1
|
Gorki V, Walter NS, Chauhan M, Dhingra N, Bagai U, Kaur S. Gefitinib as an antimalarial: unveiling its therapeutic potential. Inflammopharmacology 2025; 33:1357-1379. [PMID: 40019687 DOI: 10.1007/s10787-025-01682-5] [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: 01/27/2025] [Accepted: 01/31/2025] [Indexed: 03/01/2025]
Abstract
Resistant strains of Plasmodium spp. pose a great threat to healthcare. Drug repurposing is a smart, and an effective way to look for new alternatives for different ailments including malaria. Protein tyrosine kinases (PTKs) play a crucial role in growth, maturation as well as differentiation of Plasmodium and this study explores antimalarial activity of PTKs inhibitor gefitinib using in silico and experimental approaches. The drug showed considerable inhibitory activity against P. falciparum 3D7 (IC50 0.49 µg/mL) and RKL-9 (IC50 0.83 µg/mL) strains. Isobologram analysis revealed substantial synergism between gefitinib and artesunate. Gefitinib illustrated highest negative D-score towards phosphoethanolamine methyltransferase followed by PfPK5 and CDPK1. Its acute toxicity was 4 g/kg. Gefitinib (100 mg/kg) exhibited a dose-dependent curative activity against P. berghei with 91.09% chemo-suppression and the combination of gefitinib 100 mg/kg and AS 50 mg/kg exhibited complete parasite clearance with no recrudescence which was also evidenced by cytokine analysis, biochemical as well as histopathological studies. At length, gefitinib illustrated considerable antiplasmodial action by targeting phosphoethanolamine methyltransferase, PfPK5 and CDPK1. The combination of gefitinib (100 mg/kg) and AS (50 mg/kg) holds promise for malaria treatment. Further, research is being done to evaluate its pharmacokinetic properties.
Collapse
Affiliation(s)
- Varun Gorki
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, 160014, India
- Department of Gastroenterology, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Neha Sylvia Walter
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, 160014, India
- Department of Biophysics, Post Graduate Institute of Medical Education and Research, Chandigarh, 160012, India
| | - Monika Chauhan
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Neelima Dhingra
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | - Upma Bagai
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, 160014, India
| | - Sukhbir Kaur
- Parasitology Laboratory, Department of Zoology, Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
2
|
Chen J, Bai Y, He X, Xiao W, Chen L, Wong YK, Wang C, Gao P, Cheng G, Xu L, Yang C, Liao F, Han G, Sun J, Xu C, Wang J. The spatiotemporal transcriptional profiling of murine brain during cerebral malaria progression and after artemisinin treatment. Nat Commun 2025; 16:1540. [PMID: 39934099 PMCID: PMC11814382 DOI: 10.1038/s41467-024-52223-7] [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: 09/04/2023] [Accepted: 08/28/2024] [Indexed: 02/13/2025] Open
Abstract
Cerebral malaria (CM) is a severe encephalopathy caused by Plasmodium parasite infection, resulting in thousands of annual deaths and neuro-cognitive sequelae even after anti-malarial drugs treatment. Despite efforts to dissect the mechanism, the cellular transcriptomic reprogramming within the spatial context remains elusive. Here, we constructed single-cell and spatial transcriptome atlases of experimental CM (ECM) male murine brain tissues with or without artesunate (ART) treatment. We identified activated inflammatory endothelial cells during ECM, characterized by a disrupted blood-brain barrier, increased antigen presentation, and leukocyte adhesion. We also observed that inflammatory microglia enhance antigen presentation pathway such as MHC-I to CD8+ cytotoxic T cells. The latter underwent an inflammatory state transition with up-regulated cytokine expression and cytotoxic activity. Multi-omics analysis revealed that the activated interferon-gamma response of injured neurons during ECM and persisted after ART treatment. Overall, our research provides valuable resources for understanding malaria parasite-host interaction mechanisms and adjuvant therapy development.
Collapse
Affiliation(s)
- Jiayun Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
| | - Yunmeng Bai
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Xueling He
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Wei Xiao
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China
- Department of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China
| | - Lina Chen
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Yin Kwan Wong
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Chen Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Peng Gao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China
| | - Guangqing Cheng
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Liting Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Chuanbin Yang
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China
| | - Fulong Liao
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China
| | - Guang Han
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China
| | - Jichao Sun
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Chengchao Xu
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
| | - Jigang Wang
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Artemisinin Research Center, and Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
- Department of Critical Care Medicine, Guangdong Provincial Clinical Research Center for Geriatrics, Shenzhen Clinical Research Center for Geriatric, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, 518020, Guangdong, China.
- Center for Drug Research and Development, Guangdong Provincial Key Laboratory for Research and Evaluation of Pharmaceutical Preparations, Guangdong Pharmaceutical University, Guangzhou, 510006, Guangdong, China.
- Department of Traditional Chinese Medicine and School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, 510515, Guangdong, China.
- State Key Laboratory of Antiviral Drugs, School of Pharmacy, Henan University, Kaifeng, 475004, Henan, China.
| |
Collapse
|
3
|
Céspedes N, Tsolis RM, Piliponsky AM, Luckhart S. The type 2 immune response in gut homeostasis and parasite transmission in malaria. Trends Parasitol 2025; 41:38-51. [PMID: 39658487 DOI: 10.1016/j.pt.2024.11.007] [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: 10/31/2024] [Revised: 11/18/2024] [Accepted: 11/18/2024] [Indexed: 12/12/2024]
Abstract
Malaria predisposes to concomitant bacteremia, resulting in increased mortality risk. Previous studies indicated that malaria causes structural changes in the intestine, induces tolerogenic immune responses, inhibits neutrophil recruitment, suppresses innate synthesis of IFN-γ, dysregulates mast cells (MCs) and basophils, and induces Th2-type immune responses. These can reduce parasite control while increasing enteropathogenic dissemination. Moreover, there is growing evidence that Th2 immunity, while protecting the host from overwhelming inflammation, may also contribute to increased parasite transmission. This review explores the roles of the regulatory immune response in bacterial coinfections and parasite transmission in malaria.
Collapse
Affiliation(s)
- Nora Céspedes
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID, USA.
| | - Renée M Tsolis
- Department of Medical Microbiology and Immunology, University of California-Davis, Davis, CA, USA
| | - Adrian M Piliponsky
- Department of Pediatrics and Department of Pathology, Seattle Children's Research Institute, Seattle, WA, USA
| | - Shirley Luckhart
- Department of Entomology, Plant Pathology and Nematology, University of Idaho, Moscow, ID, USA; Department of Biological Sciences, University of Idaho, Moscow, ID, USA
| |
Collapse
|
4
|
Owumi S, Olanlokun JO, Wu B, Duro-Ladipo AM, Oyelere SE, Khan SI, Oyelere AK. Elucidation of the Active Agents in a West African Ground Herbal Medicine Formulation That Elicit Antimalarial Activities in In Vitro and In Vivo Models. Molecules 2024; 29:5658. [PMID: 39683816 DOI: 10.3390/molecules29235658] [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: 10/16/2024] [Revised: 11/25/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
Agunmu (ground herbal medicine) is a form of West African traditional medicine consisting of a cocktail of herbs. The goal of this study is to evaluate a formulation of Agunmu made from M. indica, A. repens, E. chlorantha, A. boonei, and B. ferruginea, sold in the open market and commonly used for the treatment of malaria by the locals, for its antimalarial effects and to determine the active principles that may contribute to the antimalarial effect. The ethanolic extract obtained from this formulation (Ag-Iba) was analyzed, using TLC, LC-MS, and Tandem-MS techniques, to determine its phytochemical properties. The extract was tested in vitro against representative bacteria strains, cancer and normal human cell lines, and susceptible (D6) and resistant (W2) Plasmodium falciparum. In subsequent in vivo experiments, graded doses of the extract were used to treat mice infected with chloroquine-susceptible (NK-65) and chloroquine-resistant (ANKA) strains of Plasmodium berghei. Bacteria growth was monitored with a disc diffusion assay, cancer cell viability was determined with MTS assay, and percentage parasitemia and parasite clearance were determined by microscopy. Bound heme content, host mitochondria permeability transition (mPT) pore opening, F0F1-ATPase, and lipid peroxidation were determined via spectrophotometry. Indices of oxidative stress, anti-oxidant activities, toxicity, cell death, and inflammatory responses were obtained using biochemical and ELISA techniques. The histology of the liver and spleen was performed using the standard method. We elucidated the structures of the critical active principles in the extract to be flavonoids: kaempferol, quercetin, myricetin, and their glycosides with little or no detectable levels of the toxic Aristolochic acids that are found in Aristolochia repens, one of the components of the formulation. The extract also showed anti-plasmodial activity in in vitro and in vivo models. Furthermore, the extract dose-dependently decreased mitochondrial dysfunction, cell death, and inflammatory and oxidative damage but increased antioxidant potentials. Presumably, the active principles in the extract work as a combinatorial therapy to elicit potent antimalarial activity. Overall, our study unraveled the active components from a commercial herbal formulation that could be reformulated for antimalarial therapy.
Collapse
Affiliation(s)
- Solomon Owumi
- Cancer Research and Molecular Biology Laboratories, University of Ibadan, Ibadan 200005, Nigeria
| | - John O Olanlokun
- Laboratories for Biomembrane and Biotechnology Research, Department of Biochemistry, University of Ibadan, Ibadan 200005, Nigeria
| | - Bocheng Wu
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
| | | | | | - Shabana I Khan
- NCNPR, School of Pharmacy, University of Mississippi, University, MS 38677, USA
| | - Adegboyega K Oyelere
- School of Chemistry and Biochemistry, Georgia Institute of Technology, Atlanta, GA 30332, USA
- Parker H. Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA 30332, USA
| |
Collapse
|
5
|
Cerdeira CD, Brigagão MRPL. Targeting Macrophage Polarization in Infectious Diseases: M1/M2 Functional Profiles, Immune Signaling and Microbial Virulence Factors. Immunol Invest 2024; 53:1030-1091. [PMID: 38913937 DOI: 10.1080/08820139.2024.2367682] [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] [Indexed: 06/26/2024]
Abstract
INTRODUCTION An event of increasing interest during host-pathogen interactions is the polarization of patrolling/naive monocytes (MOs) into macrophage subsets (MФs). Therapeutic strategies aimed at modulating this event are under investigation. METHODS This review focuses on the mechanisms of induction/development and profile of MФs polarized toward classically proinflammatory (M1) or alternatively anti-inflammatory (M2) phenotypes in response to bacteria, fungi, parasites, and viruses. RESULTS AND DISCUSSION It highlights nuclear, cytoplasmic, and cell surface receptors (pattern recognition receptors/PPRs), microenvironmental mediators, and immune signaling. MФs polarize into phenotypes: M1 MФs, activated by IFN-γ, pathogen-associated molecular patterns (PAMPs, e.g. lipopolysaccharide) and membrane-bound PPRs ligands (TLRs/CLRs ligands); or M2 MФs, induced by interleukins (ILs-4, -10 and -13), antigen-antibody complexes, and helminth PAMPs. Polarization toward M1 and M2 profiles evolve in a pathogen-specific manner, with or without canonicity, and can vary widely. Ultimately, this can result in varying degrees of host protection or more severe disease outcome. On the one hand, the host is driving effective MФs polarization (M1 or M2); but on the other hand, microorganisms may skew the polarization through virulence factors to increase pathogenicity. Cellular/genomic reprogramming also ensures plasticity of M1/M2 phenotypes. Because modulation of polarization can occur at multiple points, new insights and emerging perspectives may have clinical implications during the inflammation-to-resolution transition; translated into practical applications as for therapeutic/vaccine design target to boost microbicidal response (M1, e.g. triggering oxidative burst) with specifics PAMPs/IFN-γ or promote tissue repair (M2, increasing arginase activity) via immunotherapy.
Collapse
|
6
|
Pinheiro AAS, Caruso-Neves C, Rocco PRM. Extracellular vesicles in malaria: Pathogenesis, diagnosis and therapy. CURRENT TOPICS IN MEMBRANES 2024; 94:107-132. [PMID: 39370204 DOI: 10.1016/bs.ctm.2024.06.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Malaria is a life-threatening disease caused by parasites from the genus Plasmodium. Five species can cause malaria in humans, with Plasmodium vivax being the most common in many countries and Plasmodium falciparum having the highest lethality, which can lead to cerebral malaria. Extracellular vesicles (EVs) are in focus in malaria research to better understand pathogenesis, diagnosis, therapy, and prognosis. Malaria-causing parasites use EVs to transfer their molecules to host cells, a mechanism that significantly contributes to parasite survival and successful infection. EVs have thus emerged as an essential component of the immunopathological cascade of malaria, playing a pivotal role in disease progression and severity. This chapter discusses the epidemiology and pathogenesis of malaria and the role of EVs as new diagnostic and therapeutic tools, emphasizing their potential clinical significance.
