- Nanoneurobiophysics Research Group, Department of Physics, Chemistry and Mathematics, Federal University of São Carlos (UFSCAR), Sorocaba, São Paulo, Brazil
| | - Anselmo Gomes de Oliveira
- State of São Paulo University (UNESP), Drugs and Medicines Department, School of Pharmaceutical Sciences, Araraquara, São Paulo, Brazil
| | - Maura Regina Silva da Páscoa Vilela
- Neuroscience Group, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Onésia Cristina Oliveira-Lima
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Goiás (UFG), Goiânia, GO, Brazil
| | - Frederico Marianetti Soriani
- Laboratory of Functional Genetics, Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunology and Genomics of Parasites, Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Alexander Birbrair
- Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| | - Juliana Carvalho-Tavares
- Neuroscience Group, Department of Physiology and Biophysics, Institute of Biological Sciences, Federal University of Minas Gerais (UFMG), Belo Horizonte, MG, Brazil
| |
Collapse
|
3
|
Kaur M, Yardley V, Wang K, Masania J, Arroo RRJ, Turner DB, Li M. Artemisinin Cocrystals for Bioavailability Enhancement. Part 2: In Vivo Bioavailability and Physiologically Based Pharmacokinetic Modeling. Mol Pharm 2021; 18:4272-4289. [PMID: 34748332 DOI: 10.1021/acs.molpharmaceut.1c00385] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
We report the evaluation and prediction of the pharmacokinetic (PK) performance of artemisinin (ART) cocrystal formulations, that is, 1:1 artemisinin/orcinol (ART-ORC) and 2:1 artemisinin/resorcinol (ART2-RES), using in vivo murine animal and physiologically based pharmacokinetic (PBPK) models. The efficacy of the ART cocrystal formulations along with the parent drug ART was tested in mice infected with Plasmodium berghei. When given at the same dose, the ART cocrystal formulation showed a significant reduction in parasitaemia at day 4 after infection compared to ART alone. PK parameters including Cmax (maximum plasma concentration), Tmax (time to Cmax), and AUC (area under the curve) were obtained by determining drug concentrations in the plasma using liquid chromatography-high-resolution mass spectrometry (LC-HRMS), showing enhanced ART levels after dosage with the cocrystal formulations. The dose-response tests revealed that a significantly lower dose of the ART cocrystals in the formulation was required to achieve a similar therapeutic effect as ART alone. A PBPK model was developed using a PBPK mouse simulator to accurately predict the in vivo behavior of the cocrystal formulations by combining in vitro dissolution profiles with the properties of the parent drug ART. The study illustrated that information from classical in vitro and in vivo experimental investigations of the parent drug of ART formulations can be coupled with PBPK modeling to predict the PK parameters of an ART cocrystal formulation in an efficient manner. Therefore, the proposed modeling strategy could be used to establish in vitro and in vivo correlations for different cocrystals intended to improve dissolution properties and to support clinical candidate selection, contributing to the assessment of cocrystal developability and formulation development.
Collapse
Affiliation(s)
- Manreet Kaur
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - Vanessa Yardley
- Department of Infection & Immunity, Faculty of Infectious & Tropical Diseases, London School of Hygiene and Tropical Medicine, London WC1E 7HT, U.K
| | - Ke Wang
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - Jinit Masania
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - Randolph R J Arroo
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| | - David B Turner
- Certara UK Limited, Simcyp Division, Sheffield S1 2BJ, U.K
| | - Mingzhong Li
- Leicester School of Pharmacy, De Montfort University, Leicester LE1 9BH, U.K
| |
Collapse
|
4
|
Vieira-Santos F, Leal-Silva T, de Lima Silva Padrão L, Ruas ACL, Nogueira DS, Kraemer L, Oliveira FMS, Caliari MV, Russo RC, Fujiwara RT, Bueno LL. Concomitant experimental coinfection by Plasmodium berghei NK65-NY and Ascaris suum downregulates the Ascaris-specific immune response and potentiates Ascaris-associated lung pathology. Malar J 2021; 20:296. [PMID: 34210332 PMCID: PMC8248286 DOI: 10.1186/s12936-021-03824-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Accepted: 06/17/2021] [Indexed: 12/22/2022] Open
Abstract
