1
|
Onyango CO, Anyona SB, Hurwitz I, Raballah E, Wasena SA, Osata SW, Seidenberg P, McMahon BH, Lambert CG, Schneider KA, Ouma C, Cheng Q, Perkins DJ. Transcriptomic and Proteomic Insights into Host Immune Responses in Pediatric Severe Malarial Anemia: Dysregulation in HSP60-70-TLR2/4 Signaling and Altered Glutamine Metabolism. Pathogens 2024; 13:867. [PMID: 39452740 PMCID: PMC11510049 DOI: 10.3390/pathogens13100867] [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: 08/06/2024] [Revised: 09/26/2024] [Accepted: 09/27/2024] [Indexed: 10/26/2024] Open
Abstract
Severe malarial anemia (SMA, Hb < 6.0 g/dL) is a leading cause of childhood morbidity and mortality in holoendemic Plasmodium falciparum transmission zones. This study explored the entire expressed human transcriptome in whole blood from 66 Kenyan children with non-SMA (Hb ≥ 6.0 g/dL, n = 41) and SMA (n = 25), focusing on host immune response networks. RNA-seq analysis revealed 6862 differentially expressed genes, with equally distributed up-and down-regulated genes, indicating a complex host immune response. Deconvolution analyses uncovered leukocytic immune profiles indicative of a diminished antigenic response, reduced immune priming, and polarization toward cellular repair in SMA. Weighted gene co-expression network analysis revealed that immune-regulated processes are central molecular distinctions between non-SMA and SMA. A top dysregulated immune response signaling network in SMA was the HSP60-HSP70-TLR2/4 signaling pathway, indicating altered pathogen recognition, innate immune activation, stress responses, and antigen recognition. Validation with high-throughput gene expression from a separate cohort of Kenyan children (n = 50) with varying severities of malarial anemia (n = 38 non-SMA and n = 12 SMA) confirmed the RNA-seq findings. Proteomic analyses in 35 children with matched transcript and protein abundance (n = 19 non-SMA and n = 16 SMA) confirmed dysregulation in the HSP60-HSP70-TLR2/4 signaling pathway. Additionally, glutamine transporter and glutamine synthetase genes were differentially expressed, indicating altered glutamine metabolism in SMA. This comprehensive analysis underscores complex immune dysregulation and novel pathogenic features in SMA.
Collapse
Affiliation(s)
- Clinton O. Onyango
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno 40100, Kenya; (C.O.O.); (S.A.W.); (S.W.O.); (C.O.)
- Center for Global Health, Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (I.H.); (P.S.); (B.H.M.); (K.A.S.)
| | - Samuel B. Anyona
- Kenya Global Health Programs, University of New Mexico, Kisumu and Siaya 40100, Kenya; (S.B.A.); (E.R.)
- Department of Medical Biochemistry, School of Medicine, Maseno University, Maseno 40100, Kenya
| | - Ivy Hurwitz
- Center for Global Health, Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (I.H.); (P.S.); (B.H.M.); (K.A.S.)
| | - Evans Raballah
- Kenya Global Health Programs, University of New Mexico, Kisumu and Siaya 40100, Kenya; (S.B.A.); (E.R.)
- Department of Medical Laboratory Sciences, School of Public Health Biomedical Sciences and Technology, Masinde Muliro University of Science and Technology, Kakamega 50100, Kenya
| | - Sharely A. Wasena
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno 40100, Kenya; (C.O.O.); (S.A.W.); (S.W.O.); (C.O.)
- Center for Global Health, Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (I.H.); (P.S.); (B.H.M.); (K.A.S.)
| | - Shamim W. Osata
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno 40100, Kenya; (C.O.O.); (S.A.W.); (S.W.O.); (C.O.)
- Center for Global Health, Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (I.H.); (P.S.); (B.H.M.); (K.A.S.)
| | - Philip Seidenberg
- Center for Global Health, Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (I.H.); (P.S.); (B.H.M.); (K.A.S.)
- Department of Emergency Medicine, School of Medicine, University of New Mexico, Albuquerque, NM 87131, USA
| | - Benjamin H. McMahon
- Center for Global Health, Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (I.H.); (P.S.); (B.H.M.); (K.A.S.)
- Theoretical Biology and Biophysics Group, Los Alamos National Laboratory, Theoretical Division, Los Alamos, NM 87545, USA
| | - Christophe G. Lambert
- Center for Global Health, Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (I.H.); (P.S.); (B.H.M.); (K.A.S.)
- Department of Internal Medicine, Division of Translational Informatics, University of New Mexico, Albuquerque, NM 87131, USA
| | - Kristan A. Schneider
- Center for Global Health, Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (I.H.); (P.S.); (B.H.M.); (K.A.S.)
- Department of Internal Medicine, Division of Translational Informatics, University of New Mexico, Albuquerque, NM 87131, USA
| | - Collins Ouma
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno 40100, Kenya; (C.O.O.); (S.A.W.); (S.W.O.); (C.O.)
- Center for Global Health, Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (I.H.); (P.S.); (B.H.M.); (K.A.S.)
| | - Qiuying Cheng
- Center for Global Health, Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (I.H.); (P.S.); (B.H.M.); (K.A.S.)
- Kenya Global Health Programs, University of New Mexico, Kisumu and Siaya 40100, Kenya; (S.B.A.); (E.R.)
| | - Douglas J. Perkins
- Center for Global Health, Internal Medicine, University of New Mexico, Albuquerque, NM 87131, USA; (I.H.); (P.S.); (B.H.M.); (K.A.S.)