Collapse
Affiliation(s)
- Ana Acacia S Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil
| | - Celso Caruso-Neves
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil
| | - Patricia R M Rocco
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, Brazil; Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/Fundação Carlos Chagas Filho de Amparo à Pesquisa do Estado do Rio de Janeiro (FAPERJ), Rio de Janeiro, RJ, Brazil; National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, RJ, Brazil.
| |
Collapse
|
7
|
Chen P, Ding N, Pan D, Chen X, Li S, Luo Y, Chen Z, Xu Y, Zhu X, Wang K, Zou W. PET imaging for the early evaluation of ocular inflammation in diabetic rats by using [ 18F]-DPA-714. Exp Eye Res 2024; 245:109986. [PMID: 38945519 DOI: 10.1016/j.exer.2024.109986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 06/21/2024] [Accepted: 06/27/2024] [Indexed: 07/02/2024]
Abstract
Ocular complications of diabetes mellitus (DM) are the leading cause of vision loss. Ocular inflammation often occurs in the early stage of DM; however, there are no proven quantitative methods to evaluate the inflammatory status of eyes in DM. The 18 kDa translocator protein (TSPO) is an evolutionarily conserved cholesterol binding protein localized in the outer mitochondrial membrane. It is a biomarker of activated microglia/macrophages; however, its role in ocular inflammation is unclear. In this study, fluorine-18-DPA-714 ([18F]-DPA-714) was evaluated as a specific TSPO probe by cell uptake, cell binding assays and micro positron emission tomography (microPET) imaging in both in vitro and in vivo models. Primary microglia/macrophages (PMs) extracted from the cornea, retina, choroid or sclera of neonatal rats with or without high glucose (50 mM) treatment were used as the in vitro model. Sprague-Dawley (SD) rats that received an intraperitoneal administration of streptozotocin (STZ, 60 mg/kg once) were used as the in vivo model. Increased cell uptake and high binding affinity of [18F]-DPA-714 were observed in primary PMs under hyperglycemic stress. These findings were consistent with cellular morphological changes, cell activation, and TSPO up-regulation. [18F]-DPA-714 PET imaging and biodistribution in the eyes of DM rats revealed that inflammation initiates in microglia/macrophages in the early stages (3 weeks and 6 weeks), corresponding with up-regulated TSPO levels. Thus, [18F]-DPA-714 microPET imaging may be an effective approach for the early evaluation of ocular inflammation in DM.
Collapse
Affiliation(s)
- Peng Chen
- Department of Ophthalmology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China; Department of Ophthalmology, Jintan Affiliated Hospital of Jiangsu University, Changzhou, Jiangsu, China
| | - Nannan Ding
- Department of Ophthalmology, Wuxi No.2 People's Hospital, Jiangnan University Medical Center (JUMC), Wuxi, Jiangsu, China; Department of Ophthalmology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China; Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong Medical University, Wuxi, Jiangsu, China
| | - Donghui Pan
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xuelian Chen
- Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong Medical University, Wuxi, Jiangsu, China; Department of Ophthalmology, PuNan Branch of Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - ShiYi Li
- Department of Ophthalmology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China; Department of Ophthalmology, Jingjiang People's Hospital Affiliated to Yangzhou University, Taizhou, Jiangsu, China
| | - Yidan Luo
- Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong Medical University, Wuxi, Jiangsu, China
| | - Ziqing Chen
- Department of Ophthalmology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China
| | - Yuping Xu
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xue Zhu
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ke Wang
- National Health Commission (NHC) Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, Jiangsu, China; Department of Radiopharmaceuticals, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Wenjun Zou
- Department of Ophthalmology, Wuxi No.2 People's Hospital, Jiangnan University Medical Center (JUMC), Wuxi, Jiangsu, China; Department of Ophthalmology, The Affiliated Wuxi No.2 People's Hospital of Nanjing Medical University, Wuxi, Jiangsu, China; Department of Ophthalmology, Affiliated Wuxi Clinical College of Nantong Medical University, Wuxi, Jiangsu, China.
| |
Collapse
|
8
|
Dooley NL, Chabikwa TG, Pava Z, Loughland JR, Hamelink J, Berry K, Andrew D, Soon MSF, SheelaNair A, Piera KA, William T, Barber BE, Grigg MJ, Engwerda CR, Lopez JA, Anstey NM, Boyle MJ. Single cell transcriptomics shows that malaria promotes unique regulatory responses across multiple immune cell subsets. Nat Commun 2023; 14:7387. [PMID: 37968278 PMCID: PMC10651914 DOI: 10.1038/s41467-023-43181-7] [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: 11/23/2022] [Accepted: 11/02/2023] [Indexed: 11/17/2023] Open
Abstract
Plasmodium falciparum malaria drives immunoregulatory responses across multiple cell subsets, which protects from immunopathogenesis, but also hampers the development of effective anti-parasitic immunity. Understanding malaria induced tolerogenic responses in specific cell subsets may inform development of strategies to boost protective immunity during drug treatment and vaccination. Here, we analyse the immune landscape with single cell RNA sequencing during P. falciparum malaria. We identify cell type specific responses in sub-clustered major immune cell types. Malaria is associated with an increase in immunosuppressive monocytes, alongside NK and γδ T cells which up-regulate tolerogenic markers. IL-10-producing Tr1 CD4 T cells and IL-10-producing regulatory B cells are also induced. Type I interferon responses are identified across all cell types, suggesting Type I interferon signalling may be linked to induction of immunoregulatory networks during malaria. These findings provide insights into cell-specific and shared immunoregulatory changes during malaria and provide a data resource for further analysis.
Collapse
Affiliation(s)
- Nicholas L Dooley
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Environment and Sciences, Griffith University, Brisbane, QLD, Australia
| | | | - Zuleima Pava
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | | | - Julianne Hamelink
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- University of Queensland, Brisbane, QLD, Australia
| | - Kiana Berry
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Queensland University of Technology, Brisbane, QLD, Australia
| | - Dean Andrew
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Megan S F Soon
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Arya SheelaNair
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Kim A Piera
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
| | - Timothy William
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Program, Kota Kinabalu, Sabah, Malaysia
- Subang Jaya Medical Centre, Selangor, Malaysia
| | - Bridget E Barber
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Program, Kota Kinabalu, Sabah, Malaysia
| | - Matthew J Grigg
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Program, Kota Kinabalu, Sabah, Malaysia
| | | | - J Alejandro Lopez
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
- School of Environment and Sciences, Griffith University, Brisbane, QLD, Australia
| | - Nicholas M Anstey
- Menzies School of Health Research, Charles Darwin University, Darwin, NT, Australia
- Infectious Diseases Society Kota Kinabalu Sabah-Menzies School of Health Research Program, Kota Kinabalu, Sabah, Malaysia
| | - Michelle J Boyle
- QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia.
- School of Environment and Sciences, Griffith University, Brisbane, QLD, Australia.
- University of Queensland, Brisbane, QLD, Australia.
- Queensland University of Technology, Brisbane, QLD, Australia.
- Burnet Institute, Melbourne, VIC, Australia.
| |
Collapse
|
9
|
Szymula A, Samayoa-Reyes G, Ogolla S, Liu B, Li S, George A, Van Sciver N, Rochford R, Simas JP, Kaye KM. Macrophages drive KSHV B cell latency. Cell Rep 2023; 42:112767. [PMID: 37440412 PMCID: PMC10528218 DOI: 10.1016/j.celrep.2023.112767] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Revised: 06/06/2023] [Accepted: 06/22/2023] [Indexed: 07/15/2023] Open
Abstract
Kaposi's sarcoma herpesvirus (KSHV) establishes lifelong infection and persists in latently infected B cells. Paradoxically, in vitro B cell infection is inefficient, and cells rapidly die, suggesting the absence of necessary factor(s). KSHV epidemiology unexpectedly mirrors that of malaria and certain helminthic infections, while other herpesviruses are ubiquitous. Elevated circulating monocytes are common in these parasitic infections. Here, we show that KSHV infection of monocytes or M-CSF-differentiated (M2) macrophages is highly efficient. Proteomic analyses demonstrate that infection induces macrophage production of B cell chemoattractants and activating factor. We find that KSHV acts with monocytes or M2 macrophages to stimulate B cell survival, proliferation, and plasmablast differentiation. Further, macrophages drive infected plasma cell differentiation and long-term viral latency. In Kenya, where KSHV is endemic, we find elevated monocyte levels in children with malaria. These findings demonstrate a role for mononuclear phagocytes in KSHV B cell latency and suggest that mononuclear phagocyte abundance may underlie KSHV's geographic disparity.
Collapse
Affiliation(s)
- Agnieszka Szymula
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Gabriela Samayoa-Reyes
- Department of Immunology and Microbiology, School of Medicine, University of Colorado, Aurora, CO 80045, USA
| | - Sidney Ogolla
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu 40100, Kenya
| | - Bing Liu
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Shijun Li
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Athira George
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Nicholas Van Sciver
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Program in Virology, Harvard Medical School, Boston, MA 02115, USA
| | - Rosemary Rochford
- Center for Global Health Research, Kenya Medical Research Institute, Kisumu 40100, Kenya
| | - J Pedro Simas
- Instituto de Medicina Molecular, Avenida Professor Egas Moniz, 1649-028 Lisboa, Portugal; Católica Biomedical Research, Católica Medical School, Universidade Católica Portuguesa, Palma de Cima, 1649-023 Lisboa, Portugal.
| | - Kenneth M Kaye
- Departments of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA; Program in Virology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA 02142, USA.
| |
Collapse
|
10
|
Starikova EA, Rubinstein AA, Mammedova JT, Isakov DV, Kudryavtsev IV. Regulated Arginine Metabolism in Immunopathogenesis of a Wide Range of Diseases: Is There a Way to Pass between Scylla and Charybdis? Curr Issues Mol Biol 2023; 45:3525-3551. [PMID: 37185755 PMCID: PMC10137093 DOI: 10.3390/cimb45040231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 04/12/2023] [Accepted: 04/14/2023] [Indexed: 05/17/2023] Open
Abstract
More than a century has passed since arginine was discovered, but the metabolism of the amino acid never ceases to amaze researchers. Being a conditionally essential amino acid, arginine performs many important homeostatic functions in the body; it is involved in the regulation of the cardiovascular system and regeneration processes. In recent years, more and more facts have been accumulating that demonstrate a close relationship between arginine metabolic pathways and immune responses. This opens new opportunities for the development of original ways to treat diseases associated with suppressed or increased activity of the immune system. In this review, we analyze the literature describing the role of arginine metabolism in the immunopathogenesis of a wide range of diseases, and discuss arginine-dependent processes as a possible target for therapeutic approaches.
Collapse
Affiliation(s)
- Eleonora A Starikova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L'va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Artem A Rubinstein
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
| | - Jennet T Mammedova
- Laboratory of General Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
| | - Dmitry V Isakov
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L'va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| | - Igor V Kudryavtsev
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia
- School of Biomedicine, Far Eastern Federal University, FEFU Campus, 10 Ajax Bay, Russky Island, 690922 Vladivostok, Russia
| |
Collapse
|
11
|
Khowawisetsut L, Vimonpatranon S, Lekmanee K, Sawasdipokin H, Srimark N, Chotivanich K, Pattanapanyasat K. Differential Effect of Extracellular Vesicles Derived from Plasmodium falciparum-Infected Red Blood Cells on Monocyte Polarization. Int J Mol Sci 2023; 24:2631. [PMID: 36768950 PMCID: PMC9916780 DOI: 10.3390/ijms24032631] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/22/2023] [Accepted: 01/25/2023] [Indexed: 01/31/2023] Open
Abstract
Malaria is a life-threatening tropical arthropod-borne disease caused by Plasmodium spp. Monocytes are the primary immune cells to eliminate malaria-infected red blood cells. Thus, the monocyte's functions are one of the crucial factors in controlling parasite growth. It is reasoned that the activation or modulation of monocyte function by parasite products might dictate the rate of disease progression. Extracellular vesicles (EVs), microvesicles, and exosomes, released from infected red blood cells, mediate intercellular communication and control the recipient cell function. This study aimed to investigate the physical characteristics of EVs derived from culture-adapted P. falciparum isolates (Pf-EVs) from different clinical malaria outcomes and their impact on monocyte polarization. The results showed that all P. falciparum strains released similar amounts of EVs with some variation in size characteristics. The effect of Pf-EV stimulation on M1/M2 monocyte polarization revealed a more pronounced effect on CD14+CD16+ intermediate monocytes than the CD14+CD16- classical monocytes with a marked induction of Pf-EVs from a severe malaria strain. However, no difference in the levels of microRNAs (miR), miR-451a, miR-486, and miR-92a among Pf-EVs derived from virulent and nonvirulent strains was found, suggesting that miR in Pf-EVs might not be a significant factor in driving M2-like monocyte polarization. Future studies on other biomolecules in Pf-EVs derived from the P. falciparum strain with high virulence that induce M2-like polarization are therefore recommended.