Background Ascariasis and malaria are highly prevalent parasitic diseases in tropical regions and often have overlapping endemic areas, contributing to high morbidity and mortality rates in areas with poor sanitary conditions. Several studies have previously aimed to correlate the effects of Ascaris-Plasmodium coinfections but have obtained contradictory and inconclusive results. Therefore, the present study aimed to investigate parasitological and immunopathological aspects of the lung during murine experimental concomitant coinfection by Plasmodium berghei and Ascaris suum during larvae ascariasis. Methods C57BL/6J mice were inoculated with 1 × 104P. berghei strain NK65-NY-infected red blood cells (iRBCs) intraperitoneally and/or 2500 embryonated eggs of A. suum by oral gavage. P. berghei parasitaemia, morbidity and the survival rate were assessed. On the seventh day postinfection (dpi), A. suum lung burden analysis; bronchoalveolar lavage (BAL); histopathology; NAG, MPO and EPO activity measurements; haematological analysis; and respiratory mechanics analysis were performed. The concentrations of interleukin (IL)-1β, IL-12/IL-23p40, IL-6, IL-4, IL-33, IL-13, IL-5, IL-10, IL-17A, IFN-γ, TNF and TGF-β were assayed by sandwich ELISA. Results Animals coinfected with P. berghei and A. suum show decreased production of type 1, 2, and 17 and regulatory cytokines; low leukocyte recruitment in the tissue; increased cellularity in the circulation; and low levels of NAG, MPO and EPO activity that lead to an increase in larvae migration, as shown by the decrease in larvae recovered in the lung parenchyma and increase in larvae recovered in the airway. This situation leads to severe airway haemorrhage and, consequently, an impairment respiratory function that leads to high morbidity and early mortality. Conclusions This study demonstrates that the Ascaris-Plasmodium interaction is harmful to the host and suggests that this coinfection may potentiate Ascaris-associated pathology by dampening the Ascaris-specific immune response, resulting in the early death of affected animals. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03824-w.
Collapse
Affiliation(s)
- Flaviane Vieira-Santos
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Thaís Leal-Silva
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Luiza de Lima Silva Padrão
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ana Cristina Loiola Ruas
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Denise Silva Nogueira
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lucas Kraemer
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Fabrício Marcus Silva Oliveira
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Marcelo Vidigal Caliari
- Laboratory of Protozooses, Institute of Biological Sciences, Department of General Pathology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Remo Castro Russo
- Laboratory of Pulmonary Immunology and Mechanics, Institute of Biological Sciences, Department of Physiology and Biophysics, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Ricardo Toshio Fujiwara
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil
| | - Lilian Lacerda Bueno
- Laboratory of Immunology and Genomics of Parasites, Institute of Biological Sciences, Department of Parasitology, Universidade Federal de Minas Gerais, Belo Horizonte, Brazil.
| |
Collapse
|
5
|
Su XZ, Zhang C, Joy DA. Host-Malaria Parasite Interactions and Impacts on Mutual Evolution. Front Cell Infect Microbiol 2020; 10:587933. [PMID: 33194831 PMCID: PMC7652737 DOI: 10.3389/fcimb.2020.587933] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 09/22/2020] [Indexed: 12/22/2022] Open
Abstract
Malaria is the most deadly parasitic disease, affecting hundreds of millions of people worldwide. Malaria parasites have been associated with their hosts for millions of years. During the long history of host-parasite co-evolution, both parasites and hosts have applied pressure on each other through complex host-parasite molecular interactions. Whereas the hosts activate various immune mechanisms to remove parasites during an infection, the parasites attempt to evade host immunity by diversifying their genome and switching expression of targets of the host immune system. Human intervention to control the disease such as antimalarial drugs and vaccination can greatly alter parasite population dynamics and evolution, particularly the massive applications of antimalarial drugs in recent human history. Vaccination is likely the best method to prevent the disease; however, a partially protective vaccine may have unwanted consequences that require further investigation. Studies of host-parasite interactions and co-evolution will provide important information for designing safe and effective vaccines and for preventing drug resistance. In this essay, we will discuss some interesting molecules involved in host-parasite interactions, including important parasite antigens. We also discuss subjects relevant to drug and vaccine development and some approaches for studying host-parasite interactions.