- Kenya Global Health Programs, University of New Mexico, Kisumu and Siaya 40100, Kenya; (S.B.A.); (E.R.)
| |
Collapse
|
2
|
Boyle MJ, Engwerda CR, Jagannathan P. The impact of Plasmodium-driven immunoregulatory networks on immunity to malaria. Nat Rev Immunol 2024; 24:637-653. [PMID: 38862638 DOI: 10.1038/s41577-024-01041-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2024] [Indexed: 06/13/2024]
Abstract
Malaria, caused by infection with Plasmodium parasites, drives multiple regulatory responses across the immune landscape. These regulatory responses help to protect against inflammatory disease but may in some situations hamper the acquisition of adaptive immune responses that clear parasites. In addition, the regulatory responses that occur during Plasmodium infection may negatively affect malaria vaccine efficacy in the most at-risk populations. Here, we discuss the specific cellular mechanisms of immunoregulatory networks that develop during malaria, with a focus on knowledge gained from human studies and studies that involve the main malaria parasite to affect humans, Plasmodium falciparum. Leveraging this knowledge may lead to the development of new therapeutic approaches to increase protective immunity to malaria during infection or after vaccination.
Collapse
Affiliation(s)
- Michelle J Boyle
- Life Sciences Division, Burnet Institute, Melbourne, Victoria, Australia.
- QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| | | | - Prasanna Jagannathan
- Department of Medicine, Stanford University, Stanford, CA, USA.
- Department of Microbiology and Immunology, Stanford University, Stanford, CA, USA.
| |
Collapse
|
3
|
Silva NI, Souza PFL, Silva BF, Fonseca SG, Gardinassi LG. Host Transcriptional Meta-signatures Reveal Diagnostic Biomarkers for Plasmodium falciparum Malaria. J Infect Dis 2024; 230:e474-e485. [PMID: 38271704 PMCID: PMC11326815 DOI: 10.1093/infdis/jiae041] [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: 06/21/2023] [Revised: 11/28/2023] [Accepted: 01/24/2024] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND Transcriptomics has been used to evaluate immune responses during malaria in diverse cohorts worldwide. However, the high heterogeneity of cohorts and poor generalization of transcriptional signatures reported in each study limit their potential clinical applications. METHODS We compiled 28 public data sets containing 1556 whole-blood or peripheral blood mononuclear cell transcriptome samples. We estimated effect sizes with Hedge's g value and the DerSimonian-Laird random-effects model for meta-analyses of uncomplicated malaria. Random forest models identified gene signatures that discriminate malaria from bacterial infections or malaria severity. Parasitological, hematological, immunological, and metabolomics data were used for validation. RESULTS We identified 3 gene signatures: the uncomplicated Malaria Meta-Signature, which discriminates Plasmodium falciparum malaria from uninfected controls; the Malaria or Bacteria Signature, which distinguishes malaria from sepsis and enteric fever; and the cerebral Malaria Meta-Signature, which characterizes individuals with cerebral malaria. These signatures correlate with clinical hallmark features of malaria. Blood transcription modules indicate immune regulation by glucocorticoids, whereas cell development and adhesion are associated with cerebral malaria. CONCLUSIONS Transcriptional meta-signatures reflecting immune cell responses provide potential biomarkers for translational innovation and suggest critical roles for metabolic regulators of inflammation during malaria.
Collapse
Affiliation(s)
- Nágila Isleide Silva
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Pedro Felipe Loyola Souza
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Bárbara Fernandes Silva
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Simone Gonçalves Fonseca
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
| | - Luiz Gustavo Gardinassi
- Departamento de Biociências e Tecnologia, Instituto de Patologia Tropical e Saúde Pública, Universidade Federal de Goiás, Goiânia, Brazil
- Departamento de Enfermagem Materno-Infantil e Saúde Pública, Escola de Enfermagem de Ribeirão Preto, Universidade de São Paulo, Ribeirão Preto, Brazil
| |
Collapse
|
4
|
Nideffer J, Ty M, Donato M, John R, Kajubi R, Ji X, Nankya F, Musinguzi K, Press KD, Yang N, Camanag K, Greenhouse B, Kamya M, Feeney ME, Dorsey G, Utz PJ, Pulendran B, Khatri P, Jagannathan P. Clinical immunity to malaria involves epigenetic reprogramming of innate immune cells. PNAS NEXUS 2024; 3:pgae325. [PMID: 39161730 PMCID: PMC11331423 DOI: 10.1093/pnasnexus/pgae325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 07/23/2024] [Indexed: 08/21/2024]
Abstract
The regulation of inflammation is a critical aspect of disease tolerance and naturally acquired clinical immunity to malaria. Here, we demonstrate using RNA sequencing and epigenetic landscape profiling by cytometry by time-of-flight, that the regulation of inflammatory pathways during asymptomatic parasitemia occurs downstream of pathogen sensing-at the epigenetic level. The abundance of certain epigenetic markers (methylation of H3K27 and dimethylation of arginine residues) and decreased prevalence of histone variant H3.3 correlated with suppressed cytokine responses among monocytes of Ugandan children. Such an epigenetic signature was observed across diverse immune cell populations and not only characterized active asymptomatic parasitemia but also correlated with future long-term disease tolerance and clinical immunity when observed in uninfected children. Pseudotime analyses revealed a potential trajectory of epigenetic change that correlated with a child's age and recent parasite exposure and paralleled the acquisition of clinical immunity. Thus, our data support a model whereby exposure to Plasmodium falciparum induces epigenetic changes that regulate excessive inflammation and contribute to naturally acquire clinical immunity to malaria.