Collapse
Affiliation(s)
- Ladawan Khowawisetsut
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Sinmanus Vimonpatranon
- Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kittima Lekmanee
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Hathai Sawasdipokin
- Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Narinee Srimark
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kesinee Chotivanich
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok 10400, Thailand
| | - Kovit Pattanapanyasat
- Siriraj Center of Research Excellence for Microparticle and Exosome in Diseases, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| |
Collapse
|
12
|
Varela ELP, Gomes ARQ, da Silva Barbosa dos Santos A, de Carvalho EP, Vale VV, Percário S. Potential Benefits of Lycopene Consumption: Rationale for Using It as an Adjuvant Treatment for Malaria Patients and in Several Diseases. Nutrients 2022; 14:5303. [PMID: 36558462 PMCID: PMC9787606 DOI: 10.3390/nu14245303] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 12/10/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022] Open
Abstract
Malaria is a disease that affects thousands of people around the world every year. Its pathogenesis is associated with the production of reactive oxygen and nitrogen species (RONS) and lower levels of micronutrients and antioxidants. Patients under drug treatment have high levels of oxidative stress biomarkers in the body tissues, which limits the use of these drugs. Therefore, several studies have suggested that RONS inhibition may represent an adjuvant therapeutic strategy in the treatment of these patients by increasing the antioxidant capacity of the host. In this sense, supplementation with antioxidant compounds such as zinc, selenium, and vitamins A, C, and E has been suggested as part of the treatment. Among dietary antioxidants, lycopene is the most powerful antioxidant among the main carotenoids. This review aimed to describe the main mechanisms inducing oxidative stress during malaria, highlighting the production of RONS as a defense mechanism against the infection induced by the ischemia-reperfusion syndrome, the metabolism of the parasite, and the metabolism of antimalarial drugs. Furthermore, the effects of lycopene on several diseases in which oxidative stress is implicated as a cause are outlined, providing information about its mechanism of action, and providing an evidence-based justification for its supplementation in malaria.
Collapse
Affiliation(s)
- Everton Luiz Pompeu Varela
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
- Post-Graduate Program in Biodiversity and Biotechnology of the BIONORTE Network, Federal University of Pará, Belém 66075-110, Brazil
| | - Antônio Rafael Quadros Gomes
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
- Post-Graduate Program in Pharmaceutical Innovation, Federal University of Pará, Belém 66075-110, Brazil
| | - Aline da Silva Barbosa dos Santos
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
- Post-Graduate Program in Biodiversity and Biotechnology of the BIONORTE Network, Federal University of Pará, Belém 66075-110, Brazil
| | - Eliete Pereira de Carvalho
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
- Post-Graduate Program in Biodiversity and Biotechnology of the BIONORTE Network, Federal University of Pará, Belém 66075-110, Brazil
| | - Valdicley Vieira Vale
- Post-Graduate Program in Pharmaceutical Innovation, Federal University of Pará, Belém 66075-110, Brazil
| | - Sandro Percário
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, Brazil
- Post-Graduate Program in Biodiversity and Biotechnology of the BIONORTE Network, Federal University of Pará, Belém 66075-110, Brazil
| |
Collapse
|
13
|
Conroy AL, Tran TM, Bond C, Opoka RO, Datta D, Liechty EA, Bangirana P, Namazzi R, Idro R, Cusick S, Ssenkusu JM, John CC. Plasma Amino Acid Concentrations in Children With Severe Malaria Are Associated With Mortality and Worse Long-term Kidney and Cognitive Outcomes. J Infect Dis 2022; 226:2215-2225. [PMID: 36179241 PMCID: PMC10205609 DOI: 10.1093/infdis/jiac392] [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: 06/14/2022] [Revised: 09/14/2022] [Accepted: 09/20/2022] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND Global changes in amino acid levels have been described in severe malaria (SM), but the relationship between amino acids and long-term outcomes in SM has not been evaluated. METHODS We measured enrollment plasma concentrations of 20 amino acids using high-performance liquid chromatography in 500 Ugandan children aged 18 months to 12 years, including 122 community children and 378 children with SM. The Kidney Disease: Improving Global Outcomes criteria were used to define acute kidney injury (AKI) at enrollment and chronic kidney disease (CKD) at 1-year follow-up. Cognition was assessed over 2 years of follow-up. RESULTS Compared to laboratory-defined, age-specific reference ranges, there were deficiencies in sulfur-containing amino acids (methionine, cysteine) in both community children and children with SM. Among children with SM, global changes in amino acid concentrations were observed in the context of metabolic complications including acidosis and AKI. Increases in threonine, leucine, and valine were associated with in-hospital mortality, while increases in methionine, tyrosine, lysine, and phenylalanine were associated with postdischarge mortality and CKD. Increases in glycine and asparagine were associated with worse attention in children <5 years of age. CONCLUSIONS Among children with SM, unique amino acid profiles are associated with mortality, CKD, and worse attention.
Collapse
Affiliation(s)
- Andrea L Conroy
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Tuan M Tran
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Caitlin Bond
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Robert O Opoka
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Global Health Uganda, Kampala, Uganda
| | - Dibyadyuti Datta
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Edward A Liechty
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Paul Bangirana
- Department of Psychiatry, Makerere University College of Health Sciences, Global Health Uganda, Kampala, Uganda
| | - Ruth Namazzi
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Global Health Uganda, Kampala, Uganda
| | - Richard Idro
- Department of Paediatrics and Child Health, Makerere University College of Health Sciences, Global Health Uganda, Kampala, Uganda
| | - Sarah Cusick
- Division of Pediatric Epidemiology and Clinical Research, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - John M Ssenkusu
- Department of Epidemiology and Biostatistics, Makerere University School of Public Health, Kampala, Uganda
| | - Chandy C John
- Ryan White Center for Pediatric Infectious Disease and Global Health, Indiana University School of Medicine, Indianapolis, Indiana, USA
| |
Collapse
|
14
|
Gomes ARQ, Cunha N, Varela ELP, Brígido HPC, Vale VV, Dolabela MF, de Carvalho EP, Percário S. Oxidative Stress in Malaria: Potential Benefits of Antioxidant Therapy. Int J Mol Sci 2022; 23:ijms23115949. [PMID: 35682626 PMCID: PMC9180384 DOI: 10.3390/ijms23115949] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 05/17/2022] [Accepted: 05/19/2022] [Indexed: 01/07/2023] Open
Abstract
Malaria is an infectious disease and a serious public health problem in the world, with 3.3 billion people in endemic areas in 100 countries and about 200 million new cases each year, resulting in almost 1 million deaths in 2018. Although studies look for strategies to eradicate malaria, it is necessary to know more about its pathophysiology to understand the underlying mechanisms involved, particularly the redox balance, to guarantee success in combating this disease. In this review, we addressed the involvement of oxidative stress in malaria and the potential benefits of antioxidant supplementation as an adjuvant antimalarial therapy.
Collapse
Affiliation(s)
- Antonio Rafael Quadros Gomes
- Post-Graduate Program in Pharmaceutica Innovation, Institute of Health Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (A.R.Q.G.); (H.P.C.B.); (V.V.V.); (M.F.D.)
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (N.C.); (E.L.P.V.); (E.P.d.C.)
| | - Natasha Cunha
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (N.C.); (E.L.P.V.); (E.P.d.C.)
| | - Everton Luiz Pompeu Varela
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (N.C.); (E.L.P.V.); (E.P.d.C.)
- Post-graduate Program in Biodiversity and Biotechnology (BIONORTE), Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - Heliton Patrick Cordovil Brígido
- Post-Graduate Program in Pharmaceutica Innovation, Institute of Health Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (A.R.Q.G.); (H.P.C.B.); (V.V.V.); (M.F.D.)
| | - Valdicley Vieira Vale
- Post-Graduate Program in Pharmaceutica Innovation, Institute of Health Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (A.R.Q.G.); (H.P.C.B.); (V.V.V.); (M.F.D.)
| | - Maria Fâni Dolabela
- Post-Graduate Program in Pharmaceutica Innovation, Institute of Health Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (A.R.Q.G.); (H.P.C.B.); (V.V.V.); (M.F.D.)
- Post-graduate Program in Biodiversity and Biotechnology (BIONORTE), Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - Eliete Pereira de Carvalho
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (N.C.); (E.L.P.V.); (E.P.d.C.)
- Post-graduate Program in Biodiversity and Biotechnology (BIONORTE), Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
| | - Sandro Percário
- Oxidative Stress Research Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil; (N.C.); (E.L.P.V.); (E.P.d.C.)
- Post-graduate Program in Biodiversity and Biotechnology (BIONORTE), Institute of Biological Sciences, Federal University of Pará, Belém 66075-110, PA, Brazil
- Correspondence:
| |
Collapse
|
15
|
Metabolomic Analysis of Diverse Mice Reveals Hepatic Arginase-1 as Source of Plasma Arginase in Plasmodium chabaudi Infection. mBio 2021; 12:e0242421. [PMID: 34607466 PMCID: PMC8546868 DOI: 10.1128/mbio.02424-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Infections disrupt host metabolism, but the factors that dictate the nature and magnitude of metabolic change are incompletely characterized. To determine how host metabolism changes in relation to disease severity in murine malaria, we performed plasma metabolomics on eight Plasmodium chabaudi-infected mouse strains with diverse disease phenotypes. We identified plasma metabolic biomarkers for both the nature and severity of different malarial pathologies. A subset of metabolic changes, including plasma arginine depletion, match the plasma metabolomes of human malaria patients, suggesting new connections between pathology and metabolism in human malaria. In our malarial mice, liver damage, which releases hepatic arginase-1 (Arg1) into circulation, correlated with plasma arginine depletion. We confirmed that hepatic Arg1 was the primary source of increased plasma arginase activity in our model, which motivates further investigation of liver damage in human malaria patients. More broadly, our approach shows how leveraging phenotypic diversity can identify and validate relationships between metabolism and the pathophysiology of infectious disease. IMPORTANCE Malaria is a severe and sometimes fatal infectious disease endemic to tropical and subtropical regions. Effective vaccines against malaria-causing Plasmodium parasites remain elusive, and malaria treatments often fail to prevent severe disease. Small molecules that target host metabolism have recently emerged as candidates for therapeutics in malaria and other diseases. However, our limited understanding of how metabolites affect pathophysiology limits our ability to develop new metabolite therapies. By providing a rich data set of metabolite-pathology correlations and by validating one of those correlations, our work is an important step toward harnessing metabolism to mitigate disease. Specifically, we showed that liver damage in P. chabaudi-infected mice releases hepatic arginase-1 into circulation, where it may deplete plasma arginine, a candidate malaria therapeutic that mitigates vascular stress. Our data suggest that liver damage may confound efforts to increase levels of arginine in human malaria patients.
Collapse
|
16
|
Adams KJ, Wilson JG, Millington DS, Moseley MA, Colton CA, Thompson JW, Gottschalk WK. Capillary Electrophoresis-High Resolution Mass Spectrometry for Measuring In Vivo Arginine Isotope Incorporation in Alzheimer's Disease Mouse Models. JOURNAL OF THE AMERICAN SOCIETY FOR MASS SPECTROMETRY 2021; 32:1448-1458. [PMID: 34028275 DOI: 10.1021/jasms.1c00055] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Immune-based metabolic reprogramming of arginine utilization in the brain contributes to the neuronal pathology associated with Alzheimer's disease (AD). To enable our long-term goals of differentiation of AD mouse model genotypes, ages, and sexes based on activity of this pathway, we describe here the novel dosing (using uniformly labeled (13C615N4) arginine) and analysis methods using capillary electrophoresis high-resolution accurate-mass mass spectrometry for isotope tracing of metabolic products of arginine. We developed a pseudoprimed infusion-dosing regimen, using repeated injections, to achieve a steady state of uniformly labeled arginine in 135-195 min post bolus dose. Incorporation of stable isotope labeled carbon and nitrogen from uniformly labeled arginine into a host of downstream metabolites was measured in vivo in mice using serially sampled dried blood spots from the tail. In addition to the dried blood spot time course samples, total isotope incorporation into arginine-related metabolites was measured in the whole brain and plasma after 285 min. Preliminary demonstration of the technique identified differences isotope incorporation in arginine metabolites between male and female mice in a mouse-model of sporadic Alzheimer's disease (APOE4/huNOS2). The technique described herein will permit arginine pathway activity differentiation between mouse genotypes, ages, sexes, or drug treatments in order to elucidate the contribution of this pathway to Alzheimer's disease.