Collapse
Affiliation(s)
- Xin-Zhuan Su
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Cui Zhang
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Deirdre A Joy
- Parasitology and International Programs Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
6
|
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
|
7
|
Jacqueline C, Bonnefoy N, Charrière GM, Thomas F, Roche B. Personal history of infections and immunotherapy: Unexpected links and possible therapeutic opportunities. Oncoimmunology 2018; 7:e1466019. [PMID: 30221066 PMCID: PMC6136881 DOI: 10.1080/2162402x.2018.1466019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 04/09/2018] [Accepted: 04/11/2018] [Indexed: 01/17/2023] Open
Abstract
The recent breakthroughs in the understanding of tumor immune biology have given rise to a new generation of immunotherapies, harnessing the immune system to eliminate tumors. As the typology and frequency of encountered infections are susceptible to shape the immune system, it could also impact the efficiency of immunotherapy. In this review, we report evidences for an indirect link between personal history of infection and different strategies of immunotherapy. In the current context of interest rise for personalized medicine, we discuss the potential medical applications of considering personal history of infection to design immunotherapeutic strategies.
Collapse
Affiliation(s)
- Camille Jacqueline
- Centre for Ecological and Evolutionary Research on Cancer (CREEC), Montpellier, France
- MIVEGEC, IRD, CNRS, Université Montpellier, Montpellier, France
| | - Nathalie Bonnefoy
- IRCM, INSERM, Université de Montpellier, ICM, F-34298, Montpellier, France
| | - Guillaume M. Charrière
- IHPE, UMR 5244, CNRS, Ifremer, Université de Perpignan Via Domitia, Université de Montpellier, Montpellier, 34095, France
| | - Frédéric Thomas
- Centre for Ecological and Evolutionary Research on Cancer (CREEC), Montpellier, France
- MIVEGEC, IRD, CNRS, Université Montpellier, Montpellier, France
| | - Benjamin Roche
- Centre for Ecological and Evolutionary Research on Cancer (CREEC), Montpellier, France
- UMMISCO, IRD/ Sorbonne Université, Bondy, France
- Departamento de Etología, Fauna Silvestre y Animales de Laboratorio, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Ciudad de México, México
| |
Collapse
|
8
|
Vandermosten L, Pham TT, Possemiers H, Knoops S, Van Herck E, Deckers J, Franke-Fayard B, Lamb TJ, Janse CJ, Opdenakker G, Van den Steen PE. Experimental malaria-associated acute respiratory distress syndrome is dependent on the parasite-host combination and coincides with normocyte invasion. Malar J 2018; 17:102. [PMID: 29506544 PMCID: PMC5839036 DOI: 10.1186/s12936-018-2251-3] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2017] [Accepted: 02/27/2018] [Indexed: 12/13/2022] Open
Abstract
Background Malaria-associated acute respiratory distress syndrome (MA-ARDS) is a complication of malaria with a lethality rate of up to 80% despite anti-malarial treatment. It is characterized by a vast infiltration of leukocytes, microhaemorrhages and vasogenic oedema in the lungs. Previously, a mouse model for MA-ARDS was developed by infection of C57BL/6 mice with the Edinburgh line NK65-E of Plasmodium berghei. Results Here, both host and parasite factors were demonstrated to play crucial roles in the development and severity of lung pathology. In particular, the genetic constitution of the host was an important determinant in the development of MA-ARDS. Both male and female C57BL/6, but not BALB/c, mice developed MA-ARDS when infected with P. berghei NK65-E. However, the New York line of P. berghei NK65 (NK65-NY) did not induce demonstrable MA-ARDS, despite its accumulation in the lungs and fat tissue to a similar or even higher extent as P. berghei NK65-E. These two commonly used lines of P. berghei differ in their red blood cell preference. P. berghei NK65-NY showed a stronger predilection for reticulocytes than P. berghei NK65-E and this appeared to be associated with a lower pathogenicity in the lungs. The pulmonary pathology in the C57BL/6/P. berghei NK65-E model was more pronounced than in the model with infection of DBA/2 mice with P. berghei strain ANKA. The transient lung pathology in DBA/2 mice infected with P. berghei ANKA coincided with the infection phase in which parasites mainly infected normocytes. This phase was followed by a less pathogenic phase in which P. berghei ANKA mainly infected reticulocytes. Conclusions The propensity of mice to develop MA-ARDS during P. berghei infection depends on both host and parasite factors and appears to correlate with RBC preference. These data provide insights in induction of MA-ARDS and may guide the choice of different mouse-parasite combinations to study lung pathology.
