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] [Grants] [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
|
McNitt SA, Dick JK, Hernandez Castaneda M, Sangala JA, Pierson M, Macchietto M, Burrack KS, Crompton PD, Seydel KB, Hamilton SE, Hart GT. Phenotype and function of IL-10 producing NK cells in individuals with malaria experience. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.593687. [PMID: 38798324 PMCID: PMC11118352 DOI: 10.1101/2024.05.11.593687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Plasmodium falciparum infection can trigger high levels of inflammation that lead to fever and sometimes severe disease. People living in malaria-endemic areas gradually develop resistance to symptomatic malaria and control both parasite numbers and the inflammatory response. We previously found that adaptive natural killer (NK) cells correlate with reduced parasite load and protection from symptoms. We also previously found that murine NK cell production of IL-10 can protect mice from experimental cerebral malaria. Human NK cells can also secrete IL-10, but it was unknown what NK cell subsets produce IL-10 and if this is affected by malaria experience. We hypothesize that NK cell immunoregulation may lower inflammation and reduce fever induction. Here, we show that NK cells from subjects with malaria experience make significantly more IL-10 than subjects with no malaria experience. We then determined the proportions of NK cells that are cytotoxic and produce interferon gamma and/or IL-10 and identified a signature of adaptive and checkpoint molecules on IL-10-producing NK cells. Lastly, we find that co-culture with primary monocytes, Plasmodium -infected RBCs, and antibody induces IL-10 production by NK cells. These data suggest that NK cells may contribute to protection from malaria symptoms via IL-10 production.
Collapse
|
3
|
Carpinter BA, Renhe DC, Bellei JCB, Vieira CD, Rodolphi CM, Ferreira MVR, de Freitas CS, Neto AFDS, Coelho EAF, Mietto BDS, Gomes FLR, Rocha VN, Scopel KKG. DHA-rich fish oil plays a protective role against experimental cerebral malaria by controlling inflammatory and mechanical events from infection. J Nutr Biochem 2024; 123:109492. [PMID: 37866427 DOI: 10.1016/j.jnutbio.2023.109492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 05/17/2023] [Accepted: 10/17/2023] [Indexed: 10/24/2023]
Abstract
Every year, thousands of children, particularly those under 5 years old, die because of cerebral malaria (CM). Following conventional treatment, approximately 25% of surviving individuals have lifelong severe neurocognitive sequelae. Therefore, improved conventional therapies or effective alternative therapies that prevent the severe infection are crucial. Omega-3 (Ω-3) polyunsaturated fatty acids (PUFAs) are known to have antioxidative and anti-inflammatory effects and protect against diverse neurological disorders, including Alzheimer's and Parkinson's diseases. However, little is known regarding the effects of Ω-3 PUFAs against parasitic infections. In this study, C57BL/6 mice received supplemental treatment of a fish oil rich in the Ω-3 PUFA, docosahexaenoic acid (DHA), which was started 15 days prior to infection with Plasmodium berghei ANKA and was maintained until the end of the study. Animals treated with the highest doses of DHA, 3.0 and 6.0 g/kg body weight, had 60 and 80% chance of survival, respectively, while all nontreated mice died by the 7th day postinfection due to CM. Furthermore, the parasite load during the critical period for CM development (5th to 11th day postinfection) was controlled in treated mice. However, after this period all animals developed high levels of parasitemia until the 20th day of infection. DHA treatment also effectively reduced blood-brain barrier (BBB) damage and brain edema and completely prevented brain hemorrhage and vascular occlusion. A strong anti-inflammatory profile was observed in the brains of DHA-treated mice, as well as, an increased number of neutrophil and reduced number of CD8+ T leukocytes in the spleen. Thus, this is the first study to demonstrate that the prophylactic use of DHA-rich fish oil exerts protective effects against experimental CM, reducing the mechanical and immunological events caused by the P. berghei ANKA infection.
Collapse
Affiliation(s)
- Bárbara Albuquerque Carpinter
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Daniela Chaves Renhe
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Jéssica Correa Bezerra Bellei
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Carolina David Vieira
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Cinthia Magalhães Rodolphi
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Camila Simões de Freitas
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Adolfo Firmino da Silva Neto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eduardo Antônio Ferraz Coelho
- Post-graduation Program in Health Sciences, Infectology and Tropical Medicine, Faculty of Medicine, Federal University of Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Bruno de Siqueira Mietto
- Department of Biology, Research Centre of Cellular Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | | | - Vinicius Novaes Rocha
- Department of Veterinary Medicine, Research Centre of Pathology and Veterinary Histology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Kézia Katiani Gorza Scopel
- Department of Parasitology, Microbiology and Immunology and Post-Graduation Program in Biological Science, Research Centre of Parasitology, Federal University of Juiz de Fora, Juiz de Fora, Brazil.