Collapse
Affiliation(s)
- Kendra J Adams
- Proteomics and Metabolomics Shared Resource, Duke University, Durham, North Carolina 27710, United States
| | - Joan G Wilson
- Department of Neurology, Duke University, Durham, North Carolina 27710, United States
| | - David S Millington
- Department of Pediatrics, Duke University School of Medicine, Durham, North Carolina 27710, United States
| | - M Arthur Moseley
- Proteomics and Metabolomics Shared Resource, Duke University, Durham, North Carolina 27710, United States
| | - Carol A Colton
- Department of Neurology, Duke University, Durham, North Carolina 27710, United States
| | - J Will Thompson
- Proteomics and Metabolomics Shared Resource, Duke University, Durham, North Carolina 27710, United States
- Department of Pharmacology and Cancer Biology, Duke University, Durham, North Carolina 27710, United States
| | - W Kirby Gottschalk
- Department of Neurology, Duke University, Durham, North Carolina 27710, United States
| |
Collapse
|
17
|
Blum L, Ulshöfer T, Henke M, Krieg R, Berneburg I, Geisslinger G, Becker K, Parnham MJ, Schiffmann S. The immunomodulatory potential of the arylmethylaminosteroid sc1o. J Mol Med (Berl) 2020; 99:261-272. [PMID: 33330947 PMCID: PMC7819914 DOI: 10.1007/s00109-020-02024-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 11/18/2020] [Accepted: 12/11/2020] [Indexed: 11/03/2022]
Abstract
Developing resistance mechanisms of pathogens against established and frequently used drugs are a growing global health problem. Besides the development of novel drug candidates per se, new approaches to counteract resistance mechanisms are needed. Drug candidates that not only target the pathogens directly but also modify the host immune system might boost anti-parasitic defence and facilitate clearance of pathogens. In this study, we investigated whether the novel anti-parasitic steroid compound 1o (sc1o), effective against the parasites Plasmodium falciparum and Schistosoma mansoni, might exhibit immunomodulatory properties. Our results reveal that 50 μM sc1o amplified the inflammatory potential of M1 macrophages and shifted M2 macrophages in a pro-inflammatory direction. Since M1 macrophages used predominantly glycolysis as an energy source, it is noteworthy that sc1o increased glycolysis and decreased oxidative phosphorylation in M2 macrophages. The effect of sc1o on the differentiation and activation of dendritic cells was ambiguous, since both pro- and anti-inflammatory markers were regulated. In conclusion, sc1o has several immunomodulatory effects that could possibly assist the immune system by counteracting the anti-inflammatory immune escape strategy of the parasite P. falciparum or by increasing pro-inflammatory mechanisms against pathogens, albeit at a higher concentration than that required for the anti-parasitic effect. KEY MESSAGES: • The anti-parasitic steroid compound 1o (sc1o) can modulate human immune cells. • Sc1o amplified the potential of M1 macrophages. • Sc1o shifts M2 macrophages to a M1 phenotype. • Dendritic cell differentiation and activation was ambiguously modulated. • Administration of sc1o could possibly assist the anti-parasitic defence.
Collapse
Affiliation(s)
- Leonard Blum
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology (TMP), Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany.,pharmazentrum frankfurt/ZAFES, Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt/Main, Germany
| | - Thomas Ulshöfer
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology (TMP), Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| | - Marina Henke
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology (TMP), Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| | - Reimar Krieg
- Department of Anatomy II, University Hospital Jena, Teichgraben 7, 07743, Jena, Germany
| | - Isabell Berneburg
- Department of Anatomy II, University Hospital Jena, Teichgraben 7, 07743, Jena, Germany
| | - Gerd Geisslinger
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology (TMP), Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany.,pharmazentrum frankfurt/ZAFES, Department of Clinical Pharmacology, Goethe-University Hospital Frankfurt, Theodor-Stern-Kai 7, 60590, Frankfurt/Main, Germany
| | - Katja Becker
- Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus-Liebig-University, Heinrich-Buff-Ring 26-32, 35392, Giessen, Germany
| | - Michael J Parnham
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology (TMP), Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany
| | - Susanne Schiffmann
- Fraunhofer Institute for Molecular Biology and Applied Ecology IME, Branch for Translational Medicine and Pharmacology (TMP), Theodor-Stern-Kai 7, 60596, Frankfurt am Main, Germany.
| |
Collapse
|
18
|
Andrade CM, Fleckenstein H, Thomson-Luque R, Doumbo S, Lima NF, Anderson C, Hibbert J, Hopp CS, Tran TM, Li S, Niangaly M, Cisse H, Doumtabe D, Skinner J, Sturdevant D, Ricklefs S, Virtaneva K, Asghar M, Homann MV, Turner L, Martins J, Allman EL, N'Dri ME, Winkler V, Llinás M, Lavazec C, Martens C, Färnert A, Kayentao K, Ongoiba A, Lavstsen T, Osório NS, Otto TD, Recker M, Traore B, Crompton PD, Portugal S. Increased circulation time of Plasmodium falciparum underlies persistent asymptomatic infection in the dry season. Nat Med 2020; 26:1929-1940. [PMID: 33106664 DOI: 10.1038/s41591-020-1084-0] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/27/2020] [Indexed: 12/25/2022]
Abstract
The dry season is a major challenge for Plasmodium falciparum parasites in many malaria endemic regions, where water availability limits mosquito vectors to only part of the year. How P. falciparum bridges two transmission seasons months apart, without being cleared by the human host or compromising host survival, is poorly understood. Here we show that low levels of P. falciparum parasites persist in the blood of asymptomatic Malian individuals during the 5- to 6-month dry season, rarely causing symptoms and minimally affecting the host immune response. Parasites isolated during the dry season are transcriptionally distinct from those of individuals with febrile malaria in the transmission season, coinciding with longer circulation within each replicative cycle of parasitized erythrocytes without adhering to the vascular endothelium. Low parasite levels during the dry season are not due to impaired replication but rather to increased splenic clearance of longer-circulating infected erythrocytes, which likely maintain parasitemias below clinical and immunological radar. We propose that P. falciparum virulence in areas of seasonal malaria transmission is regulated so that the parasite decreases its endothelial binding capacity, allowing increased splenic clearance and enabling several months of subclinical parasite persistence.
Collapse
Affiliation(s)
- Carolina M Andrade
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Hannah Fleckenstein
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Richard Thomson-Luque
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Safiatou Doumbo
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Nathalia F Lima
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Carrie Anderson
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Julia Hibbert
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany
| | - Christine S Hopp
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Tuan M Tran
- Division of Infectious Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shanping Li
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Moussa Niangaly
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Hamidou Cisse
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Didier Doumtabe
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Jeff Skinner
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Dan Sturdevant
- Rocky Mountain Laboratory Research Technologies Section, Genomics Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Stacy Ricklefs
- Rocky Mountain Laboratory Research Technologies Section, Genomics Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Kimmo Virtaneva
- Rocky Mountain Laboratory Research Technologies Section, Genomics Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Muhammad Asghar
- Department of Medicine Solna, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Manijeh Vafa Homann
- Department of Medicine Solna, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Louise Turner
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, København N, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Joana Martins
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Portugal and ICVS/3B's -PT Government Associate Laboratory, Braga, Portugal
| | - Erik L Allman
- Department of Biochemistry and Molecular Biology, Huck Center for Malaria Research, The Pennsylvania State University, State College, PA, USA
| | | | - Volker Winkler
- Institute of Global Health, Heidelberg University Hospital, Heidelberg, Germany
| | - Manuel Llinás
- Department of Biochemistry and Molecular Biology, Huck Center for Malaria Research, The Pennsylvania State University, State College, PA, USA.,Department of Chemistry, The Pennsylvania State University, State College, PA, USA
| | | | - Craig Martens
- Rocky Mountain Laboratory Research Technologies Section, Genomics Unit, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Anna Färnert
- Department of Medicine Solna, Division of Infectious Diseases, Karolinska Institutet, Stockholm, Sweden.,Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Kassoum Kayentao
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Aissata Ongoiba
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Thomas Lavstsen
- Department of Immunology and Microbiology, Centre for Medical Parasitology, Faculty of Health and Medical Sciences, University of Copenhagen, København N, Denmark.,Department of Infectious Diseases, Copenhagen University Hospital (Rigshospitalet), Copenhagen, Denmark
| | - Nuno S Osório
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Portugal and ICVS/3B's -PT Government Associate Laboratory, Braga, Portugal
| | - Thomas D Otto
- Institute of Infection, Immunity & Inflammation, MVLS, University of Glasgow, Glasgow, UK
| | - Mario Recker
- Centre for Mathematics & the Environment, University of Exeter, Penryn Campus, Penryn, UK
| | - Boubacar Traore
- Mali International Center of Excellence in Research, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Peter D Crompton
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Silvia Portugal
- Center for Infectious Diseases, Parasitology, Heidelberg University Hospital, Heidelberg, Germany. .,German Center for Infection Research (DZIF), Heidelberg, Heidelberg, Germany. .,Max Planck Institute for Infection Biology, Berlin, Germany.
| |
Collapse
|
19
|
Skytthe MK, Graversen JH, Moestrup SK. Targeting of CD163 + Macrophages in Inflammatory and Malignant Diseases. Int J Mol Sci 2020; 21:E5497. [PMID: 32752088 PMCID: PMC7432735 DOI: 10.3390/ijms21155497] [Citation(s) in RCA: 144] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 07/28/2020] [Accepted: 07/29/2020] [Indexed: 02/07/2023] Open
Abstract
The macrophage is a key cell in the pro- and anti-inflammatory response including that of the inflammatory microenvironment of malignant tumors. Much current drug development in chronic inflammatory diseases and cancer therefore focuses on the macrophage as a target for immunotherapy. However, this strategy is complicated by the pleiotropic phenotype of the macrophage that is highly responsive to its microenvironment. The plasticity leads to numerous types of macrophages with rather different and, to some extent, opposing functionalities, as evident by the existence of macrophages with either stimulating or down-regulating effect on inflammation and tumor growth. The phenotypes are characterized by different surface markers and the present review describes recent progress in drug-targeting of the surface marker CD163 expressed in a subpopulation of macrophages. CD163 is an abundant endocytic receptor for multiple ligands, quantitatively important being the haptoglobin-hemoglobin complex. The microenvironment of inflammation and tumorigenesis is particular rich in CD163+ macrophages. The use of antibodies for directing anti-inflammatory (e.g., glucocorticoids) or tumoricidal (e.g., doxorubicin) drugs to CD163+ macrophages in animal models of inflammation and cancer has demonstrated a high efficacy of the conjugate drugs. This macrophage-targeting approach has a low toxicity profile that may highly improve the therapeutic window of many current drugs and drug candidates.
Collapse
Affiliation(s)
- Maria K. Skytthe
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (M.K.S.); (S.K.M.)
| | - Jonas Heilskov Graversen
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (M.K.S.); (S.K.M.)
| | - Søren K. Moestrup
- Department of Molecular Medicine, University of Southern Denmark, 5000 Odense, Denmark; (M.K.S.); (S.K.M.)
- Department of Biomedicine, Aarhus University, 8200 Aarhus, Denmark
| |
Collapse
|
20
|
Understanding Human Cerebral Malaria through a Blood Transcriptomic Signature: Evidences for Erythrocyte Alteration, Immune/Inflammatory Dysregulation, and Brain Dysfunction. Mediators Inflamm 2020; 2020:3280689. [PMID: 32801995 PMCID: PMC7327554 DOI: 10.1155/2020/3280689] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 05/08/2020] [Indexed: 12/26/2022] Open
Abstract
Background Cerebral malaria (CM), a reversible encephalopathy affecting young children, is a medical emergency requiring rapid clinical assessment and treatment. However, understanding of the genes/proteins and the biological pathways involved in the disease outcome is still limited. Methods We have performed a whole transcriptomic analysis of blood samples from Malian children with CM or uncomplicated malaria (UM). Hierarchical clustering and pathway, network, and upstream regulator analyses were performed to explore differentially expressed genes (DEGs). We validated gene expression for 8 genes using real-time quantitative PCR (RT-qPCR). Plasma levels were measured for IP-10/CXCL10 and IL-18. Results A blood RNA signature including 538 DEGs (∣FC | ≥2.0, adjusted P value ≤ 0.01) allowed to discriminate between CM and UM. Ingenuity Pathway Analysis (IPA) and Kyoto Encyclopedia of Genes and Genomes (KEGG) revealed novel genes and biological pathways related to immune/inflammatory responses, erythrocyte alteration, and neurodegenerative disorders. Gene expressions of CXCL10, IL12RB2, IL18BP, IL2RA, AXIN2, and NET were significantly lower in CM whereas ARG1 and SLC6A9 were higher in CM compared to UM. Plasma protein levels of IP-10/CXCL10 were significantly lower in CM than in UM while levels of IL-18 were higher. Interestingly, among children with CM, those who died from a complication of malaria tended to have higher concentrations of IP-10/CXCL10 and IFN-γ than those who recovered. Conclusions This study identified some new factors and mechanisms that play crucial roles in CM and characterized their respective biological pathways as well as some upstream regulators.