Collapse
Affiliation(s)
- Jason Nideffer
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Maureen Ty
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Michele Donato
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Rek John
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Richard Kajubi
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Xuhuai Ji
- Institute for Immunity, Infection, and Transplantation, Stanford University, Stanford, CA 94305, USA
| | | | | | | | - Nora Yang
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Kylie Camanag
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Bryan Greenhouse
- Department of Medicine, University of California, San Francisco, CA 94142, USA
| | - Moses Kamya
- School of Medicine, Makerere University, Kampala, Uganda
| | - Margaret E Feeney
- Department of Pediatrics, University of California, San Francisco, CA 94142, USA
| | - Grant Dorsey
- Department of Medicine, University of California, San Francisco, CA 94142, USA
| | - Paul J Utz
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Bali Pulendran
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Purvesh Khatri
- Department of Medicine, Stanford University, Stanford, CA 94305, USA
| | | |
Collapse
|
5
|
Anyona SB, Cheng Q, Wasena SA, Osata SW, Guo Y, Raballah E, Hurwitz I, Onyango CO, Ouma C, Seidenberg PD, McMahon BH, Lambert CG, Schneider KA, Perkins DJ. Entire expressed peripheral blood transcriptome in pediatric severe malarial anemia. Nat Commun 2024; 15:5037. [PMID: 38866743 PMCID: PMC11169501 DOI: 10.1038/s41467-024-48259-4] [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: 07/07/2023] [Accepted: 04/25/2024] [Indexed: 06/14/2024] Open
Abstract
This study on severe malarial anemia (SMA: Hb < 6.0 g/dL), a leading global cause of childhood morbidity and mortality, compares the entire expressed whole blood host transcriptome between Kenyan children (3-48 mos.) with non-SMA (Hb ≥ 6.0 g/dL, n = 39) and SMA (n = 18). Differential expression analyses reveal 1403 up-regulated and 279 down-regulated transcripts in SMA, signifying impairments in host inflammasome activation, cell death, and innate immune and cellular stress responses. Immune cell profiling shows decreased memory responses, antigen presentation, and immediate pathogen clearance, suggesting an immature/improperly regulated immune response in SMA. Module repertoire analysis of blood-specific gene signatures identifies up-regulation of erythroid genes, enhanced neutrophil activation, and impaired inflammatory responses in SMA. Enrichment analyses converge on disruptions in cellular homeostasis and regulatory pathways for the ubiquitin-proteasome system, autophagy, and heme metabolism. Pathway analyses highlight activation in response to hypoxic conditions [Hypoxia Inducible Factor (HIF)-1 target and Reactive Oxygen Species (ROS) signaling] as a central theme in SMA. These signaling pathways are also top-ranking in protein abundance measures and a Ugandan SMA cohort with available transcriptomic data. Targeted RNA-Seq validation shows strong concordance with our entire expressed transcriptome data. These findings identify key molecular themes in SMA pathogenesis, offering potential targets for new malaria therapies.
Collapse
Affiliation(s)
- Samuel B Anyona
- Department of Medical Biochemistry, School of Medicine, Maseno University, Maseno, 40105, Kenya.
- University of New Mexico-Kenya Global Health Programs, Kisumu and Siaya, 40100, Kenya.
| | - Qiuying Cheng
- Department of Internal Medicine, Center for Global Health, University of New Mexico, Albuquerque, NM, 87131-0001, USA
| | - Sharley A Wasena
- University of New Mexico-Kenya Global Health Programs, Kisumu and Siaya, 40100, Kenya
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, 40105, Kenya
| | - Shamim W Osata
- University of New Mexico-Kenya Global Health Programs, Kisumu and Siaya, 40100, Kenya
| | - Yan Guo
- Department of Public Health Sciences, University of Miami, Miami, 33136, USA
| | - Evans Raballah
- University of New Mexico-Kenya Global Health Programs, Kisumu and Siaya, 40100, Kenya
- Department of Medical Laboratory Sciences, School of Public Health, Biomedical Sciences and Technology, Masinde Muliro University of Science and Technology, Kakamega, 50100, Kenya
| | - Ivy Hurwitz
- Department of Internal Medicine, Center for Global Health, University of New Mexico, Albuquerque, NM, 87131-0001, USA
| | - Clinton O Onyango
- University of New Mexico-Kenya Global Health Programs, Kisumu and Siaya, 40100, Kenya
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, 40105, Kenya
| | - Collins Ouma
- University of New Mexico-Kenya Global Health Programs, Kisumu and Siaya, 40100, Kenya
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, 40105, Kenya
| | - Philip D Seidenberg
- Department of Emergency Medicine, School of Medicine, University of New Mexico, Albuquerque, NM, 87131-0001, USA
| | - Benjamin H McMahon
- Theoretical Biology and Biophysics Group, Theoretical Division, Los Alamos National Laboratory, Los Alamos, NM, 87545, USA
| | - Christophe G Lambert
- Department of Internal Medicine, Division of Translational Informatics, University of New Mexico, Albuquerque, NM, 87131-0001, USA
| | - Kristan A Schneider
- Department of Internal Medicine, Division of Translational Informatics, University of New Mexico, Albuquerque, NM, 87131-0001, USA
- Department Applied Computer and Bio-Sciences, University of Applied Sciences Mittweida, Mittweida, 09648, Germany
| | - Douglas J Perkins
- University of New Mexico-Kenya Global Health Programs, Kisumu and Siaya, 40100, Kenya.