| |
Collapse
|
4
|
Obeng-Aboagye E, Frimpong A, Amponsah JA, Danso SE, Owusu EDA, Ofori MF. Inflammatory cytokines as potential biomarkers for early diagnosis of severe malaria in children in Ghana. Malar J 2023; 22:220. [PMID: 37525227 PMCID: PMC10388454 DOI: 10.1186/s12936-023-04652-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Accepted: 07/22/2023] [Indexed: 08/02/2023] Open
Abstract
BACKGROUND Severe malaria (SM) is a fatal multi-system disease which accounted for an estimated 619,000 deaths in 2021. Less than 30% of children presenting with SM are diagnosed and treated promptly, resulting in increased mortality and neurologic impairments in survivors. Studies have identified cytokine profiles that differentiate the various clinical manifestations of malaria (severe and uncomplicated). However, the diagnostic capability of these cytokines in differentiating between the disease states in terms of cut-off values has not yet been determined. METHODS The plasma levels of 22 pro-inflammatory cytokines (Eotaxin/CCL 11, interferon-gamma (IFN-γ), interleukin (IL)- 2, IL-6, IL-1β, IL-12p40/p70, IL-17A, RANTES, MCP-1, IL-15, IL-5, IL-1RA, IL-2R, IFN-α, IP-10, TNF, MIG, MIP-1α, MIP-1β, IL-7, IL-8 and Granulocyte Macrophage-Colony Stimulating Factor (GM-CSF), and 3 anti-inflammatory cytokines-(IL-4, IL-13 and IL-10) in patients with SM, uncomplicated malaria (UM) and other febrile conditions, were measured and compared using the Human Cytokine Magnetic 25-Plex Panel. The receiver operating characteristic (ROC) curve analysis was used to determine the diagnostic value of these cytokines. RESULTS The level of the pro-inflammatory cytokine, IL-17A, was significantly higher in the SM group as compared to the UM group. Levels of the anti-inflammatory cytokines however did not differ significantly among the SM and UM groups. Only IL-1β and IL-17A showed good diagnostic potential after ROC curve analysis. CONCLUSION The data show that levels of pro-inflammatory cytokines correlate with malaria disease severity. IL-1β and IL-17A showed good diagnostic potentials and can be considered for use in clinical practice to target treatment.
Collapse
Affiliation(s)
- Elizabeth Obeng-Aboagye
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Augustina Frimpong
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana.
| | - Jones Amo Amponsah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | | | - Ewurama D A Owusu
- Department of Medical Laboratory Sciences, School of Biomedical and Allied Health Sciences, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Michael Fokuo Ofori
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana.
| |
Collapse
|
5
|
Cimperman CK, Pena M, Gokcek SM, Theall BP, Patel MV, Sharma A, Qi C, Sturdevant D, Miller LH, Collins PL, Pierce SK, Akkaya M. Cerebral Malaria Is Regulated by Host-Mediated Changes in Plasmodium Gene Expression. mBio 2023; 14:e0339122. [PMID: 36852995 PMCID: PMC10127683 DOI: 10.1128/mbio.03391-22] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 02/06/2023] [Indexed: 03/01/2023] Open
Abstract
Cerebral malaria (CM), the deadliest complication of Plasmodium infection, is a complex and unpredictable disease. However, our understanding of the host and parasite factors that cause CM is limited. Using a mouse model of CM, experimental CM (ECM), we performed a three-way comparison between ECM-susceptible C57BL/6 mice infected with ECM-causing Plasmodium ANKA parasites [ANKA(C57BL/6)], ECM-resistant BALB/c mice infected with Plasmodium ANKA [ANKA(BALB/c)], and C57BL/6 mice infected with Plasmodium NK65 that does not cause ECM [NK65(C57BL/6)]. All ANKA(C57BL/6) mice developed CM. In contrast, in ANKA(BALB/c) and NK65(C57BL/6), infections do not result in CM and proceed similarly in terms of parasite growth, disease course, and host immune response. However, parasite gene expression in ANKA(BALB/c) was remarkably different than that in ANKA(C57BL/6) but similar to the gene expression in NK65(C57BL/6). Thus, Plasmodium ANKA has an ECM-specific gene expression profile that is activated only in susceptible hosts, providing evidence that the host has a critical influence on the outcome of infection. IMPORTANCE Hundreds of thousands of lives are lost each year due to the brain damage caused by malaria disease. The overwhelming majority of these deaths occur in young children living in sub-Saharan Africa. Thus far, there are no vaccines against this deadly disease, and we still do not know why fatal brain damage occurs in some children while others have milder, self-limiting disease progression. Our research provides an important clue to this problem. Here, we showed that the genetic background of the host has an important role in determining the course and the outcome of the disease. Our research also identified parasite molecules that can potentially be targeted in vaccination and therapy approaches.
Collapse
Affiliation(s)
- Clare K. Cimperman
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Mirna Pena
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Sohret M. Gokcek
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Brandon P. Theall
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Meha V. Patel
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Anisha Sharma
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - ChenFeng Qi
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Daniel Sturdevant
- Research Technologies Branch, Rocky Mountain Laboratories, National Institute of Allergy and Infectious Diseases, Hamilton, Montana, USA
| | - Louis H. Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Patrick L. Collins
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| | - Susan K. Pierce
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, Rockville, Maryland, USA
| | - Munir Akkaya
- Division of Rheumatology and Immunology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA
- Pelotonia Institute for Immuno-Oncology, The James Comprehensive Cancer Center, College of Medicine, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
6
|
Storm J, Camarda G, Haley MJ, Brough D, Couper KN, Craig AG. Plasmodium falciparum-infected erythrocyte co-culture with the monocyte cell line THP-1 does not trigger production of soluble factors reducing brain microvascular barrier function. PLoS One 2023; 18:e0285323. [PMID: 37141324 PMCID: PMC10159134 DOI: 10.1371/journal.pone.0285323] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Accepted: 04/19/2023] [Indexed: 05/06/2023] Open
Abstract
Monocytes contribute to the pro-inflammatory immune response during the blood stage of a Plasmodium falciparum infection, but their precise role in malaria pathology is not clear. Besides phagocytosis, monocytes are activated by products from P. falciparum infected erythrocytes (IE) and one of the activation pathways is potentially the NLR family pyrin domain containing 3 (NLRP3) inflammasome, a multi-protein complex that leads to the production of interleukin (IL)-1β. In cerebral malaria cases, monocytes accumulate at IE sequestration sites in the brain microvascular and the locally produced IL-1β, or other secreted molecules, could contribute to leakage of the blood-brain barrier. To study the activation of monocytes by IE within the brain microvasculature in an in vitro model, we co-cultured IT4var14 IE and the monocyte cell line THP-1 for 24 hours and determined whether generated soluble molecules affect barrier function of human brain microvascular endothelial cells, measured by real time trans-endothelial electrical resistance. The medium produced after co-culture did not affect endothelial barrier function and similarly no effect was measured after inducing oxidative stress by adding xanthine oxidase to the co-culture. While IL-1β does decrease barrier function, barely any IL-1β was produced in the co- cultures, indicative of a lack of or incomplete THP-1 activation by IE in this co-culture model.