Collapse
Affiliation(s)
- Leen Vandermosten
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Thao-Thy Pham
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Hendrik Possemiers
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Sofie Knoops
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Evelien Van Herck
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Julie Deckers
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium.,Laboratory of Immunoregulation, VIB Center for Inflammation Research, Department of Internal Medicine, Ghent University, Technologiepark 927, 9052, Ghent, Belgium
| | - Blandine Franke-Fayard
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Tracey J Lamb
- Department of Pathology, University of Utah, 15 N Medical Drive E, Salt Lake City, UT, 84112, USA
| | - Chris J Janse
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center (LUMC), Albinusdreef 2, 2333 ZA, Leiden, The Netherlands
| | - Ghislain Opdenakker
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium
| | - Philippe E Van den Steen
- Laboratory of Immunobiology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven-University of Leuven, Herestraat 49, box 1044, 3000, Leuven, Belgium.
| |
Collapse
|
9
|
Nair SC, Xu R, Pattaradilokrat S, Wu J, Qi Y, Zilversmit M, Ganesan S, Nagarajan V, Eastman RT, Orandle MS, Tan JC, Myers TG, Liu S, Long CA, Li J, Su XZ. A Plasmodium yoelii HECT-like E3 ubiquitin ligase regulates parasite growth and virulence. Nat Commun 2017; 8:223. [PMID: 28790316 PMCID: PMC5548792 DOI: 10.1038/s41467-017-00267-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 06/12/2017] [Indexed: 01/18/2023] Open
Abstract
Infection of mice with strains of Plasmodium yoelii parasites can result in different pathology, but molecular mechanisms to explain this variation are unclear. Here we show that a P. yoelii gene encoding a HECT-like E3 ubiquitin ligase (Pyheul) influences parasitemia and host mortality. We genetically cross two lethal parasites with distinct disease phenotypes, and identify 43 genetically diverse progeny by typing with microsatellites and 9230 single-nucleotide polymorphisms. A genome-wide quantitative trait loci scan links parasite growth and host mortality to two major loci on chromosomes 1 and 7 with LOD (logarithm of the odds) scores = 6.1 and 8.1, respectively. Allelic exchange of partial sequences of Pyheul in the chromosome 7 locus and modification of the gene expression alter parasite growth and host mortality. This study identifies a gene that may have a function in parasite growth, virulence, and host–parasite interaction, and therefore could be a target for drug or vaccine development. Many strains of Plasmodium differ in virulence, but factors that control these distinctions are not known. Here the authors comparatively map virulence loci using the offspring from a P. yoelii YM and N67 genetic cross, and identify a putative HECT E3 ubiquitin ligase that may explain the variance.
Collapse
Affiliation(s)
- Sethu C Nair
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Ruixue Xu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Sittiporn Pattaradilokrat
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA.,Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Jian Wu
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yanwei Qi
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA.,State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Martine Zilversmit
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Sundar Ganesan
- Biological Imaging Section, Research Technology Branch, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Vijayaraj Nagarajan
- Bioinformatics and Computational Biosciences Branch, Office of Cyber Infrastructure and Computational Biology, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Richard T Eastman
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Marlene S Orandle
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - John C Tan
- The Eck Institute of Global Health, Department of Biological Sciences, University of Notre Dame, Indiana, 46556, USA
| | - Timothy G Myers
- Genomic Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Shengfa Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China
| | - Carole A Long
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jian Li
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China.
| | - Xin-Zhuan Su
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892, USA. .,State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, 361005, China.
| |
Collapse
|
10
|
Van Raemdonck K, Van den Steen PE, Liekens S, Van Damme J, Struyf S. CXCR3 ligands in disease and therapy. Cytokine Growth Factor Rev 2015; 26:311-27. [DOI: 10.1016/j.cytogfr.2014.11.009] [Citation(s) in RCA: 167] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Accepted: 11/05/2014] [Indexed: 12/19/2022]
|
11
|
Frevert U, Nacer A. Immunobiology of Plasmodium in liver and brain. Parasite Immunol 2014; 35:267-82. [PMID: 23631610 DOI: 10.1111/pim.12039] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2013] [Accepted: 04/17/2013] [Indexed: 12/11/2022]
Abstract
Malaria remains one of the most serious health problems globally, but our understanding of the biology of the parasite and the pathogenesis of severe disease is still limited. Multiple cellular effector mechanisms that mediate parasite elimination from the liver have been described, but how effector cells use classical granule-mediated cytotoxicity to attack infected hepatocytes and how cytokines and chemokines spread via the unique fluid pathways of the liver to reach the parasites over considerable distances remains unknown. Similarly, a wealth of information on cerebral malaria (CM), one of the most severe manifestations of the disease, was gained from post-mortem analyses of human brain and murine disease models, but the cellular processes that ultimately cause disease are not fully understood. Here, we discuss how imaging of the local dynamics of parasite infection and host response as well as consideration of anatomical and physiological features of liver and brain can provide a better understanding of the initial asymptomatic hepatic phase of the infection and the cascade of events leading to CM. Given the increasing drug resistance of both parasite and vector and the unavailability of a protective vaccine, the urgency to reduce the tremendous morbidity and mortality associated with severe malaria is obvious.