Collapse
|
21
|
Loughland JR, Woodberry T, Field M, Andrew DW, SheelaNair A, Dooley NL, Piera KA, Amante FH, Kenangalem E, Price RN, Engwerda CR, Anstey NM, McCarthy JS, Boyle MJ, Minigo G. Transcriptional profiling and immunophenotyping show sustained activation of blood monocytes in subpatent Plasmodium falciparum infection. Clin Transl Immunology 2020; 9:e1144. [PMID: 32566226 PMCID: PMC7302943 DOI: 10.1002/cti2.1144] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/17/2020] [Accepted: 05/18/2020] [Indexed: 01/13/2023] Open
Abstract
OBJECTIVES Malaria, caused by Plasmodium infection, remains a major global health problem. Monocytes are integral to the immune response, yet their transcriptional and functional responses in primary Plasmodium falciparum infection and in clinical malaria are poorly understood. METHODS The transcriptional and functional profiles of monocytes were examined in controlled human malaria infection with P. falciparum blood stages and in children and adults with acute malaria. Monocyte gene expression and functional phenotypes were examined by RNA sequencing and flow cytometry at peak infection and compared to pre-infection or at convalescence in acute malaria. RESULTS In subpatent primary infection, the monocyte transcriptional profile was dominated by an interferon (IFN) molecular signature. Pathways enriched included type I IFN signalling, innate immune response and cytokine-mediated signalling. Monocytes increased TNF and IL-12 production upon in vitro toll-like receptor stimulation and increased IL-10 production upon in vitro parasite restimulation. Longitudinal phenotypic analyses revealed sustained significant changes in the composition of monocytes following infection, with increased CD14+CD16- and decreased CD14-CD16+ subsets. In acute malaria, monocyte CD64/FcγRI expression was significantly increased in children and adults, while HLA-DR remained stable. Although children and adults showed a similar pattern of differentially expressed genes, the number and magnitude of gene expression change were greater in children. CONCLUSIONS Monocyte activation during subpatent malaria is driven by an IFN molecular signature with robust activation of genes enriched in pathogen detection, phagocytosis, antimicrobial activity and antigen presentation. The greater magnitude of transcriptional changes in children with acute malaria suggests monocyte phenotypes may change with age or exposure.
Collapse
Affiliation(s)
- Jessica R Loughland
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia,Menzies School of Health ResearchDarwinNTAustralia
| | - Tonia Woodberry
- Menzies School of Health ResearchDarwinNTAustralia,Charles Darwin UniversityDarwinNTAustralia,Present address:
The University of NewcastleCallaghanNSWAustralia
| | - Matt Field
- Australian Institute of Tropical Health and Medicine and Centre for Tropical Bioinformatics and Molecular BiologyJames Cook UniversityCairnsQLDAustralia,John Curtin School of Medical ResearchAustralian National UniversityCanberraACTAustralia
| | - Dean W Andrew
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Arya SheelaNair
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | | | - Kim A Piera
- Menzies School of Health ResearchDarwinNTAustralia,Charles Darwin UniversityDarwinNTAustralia
| | - Fiona H Amante
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia
| | - Enny Kenangalem
- Timika Malaria Research ProgramPapuan Health and Community Development FoundationTimikaIndonesia,District Health AuthorityTimikaIndonesia
| | - Ric N Price
- Menzies School of Health ResearchDarwinNTAustralia,Charles Darwin UniversityDarwinNTAustralia,Centre for Tropical Medicine and Global HealthNuffield Department of Clinical MedicineUniversity of OxfordOxfordUK,Mahidol‐Oxford Tropical Medicine Research UnitFaculty of Tropical MedicineMahidol UniversityBangkokThailand
| | | | - Nicholas M Anstey
- Menzies School of Health ResearchDarwinNTAustralia,Charles Darwin UniversityDarwinNTAustralia
| | | | - Michelle J Boyle
- QIMR Berghofer Medical Research InstituteBrisbaneQLDAustralia,Menzies School of Health ResearchDarwinNTAustralia
| | - Gabriela Minigo
- Menzies School of Health ResearchDarwinNTAustralia,Charles Darwin UniversityDarwinNTAustralia,College of Health and Human SciencesCharles Darwin UniversityDarwinNTAustralia
| |
Collapse
|
22
|
Pereira DMS, Carvalho Júnior AR, Lacerda EMDCB, da Silva LCN, Marinho CRF, André E, Fernandes ES. Oxidative and nitrosative stresses in cerebral malaria: can we target them to avoid a bad prognosis? J Antimicrob Chemother 2020; 75:1363-1373. [PMID: 32105324 DOI: 10.1093/jac/dkaa032] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
There is currently a global effort to reduce malaria morbidity and mortality. However, malaria still results in the deaths of thousands of people every year. Malaria is caused by Plasmodium spp., parasites transmitted through the bite of an infected female Anopheles mosquito. Treatment timing plays a decisive role in reducing mortality and sequelae associated with the severe forms of the disease such as cerebral malaria (CM). The available antimalarial therapy is considered effective but parasite resistance to these drugs has been observed in some countries. Antimalarial drugs act by increasing parasite lysis, especially through targeting oxidative stress pathways. Here we discuss the roles of reactive oxygen species and reactive nitrogen intermediates in CM as a result of host-parasite interactions. We also present evidence of the potential contribution of oxidative and nitrosative stress-based antimalarial drugs to disease treatment and control.
Collapse
Affiliation(s)
| | | | | | | | | | - Eunice André
- Departamento de Farmacologia, Universidade Federal do Paraná, Curitiba, PR, Brazil
| | - Elizabeth Soares Fernandes
- Programa de Pós-graduação, Universidade CEUMA, São Luís, MA, Brazil.,Instituto de Pesquisa Pelé Pequeno Príncipe, Curitiba, PR, Brazil.,Faculdades Pequeno Príncipe, Curitiba, PR, Brazil
| |
Collapse
|
23
|
Dobbs KR, Crabtree JN, Dent AE. Innate immunity to malaria-The role of monocytes. Immunol Rev 2020; 293:8-24. [PMID: 31840836 PMCID: PMC6986449 DOI: 10.1111/imr.12830] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/19/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022]
Abstract
Monocytes are innate immune cells essential for host protection against malaria. Upon activation, monocytes function to help reduce parasite burden through phagocytosis, cytokine production, and antigen presentation. However, monocytes have also been implicated in the pathogenesis of severe disease through production of damaging inflammatory cytokines, resulting in systemic inflammation and vascular dysfunction. Understanding the molecular pathways influencing the balance between protection and pathology is critical. In this review, we discuss recent data regarding the role of monocytes in human malaria, including studies of innate sensing of the parasite, immunometabolism, and innate immune training. Knowledge gained from these studies may guide rational development of novel antimalarial therapies and inform vaccine development.
Collapse
Affiliation(s)
- Katherine R. Dobbs
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
- Division of Pediatric Infectious Diseases, University Hospitals Rainbow Babies and Children’s Hospital, Cleveland, OH, USA
| | - Juliet N. Crabtree
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA
| | - Arlene E. Dent
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, OH, USA
- Division of Pediatric Infectious Diseases, University Hospitals Rainbow Babies and Children’s Hospital, Cleveland, OH, USA
| |
Collapse
|
24
|
Pham TT, Punsawad C, Glaharn S, De Meyer SF, Viriyavejakul P, Van den Steen PE. Release of endothelial activation markers in lungs of patients with malaria-associated acute respiratory distress syndrome. Malar J 2019; 18:395. [PMID: 31796023 PMCID: PMC6891978 DOI: 10.1186/s12936-019-3040-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Accepted: 11/26/2019] [Indexed: 11/17/2022] Open
Abstract
Background Malaria-associated acute respiratory distress syndrome (MA-ARDS) is an understudied complication of malaria and is characterized by pulmonary inflammation and disruption of the alveolar-capillary membrane. Its pathogenesis remains poorly understood. Since endothelial activation plays an important role in other malarial complications, the expression of two endothelial activation markers, von Willebrand factor (VWF) and angiopoietin-2 (ANG-2), was investigated in the lungs of patients with MA-ARDS. Methods Post-mortem lung sections of Plasmodium falciparum-infected patients without alveolar oedema (NA), P. falciparum-infected patients with alveolar oedema (MA-ARDS), and uninfected people who died accidentally with no pathological changes to the lungs (CON) were immunohistochemically stained for VWF and ANG-2, and were evaluated with semi-quantitative analysis. Results Alveolar oedematous VWF and ANG-2 and intravascular VWF staining were significantly increased in patients with MA-ARDS versus infected and uninfected control groups. The levels of VWF in the alveolar septa and endothelial lining of large blood vessels of patients with MA-ARDS was significantly decreased compared to controls. ANG-2 expression was increased in the alveolar septa of malaria patients without alveolar oedema versus control patients, while ANG-2+ leukocytes were increased in the alveoli in both infected patient groups. Conclusions This study documents a high level of VWF and ANG-2, two endothelial activation markers in the oedematous alveoli of post-mortem lung sections of Thai patients with MA-ARDS. Decreased detection of VWF in the endothelial lining of blood vessels, in parallel with an increased presence of intravascular VWF staining suggests marked endothelial activation and Weibel–Palade body release in the lungs of patients with MA-ARDS.
Collapse
Affiliation(s)
- Thao-Thy Pham
- Laboratory of Immunoparasitology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Leuven, Belgium
| | - Chuchard Punsawad
- School of Medicine, Walailak University, Nakhon Si Thammarat, Thailand
| | - Supattra Glaharn
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Simon F De Meyer
- Laboratory for Thrombosis Research, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | - Parnpen Viriyavejakul
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Philippe E Van den Steen
- Laboratory of Immunoparasitology, Department of Microbiology and Immunology, Rega Institute for Medical Research, KU Leuven, University of Leuven, Leuven, Belgium.
| |
Collapse
|
25
|
Changes in monocyte subsets are associated with clinical outcomes in severe malarial anaemia and cerebral malaria. Sci Rep 2019; 9:17545. [PMID: 31772386 PMCID: PMC6879635 DOI: 10.1038/s41598-019-52579-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 10/18/2019] [Indexed: 01/17/2023] Open
Abstract
Monocytes are plastic heterogeneous immune cells involved in host-parasite interactions critical for malaria pathogenesis. Human monocytes have been subdivided into three populations based on surface expression of CD14 and CD16. We hypothesised that proportions and phenotypes of circulating monocyte subsets can be markers of severity or fatality in children with malaria. To address this question, we compared monocytes sampled in children with uncomplicated malaria, severe malarial anaemia, or cerebral malaria. Flow cytometry was used to distinguish and phenotype monocyte subsets through CD14, CD16, CD36 and TLR2 expression. Data were first analysed by univariate analysis to evaluate their link to severity and death. Second, multinomial logistic regression was used to measure the specific effect of monocyte proportions and phenotypes on severity and death, after adjustments for other variables unrelated to monocytes. Multivariate analysis demonstrated that decreased percentages of non-classical monocytes were associated with death, suggesting that this monocyte subset has a role in resolving malaria. Using univariate analysis, we also showed that the role of non-classical monocytes involves a mostly anti-inflammatory profile and the expression of CD16. Further studies are needed to decipher the functions of this sub-population during severe malaria episodes, and understand the underlying mechanisms.