Collapse
Affiliation(s)
- Janet Storm
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Grazia Camarda
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| | - Michael J Haley
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - David Brough
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - Kevin N Couper
- Division of Immunology, Immunity to Infection and Respiratory Medicine, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom
| | - Alister G Craig
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
- Centre for Drugs and Diagnostics, Liverpool School of Tropical Medicine, Liverpool, United Kingdom
| |
Collapse
|
7
|
Qian H, Ye Z, Pi L, Ao J. Roles and current applications of S-nitrosoglutathione in anti-infective biomaterials. Mater Today Bio 2022; 16:100419. [PMID: 36105674 PMCID: PMC9465324 DOI: 10.1016/j.mtbio.2022.100419] [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: 07/12/2022] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 11/29/2022]
Abstract
Bacterial infections can compromise the physical and biological functionalities of humans and pose a huge economical and psychological burden on infected patients. Nitric oxide (NO) is a broad-spectrum antimicrobial agent, whose mechanism of action is not affected by bacterial resistance. S-nitrosoglutathione (GSNO), an endogenous donor and carrier of NO, has gained increasing attention because of its potent antibacterial activity and efficient biocompatibility. Significant breakthroughs have been made in the application of GSNO in biomaterials. This review is based on the existing evidence that comprehensively summarizes the progress of antimicrobial GSNO applications focusing on their anti-infective performance, underlying antibacterial mechanisms, and application in anti-infective biomaterials. We provide an accurate overview of the roles and applications of GSNO in antibacterial biomaterials and shed new light on the avenues for future studies.
Collapse
Key Words
- A.baumannii, Acinetobacter baumannii
- AgNPs, Silver nanoparticles
- Antibacterial property
- BMSCs, Bone marrow stem cells
- Bacterial resistance
- Biomaterials
- C.albicans, Candida albicans
- CS/GE, Chitosan/gelatin
- Cu, copper
- DMSO, Dimethyl sulfoxide
- DPA, Diethylenetriamine pentaacetic acid
- E. coli, Escherichia coli
- E.tenella, Eimeria tenella
- ECC, Extracorporeal circulation
- ECM, Experimental cerebral malaria
- GSNO, S-Nitrosoglutathione
- GSNOR, S-Nitrosoglutathione Reductase
- H.pylori, Helicobacter pylori
- HCC, Human cervical carcinoma
- HDFs, Human dermal fibroblasts
- HUVEC, Human umbilical vein endothelial cells
- ICR, Imprinted control region
- Infection
- K.Pneumonia, Klebsiella Pneumonia
- L.amazonensis, Leishmania amazonensis
- L.major, Leishmania major
- M.Tuberculosis, Mycobacterium tuberculosis
- M.smegmatis, Mycobacterium smegmatis
- MOF, Metal–organic framework
- MRPA, Multidrug-resistant Pseudomonas aeruginosa
- MRSA, Methicillin resistant Staphylococcus aureus
- N. gonorrhoeae, Neisseria gonorrhoeae
- N.meningitidis, Neisseria meningitidis
- NA, Not available
- NO-np, NO-releasing nanoparticulate platform
- NP, Nanoparticle
- P.aeruginosa, Pseudomonas aeruginosa
- P.berghei, Plasmodium berghei
- P.mirabilis, Proteus mirabilis
- PCL, Polycaprolactone
- PCVAD, Porcine circovirus-associated disease
- PDA-GSNO NPs, Polydopamine nanoparticles containing GSNO
- PDAM@Cu, polydopamine based copper coatings
- PEG, polyethylene glycol
- PHB, polyhydroxybutyrate
- PLA, polylactic acid
- PLGA, poly(lactic-co-glycolic acid)
- PTT, Photothermal therapy
- PVA, poly(vinyl alcohol)
- PVA/PEG, poly(vinyl alcohol)/poly(ethylene glycol)
- PVC, poly(vinyl chloride)
- S-nitrosoglutathione
- S. typhimurium, Salmonella typhimurium
- S.aureus, Staphylococcus aureus
- S.epidermidis, Staphylococcus epidermidis
- S.pneumoniae, Streptococcus pneumoniae
- SAKI, Septic acute kidney injury
- SCI, Spinal cord slices
- Se, Selenium
- Sp3, Specificity proteins 3
- TDC, Tunneled dialysis catheters
- TMOS, Tetramethylorthosilicate
- ZnO, Zinc oxide
- cftr, cystic fibrosis transmembrane conductance regulatory gene
- d, day
- h, hour
- min, minute
- pSiNPs, porous silicon nanoparticles
- w, week
Collapse
Affiliation(s)
- Hu Qian
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhimin Ye
- Department of Pathology, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Lanping Pi
- Nursing Department, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jun Ao
- Department of Orthopaedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| |
Collapse
|
8
|
Ramachandran A, Sharma A. Dissecting the mechanisms of pathogenesis in cerebral malaria. PLoS Pathog 2022; 18:e1010919. [PMCID: PMC9671333 DOI: 10.1371/journal.ppat.1010919] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Cerebral malaria (CM) is one of the leading causes of death due to malaria. It is characterised by coma, presence of asexual parasites in blood smear, and absence of any other reason that can cause encephalopathy. The fatality rate for CM is high, and those who survive CM often experience long-term sequelae, including cognitive and motor dysfunctions. It is unclear how parasites sequestered in the lumen of endothelial cells of the blood–brain barrier (BBB), and localised breakdown of BBB can manifest gross physiological changes across the brain. The pathological changes associated with CM are mainly due to the dysregulation of inflammatory and coagulation pathways. Other factors like host and parasite genetics, transmission intensity, and the host’s immune status are likely to play a role in the development and progression of CM. This work focuses on the pathological mechanisms underlying CM. Insights from humans, mice, and in vitro studies have been summarised to present a cohesive understanding of molecular mechanisms involved in CM pathology.