Collapse
Affiliation(s)
- U Frevert
- Division of Medical Parasitology, Department of Microbiology, New York University School of Medicine, New York, NY 10010, USA.
| | | |
Collapse
|
12
|
Auclair SR, Roth KE, Saunders BL, Ogborn KM, Sheikh AA, Naples J, Young AMP, Boisen DK, Tavangar AT, Welch JE, Lantz CS. Interleukin-3-deficient mice have increased resistance to blood-stage malaria. Infect Immun 2014; 82:1308-14. [PMID: 24379292 PMCID: PMC3957999 DOI: 10.1128/iai.01140-13] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2013] [Accepted: 12/23/2013] [Indexed: 01/05/2023] Open
Abstract
The contribution of interleukin-3 (IL-3), a hematopoietic growth factor and immunoregulatory cytokine, to resistance to blood-stage malaria was investigated by infecting IL-3-deficient (knockout [KO]) mice with Plasmodium berghei NK65. Male IL-3 KO mice, but not female mice, were more resistant to infection than wild-type (WT) mice, as evidenced by lower peak parasitemia and prolonged survival. Both male and female IL-3 KO mice had increased splenomegaly and were more anemic than corresponding WT mice. Anemia was compensated for by an increase in bone marrow and splenic erythropoiesis in IL-3 KO mice, as evidenced by higher levels of erythroid progenitors. Plasma levels of gamma interferon (IFN-γ) and CXCL9 (monokine induced by IFN-γ [MIG]) were found to be significantly reduced in IL-3 KO mice during early stages of infection. In contrast, granulocyte colony-stimulating factor (G-CSF) levels were significantly higher, and the percentage of peripheral blood neutrophils lower, in infected IL-3 KO mice than in WT counterparts. Overall, our results indicate that IL-3 plays a critical role in suppressing protective immunity to P. berghei NK65 infection and that it is involved in inhibiting the development of splenomegaly, anemia, and erythropoiesis. IL-3 also influences IFN-γ, CXCL9, and G-CSF production in response to infection. The abnormal responses seen in infected IL-3 KO mice may be due to the lack of IL-3 during development, to the lack of IL-3 in the infected mature mice, or to both.
Collapse
Affiliation(s)
- Sarah R Auclair
- Department of Biology, James Madison University, Harrisonburg, Virginia, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Polimeni M, Prato M. Host matrix metalloproteinases in cerebral malaria: new kids on the block against blood-brain barrier integrity? Fluids Barriers CNS 2014; 11:1. [PMID: 24467887 PMCID: PMC3905658 DOI: 10.1186/2045-8118-11-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2013] [Accepted: 01/24/2014] [Indexed: 12/23/2022] Open
Abstract
Cerebral malaria (CM) is a life-threatening complication of falciparum malaria, associated with high mortality rates, as well as neurological impairment in surviving patients. Despite disease severity, the etiology of CM remains elusive. Interestingly, although the Plasmodium parasite is sequestered in cerebral microvessels, it does not enter the brain parenchyma: so how does Plasmodium induce neuronal dysfunction? Several independent research groups have suggested a mechanism in which increased blood–brain barrier (BBB) permeability might allow toxic molecules from the parasite or the host to enter the brain. However, the reported severity of BBB damage in CM is variable depending on the model system, ranging from mild impairment to full BBB breakdown. Moreover, the factors responsible for increased BBB permeability are still unknown. Here we review the prevailing theories on CM pathophysiology and discuss new evidence from animal and human CM models implicating BBB damage. Finally, we will review the newly-described role of matrix metalloproteinases (MMPs) and BBB integrity. MMPs comprise a family of proteolytic enzymes involved in modulating inflammatory response, disrupting tight junctions, and degrading sub-endothelial basal lamina. As such, MMPs represent potential innovative drug targets for CM.