- Department of Internal Medicine, Center for Global Health, University of New Mexico, Albuquerque, NM, 87131-0001, USA.
| |
Collapse
|
6
|
Dunican C, Andradi-Brown C, Ebmeier S, Georgiadou A, Cunnington AJ. The malarial blood transcriptome: translational applications. Biochem Soc Trans 2024; 52:651-660. [PMID: 38421063 PMCID: PMC11088907 DOI: 10.1042/bst20230497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/02/2024]
Abstract
The blood transcriptome of malaria patients has been used extensively to elucidate the pathophysiological mechanisms and host immune responses to disease, identify candidate diagnostic and prognostic biomarkers, and reveal new therapeutic targets for drug discovery. This review gives a high-level overview of the three main translational applications of these studies (diagnostics, prognostics, and therapeutics) by summarising recent literature and outlining the main limitations and future directions of each application. It highlights the need for consistent and accurate definitions of disease states and subject groups and discusses how prognostic studies must distinguish clearly between analyses that attempt to predict future disease states and those which attempt to discriminate between current disease states (classification). Lastly it examines how many promising therapeutics fail due to the choice of imperfect animal models for pre-clinical testing and lack of appropriate validation studies in humans, and how future transcriptional studies may be utilised to overcome some of these limitations.
Collapse
Affiliation(s)
- Claire Dunican
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, U.K
- Centre for Paediatrics and Child Health, Imperial College London, London, U.K
| | - Clare Andradi-Brown
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, U.K
- Centre for Paediatrics and Child Health, Imperial College London, London, U.K
| | - Stefan Ebmeier
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, U.K
- Centre for Paediatrics and Child Health, Imperial College London, London, U.K
| | - Athina Georgiadou
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, U.K
- Centre for Paediatrics and Child Health, Imperial College London, London, U.K
| | - Aubrey J. Cunnington
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, U.K
- Centre for Paediatrics and Child Health, Imperial College London, London, U.K
| |
Collapse
|
7
|
Berna-Erro A, Granados MP, Teruel-Montoya R, Ferrer-Marin F, Delgado E, Corbacho AJ, Fenández E, Vazquez-Godoy MT, Tapia JA, Redondo PC. SARAF overexpression impairs thrombin-induced Ca 2+ homeostasis in neonatal platelets. Br J Haematol 2024; 204:988-1004. [PMID: 38062782 DOI: 10.1111/bjh.19210] [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: 01/14/2023] [Revised: 11/01/2023] [Accepted: 11/07/2023] [Indexed: 03/14/2024]
Abstract
Neonatal platelets present a reduced response to the platelet agonist, thrombin (Thr), thus resulting in a deficient Thr-induced aggregation. These alterations are more pronounced in premature newborns. Here, our aim was to uncover the causes underneath the impaired Ca2+ homeostasis described in neonatal platelets. Both Ca2+ mobilization and Ca2+ influx in response to Thr are decreased in neonatal platelets compared to maternal and control woman platelets. In neonatal platelets, we observed impaired Ca2+ mobilization in response to the PAR-1 agonist (SFLLRN) or by blocking SERCA3 function with tert-butylhydroquinone. Regarding SOCE, the STIM1 regulatory protein, SARAF, was found overexpressed in neonatal platelets, promoting an increase in STIM1/SARAF interaction even under resting conditions. Additionally, higher interaction between SARAF and PDCD61/ALG2 was also observed, reducing SARAF ubiquitination and prolonging its half-life. These results were reproduced by overexpressing SARAF in MEG01 and DAMI cells. Finally, we also observed that pannexin 1 permeability is enhanced in response to Thr in control woman and maternal platelets, but not in neonatal platelets, hence, leading to the deregulation of the Ca2+ entry found in neonatal platelets. Summarizing, we show that in neonatal platelets both Ca2+ accumulation in the intracellular stores and Thr-evoked Ca2+ entry through either capacitative channels or non-selective channels are altered in neonatal platelets, contributing to deregulated Ca2+ homeostasis in neonatal platelets and leading to the altered aggregation observed in these subjects.
Collapse
Affiliation(s)
- Alejandro Berna-Erro
- Department of Physiology (PHYCELL Group), University of Extremadura, Caceres, Spain
| | - Maria P Granados
- Pharmacy Unit of Health Center, Extremadura County Health Service, Caceres, Spain
| | - Raul Teruel-Montoya
- Hemodonation County Center, University Hospital of Morales-Meseguer, IMIB-Arrixaca, CIBERER CB55, Murcia, Spain
| | - Francisca Ferrer-Marin
- Hemodonation County Center, University Hospital of Morales-Meseguer, IMIB-Arrixaca, CIBERER CB55, Murcia, Spain
| | - Elena Delgado
- Blood Donation Center, Extremadura County Health Service, Merida, Spain
| | | | | | | | - Jose A Tapia
- Department of Physiology (PHYCELL Group), University of Extremadura, Caceres, Spain
| | - Pedro Cosme Redondo
- Department of Physiology (PHYCELL Group), University of Extremadura, Caceres, Spain
| |
Collapse
|
8
|
Duarte N, Shafi AM, Penha-Gonçalves C, Pais TF. Endothelial type I interferon response and brain diseases: identifying STING as a therapeutic target. Front Cell Dev Biol 2023; 11:1249235. [PMID: 37791071 PMCID: PMC10542901 DOI: 10.3389/fcell.2023.1249235] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 08/30/2023] [Indexed: 10/05/2023] Open
Abstract
The endothelium layer lining the inner surface of blood vessels serves relevant physiological functions in all body systems, including the exchanges between blood and extravascular space. However, endothelial cells also participate in innate and adaptive immune response that contribute to the pathophysiology of inflammatory disorders. Type I Interferon (IFN) signaling is an inflammatory response triggered by a variety of pathogens, but it can also be induced by misplaced DNA in the cytosol caused by cell stress or gene mutations. Type I IFN produced by blood leukocytes or by the endothelium itself is well-known to activate the interferon receptor (IFNAR) in endothelial cells. Here, we discuss the induction of type I IFN secretion and signaling in the endothelium, specifically in the brain microvasculature where endothelial cells participate in the tight blood-brain barrier (BBB). This barrier is targeted during neuroinflammatory disorders such as infection, multiple sclerosis, Alzheimer's disease and traumatic brain injury. We focus on type I IFN induction through the cGAS-STING activation pathway in endothelial cells in context of autoinflammatory type I interferonopathies, inflammation and infection. By comparing the pathophysiology of two separate infectious diseases-cerebral malaria induced by Plasmodium infection and COVID-19 caused by SARS-CoV-2 infection-we emphasize the relevance of type I IFN and STING-induced vasculopathy in organ dysfunction. Investigating the role of endothelial cells as active type I IFN producers and responders in disease pathogenesis could lead to new therapeutic targets. Namely, endothelial dysfunction and brain inflammation may be avoided with strategies that target excessive STING activation in endothelial cells.