Collapse
|
26
|
Yeo TW, Weinberg JB, Lampah DA, Kenangalem E, Bush P, Chen Y, Price RN, Young S, Zhang HY, Millington D, Granger DL, Anstey NM. Glycocalyx Breakdown Is Associated With Severe Disease and Fatal Outcome in Plasmodium falciparum Malaria. Clin Infect Dis 2019; 69:1712-1720. [PMID: 30753363 PMCID: PMC6821254 DOI: 10.1093/cid/ciz038] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 01/15/2019] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Interactions between the endothelium and infected erythrocytes play a major role in the pathogenesis of falciparum malaria, with microvascular dysfunction and parasite sequestration associated with worsening outcomes. The glycocalyx is a carbohydrate-rich layer that lines the endothelium, with multiple roles in vascular homeostasis. The role of the glycocalyx in falciparum malaria and the association with disease severity has not been investigated. METHODS We prospectively enrolled Indonesian inpatients (aged ≥18 years) with severe (SM) or moderately severe (MSM) falciparum malaria, as defined by World Health Organization criteria, and healthy controls (HCs). On enrollment, blood and urine samples were collected concurrently with measurements of vascular nitric oxide (NO) bioavailability. Urine was assayed for glycocalyx breakdown products (glycosaminoglycans) using a dimethylmethylene blue (GAG-DMMB) and liquid chromatography-tandem mass spectrometry (GAG-MS) assay. RESULTS A total of 129 patients (SM = 43, MSM = 57, HC=29) were recruited. GAG-DMMB and GAG-MS (g/mol creatinine) were increased in SM (mean, 95% confidence interval: 3.98, 2.44-5.53 and 6.82, 5.19-8.44) compared to MSM patients (1.78, 1.27-2.29 and 4.87, 4.27-5.46) and HCs (0.22, 0.06-0.37 and 1.24, 0.89-1.59; P < 0.001). In SM patients, GAG-DMMB and GAG-MS were increased in those with a fatal outcome (n = 3; median, interquartile range: 6.72, 3.80-27.87 and 12.15, 7.88-17.20) compared to survivors (n = 39; 3.10, 0.46-4.5 and 4.64, 2.02-15.20; P = 0.03). Glycocalyx degradation was significantly associated with parasite biomass in both MSM (r = 0.48, GAG-DMMB and r = 0.43, GAG-MS; P < 0.001) and SM patients (r = 0.47, P = 0.002 and r = 0.33, P = 0.04) and inversely associated with endothelial NO bioavailability. CONCLUSIONS Increased endothelial glycocalyx breakdown is associated with severe disease and a fatal outcome in adults with falciparum malaria.
Collapse
Affiliation(s)
- Tsin W Yeo
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore
- Institute of Infectious Diseases and Epidemiology, Tan Tock Seng Hospital, Singapore
| | - J Brice Weinberg
- Duke University School of Medicine and VA Medical Center, Durham, North Carolina
| | - Daniel A Lampah
- Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
| | - Enny Kenangalem
- Papuan Health and Community Development Foundation, Timika, Papua, Indonesia
- Mimika District Health Authority, Timika, Papua, Indonesia
| | - Peggy Bush
- Duke University School of Medicine and VA Medical Center, Durham, North Carolina
| | - Youwei Chen
- Duke University School of Medicine and VA Medical Center, Durham, North Carolina
| | - Richard N Price
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
- Centre for Tropical Medicine and Global Health, Nuffield Department of Clinical Medicine, University of Oxford, United Kingdom
| | - Sarah Young
- Duke University School of Medicine and VA Medical Center, Durham, North Carolina
| | - Hao Y Zhang
- Duke University School of Medicine and VA Medical Center, Durham, North Carolina
| | - David Millington
- Duke University School of Medicine and VA Medical Center, Durham, North Carolina
| | | | - Nicholas M Anstey
- Global and Tropical Health Division, Menzies School of Health Research, Darwin, Northern Territory, Australia
| |
Collapse
|
27
|
Yeo TW, Bush PA, Chen Y, Young SP, Zhang H, Millington DS, Granger DL, Mwaikambo ED, Anstey NM, Weinberg JB. Glycocalyx breakdown is increased in African children with cerebral and uncomplicated falciparum malaria. FASEB J 2019; 33:14185-14193. [PMID: 31658834 DOI: 10.1096/fj.201901048rr] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cerebral malaria (CM) from Plasmodium falciparum infection is associated with endothelial dysfunction and parasite sequestration. The glycocalyx (GCX), a carbohydrate-rich layer lining the endothelium, is crucial in vascular homeostasis. To evaluate the role of its loss in the pathogenesis of pediatric CM, we measured GCX degradation in Tanzanian children with World Health Organization-defined CM (n = 55), uncomplicated malaria (UM; n = 20), and healthy controls (HCs; n = 25). Urine GCX breakdown products [glycosaminoglycans (GAGs)] were quantified using dimethylmethylene blue (DMMB) and liquid chromatography-tandem mass spectrometry assays. DMMB-GAG and mass spectrometry (MS)-GAG (g/mol creatinine) were increased in CM and UM compared with HCs (P < 0.001), with no differences in DMMB-GAG and MS-GAG between CM and UM children or between those with and without a fatal outcome. In CM survivors, urinary GCX DMMB-GAG normalized by d 3. After adjusting for disease severity, DMMB-GAG was significantly associated with parasitemia [partial correlation coefficient (Pcorr) = 0.34; P = 0.01] and plasma TNF (Pcorr = 0.26; P = 0.04) and inversely with plasma and urine NO oxidation products [Pcorr = -0.31 (P = 0.01) and Pcorr = -0.26 (P = 0.03), respectively]. GCX breakdown is increased in children with falciparum malaria, with similar elevations in CM and UM. Endothelial GCX degradation may impair endothelial NO production, exacerbate adhesion-molecule expression, exposure, and parasite sequestration, and contribute to malaria pathogenesis.-Yeo, T. W., Bush, P. A., Chen, Y., Young, S. P., Zhang, H., Millington, D. S., Granger, D. L., Mwaikambo, E. D., Anstey, N. M., Weinberg, J. B. Glycocalyx breakdown is increased in African children with cerebral and uncomplicated falciparum malaria.
Collapse
Affiliation(s)
- Tsin W Yeo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,National Centre for Infectious Diseases, Tan Tock Seng Hospital, Singapore.,Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - Peggy A Bush
- Duke University-Veterans Affairs (VA) Medical Center, Durham, North Carolina, USA
| | - Youwei Chen
- Duke University-Veterans Affairs (VA) Medical Center, Durham, North Carolina, USA
| | - Sarah P Young
- Duke University-Veterans Affairs (VA) Medical Center, Durham, North Carolina, USA
| | - Haoyue Zhang
- Duke University-Veterans Affairs (VA) Medical Center, Durham, North Carolina, USA
| | - David S Millington
- Duke University-Veterans Affairs (VA) Medical Center, Durham, North Carolina, USA
| | - Donald L Granger
- University of Utah-Veterans Affairs (VA) Medical Center, Salt Lake City, Utah, USA
| | | | - Nicholas M Anstey
- Menzies School of Health Research, Charles Darwin University, Darwin, Northern Territory, Australia
| | - J Brice Weinberg
- Duke University-Veterans Affairs (VA) Medical Center, Durham, North Carolina, USA
| |
Collapse
|
28
|
Singh N, Kumar R, Chauhan SB, Engwerda C, Sundar S. Peripheral Blood Monocytes With an Antiinflammatory Phenotype Display Limited Phagocytosis and Oxidative Burst in Patients With Visceral Leishmaniasis. J Infect Dis 2019; 218:1130-1141. [PMID: 30053070 DOI: 10.1093/infdis/jiy228] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Accepted: 06/03/2018] [Indexed: 12/17/2022] Open
Abstract
Background Monocytes are important effector cells during Leishmania infection, and changes in their functions may impact development of immunity. However, functional characteristics of monocytes in patients with visceral leishmaniasis (VL) remains poorly understood. Methods Peripheral blood monocytes from patients with VL and healthy endemic controls from Muzaffarpur, India, were isolated and compared in an ex vivo setting, using cell-culture techniques, flow cytometry, and reverse transcription quantitative polymerase chain reaction analysis. Results A blood monocyte population with a gene signature comprising upregulated expression of TGM2, CTLRs, VDR, PKM, SOCS1, and CAMP1 and downregulated expression of NOS2 and HIF1A was observed in patients with VL but not in controls. Monocytes from patients with VL also had impaired expression of chemokine receptors and adhesion molecules and decreased frequencies of interleukin 1β- and interleukin 6-producing cells. Importantly, monocytes from patients with VL had a markedly reduced capacity for phagocytosis of amastigotes, p47phox and p67phox expression, and reactive oxygen species production. Conclusions Monocytes from patients with VL express antiinflammatory molecules and lack a classically activated phenotype. They have reduced expression of molecules related to activation and antiparasitic effector functions, indicating that monocytes are skewed toward an antiinflammatory phenotype. These findings provide insights into the functional status of monocytes during VL and advise that therapeutic manipulation of this important cell population may result in favorable patient outcomes.
Collapse
Affiliation(s)
- Neetu Singh
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajiv Kumar
- Department of Biochemistry, Banaras Hindu University, Varanasi, India
| | | | - Christian Engwerda
- Immunology and Infection Laboratory, QIMR Berghofer Medical Research Institute, Herston, Australia
| | - Shyam Sundar
- Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
29
|
Penha-Gonçalves C. Genetics of Malaria Inflammatory Responses: A Pathogenesis Perspective. Front Immunol 2019; 10:1771. [PMID: 31417551 PMCID: PMC6682681 DOI: 10.3389/fimmu.2019.01771] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2018] [Accepted: 07/15/2019] [Indexed: 12/27/2022] Open
Abstract
Despite significant progress in combating malaria in recent years the burden of severe disease and death due to Plasmodium infections remains a global public health concern. Only a fraction of infected people develops severe clinical syndromes motivating a longstanding search for genetic determinants of malaria severity. Strong genetic effects have been repeatedly ascribed to mutations and allelic variants of proteins expressed in red blood cells but the role of inflammatory response genes in disease pathogenesis has been difficult to discern. We revisited genetic evidence provided by inflammatory response genes that have been repeatedly associated to malaria, namely TNF, NOS2, IFNAR1, HMOX1, TLRs, CD36, and CD40LG. This highlighted specific genetic variants having opposing roles in the development of distinct malaria clinical outcomes and unveiled diverse levels of genetic heterogeneity that shaped the complex association landscape of inflammatory response genes with malaria. However, scrutinizing genetic effects of individual variants corroborates a pathogenesis model where pro-inflammatory genetic variants acting in early infection stages contribute to resolve infection but at later stages confer increased vulnerability to severe organ dysfunction driven by tissue inflammation. Human genetics studies are an invaluable tool to find genes and molecular pathways involved in the inflammatory response to malaria but their precise roles in disease pathogenesis are still unexploited. Genome editing in malaria experimental models and novel genotyping-by-sequencing techniques are promising approaches to delineate the relevance of inflammatory response gene variants in the natural history of infection thereby will offer new rational angles on adjuvant therapeutics for prevention and clinical management of severe malaria.
Collapse
|
30
|
Reactive nitrogen species in host-bacterial interactions. Curr Opin Immunol 2019; 60:96-102. [PMID: 31200187 DOI: 10.1016/j.coi.2019.05.008] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Revised: 04/29/2019] [Accepted: 05/11/2019] [Indexed: 12/11/2022]
Abstract
Reactive nitrogen species play diverse and essential roles in host-pathogen interactions. Here, we review selected recent discoveries regarding nitric oxide (NO) in host defense and the pathogenesis of infection, mechanisms of bacterial NO resistance, production of NO by human macrophages, NO-based antimicrobial therapeutics and NO interactions with the gut microbiota.