Collapse
Affiliation(s)
- Arathy Ramachandran
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
| | - Amit Sharma
- Molecular Medicine Group, International Centre for Genetic Engineering and Biotechnology, New Delhi, India
- * E-mail:
| |
Collapse
|
9
|
Plasmodium falciparum and TNF-α Differentially Regulate Inflammatory and Barrier Integrity Pathways in Human Brain Endothelial Cells. mBio 2022; 13:e0174622. [PMID: 36036514 PMCID: PMC9601155 DOI: 10.1128/mbio.01746-22] [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] [Indexed: 11/20/2022] Open
Abstract
Cerebral malaria is a severe complication of Plasmodium falciparum infection characterized by the loss of blood-brain barrier (BBB) integrity, which is associated with brain swelling and mortality in patients. P. falciparum-infected red blood cells and inflammatory cytokines, like tumor necrosis factor alpha (TNF-α), have been implicated in the development of cerebral malaria, but it is still unclear how they contribute to the loss of BBB integrity. Here, a combination of transcriptomic analysis and cellular assays detecting changes in barrier integrity and endothelial activation were used to distinguish between the effects of P. falciparum and TNF-α on a human brain microvascular endothelial cell (HBMEC) line and in primary human brain microvascular endothelial cells. We observed that while TNF-α induced high levels of endothelial activation, it only caused a small increase in HBMEC permeability. Conversely, P. falciparum-infected red blood cells (iRBCs) led to a strong increase in HBMEC permeability that was not mediated by cell death. Distinct transcriptomic profiles of TNF-α and P. falciparum in HBMECs confirm the differential effects of these stimuli, with the parasite preferentially inducing an endoplasmic reticulum stress response. Our results establish that there are fundamental differences in the responses induced by TNF-α and P. falciparum on brain endothelial cells and suggest that parasite-induced signaling is a major component driving the disruption of the BBB during cerebral malaria, proposing a potential target for much needed therapeutics.
Collapse
|
10
|
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
|
11
|
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: 3.3] [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
|
12
|
Matsuo-Dapaah J, Lee MSJ, Ishii KJ, Tainaka K, Coban C. Using a new three-dimensional CUBIC tissue-clearing method to examine the brain during experimental cerebral malaria. Int Immunol 2021; 33:587-594. [PMID: 34455438 DOI: 10.1093/intimm/dxab060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 08/28/2021] [Indexed: 11/13/2022] Open
Abstract
Cerebral malaria (CM) is a life-threatening complication of the malaria disease caused by Plasmodium falciparum infection and is responsible for the death of half a million people annually. The molecular pathogenesis underlying CM in humans is not completely understood, although sequestration of infected erythrocytes in cerebral microvessels is thought to play a major role. In contrast, experimental cerebral malaria (ECM) models in mice have been thought to be distinct from human CM, and are mainly caused by inflammatory mediators. Here, to understand the spatial distribution and the potential sequestration of parasites in the whole-brain microvessels during a mouse model of ECM, we utilized the new tissue-clearing method CUBIC (Clear, Unobstructed, Brain/Body Imaging Cocktails and Computational analysis) with light-sheet fluorescent microscopy (LSFM), and reconstructed images in three dimensions (3D). We demonstrated significantly greater accumulation of Plasmodium berghei ANKA (PbANKA) parasites in the olfactory bulb (OB) of mice, compared with the other parts of the brain, including the cerebral cortex, cerebellum and brainstem. Furthermore, we show that PbANKA parasites preferentially accumulate in the brainstem when the OB is surgically removed. This study therefore not only highlights a successful application of CUBIC tissue-clearing technology to visualize the whole brain and its microvessels during ECM, but it also shows CUBIC's future potential for visualizing pathological events in the whole ECM brain at the cellular level, an achievement that would greatly advance our understanding of human cerebral malaria.