Collapse
|
9
|
Prah DA, Dunican C, Amoah LE, Marjaneh MM, Kaforou M, Nordgren A, Jones-Warner W, Aniweh Y, Awandare GA, Cunnington AJ, Hafalla JC. Asymptomatic Plasmodium falciparum infection evades triggering a host transcriptomic response. J Infect 2023; 87:259-262. [PMID: 37356628 DOI: 10.1016/j.jinf.2023.06.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/17/2023] [Accepted: 06/21/2023] [Indexed: 06/27/2023]
Affiliation(s)
- Diana Ahu Prah
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Ghana; Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Ghana; Department of Infection Biology, Faculty of Infectious and Tropical Medicine, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Claire Dunican
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, United Kingdom
| | - Linda Eva Amoah
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Ghana; Immunology Department, Noguchi Memorial Institute for Medical Research, University of Ghana, Legon, Ghana
| | - Mahdi Moradi Marjaneh
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, United Kingdom
| | - Myrsini Kaforou
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, United Kingdom; Centre for Paediatrics and Child Health, Imperial College London, United Kingdom
| | - Asa Nordgren
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, United Kingdom
| | - William Jones-Warner
- Department of Infection Biology, Faculty of Infectious and Tropical Medicine, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Yaw Aniweh
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Ghana; Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Ghana
| | - Gordon A Awandare
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Ghana; Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Legon, Ghana
| | - Aubrey J Cunnington
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, United Kingdom; Centre for Paediatrics and Child Health, Imperial College London, United Kingdom.
| | - Julius Clemence Hafalla
- Department of Infection Biology, Faculty of Infectious and Tropical Medicine, London School of Hygiene and Tropical Medicine, London, United Kingdom.
| |
Collapse
|
10
|
Anyona S, Cheng Q, Guo Y, Raballah E, Hurwitz I, Onyango C, Seidenberg P, Schneider K, Lambert C, McMahon B, Ouma C, Perkins D. Entire Expressed Peripheral Blood Transcriptome in Pediatric Severe Malarial Anemia. RESEARCH SQUARE 2023:rs.3.rs-3150748. [PMID: 37503086 PMCID: PMC10371159 DOI: 10.21203/rs.3.rs-3150748/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/29/2023]
Abstract
This study on severe malarial anemia (SMA: Hb < 6.0 g/dL), a leading global cause of childhood morbidity and mortality, analyzed the entire expressed transcriptome in whole blood from children with non-SMA (Hb ≥ 6.0 g/dL, n = 41) and SMA (n = 25). Analyses revealed 3,420 up-regulated and 3,442 down-regulated transcripts, signifying impairments in host inflammasome activation, cell death, innate immune responses, and cellular stress responses in SMA. Immune cell profiling showed a decreased antigenic and immune priming response in children with SMA, favoring polarization toward cellular proliferation and repair. Enrichment analysis further identified altered neutrophil and autophagy-related processes, consistent with neutrophil degranulation and altered ubiquitination and proteasome degradation. Pathway analyses highlighted SMA-related alterations in cellular homeostasis, signaling, response to environmental cues, and cellular and immune stress responses. Validation with a qRT-PCR array showed strong concordance with the sequencing data. These findings identify key molecular themes in SMA pathogenesis, providing potential targets for new malaria therapies.