Collapse
Affiliation(s)
| | - Mauro Prato
- Dipartimento di Neuroscienze, Università di Torino, C,so Raffaello 30, 10125 Torino, Italy.
| |
Collapse
|
14
|
Tripathy S, Mahapatra SK, Chattopadhyay S, Das S, Dash SK, Majumder S, Pramanik P, Roy S. A novel chitosan based antimalarial drug delivery against Plasmodium berghei infection. Acta Trop 2013; 128:494-503. [PMID: 23906613 DOI: 10.1016/j.actatropica.2013.07.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2013] [Revised: 07/03/2013] [Accepted: 07/19/2013] [Indexed: 02/06/2023]
Abstract
Chitosan is a natural polysaccharide that has attracted significant scientific interest during the last two decades and chitosan based nanodrug delivery systems seem to be a hopeful and viable strategy for improving disease treatment. This study aims to evaluate the potency of the polymer based nanochloroquine in application for attenuation of Plasmodium berghei infection in Swiss mice and effectiveness against the parasite induced oxidative stress and deoxyribo nucleic acid (DNA) damage in lymphocytes. Nanoparticle was prepared by ionotropic gelation between chitosan and sodium tripolyphosphate. The chloroquine was treated by the actual drug content of effective nanochloroquine and the nanodrug was charged with its effective dose for fifteen days, after successive infection development in Swiss mice. Gimsa staining of thin smear and flow cytometry analysis was pursued to reveal the parasitemia. Different oxidative markers, inflammatory markers, antioxidant enzymes level and also lymphocytic deoxyribo nucleic acid damage study were performed. The present study reveals the potency of the nanodrug which has been found as more prospective than only chloroquine treatment to combat the parasite infection, oxidative stress as well as inflammation and DNA damage. From the study, we conclude this nanodrug may be applicable as potent therapeutic agent than only chloroquine.
Collapse
Affiliation(s)
- Satyajit Tripathy
- Immunology and Microbiology Laboratory, Department of Human Physiology with Community Health, Vidyasagar University, Midnapore 721 102, West Bengal, India
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Frevert U, Nacer A, Cabrera M, Movila A, Leberl M. Imaging Plasmodium immunobiology in the liver, brain, and lung. Parasitol Int 2013; 63:171-86. [PMID: 24076429 DOI: 10.1016/j.parint.2013.09.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2012] [Revised: 08/28/2013] [Accepted: 09/18/2013] [Indexed: 01/10/2023]
Abstract
Plasmodium falciparum malaria is responsible for the deaths of over half a million African children annually. Until a decade ago, dynamic analysis of the malaria parasite was limited to in vitro systems with the typical limitations associated with 2D monocultures or entirely artificial surfaces. Due to extremely low parasite densities, the liver was considered a black box in terms of Plasmodium sporozoite invasion, liver stage development, and merozoite release into the blood. Further, nothing was known about the behavior of blood stage parasites in organs such as the brain where clinical signs manifest and the ensuing immune response of the host that may ultimately result in a fatal outcome. The advent of fluorescent parasites, advances in imaging technology, and availability of an ever-increasing number of cellular and molecular probes have helped illuminate many steps along the pathogenetic cascade of this deadly tropical parasite.
Collapse
Affiliation(s)
- Ute Frevert
- Division of Medical Parasitology, Department of Microbiology, New York University School of Medicine, 341 E 25 Street, New York, NY 10010, USA.
| | | | | | | | | |
Collapse
|
16
|
Oliveira-Lima OC, Bernardes D, Xavier Pinto MC, Esteves Arantes RM, Carvalho-Tavares J. Mice lacking inducible nitric oxide synthase develop exacerbated hepatic inflammatory responses induced by Plasmodium berghei NK65 infection. Microbes Infect 2013; 15:903-10. [PMID: 23988520 DOI: 10.1016/j.micinf.2013.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Revised: 08/03/2013] [Accepted: 08/06/2013] [Indexed: 02/06/2023]
Abstract
Infection of mice with Plasmodium berghei NK65 represents a well-recognized malaria model in which infection is accompanied by an intense hepatic inflammatory response. Enzyme-inducible nitric oxide synthase is an important regulator of inflammation and leukocyte recruitment in microvessels, but these functions have yet to be evaluated in experimental malaria. In this study, we assessed the involvement of inducible nitric oxide synthase in inflammatory responses to murine experimental malaria induced by P. berghei NK65. We observed that wild type (WT) and nitric oxide synthase (iNOS)-deficient mice (iNOS(-/-)) mice showed similar levels of parasitemia following P. berghei NK65 infection, although infected iNOS(-/-) mice presented early mortality. Inducible nitric oxide synthase deficiency led to increased leukocyte rolling and adhesion to the liver in iNOS(-/-) mice relative to the WT animals, as observed via intravital microscopy. Infected iNOS(-/-) mice also exhibited increased hepatic leukocyte migration and subsequent liver damage, which was associated with high serum levels of the cytokines TNF-α, IL-6 and IL-10. Our data suggest potential role for the iNOS enzyme as a regulator of hepatic inflammatory response induced by P. berghei NK65-infection, and its absence leads to exacerbated inflammation and sequential associated-hepatic damage in the animals.