Collapse
|
31
|
Kinetic and Cross-Sectional Studies on the Genesis of Hypoargininemia in Severe Pediatric Plasmodium falciparum Malaria. Infect Immun 2019; 87:IAI.00655-18. [PMID: 30718287 DOI: 10.1128/iai.00655-18] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 01/05/2019] [Indexed: 12/12/2022] Open
Abstract
The low bioavailability of nitric oxide (NO) and its precursor, arginine, contributes to the microvascular pathophysiology of severe falciparum malaria. To better characterize the mechanisms underlying hypoargininemia in severe malaria, we measured the plasma concentrations of amino acids involved in de novo arginine synthesis in children with uncomplicated falciparum malaria (UM; n = 61), children with cerebral falciparum malaria (CM; n = 45), and healthy children (HC; n = 109). We also administered primed infusions of l-arginine uniformly labeled with 13C6 and 15N4 to 8 children with severe falciparum malaria (SM; age range, 4 to 9 years) and 7 healthy children (HC; age range, 4 to 8 years) to measure the metabolic flux of arginine, hypothesizing that arginine flux is increased in SM. Using two different tandem mass spectrometric methods, we measured the isotopic enrichment of arginine in plasma obtained at 0, 60, 90, 120, 150, and 180 min during the infusion. The plasma concentrations of glutamine, glutamate, proline, ornithine, citrulline, and arginine were significantly lower in UM and CM than in HC (P ≤ 0.04 for all pairwise comparisons). Of these, glutamine concentrations were the most markedly decreased: median, 457 μM (interquartile range [IQR], 400 to 508 μM) in HC, 300 μM (IQR, 256 to 365 μM) in UM, and 257 μM (IQR, 195 to 320 μM) in CM. Arginine flux during steady state was not significantly different in SM than in HC by the respective mass spectrometric methods: 93.2 μmol/h/kg of body weight (IQR, 84.4 to 129.3 μmol/h/kg) versus 88.0 μmol/h/kg (IQR, 73.0 to 102.2 μmol/h/kg) (P = 0.247) by the two mass spectrometric methods in SM and 93.7 μmol/h/kg (IQR, 79.1 to 117.8 μmol/h/kg) versus 81.0 μmol/h/kg (IQR, 75.9 to 88.6 μmol/h/kg) (P = 0.165) by the two mass spectrometric methods in HC. A limited supply of amino acid precursors for arginine synthesis likely contributes to the hypoargininemia and NO insufficiency in falciparum malaria in children.
Collapse
|
32
|
Wang Q, Feng Y, Sun X, Pang W, Fu W, Cao Y. Prophylactic treatment of L-Arg improves malaria outcomes by regulating host immune responses during Plasmodium yoelii 17XL infection. Exp Parasitol 2018; 195:1-7. [DOI: 10.1016/j.exppara.2018.09.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 02/23/2018] [Accepted: 09/20/2018] [Indexed: 12/24/2022]
|
33
|
Bobade D, Khandare AV, Deval M, Shastry P, Deshpande P. Hemozoin-induced activation of human monocytes toward M2-like phenotype is partially reversed by antimalarial drugs-chloroquine and artemisinin. Microbiologyopen 2018; 8:e00651. [PMID: 29877619 PMCID: PMC6436431 DOI: 10.1002/mbo3.651] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 04/08/2018] [Accepted: 04/16/2018] [Indexed: 12/20/2022] Open
Abstract
Plasmodium falciparum malaria is the most severe form of malaria with several complications. The malaria pigment‐hemozoin (Hz) is associated with severe anemia, cytokine dysfunction, and immunosuppression, thus making it an interesting target for developing new strategies for antimalarial therapy. Monocytes (MO) in circulation actively ingest Hz released by Plasmodium parasites and secrete pro‐ and anti‐inflammatory cytokines. M1 and M2 types represent the two major forms of MO/macrophages (MQ) with distinct phenotypes and opposing functions. Imbalance in the polarization of these types is reported in many infectious diseases. Though the association of Hz with immunosuppression is well documented, its role in activation of MO in context of M1/M2 phenotypes remains to be addressed. We report here that natural Hz drives human MO toward M2‐like phenotype as evidenced by the expression of M2 signature markers. Hz‐fed MO showed elevated transcript and secreted level of IL‐10, CCL17, CCL1, expression of mannose‐binding lectin receptor (CD206), and arginase activity. Hz attenuated HLA‐DR expression, nitric oxide, and reactive oxygen species production, which are the features of M1 phenotype. Our data also implicate the involvement of p38 MAPK, PI3K/AKT, and NF‐κB signaling pathways in skewing of Hz‐fed MO toward M2‐like type and suppression of mitogen‐stimulated lymphocyte proliferation. Importantly, antimalarial drugs—chloroquine and artemisinin—partially reversed activation of Hz‐induced MO toward M2‐like phenotype. Considering the limitations in the current therapeutic options for malaria, we propose that these drugs may be re‐examined for their potential as immunomodulators and candidates for adjunctive treatment in malaria.
Collapse
Affiliation(s)
| | | | - Mangesh Deval
- National Centre for Cell Science (NCCS), Pune, India
| | - Padma Shastry
- National Centre for Cell Science (NCCS), Pune, India
| | | |
Collapse
|
34
|
Kumar A, Singh KP, Bali P, Anwar S, Kaul A, Singh OP, Gupta BK, Kumari N, Noor Alam M, Raziuddin M, Sinha MP, Gourinath S, Sharma AK, Sohail M. iNOS polymorphism modulates iNOS/NO expression via impaired antioxidant and ROS content in P. vivax and P. falciparum infection. Redox Biol 2018; 15:192-206. [PMID: 29268202 PMCID: PMC5738204 DOI: 10.1016/j.redox.2017.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Revised: 11/30/2017] [Accepted: 12/12/2017] [Indexed: 01/09/2023] Open
Abstract
Nitric oxide (NO) has dicotomic influence on modulating host-parasite interplay, synchronizing physiological orchestrations and diagnostic potential; instigated us to investigate the plausible association and genetic regulation among NO level, components of oxidative stress, iNOS polymorphisms and risk of malaria. Here, we experimentally elucidate that iNOS promoter polymorphisms are associated with risk of malaria; employing mutation specific genotyping, functional interplay using western blot and RT-PCR, quantitative estimation of NO, total antioxidant content (TAC) and reactive oxygen species (ROS). Genotyping revealed significantly associated risk of P. vivax (adjusted OR = 1.92 and 1.72) and P. falciparum (adjusted OR = 1.68 and 1.75) infection with SNP at iNOS-954G/C and iNOS-1173C/T positions, respectively; though vivax showed higher risk of infection. Intriguingly, mutation and infection specific differential upregulation of iNOS expression/NO level was observed and found to be significantly associated with mutant genotypes. Moreover, P. vivax showed pronounced iNOS protein (2.4 fold) and mRNA (2.5 fold) expression relative to healthy subjects. Furthermore, TAC and ROS were significantly decreased in infection; and differentially decreased in mutant genotypes. Our findings endorse polymorphic regulation of iNOS expression, altered oxidant-antioxidant components and evidences of risk association as the hallmark of malaria pathogenesis. iNOS/NO may serve as potential diagnostic marker in assessing clinical malaria.
Collapse
Affiliation(s)
- Amod Kumar
- University Department of Zoology, Vinoba Bhave University, Hazaribag, Jharkhand, India
| | - Krishn Pratap Singh
- University Department of Zoology, Vinoba Bhave University, Hazaribag, Jharkhand, India
| | - Prerna Bali
- National Institute of Malaria Research, Dawarka, Delhi, India
| | - Shadab Anwar
- School of Life Sciences, Jawaharlal Nehru University, New Delhi, India
| | - Asha Kaul
- National Institute of Malaria Research, Dawarka, Delhi, India
| | - Om P Singh
- National Institute of Malaria Research, Dawarka, Delhi, India
| | - Birendra Kumar Gupta
- University Department of Zoology, Vinoba Bhave University, Hazaribag, Jharkhand, India
| | - Nutan Kumari
- Department of Physiology, Patna Medical College and Hospital, Patna, India
| | - Md Noor Alam
- University Department of Zoology, Vinoba Bhave University, Hazaribag, Jharkhand, India
| | - Mohammad Raziuddin
- University Department of Zoology, Vinoba Bhave University, Hazaribag, Jharkhand, India
| | | | | | - Ajay Kumar Sharma
- University Department of Zoology, Vinoba Bhave University, Hazaribag, Jharkhand, India.
| | - Mohammad Sohail
- University Department of Zoology, Vinoba Bhave University, Hazaribag, Jharkhand, India.
| |
Collapse
|
35
|
Conroy AL, Hawkes MT, Elphinstone R, Opoka RO, Namasopo S, Miller C, John CC, Kain KC. Chitinase-3-like 1 is a biomarker of acute kidney injury and mortality in paediatric severe malaria. Malar J 2018; 17:82. [PMID: 29448936 PMCID: PMC5815237 DOI: 10.1186/s12936-018-2225-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Accepted: 02/07/2018] [Indexed: 12/16/2022] Open
Abstract
Background Chitinase-3-like 1 (CHI3L1) is a glycoprotein elevated in paediatric severe malaria, and an emerging urinary biomarker of acute kidney injury (AKI). Based on the hypothesis that elevated CHI3L1 levels in malaria are associated with disease severity, the relationship between plasma CHI3L1 levels, AKI and mortality was investigated in Ugandan children enrolled in a clinical trial evaluating inhaled nitric oxide (iNO) as an adjunctive therapy for severe malaria. Methods Plasma CHI3L1 levels were measured daily for 4 days in children admitted to hospital with severe malaria and at day 14 follow up. AKI was defined using the Kidney Disease: Improving Global Outcomes consensus criteria. This is a secondary analysis of a randomized double-blind placebo-controlled trial of iNO versus placebo as an adjunctive therapy for severe malaria. Inclusion criteria were: age 1–10 years, and selected criteria for severe malaria. Exclusion criteria included suspected bacterial meningitis, known chronic illness including renal disease, haemoglobinopathy, or severe malnutrition. iNO was administered by non-rebreather mask for up to 72 h at 80 ppm. Results CHI3L1 was elevated in patients with AKI and remained higher over hospitalization (p < 0.0001). Admission CHI3L1 levels were elevated in children who died. By multivariable analysis logCHI3L1 levels were associated with increased risk of in-hospital death (relative risk, 95% CI 4.10, 1.32–12.75, p = 0.015) and all-cause 6 month mortality (3.21, 1.47–6.98, p = 0.003) following correction for iNO and AKI. Treatment with iNO was associated with delayed CHI3L1 recovery with a daily decline of 34% in the placebo group versus 29% in the iNO group (p = 0.012). CHI3L1 levels correlated with markers of inflammation (CRP, sTREM-1, CXCL10), endothelial activation (Ang-2, sICAM-1) and intravascular haemolysis (LDH, haem, haemopexin). Conclusions CHI3L1 is a novel biomarker of malaria-associated AKI and an independent risk factor for mortality that is associated with well-established pathways of severe malaria pathogenesis including inflammation, endothelial activation, and haemolysis. Trial registration Clinicaltrials.gov, NCT01255215. Registered December 7th 2010 Electronic supplementary material The online version of this article (10.1186/s12936-018-2225-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Andrea L Conroy
- Department of Pediatrics, Indiana University School of Medicine, 1044 West Walnut St., Building 4, Indianapolis, IN, 46202, USA. .,Sandra Rotman Centre for Global Health, Toronto General Hospital, University Health Network, MaRS Centre, 101 College St. TMDT 10-360A, Toronto, ON, M5G 1L7, Canada. .,Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, ON, Canada.
| | - Michael T Hawkes
- Division of Pediatric Infectious Diseases, 3-593 Edmonton Clinic Health Academy, University of Alberta, Edmonton, AB, T6G1C9, Canada
| | - Robyn Elphinstone
- Sandra Rotman Centre for Global Health, Toronto General Hospital, University Health Network, MaRS Centre, 101 College St. TMDT 10-360A, Toronto, ON, M5G 1L7, Canada
| | - Robert O Opoka
- Department of Pediatrics and Child Health, Makerere University, Kampala, Uganda
| | - Sophie Namasopo
- Department of Pediatrics, Jinja Regional Referral Hospital, P.O. Box 43, Jinja, Uganda
| | | | - Chandy C John
- Department of Pediatrics, Indiana University School of Medicine, 1044 West Walnut St., Building 4, Indianapolis, IN, 46202, USA
| | - Kevin C Kain
- Sandra Rotman Centre for Global Health, Toronto General Hospital, University Health Network, MaRS Centre, 101 College St. TMDT 10-360A, Toronto, ON, M5G 1L7, Canada.,Division of Infectious Diseases, Department of Medicine, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
36
|
Dobbs KR, Embury P, Vulule J, Odada PS, Rosa BA, Mitreva M, Kazura JW, Dent AE. Monocyte dysregulation and systemic inflammation during pediatric falciparum malaria. JCI Insight 2017; 2:95352. [PMID: 28931756 DOI: 10.1172/jci.insight.95352] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Accepted: 08/16/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Inflammation and monocytes are thought to be important to human malaria pathogenesis. However, the relationship of inflammation and various monocyte functions to acute malaria, recovery from acute malaria, and asymptomatic parasitemia in endemic populations is poorly understood. METHODS We evaluated plasma cytokine levels, monocyte subsets, monocyte functional responses, and monocyte inflammatory transcriptional profiles of 1- to 10-year-old Kenyan children at the time of presentation with acute uncomplicated malaria and at recovery 6 weeks later; these results were compared with analogous data from asymptomatic children and adults in the same community. RESULTS Acute malaria was marked by elevated levels of proinflammatory and regulatory cytokines and expansion of the inflammatory "intermediate" monocyte subset that returned to levels of healthy asymptomatic children 6 weeks later. Monocytes displayed activated phenotypes during acute malaria, with changes in surface expression of markers important to innate and adaptive immunity. Functionally, acute malaria monocytes and monocytes from asymptomatic infected children had impaired phagocytosis of P. falciparum-infected erythrocytes relative to asymptomatic children with no blood-stage infection. Monocytes from both acute malaria and recovery time points displayed strong and equivalent cytokine responsiveness to innate immune agonists that were independent of infection status. Monocyte transcriptional profiles revealed regulated and balanced proinflammatory and antiinflammatory and altered phagocytosis gene expression patterns distinct from malaria-naive monocytes. CONCLUSION These observations provide insights into monocyte functions and the innate immune response during uncomplicated malaria and suggest that asymptomatic parasitemia in children is not clinically benign. FUNDING Support for this work was provided by NIH/National Institute of Allergy and Infectious Diseases (R01AI095192-05), the Burroughs Wellcome Fund/American Society of Tropical Medicine and Hygiene, and the Rainbow Babies & Children's Foundation.