Collapse
Affiliation(s)
- Julia Matsuo-Dapaah
- Division of Malaria Immunology, Department of Microbiology and Immunology, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan.,Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Michelle Sue Jann Lee
- Division of Malaria Immunology, Department of Microbiology and Immunology, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan
| | - Ken J Ishii
- Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,Division of Vaccine Science, Department of Microbiology and Immunology, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan.,International Vaccine Design Center, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan.,Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| | - Kazuki Tainaka
- Department of System Pathology for Neurological Disorders, Center for Bioresources, Brain Research Institute, Niigata University, Niigata, Japan.,Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Osaka, Japan
| | - Cevayir Coban
- Division of Malaria Immunology, Department of Microbiology and Immunology, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan.,Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,International Vaccine Design Center, Institute of Medical Science (IMSUT), University of Tokyo, Tokyo, Japan.,Immunology Frontier Research Center (IFReC), Osaka University, Osaka, Japan
| |
Collapse
|
13
|
Amoah LE, Asare KK, Dickson D, Abankwa J, Busayo A, Bredu D, Annan S, Asumah GA, Peprah NY, Asamoah A, Laurencia Malm K. Genotypic glucose-6-phosphate dehydrogenase (G6PD) deficiency protects against Plasmodium falciparum infection in individuals living in Ghana. PLoS One 2021; 16:e0257562. [PMID: 34570821 PMCID: PMC8476035 DOI: 10.1371/journal.pone.0257562] [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: 06/20/2021] [Accepted: 09/05/2021] [Indexed: 11/18/2022] Open
Abstract
INTRODUCTION The global effort to eradicate malaria requires a drastic measure to terminate relapse from hypnozoites as well as transmission via gametocytes in malaria-endemic areas. Primaquine has been recommended for the treatment of P. falciparum gametocytes and P. vivax hypnozoites, however, its implementation is challenged by the high prevalence of G6PD deficient (G6PDd) genotypes in malaria endemic countries. The objective of this study was to profile G6PDd genotypic variants and correlate them with malaria prevalence in Ghana. METHODS A cross-sectional survey of G6PDd genotypic variants was conducted amongst suspected malaria patients attending health care facilities across the entire country. Malaria was diagnosed using microscopy whilst G6PD deficiency was determined using restriction fragment length polymorphisms at position 376 and 202 of the G6PD gene. The results were analysed using GraphPad prism. RESULTS A total of 6108 subjects were enrolled in the study with females representing 65.59% of the population. The overall prevalence of malaria was 36.31%, with malaria prevalence among G6PDd genotypic variants were 0.07% for A-A- homozygous deficient females, 1.31% and 3.03% for AA- and BA- heterozygous deficient females respectively and 2.03% for A- hemizygous deficient males. The odd ratio (OR) for detecting P. falciparum malaria infection in the A-A- genotypic variant was 0.0784 (95% CI: 0.0265-0.2319, p<0.0001). Also, P. malariae and P. ovale parasites frequently were observed in G6PD B variants relative to G6PD A- variants. CONCLUSION G6PDd genotypic variants, A-A-, AA- and A- protect against P. falciparum, P. ovale and P. malariae infection in Ghana.
Collapse
Affiliation(s)
- Linda Eva Amoah
- Dept. of Immunology, Noguchi Memorial Institute of Medical Research, University of Ghana, Accra, Ghana
| | - Kwame Kumi Asare
- Dept. of Biomedical Science, School of Allied Health Sciences, College of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Donu Dickson
- Dept. of Immunology, Noguchi Memorial Institute of Medical Research, University of Ghana, Accra, Ghana
| | - Joana Abankwa
- Dept. of Immunology, Noguchi Memorial Institute of Medical Research, University of Ghana, Accra, Ghana
| | - Abena Busayo
- Dept. of Immunology, Noguchi Memorial Institute of Medical Research, University of Ghana, Accra, Ghana
| | - Dorcas Bredu
- Dept. of Immunology, Noguchi Memorial Institute of Medical Research, University of Ghana, Accra, Ghana
| | - Sherifa Annan
- Dept. of Immunology, Noguchi Memorial Institute of Medical Research, University of Ghana, Accra, Ghana
| | | | | | | | | |
Collapse
|
14
|
Shinjyo N, Kagaya W, Pekna M. Interaction Between the Complement System and Infectious Agents - A Potential Mechanistic Link to Neurodegeneration and Dementia. Front Cell Neurosci 2021; 15:710390. [PMID: 34408631 PMCID: PMC8365172 DOI: 10.3389/fncel.2021.710390] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2021] [Accepted: 07/09/2021] [Indexed: 12/24/2022] Open
Abstract
As part of the innate immune system, complement plays a critical role in the elimination of pathogens and mobilization of cellular immune responses. In the central nervous system (CNS), many complement proteins are locally produced and regulate nervous system development and physiological processes such as neural plasticity. However, aberrant complement activation has been implicated in neurodegeneration, including Alzheimer's disease. There is a growing list of pathogens that have been shown to interact with the complement system in the brain but the short- and long-term consequences of infection-induced complement activation for neuronal functioning are largely elusive. Available evidence suggests that the infection-induced complement activation could be protective or harmful, depending on the context. Here we summarize how various infectious agents, including bacteria (e.g., Streptococcus spp.), viruses (e.g., HIV and measles virus), fungi (e.g., Candida spp.), parasites (e.g., Toxoplasma gondii and Plasmodium spp.), and prion proteins activate and manipulate the complement system in the CNS. We also discuss the potential mechanisms by which the interaction between the infectious agents and the complement system can play a role in neurodegeneration and dementia.