Collapse
Affiliation(s)
- Onésia Cristina Oliveira-Lima
- Núcleo de Neurociências, Departamento de Fisiologia e Biofísica, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, MG 31270-901, Brazil
| | | | | | | | | |
Collapse
|
17
|
Martins YC, Daniel-Ribeiro CT. A new hypothesis on the manifestation of cerebral malaria: the secret is in the liver. Med Hypotheses 2013; 81:777-83. [PMID: 23978689 DOI: 10.1016/j.mehy.2013.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2013] [Accepted: 08/06/2013] [Indexed: 02/06/2023]
Abstract
Despite the abundance of information on cerebral malaria (CM), the pathogenesis of this disease is not completely understood. At present, two nonexclusive dominant hypotheses exist to explain how the neurological syndrome manifests: the sequestration (or mechanical) hypothesis and the inflammatory hypothesis. The sequestration hypothesis states that sequestration of Plasmodium falciparum-parasitized red blood cells (pRBCs) to brain capillary endothelia causes obstruction of capillary blood flow followed by brain tissue anoxia and coma. The inflammatory hypothesis postulates that P. falciparum infection releases toxic molecules in the circulation, inducing an imbalanced systemic inflammatory response that leads to coagulopathy, brain endothelial cell dysfunction, accumulation of leukocytes in the brain microcirculation, blood brain barrier (BBB) leakage, cerebral vasoconstriction, edema, and coma. However, both hypotheses, even when considered together, are not sufficient to fully explain the pathogenesis of CM. Here, we propose that the development of acute liver failure (ALF) together with BBB breakdown are the necessary and sufficient conditions for the genesis of CM. ALF is characterized by coagulopathy and hepatic encephalopathy (HE) in a patient without pre-existing liver disease. Signs of hepatic dysfunction have been shown to occur in 2.5-40% of CM patients. In addition, recent studies with murine models demonstrated that mice presenting experimental cerebral malaria (ECM) had hepatic damage and brain metabolic changes characteristic of HE. However, the occurrence of CM in patients with mild or without apparent hepatocellular liver damage and the presence of liver damage in non-CM murine models indicate that the development of ALF during malaria infection is not the single factor responsible for neuropathology. To solve this problem, we also propose that BBB breakdown contributes to the pathogenesis of CM and synergizes with hepatic failure to cause neurological signs and symptoms. BBB dysfunction would thus occur in CM by a mechanism similar to the one occurring in sepsis and is in agreement with the inflammatory hypothesis. Nevertheless, differently from in the inflammatory hypothesis, BBB leakage would facilitate the penetration of ammonia and other toxins into the brain parenchyma, but would not be sufficient to cause CM when occurring alone. We believe our hypothesis better explains the pathogenesis of CM, does not have problems to deal with the exception data not explained by the previous hypotheses, and reveals new targets for adjunctive therapy.
Collapse
Affiliation(s)
- Yuri Chaves Martins
- Laboratório de Pesquisas em Malária, Instituto Oswaldo Cruz and Centro de Pesquisa Diagnóstico e Treinamento em Malária, Fiocruz, Rio de Janeiro, RJ, Brazil; Department of Pathology, Albert Einstein College of Medicine, The Bronx, NY, USA.
| | | |
Collapse
|
18
|
Hawkes M, Elphinstone RE, Conroy AL, Kain KC. Contrasting pediatric and adult cerebral malaria: the role of the endothelial barrier. Virulence 2013; 4:543-55. [PMID: 23924893 PMCID: PMC5359751 DOI: 10.4161/viru.25949] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Malaria affects millions of people around the world and a small subset of those infected develop cerebral malaria. The clinical presentation of cerebral malaria differs between children and adults, and it has been suggested that age-related changes in the endothelial response may account for some of these differences. During cerebral malaria, parasites sequester within the brain microvasculature but do not penetrate into the brain parenchyma and yet, the infection causes severe neurological symptoms. Endothelial dysfunction is thought to play an important role in mediating these adverse clinical outcomes. During infection, the endothelium becomes activated and more permeable, which leads to increased inflammation, hemorrhages, and edema in the surrounding tissue. We hypothesize that post-natal developmental changes, occurring in both endothelial response and the neurovascular unit, account for the differences observed in the clinical presentations of cerebral malaria in children compared with adults.