Collapse
Affiliation(s)
| | | | | | - Evans Raballah
- School of Public Health, Biomedical Sciences and Technology, Masinde Muliro University of Science and Technology
| | - Ivy Hurwitz
- Center for Global Health, University of New Mexico
| | - Clinton Onyango
- School of Public Health and Community Development, Maseno University
| | | | | | | | | | | | | |
Collapse
|
11
|
Li JX, Liao WZ, Huang ZM, Yin X, Ouyang S, Gu B, Guo XG. Identifying effective diagnostic biomarkers for childhood cerebral malaria in Africa integrating coexpression analysis with machine learning algorithm. Eur J Med Res 2023; 28:76. [PMID: 36782344 PMCID: PMC9926768 DOI: 10.1186/s40001-022-00980-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Accepted: 12/30/2022] [Indexed: 02/15/2023] Open
Abstract
BACKGROUND Cerebral malaria (CM) is a manifestation of malaria caused by plasmodium infection. It has a high mortality rate and severe neurological sequelae, existing a significant research gap and requiring further study at the molecular level. METHODS We downloaded the GSE117613 dataset from the Gene Expression Omnibus (GEO) database to determine the differentially expressed genes (DEGs) between the CM group and the control group. Weighted gene coexpression network analysis (WGCNA) was applied to select the module and hub genes most relevant to CM. The common genes of the key module and DEGs were selected to perform further analysis. The least absolute shrinkage and selection operator (LASSO) logistic regression and support vector machine recursive feature elimination (SVM-RFE) were applied to screen and verify the diagnostic markers of CM. Eventually, the hub genes were validated in the external dataset. Gene set enrichment analysis (GSEA) was applied to investigate the possible roles of the hub genes. RESULTS The GO and KEGG results showed that DEGs were enriched in some neutrophil-mediated pathways and associated with some lumen structures. Combining LASSO and the SVM-RFE algorithms, LEF1 and IRAK3 were identified as potential hub genes in CM. Through the GSEA enrichment results, we found that LEF1 and IRAK3 participated in maintaining the integrity of the blood-brain barrier (BBB), which contributed to improving the prognosis of CM. CONCLUSIONS This study may help illustrate the pathophysiology of CM at the molecular level. LEF1 and IRAK3 can be used as diagnostic biomarkers, providing new insight into the diagnosis and prognosis prediction in pediatric CM.
Collapse
Affiliation(s)
- Jia-Xin Li
- grid.417009.b0000 0004 1758 4591Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150 China ,grid.410737.60000 0000 8653 1072Department of Clinical Medicine, The First Clinical School of Guangzhou Medical University, Guangzhou, 511436 China
| | - Wan-Zhe Liao
- grid.417009.b0000 0004 1758 4591Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150 China ,grid.410737.60000 0000 8653 1072Department of Clinical Medicine, The Nanshan College of Guangzhou Medical University, Guangzhou, 511436 China
| | - Ze-Min Huang
- grid.417009.b0000 0004 1758 4591Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150 China ,grid.410737.60000 0000 8653 1072Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, 511436 China
| | - Xin Yin
- grid.417009.b0000 0004 1758 4591Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150 China ,grid.410737.60000 0000 8653 1072Department of Pediatrics, The Pediatrics School of Guangzhou Medical University, Guangzhou, 511436 China
| | - Shi Ouyang
- grid.410737.60000 0000 8653 1072Department of Infectious Disease, The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150 China
| | - Bing Gu
- Laboratory Medicine, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510000, China.
| | - Xu-Guang Guo
- Department of Clinical Laboratory Medicine, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China. .,Guangdong Provincial Key Laboratory of Major Obstetric Diseases, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China. .,Guangzhou Key Laboratory for Clinical Rapid Diagnosis and Early Warning of Infectious Diseases, KingMed School of Laboratory Medicine, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
12
|
Muppidi P, Wright E, Wassmer SC, Gupta H. Diagnosis of cerebral malaria: Tools to reduce Plasmodium falciparum associated mortality. Front Cell Infect Microbiol 2023; 13:1090013. [PMID: 36844403 PMCID: PMC9947298 DOI: 10.3389/fcimb.2023.1090013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Accepted: 01/24/2023] [Indexed: 02/11/2023] Open
Abstract
Cerebral malaria (CM) is a major cause of mortality in Plasmodium falciparum (Pf) infection and is associated with the sequestration of parasitised erythrocytes in the microvasculature of the host's vital organs. Prompt diagnosis and treatment are key to a positive outcome in CM. However, current diagnostic tools remain inadequate to assess the degree of brain dysfunction associated with CM before the window for effective treatment closes. Several host and parasite factor-based biomarkers have been suggested as rapid diagnostic tools with potential for early CM diagnosis, however, no specific biomarker signature has been validated. Here, we provide an updated review on promising CM biomarker candidates and evaluate their applicability as point-of-care tools in malaria-endemic areas.
Collapse
Affiliation(s)
- Pranavi Muppidi
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Emily Wright
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Samuel C. Wassmer
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Himanshu Gupta
- Department of Infection Biology, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, UP, India
| |
Collapse
|
13
|
Hu X, Zhao J, Zhao J, Yang E, Jia M. Genome-wide liver transcriptomic profiling of a malaria mouse model reveals disturbed immune and metabolic responses. Parasit Vectors 2023; 16:40. [PMID: 36717945 PMCID: PMC9885691 DOI: 10.1186/s13071-023-05672-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
BACKGROUND The liver is responsible for a range of functions in vertebrates, such as metabolism and immunity. In malaria, the liver plays a crucial role in the interaction between the parasite and host. Although malarial hepatitis is a common clinical complication of severe malaria, other malaria-related liver changes have been overlooked during the blood stage of the parasite life-cycle, in contrast to the many studies that have focused on parasite invasion of and replication in the liver during the hepatic stage of the parasite. METHODS A rodent model of malaria was established using Plasmodium yoelii strain 17XL, a lethal strain of rodent malaria, for liver transcriptomic profiling. RESULTS Differentially expressed messenger RNAs were associated with innate and adaptive immune responses, while differentially expressed long noncoding RNAs were enriched in the regulation of metabolism-related pathways, such as lipid metabolism. The coexpression network showed that host genes were related to cellular transport and tissue remodeling. Hub gene analysis of P. yoelii indicated that ubiquitination genes that were coexpressed with the host were evolutionarily conserved. CONCLUSIONS Our analysis yielded evidence of activated immune responses, aberrant metabolic processes and tissue remodeling changes in the livers of mice with malaria during the blood stage of the parasite, which provided a systematic outline of liver responses during Plasmodium infection.