Collapse
Affiliation(s)
- Katherine R Dobbs
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA.,Division of Pediatric Infectious Diseases, University Hospitals Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
| | - Paula Embury
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
| | - John Vulule
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Peter S Odada
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Bruce A Rosa
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Makedonka Mitreva
- McDonnell Genome Institute, Washington University School of Medicine, St. Louis, Missouri, USA
| | - James W Kazura
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA
| | - Arlene E Dent
- Center for Global Health and Diseases, Case Western Reserve University, Cleveland, Ohio, USA.,Division of Pediatric Infectious Diseases, University Hospitals Rainbow Babies and Children's Hospital, Cleveland, Ohio, USA
| |
Collapse
|
37
|
Alkaitis MS, Wang H, Ikeda AK, Rowley CA, MacCormick IJC, Chertow JH, Billker O, Suffredini AF, Roberts DJ, Taylor TE, Seydel KB, Ackerman HC. Decreased Rate of Plasma Arginine Appearance in Murine Malaria May Explain Hypoargininemia in Children With Cerebral Malaria. J Infect Dis 2017; 214:1840-1849. [PMID: 27923948 DOI: 10.1093/infdis/jiw452] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 09/16/2016] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Plasmodium infection depletes arginine, the substrate for nitric oxide synthesis, and impairs endothelium-dependent vasodilation. Increased conversion of arginine to ornithine by parasites or host arginase is a proposed mechanism of arginine depletion. METHODS We used high-performance liquid chromatography to measure plasma arginine, ornithine, and citrulline levels in Malawian children with cerebral malaria and in mice infected with Plasmodium berghei ANKA with or without the arginase gene. Heavy isotope-labeled tracers measured by quadrupole time-of-flight liquid chromatography-mass spectrometry were used to quantify the in vivo rate of appearance and interconversion of plasma arginine, ornithine, and citrulline in infected mice. RESULTS Children with cerebral malaria and P. berghei-infected mice demonstrated depletion of plasma arginine, ornithine, and citrulline. Knock out of Plasmodium arginase did not alter arginine depletion in infected mice. Metabolic tracer analysis demonstrated that plasma arginase flux was unchanged by P. berghei infection. Instead, infected mice exhibited decreased rates of plasma arginine, ornithine, and citrulline appearance and decreased conversion of plasma citrulline to arginine. Notably, plasma arginine use by nitric oxide synthase was decreased in infected mice. CONCLUSIONS Simultaneous arginine and ornithine depletion in malaria parasite-infected children cannot be fully explained by plasma arginase activity. Our mouse model studies suggest that plasma arginine depletion is driven primarily by a decreased rate of appearance.
Collapse
Affiliation(s)
- Matthew S Alkaitis
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville.,Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington
| | - Honghui Wang
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - Allison K Ikeda
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville
| | - Carol A Rowley
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville
| | - Ian J C MacCormick
- Department of Eye and Vision Science, University of Liverpool.,Centre for Clinical Brain Sciences, University of Edinburgh.,Malawi-Liverpool-Wellcome Trust Clinical Research Programme
| | - Jessica H Chertow
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville
| | | | - Anthony F Suffredini
- Critical Care Medicine Department, Clinical Center, National Institutes of Health, Bethesda, Maryland
| | - David J Roberts
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington.,National Health Service Blood and Transplant, John Radcliffe Hospital, Oxford, United Kingdom
| | - Terrie E Taylor
- Michigan State University, East Lansing.,Blantyre Malaria Project, Malawi
| | - Karl B Seydel
- Michigan State University, East Lansing.,Blantyre Malaria Project, Malawi
| | - Hans C Ackerman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville
| |
Collapse
|
38
|
Yeo TW, Florence SM, Kalingonji AR, Chen Y, Granger DL, Anstey NM, Mwaikambo ED, Weinberg JB. Decreased Microvascular Function in Tanzanian Children With Severe and Uncomplicated Falciparum Malaria. Open Forum Infect Dis 2017; 4:ofx079. [PMID: 28852670 PMCID: PMC5569702 DOI: 10.1093/ofid/ofx079] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Accepted: 04/19/2017] [Indexed: 11/13/2022] Open
Abstract
Microvascular function and oxygen consumption affect oxygen homeostasis but have not been assessed in African children with malaria. Microvascular function in Tanzanian children with severe malaria (SM) or uncomplicated malaria were 39% and 72%, respectively, of controls (P < .001). Uncomplicated malaria (P = .04), not SM (P = .06), children had increased oxygen consumption compared with controls.
Collapse
Affiliation(s)
- Tsin W Yeo
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Communicable Disease Centre, Institute of Infectious Diseases and Epidemiology, Tan Tock Seng Hospital, Singapore.,Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| | | | | | - Youwei Chen
- Duke University and V.A. Medical Centers, Durham, North Carolina; and
| | | | - Nicholas M Anstey
- Menzies School of Health Research and Charles Darwin University, Darwin, Australia
| | | | - J Brice Weinberg
- Duke University and V.A. Medical Centers, Durham, North Carolina; and
| |
Collapse
|
39
|
Gillrie MR, Ho M. Dynamic interactions of Plasmodium spp. with vascular endothelium. Tissue Barriers 2017; 5:e1268667. [PMID: 28452684 PMCID: PMC5362994 DOI: 10.1080/21688370.2016.1268667] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 11/24/2016] [Accepted: 11/30/2016] [Indexed: 12/18/2022] Open
Abstract
Plasmodial species are protozoan parasites that infect erythrocytes. As such, they are in close contact with microvascular endothelium for most of the life cycle in the mammalian host. The host-parasite interactions of this stage of the infection are responsible for the clinical manifestations of the disease that range from a mild febrile illness to severe and frequently fatal syndromes such as cerebral malaria and multi-organ failure. Plasmodium falciparum, the causative agent of the most severe form of malaria, is particularly predisposed to modulating endothelial function through either direct adhesion to endothelial receptor molecules, or by releasing potent host and parasite products that can stimulate endothelial activation and/or disrupt barrier function. In this review, we provide a critical analysis of the current clinical and laboratory evidence for endothelial dysfunction during severe P. falciparum malaria. Future investigations using state-of-the-art technologies such as mass cytometry and organs-on-chips to further delineate parasite-endothelial cell interactions are also discussed.
Collapse
Affiliation(s)
- Mark R. Gillrie
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
- Department of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - May Ho
- Department of Microbiology, Immunology and Infectious Diseases, University of Calgary, Calgary, Alberta, Canada
| |
Collapse
|
40
|
Alkaitis MS, Ackerman HC. Tetrahydrobiopterin Supplementation Improves Phenylalanine Metabolism in a Murine Model of Severe Malaria. ACS Infect Dis 2016; 2:827-838. [PMID: 27641435 PMCID: PMC6289270 DOI: 10.1021/acsinfecdis.6b00124] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Tetrahydrobiopterin (BH4) is an essential cofactor for both phenylalanine hydroxylase and nitric oxide synthase. Patients with severe malaria have low urinary BH4, elevated plasma phenylalanine, and impaired endothelium-dependent vasodilation, suggesting that BH4 depletion may limit phenylalanine metabolism and nitric oxide synthesis. We infected C57BL/6 mice with Plasmodium berghei ANKA to characterize BH4 availability and to investigate the effects of BH4 supplementation. P. berghei ANKA infection lowered BH4 in plasma, erythrocytes, and brain tissue but raised it in aorta and liver tissue. The ratio of BH4 to 7,8-BH2 (the major product of BH4 oxidation) was decreased in plasma, erythrocytes, and brain tissue, suggesting that oxidation contributes to BH4 depletion. The continuous infusion of sepiapterin (a BH4 precursor) and citrulline (an arginine precursor) raised the concentrations of BH4 and arginine in both blood and tissue compartments. The restoration of systemic BH4 and arginine availability in infected mice produced only a minor improvement in whole blood nitrite concentrations, a biomarker of NO synthesis, and failed to prevent the onset of severe disease symptoms. However, sepiapterin and citrulline infusion reduced the ratio of phenylalanine to tyrosine in plasma, aortic tissue, and brain tissue. In summary, BH4 depletion in P. berghei infection may compromise both nitric oxide synthesis and phenylalanine metabolism; however, these findings require further investigation in human patients with severe malaria.
Collapse
Affiliation(s)
- Matthew S. Alkaitis
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
- Radcliffe Department of Medicine, University of Oxford, John Radcliffe Hospital, Headington Oxford, United Kingdom
| | - Hans C. Ackerman
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, United States
| |
Collapse
|
41
|
Barber BE, William T, Grigg MJ, Piera KA, Chen Y, Wang H, Weinberg JB, Yeo TW, Anstey NM. Nitric Oxide-Dependent Endothelial Dysfunction and Reduced Arginine Bioavailability in Plasmodium vivax Malaria but No Greater Increase in Intravascular Hemolysis in Severe Disease. J Infect Dis 2016; 214:1557-1564. [PMID: 27630198 DOI: 10.1093/infdis/jiw427] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/06/2016] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Pathogenesis of severe Plasmodium vivax malaria is poorly understood. Endothelial dysfunction and reduced nitric oxide (NO) bioavailability characterize severe falciparum malaria, but have not been assessed in severe vivax malaria. METHODS In patients with severe vivax malaria (n = 9), patients with nonsevere vivax malaria (n = 58), and healthy controls (n = 79), we measured NO-dependent endothelial function by using reactive hyperemia-peripheral arterial tonometry (RH-PAT) and assessed associations with arginine, asymmetric dimethylarginine (ADMA), and hemolysis. RESULTS The L-arginine level and the L-arginine to ADMA ratio (a measure of L-arginine bioavailability) were reduced in patients with severe vivax malaria and those with nonsevere vivax malaria, compared with healthy controls (median L-arginine level, 65, 66, and 98 µmol/mL, respectively [P = .0001]; median L-arginine to ADMA ratio, 115, 125, and 187, respectively [P = .0001]). Endothelial function was impaired in proportion to disease severity (median RH-PAT index, 1.49, 1.73, and 1.97 in patients with severe vivax malaria, those with nonsevere vivax malaria, and healthy controls, respectively; P = .018) and was associated with the L-arginine to ADMA ratio. While the posttreatment fall in hemoglobin level was greater in severe vivax malaria as compared to nonsevere vivax malaria (2.5 vs 1 g/dL; P = .0001), markers of intravascular hemolysis were not higher in severe disease. CONCLUSIONS Endothelial function is impaired in nonsevere and severe vivax malaria, is associated with reduced L-arginine bioavailability, and may contribute to microvascular pathogenesis. Severe disease appears to be more associated with extravascular hemolysis than with intravascular hemolysis.
Collapse
Affiliation(s)
- Bridget E Barber
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University.,Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit
| | - Timothy William
- Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit.,Jesselton Medical Center, Kota Kinabalu, Malaysia
| | - Matthew J Grigg
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University.,Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit
| | - Kim A Piera
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University
| | - Youwei Chen
- Duke University Medical Center.,VA Medical Center, Durham, North Carolina
| | - Hao Wang
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University
| | - J Brice Weinberg
- Duke University Medical Center.,VA Medical Center, Durham, North Carolina
| | - Tsin W Yeo
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University.,Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit.,Lee Kong Chian School of Medicine, Nanyang Technological University.,Institute of Infectious Disease and Epidemiology, Tan Tock Seng Hospital, Singapore
| | - Nicholas M Anstey
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University.,Department of Infectious Diseases, Royal Darwin Hospital, Darwin, Australia.,Infectious Diseases Society Sabah-Menzies School of Health Research Clinical Research Unit
| |
Collapse
|