Collapse
Affiliation(s)
- Noriko Shinjyo
- Laboratory of Immune Homeostasis, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- School of Tropical Medicine and Global Health, Nagasaki University, Nagasaki, Japan
| | - Wataru Kagaya
- Department of Parasitology and Research Center for Infectious Disease Sciences, Graduate School of Medicine, Osaka City University, Osaka, Japan
| | - Marcela Pekna
- Laboratory of Regenerative Neuroimmunology, Center for Brain Repair, Department of Clinical Neuroscience, Institute of Neuroscience and Physiology, Sahlgrenska Academy at the University of Gothenburg, Gothenburg, Sweden
- Florey Institute of Neuroscience and Mental Health, Parkville, VIC, Australia
| |
Collapse
|
15
|
Sahra S, Jahangir A, Iqbal QZ, Mobarakai N, Glaser A, Jahangir A. Co-infection of hepatitis E virus and Plasmodium falciparum malaria: A genuine risk in sub-Saharan Africa. Parasit Vectors 2021; 14:215. [PMID: 33879247 PMCID: PMC8056203 DOI: 10.1186/s13071-021-04723-4] [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: 02/16/2021] [Accepted: 04/07/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND There is a high prevalence of malaria and viral hepatitis in South Africa. Co-infection with Plasmodium malaria (leading to cerebral malaria) and hepatitis E virus (HEV) is a rare phenomenon. CASE PRESENTATION A 33-year-old African American male with no past medical history developed altered mental status on his return from Ivory Coast. His blood tests were significant for renal and liver failure and a high Plasmodium parasite burden of 33% on the blood smear. Interestingly, he also had a positive result for hepatitis E IgM. The patient was effectively treated with aggressive hydration and intravenous (IV) artesunate. CONCLUSION Our report is the first to our knowledge in the cerebral malaria literature on a patient with hepatitis E co-infection. This exciting case emphasizes the importance of considering all kinds of endemic infectious diseases when evaluating sick returning travelers presenting to the emergency department.
Collapse
Affiliation(s)
- Syeda Sahra
- Staten Island University Hospital, Staten Island, NY, 10305, USA. .,Department of Internal Medicine, Hofstra School of Medicine, Staten Island University Hospital, 475 Seaview Avenue, Staten Island, NY, 10305, USA.
| | | | | | | | - Allison Glaser
- Staten Island University Hospital, Staten Island, NY, 10305, USA
| | | |
Collapse
|
16
|
Jajosky RP, Jajosky AN, Jajosky PG. Therapeutically-rational exchange (T-REX) of Gerbich-negative red blood cells can be evaluated in Papua New Guinea as "a rescue adjunct" for patients with Plasmodium falciparum malaria. Ther Apher Dial 2021; 25:242-247. [PMID: 32567190 PMCID: PMC11720888 DOI: 10.1111/1744-9987.13551] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/13/2020] [Accepted: 06/19/2020] [Indexed: 12/01/2022]
Abstract
"Conventional exchange transfusion"-that delivers nondescript "standard issue" units of red blood cells (RBCs)-is used worldwide to rescue dying Plasmodium falciparum (Pf) malaria patients. Recently, exchanging special malaria-resistant RBCs (T-REX) has been recommended to prevent random delivery of malaria-susceptible RBCs that promote Pf infection. Fortunately, Papua New Guinea (PNG) is well positioned to help optimize exchange as "a rescue adjunct" because (a) Gerbich-negative (GN) RBCs that resist Pf invasion are prevalent in PNG; (b) with international support, PNG has conducted outstanding malaria research; (c) PNG's scientists feel studies of GN RBCs can advance malaria therapeutics; and (d) with blood-bank support, evaluating exchange of GN RBCs is feasible in PNG. An exchange-transfusion study of GN RBCs might attract international sponsorship given the threat of expanding drug-resistance as well as growing recognition that advancing transfusion medicine and expanding blood donation could especially help Pf-infected children-immediately.
Collapse
Affiliation(s)
- Ryan Philip Jajosky
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
- Biconcavity Inc., Lilburn, Georgia
| | - Audrey N. Jajosky
- Department of Pathology, Case Western Reserve University, University Hospitals Cleveland Medical Center, Cleveland, Ohio
| | - Philip G. Jajosky
- Biconcavity Inc., Lilburn, Georgia
- Retired USPHS Officer, Centers for Disease Control and Prevention, Atlanta, Georgia
| |
Collapse
|
17
|
Lima MNN, Borba JVB, Cassiano GC, Mottin M, Mendonça SS, Silva AC, Tomaz KCP, Calit J, Bargieri DY, Costa FTM, Andrade CH. Artificial Intelligence Applied to the Rapid Identification of New Antimalarial Candidates with Dual-Stage Activity. ChemMedChem 2021; 16:1093-1103. [PMID: 33247522 DOI: 10.1002/cmdc.202000685] [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: 08/31/2002] [Revised: 11/16/2020] [Indexed: 01/06/2023]
Abstract
Increasing reports of multidrug-resistant malaria parasites urge the discovery of new effective drugs with different chemical scaffolds. Protein kinases play a key role in many cellular processes such as signal transduction and cell division, making them interesting targets in many diseases. Protein kinase 7 (PK7) is an orphan kinase from the Plasmodium genus, essential for the sporogonic cycle of these parasites. Here, we applied a robust and integrative artificial intelligence-assisted virtual-screening (VS) approach using shape-based and machine learning models to identify new potential PK7 inhibitors with in vitro antiplasmodial activity. Eight virtual hits were experimentally evaluated, and compound LabMol-167 inhibited ookinete conversion of Plasmodium berghei and blood stages of Plasmodium falciparum at nanomolar concentrations with low cytotoxicity in mammalian cells. As PK7 does not have an essential role in the Plasmodium blood stage and our virtual screening strategy aimed for both PK7 and blood-stage inhibition, we conducted an in silico target fishing approach and propose that this compound might also inhibit P. falciparum PK5, acting as a possible dual-target inhibitor. Finally, docking studies of LabMol-167 with P. falciparum PK7 and PK5 proteins highlighted key interactions for further hit-to lead optimization.