Collapse
Affiliation(s)
- Xueyan Hu
- grid.11135.370000 0001 2256 9319Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China
| | - Jie Zhao
- grid.11135.370000 0001 2256 9319Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China
| | - Junhui Zhao
- grid.11135.370000 0001 2256 9319Department of Physiology and Pathophysiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China
| | - Ence Yang
- grid.11135.370000 0001 2256 9319Department of Medical Bioinformatics, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China ,grid.11135.370000 0001 2256 9319Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China
| | - Mozhi Jia
- grid.11135.370000 0001 2256 9319Department of Microbiology & Infectious Disease Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191 China
| |
Collapse
|
14
|
Olupot-Olupot P, Okiror W, Mnjalla H, Muhindo R, Uyoga S, Mpoya A, Williams TN, terHeine R, Burger DM, Urban B, Connon R, George EC, Gibb DM, Walker AS, Maitland K. Pharmacokinetics and pharmacodynamics of azithromycin in severe malaria bacterial co-infection in African children (TABS-PKPD): a protocol for a Phase II randomised controlled trial. Wellcome Open Res 2023; 6:161. [PMID: 37519413 PMCID: PMC10382785 DOI: 10.12688/wellcomeopenres.16968.2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/19/2022] [Indexed: 08/01/2023] Open
Abstract
Background: African children with severe malaria are susceptible to Gram-negative bacterial co-infection, largely non-typhoidal Salmonellae, leading to a substantially higher rates of in-hospital and post-discharge mortality than those without bacteraemia. Current evidence for treating co-infection is lacking, and there is no consensus on the dosage or length of treatment required. We therefore aimed to establish the appropriate dose of oral dispersible azithromycin as an antimicrobial treatment for children with severe malaria and to investigate whether antibiotics can be targeted to those at greatest risk of bacterial co-infection using clinical criteria alone or in combination with rapid diagnostic biomarker tests. Methods: A Phase I/II open-label trial comparing three doses of azithromycin: 10, 15 and 20 mg/kg spanning the lowest to highest mg/kg doses previously demonstrated to be equally effective as parenteral treatment for other salmonellae infection. Children with the highest risk of bacterial infection will receive five days of azithromycin and followed for 90 days. We will generate relevant pharmacokinetic data by sparse sampling during dosing intervals. We will use population pharmacokinetic modelling to determine the optimal azithromycin dose in severe malaria and investigate azithromycin exposure to change in C-reactive protein, a putative marker of sepsis at 72 hours, and microbiological cure (seven-day), alone and as a composite with seven-day survival. We will also evaluate whether a combination of clinical, point-of-care diagnostic tests, and/or biomarkers can accurately identify the sub-group of severe malaria with culture-proven bacteraemia by comparison with a control cohort of children hospitalized with severe malaria at low risk of bacterial co-infection. Discussion: We plan to study azithromycin because of its favourable microbiological spectrum, its inherent antimalarial and immunomodulatory properties and dosing and safety profile. This study will generate new data to inform the design and sample size for definitive Phase III trial evaluation. Registration: ISRCTN49726849 (27 th October 2017).
Collapse
Affiliation(s)
- Peter Olupot-Olupot
- Mbale Clinical Research Institute, Pallisa Road, PO Box 291, Mbale, Uganda
- Busitema University Faculty of Health Sciences, Mbale Regional Referral Hospital, Mbale, Uganda
| | - William Okiror
- Mbale Clinical Research Institute, Pallisa Road, PO Box 291, Mbale, Uganda
- Busitema University Faculty of Health Sciences, Mbale Regional Referral Hospital, Mbale, Uganda
| | - Hellen Mnjalla
- KEMRI Wellcome Trust Research Programme, PO Box 230, Kilifi, Kenya
| | - Rita Muhindo
- Mbale Clinical Research Institute, Pallisa Road, PO Box 291, Mbale, Uganda
- Busitema University Faculty of Health Sciences, Mbale Regional Referral Hospital, Mbale, Uganda
| | - Sophie Uyoga
- KEMRI Wellcome Trust Research Programme, PO Box 230, Kilifi, Kenya
| | - Ayub Mpoya
- KEMRI Wellcome Trust Research Programme, PO Box 230, Kilifi, Kenya
| | - Thomas N Williams
- KEMRI Wellcome Trust Research Programme, PO Box 230, Kilifi, Kenya
- Department of Infectious Disease and Institute of Global Health and Innovation, Division of Medicine, Imperial College, London, UK
| | - Rob terHeine
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - David M Burger
- Department of Pharmacy, Radboud University Medical Center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | - Britta Urban
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool, L3 5QA, UK
| | - Roisin Connon
- MRC Clinical Trials Unit, University College London, Aviation House, 125 Kingsway, London, WC28 6NH, UK
| | - Elizabeth C George
- MRC Clinical Trials Unit, University College London, Aviation House, 125 Kingsway, London, WC28 6NH, UK
| | - Diana M Gibb
- MRC Clinical Trials Unit, University College London, Aviation House, 125 Kingsway, London, WC28 6NH, UK
| | - A Sarah Walker
- MRC Clinical Trials Unit, University College London, Aviation House, 125 Kingsway, London, WC28 6NH, UK
| | - Kathryn Maitland
- KEMRI Wellcome Trust Research Programme, PO Box 230, Kilifi, Kenya
- Department of Infectious Disease and Institute of Global Health and Innovation, Division of Medicine, Imperial College, London, UK
| |
Collapse
|
15
|
Tebben K, Yirampo S, Coulibaly D, Koné AK, Laurens MB, Stucke EM, Dembélé A, Tolo Y, Traoré K, Niangaly A, Berry AA, Kouriba B, Plowe CV, Doumbo OK, Lyke KE, Takala-Harrison S, Thera MA, Travassos MA, Serre D. Malian children infected with Plasmodium ovale and Plasmodium falciparum display very similar gene expression profiles. PLoS Negl Trop Dis 2023; 17:e0010802. [PMID: 36696438 PMCID: PMC9901758 DOI: 10.1371/journal.pntd.0010802] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 02/06/2023] [Accepted: 01/16/2023] [Indexed: 01/26/2023] Open
Abstract
Plasmodium parasites caused 241 million cases of malaria and over 600,000 deaths in 2020. Both P. falciparum and P. ovale are endemic to Mali and cause clinical malaria, with P. falciparum infections typically being more severe. Here, we sequenced RNA from nine pediatric blood samples collected during infections with either P. falciparum or P. ovale, and characterized the host and parasite gene expression profiles. We found that human gene expression varies more between individuals than according to the parasite species causing the infection, while parasite gene expression profiles cluster by species. Additionally, we characterized DNA polymorphisms of the parasites directly from the RNA-seq reads and found comparable levels of genetic diversity in both species, despite dramatic differences in prevalence. Our results provide unique insights into host-pathogen interactions during malaria infections and their variations according to the infecting Plasmodium species, which will be critical to develop better elimination strategies against all human Plasmodium parasites.