Collapse
|
19
|
Dai M, Freeman B, Shikani HJ, Bruno FP, Collado JE, Macias R, Reznik SE, Davies P, Spray DC, Tanowitz HB, Weiss LM, Desruisseaux MS. Altered regulation of Akt signaling with murine cerebral malaria, effects on long-term neuro-cognitive function, restoration with lithium treatment. PLoS One 2012; 7:e44117. [PMID: 23082110 PMCID: PMC3474787 DOI: 10.1371/journal.pone.0044117] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2011] [Accepted: 07/31/2012] [Indexed: 01/29/2023] Open
Abstract
Neurological and cognitive impairment persist in more than 20% of cerebral malaria (CM) patients long after successful anti-parasitic treatment. We recently reported that long term memory and motor coordination deficits are also present in our experimental cerebral malaria model (ECM). We also documented, in a murine model, a lack of obvious pathology or inflammation after parasite elimination, suggesting that the long-term negative neurological outcomes result from potentially reversible biochemical and physiological changes in brains of ECM mice, subsequent to acute ischemic and inflammatory processes. Here, we demonstrate for the first time that acute ECM results in significantly reduced activation of protein kinase B (PKB or Akt) leading to decreased Akt phosphorylation and inhibition of the glycogen kinase synthase (GSK3β) in the brains of mice infected with Plasmodium berghei ANKA (PbA) compared to uninfected controls and to mice infected with the non-neurotrophic P. berghei NK65 (PbN). Though Akt activation improved to control levels after chloroquine treatment in PbA-infected mice, the addition of lithium chloride, a compound which inhibits GSK3β activity and stimulates Akt activation, induced a modest, but significant activation of Akt in the brains of infected mice when compared to uninfected controls treated with chloroquine with and without lithium. In addition, lithium significantly reversed the long-term spatial and visual memory impairment as well as the motor coordination deficits which persisted after successful anti-parasitic treatment. GSK3β inhibition was significantly increased after chloroquine treatment, both in lithium and non-lithium treated PbA-infected mice. These data indicate that acute ECM is associated with abnormalities in cell survival pathways that result in neuronal damage. Regulation of Akt/GSK3β with lithium reduces neuronal degeneration and may have neuroprotective effects in ECM. Aberrant regulation of Akt/GSK3β signaling likely underlies long-term neurological sequelae observed in ECM and may yield adjunctive therapeutic targets for the management of CM.
Collapse
Affiliation(s)
- Minxian Dai
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St John's University, Queens, New York, United States of America
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Brandi Freeman
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Henry J. Shikani
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Fernando Pereira Bruno
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - J. Elias Collado
- Pontificia Universidad Catolica Madrey Maestra, Santiago, Dominican Republic
| | - Rolando Macias
- Meharry Medical College, Nashville, Tennessee, United States of America
| | - Sandra E. Reznik
- Department of Pharmaceutical Sciences, College of Pharmacy and Allied Health Professions, St John's University, Queens, New York, United States of America
| | - Peter Davies
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- The Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Litwin-Zucker Center for the Study of Alzheimer's Disease and Memory Disorders, Feinstein Institute for Medical Research, Manhasset, New York, United States of America
| | - David Conover Spray
- The Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Herbert Bernard Tanowitz
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Louis Martin Weiss
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| | - Mahalia Sabrina Desruisseaux
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York, United States of America
- Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, United States of America
| |
Collapse
|
20
|
Spindler KR, Hsu TH. Viral disruption of the blood-brain barrier. Trends Microbiol 2012; 20:282-90. [PMID: 22564250 DOI: 10.1016/j.tim.2012.03.009] [Citation(s) in RCA: 152] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2012] [Revised: 03/16/2012] [Accepted: 03/27/2012] [Indexed: 12/25/2022]
Abstract
The blood-brain barrier (BBB) provides significant protection against microbial invasion of the brain. However, the BBB is not impenetrable, and mechanisms by which viruses breach it are becoming clearer. In vivo and in vitro model systems are enabling identification of host and viral factors contributing to breakdown of the unique BBB tight junctions. Key mechanisms of tight junction damage from inside and outside cells are disruption of the actin cytoskeleton and matrix metalloproteinase activity, respectively. Viral proteins acting in BBB disruption are described for HIV-1, currently the most studied encephalitic virus; other viruses are also discussed.
Collapse
Affiliation(s)
- Katherine R Spindler
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109-5620, USA.
| | | |
Collapse
|