Collapse
Affiliation(s)
- Marilia N N Lima
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Rua 240, qd. 87, Goiânia, GO, 74605-170, Brazil
| | - Joyce V B Borba
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Rua 240, qd. 87, Goiânia, GO, 74605-170, Brazil.,Laboratory of Tropical Diseases - Prof. Dr. Luiz Jacintho da Silva, Department of Genetics Evolution, Microbiology and Immunology, Institute of Biology, 13083-970, Campinas, SP, Brazil
| | - Gustavo C Cassiano
- Laboratory of Tropical Diseases - Prof. Dr. Luiz Jacintho da Silva, Department of Genetics Evolution, Microbiology and Immunology, Institute of Biology, 13083-970, Campinas, SP, Brazil.,Global Health and Tropical Medicine (GHTM), Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Lisbon, Portugal
| | - Melina Mottin
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Rua 240, qd. 87, Goiânia, GO, 74605-170, Brazil
| | - Sabrina S Mendonça
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Rua 240, qd. 87, Goiânia, GO, 74605-170, Brazil
| | - Arthur C Silva
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Rua 240, qd. 87, Goiânia, GO, 74605-170, Brazil
| | - Kaira C P Tomaz
- Laboratory of Tropical Diseases - Prof. Dr. Luiz Jacintho da Silva, Department of Genetics Evolution, Microbiology and Immunology, Institute of Biology, 13083-970, Campinas, SP, Brazil
| | - Juliana Calit
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, São Paulo, SP, Brazil
| | - Daniel Y Bargieri
- Department of Parasitology, Institute of Biomedical Sciences, University of São Paulo, 05508-000, São Paulo, SP, Brazil
| | - Fabio T M Costa
- Laboratory of Tropical Diseases - Prof. Dr. Luiz Jacintho da Silva, Department of Genetics Evolution, Microbiology and Immunology, Institute of Biology, 13083-970, Campinas, SP, Brazil
| | - Carolina H Andrade
- LabMol - Laboratory for Molecular Modeling and Drug Design, Faculty of Pharmacy, Federal University of Goias, Rua 240, qd. 87, Goiânia, GO, 74605-170, Brazil.,Laboratory of Tropical Diseases - Prof. Dr. Luiz Jacintho da Silva, Department of Genetics Evolution, Microbiology and Immunology, Institute of Biology, 13083-970, Campinas, SP, Brazil
| |
Collapse
|
18
|
Wu X, Thylur RP, Dayanand KK, Punnath K, Norbury CC, Gowda DC. IL-4 Treatment Mitigates Experimental Cerebral Malaria by Reducing Parasitemia, Dampening Inflammation, and Lessening the Cytotoxicity of T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 206:118-131. [PMID: 33239419 DOI: 10.4049/jimmunol.2000779] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/28/2020] [Indexed: 12/22/2022]
Abstract
Cytokine responses to malaria play important roles in both protective immunity development and pathogenesis. Although the roles of cytokines such as TNF-α, IL-12, IFN-γ, and IL-10 in immunity and pathogenesis to the blood stage malaria are largely known, the role of IL-4 remains less understood. IL-4 targets many cell types and induces multiple effects, including cell proliferation, gene expression, protection from apoptosis, and immune regulation. Accordingly, IL-4 has been exploited as a therapeutic for several inflammatory diseases. Malaria caused by Plasmodium falciparum manifests in many organ-specific fatal pathologies, including cerebral malaria (CM), driven by a high parasite load, leading to parasite sequestration in organs and consequent excessive inflammatory responses and endothelial damage. We investigated the therapeutic potential of IL-4 against fatal malaria in Plasmodium berghei ANKA-infected C57BL/6J mice, an experimental CM model. IL-4 treatment significantly reduced parasitemia, CM pathology, and mortality. The therapeutic effect of IL-4 is mediated through multiple mechanisms, including enhanced parasite clearance mediated by upregulation of phagocytic receptors and increased IgM production, and decreased brain inflammatory responses, including reduced chemokine (CXCL10) production, reduced chemokine receptor (CXCR3) and adhesion molecule (LFA-1) expression by T cells, and downregulation of cytotoxic T cell lytic potential. IL-4 treatment markedly reduced the infiltration of CD8+ T cells and brain pathology. STAT6, PI3K-Akt-NF-κB, and Src signaling mediated the cellular and molecular events that contributed to the IL-4-dependent decrease in parasitemia. Overall, our results provide mechanistic insights into how IL-4 treatment mitigates experimental CM and have implications in developing treatment strategies for organ-specific fatal malaria.
Collapse
Affiliation(s)
- Xianzhu Wu
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Ramesh P Thylur
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Kiran K Dayanand
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Kishore Punnath
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Christopher C Norbury
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - D Channe Gowda
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| |
Collapse
|
19
|
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: 8] [Impact Index Per Article: 1.6] [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
|
20
|
Golenser J, Salaymeh N, Higazi AA, Alyan M, Daif M, Dzikowski R, Domb AJ. Treatment of Experimental Cerebral Malaria by Slow Release of Artemisone From Injectable Pasty Formulation. Front Pharmacol 2020; 11:846. [PMID: 32595499 PMCID: PMC7303303 DOI: 10.3389/fphar.2020.00846] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 05/22/2020] [Indexed: 12/26/2022] Open
Abstract
Malaria caused by Plasmodium falciparum causes numerous cases of morbidity with about 400,000 deaths yearly owing, mainly, to inflammation leading to cerebral malaria (CM). CM conventionally is treated by repetitive administration of anti-plasmodial drugs and supportive non-specific drugs, for about a week. A mouse model of CM caused by Plasmodium berghei ANKA, in which brain and systemic clinical pathologies occur followed by sudden death within about a week, was used to study the effect of artemisone, a relatively new artemisinin, within an injectable pasty polymer formulated for its controlled release. The parasites were exposed to the drug over several days at a non-toxic concentrations for the mice but high enough to affect the parasites. Artemisone was also tested in cultures of bacteria, cancer cells and P. falciparum to evaluate the specificity and suitability of these cells for examining the release of artemisone from its carrier. Cultures of P. falciparum were the most suitable. Artemisone released from subcutaneous injected poly(sebacic acid-ricinoleic acid) (PSARA) pasty polymer, reduced parasitemias in infected mice, prolonged survival and prevented death in most of the infected mice. Successful prophylactic treatment before infection proved that there was a slow release of the drug for about a week, which contrasts with the three hour half-life that occurs after injection of just the drug. Treatment with artemisone within the polymer, even at a late stage of the disease, helped to prevent or, at least, delay accompanying severe symptoms. In some cases, treatment prevented death of CM and the mice died later of anemia. Postponing the severe clinical symptoms is also beneficial in cases of human malaria, giving more time for an appropriate diagnosis and treatment before severe symptoms appear. The method presented here may also be useful for combination therapy of anti-plasmodial and immunomodulatory drugs.
Collapse
Affiliation(s)
- Jacob Golenser
- Department of Microbiology and Molecular Genetics, The Kuvin Centre for the Study of Infectious and Tropical Diseases, Faculty of Medicine, the Hebrew University (HU), Jerusalem, Israel
| | - Nadeen Salaymeh
- Department of Microbiology and Molecular Genetics, The Kuvin Centre for the Study of Infectious and Tropical Diseases, Faculty of Medicine, the Hebrew University (HU), Jerusalem, Israel
| | | | - Mohammed Alyan
- Department of Microbiology and Molecular Genetics, The Kuvin Centre for the Study of Infectious and Tropical Diseases, Faculty of Medicine, the Hebrew University (HU), Jerusalem, Israel
- Faculty of Medicine, School of Pharmacy, Institute of Drug Research, HU, Jerusalem, Israel
| | - Mahran Daif
- Faculty of Medicine, School of Pharmacy, Institute of Drug Research, HU, Jerusalem, Israel
| | - Ron Dzikowski
- Department of Microbiology and Molecular Genetics, The Kuvin Centre for the Study of Infectious and Tropical Diseases, Faculty of Medicine, the Hebrew University (HU), Jerusalem, Israel
| | - Abraham J. Domb
- Faculty of Medicine, School of Pharmacy, Institute of Drug Research, HU, Jerusalem, Israel
| |
Collapse
|
21
|
Siddiqui AJ, Adnan M, Jahan S, Redman W, Saeed M, Patel M. Neurological disorder and psychosocial aspects of cerebral malaria: what is new on its pathogenesis and complications? A minireview. Folia Parasitol (Praha) 2020; 67. [PMID: 32636351 DOI: 10.14411/fp.2020.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Accepted: 03/06/2020] [Indexed: 01/21/2023]
Abstract
Recently, malaria is remain considered as the most prevalent infectious disease, affecting the human health globally. High morbidity and mortality worldwide is often allied with cerebral malaria (CM) based disorders of the central nervous system, especially across many tropical and sub-tropical regions. These disorders are characterised by the infection of Plasmodium species, which leads to acute or chronic neurological disorders, even after having active/effective antimalarial drugs. Furthermore, even during the treatment, individual remain sensitive for neurological impairments in the form of decrease blood flow and vascular obstruction in brain including many more other changes. This review briefly explains and update on the epidemiology, burden of disease, pathogenesis and role of CM in neurological disorders with behaviour and function in mouse and human models. Moreover, the social stigma, which plays an important role in neurological disorders and a factor for assessing CM, is also discussed in this review.
Collapse
Affiliation(s)
| | | | - Sadaf Jahan
- Department of Medical Laboratory, College of Applied Medical Sciences, Majmaah University, Majmaah city, Saudi Arabia
| | - Whitni Redman
- Surgery Department, Division of Biomedical Research, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Mοhd Saeed
- Department of Biology, College of Science, University of Hail, Hail, PO Box 2440, Saudi Arabia
| | - Mitesh Patel
- Bapalal Vaidya Botanical Research Centre, Department of Biosciences, Veer Narmad South Gujarat University, Surat, Gujarat, India
| |
Collapse
|
22
|
Oltz EM. Neuroimmunology: To Sense and Protect. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2020; 204:239-240. [PMID: 31907263 DOI: 10.4049/jimmunol.1990024] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|