Collapse
Affiliation(s)
- Kieran Tebben
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore Maryland, United States of America
| | - Salif Yirampo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Drissa Coulibaly
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Abdoulaye K. Koné
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Matthew B. Laurens
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Emily M. Stucke
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Ahmadou Dembélé
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Youssouf Tolo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Karim Traoré
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Amadou Niangaly
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Andrea A. Berry
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Bourema Kouriba
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Christopher V. Plowe
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Ogobara K. Doumbo
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Kirsten E. Lyke
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Shannon Takala-Harrison
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - Mahamadou A. Thera
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Mark A. Travassos
- Malaria Research Program, Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| | - David Serre
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore Maryland, United States of America
| |
Collapse
|
16
|
Zhang YH, Su XZ, Li J, Shi JJ, Xie LH. Multicohort transcriptome analysis of whole blood identifies robust human response signatures in Plasmodium falciparum infections. Malar J 2022; 21:333. [PMID: 36380373 PMCID: PMC9664782 DOI: 10.1186/s12936-022-04374-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 11/08/2022] [Indexed: 11/17/2022] Open
Abstract
Background To understand how Plasmodium falciparum malaria is controlled, it is essential to elucidate the transcriptomic responses of the human host in naturally-exposed populations. Various individual studies of the human transcriptomic responses to naturally transmitted P. falciparum infections have been reported with varying results. Multicohort gene expression analysis by aggregating data from diverse populations into a single analysis will increase the reproducibility and reliability of the results. Methods In this study, discovery cohorts GSE1124-GPL96, GSE34404, GSE117613, and validation cohort GSE35858 were obtained from the Gene Expression Omnibus. A meta-analysis using data from the multicohort studies was performed to identify the differentially expressed genes (DEGs) between malaria-infected and noninfected individuals using the MetaIntegrator R package. Subsequently, the protein–protein interaction (PPI) networks of the DEGs were constructed using Cytoscape software. Significant modules were selected, and the hub genes were identified using the CytoHubba and MCODE plug-ins. Multicohort WGCNA was conducted to find a correlation between modules and malaria infection. Furthermore, the immune cell profile of the peripheral blood in different groups was identified using ssGSEA. Results These analyses reveal that neutrophil activation, neutrophil-mediated immunity, and neutrophil degranulation are involved in the human response to natural malaria infection. However, neutrophil cell enrichment and activation were not significantly different between mild malaria and severe malaria groups. Malaria infection also downregulates host genes in ribosome synthesis and protein translation and upregulates host cell division-related genes. Furthermore, immune cell profiling analysis shows that activated dendritic cells and type 2 T helper cells are upregulated, while activated B cells, immature B cells, and monocytes are downregulated in the malaria-infected patients relative to the noninfected individuals. Significantly higher enrichment of activated dendritic cell-related genes and significantly lower enrichment of monocyte-related genes are also observed in the peripheral blood of the severe malaria group than in the mild malaria group. Conclusion These results reveal important molecular signatures of host responses to malaria infections, providing some bases for developing malaria control strategies and protective vaccines. Supplementary Information The online version contains supplementary material available at 10.1186/s12936-022-04374-5.
Collapse
Affiliation(s)
- Yan-hui Zhang
- grid.419897.a0000 0004 0369 313XKey Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Xin-zhuan Su
- grid.94365.3d0000 0001 2297 5165Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD USA
| | - Jian Li
- grid.12955.3a0000 0001 2264 7233State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian China
| | - Jia-jian Shi
- grid.419897.a0000 0004 0369 313XKey Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| | - Li-hua Xie
- grid.419897.a0000 0004 0369 313XKey Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, Fuzhou, China
| |
Collapse
|
17
|
Dey S, Kaur H, Mazumder M, Brodsky E. Analysis of gene expression profiles to study malaria vaccine dose efficacy and immune response modulation. Genomics Inform 2022; 20:e32. [PMID: 36239109 PMCID: PMC9576474 DOI: 10.5808/gi.22049] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2002] [Accepted: 09/04/2022] [Indexed: 11/20/2022] Open
Abstract
Malaria is a life-threatening disease, and Africa is still one of the most affected endemic regions despite years of policy to limit infection and transmission rates. Further, studies into the variable efficacy of the vaccine are needed to provide a better understanding of protective immunity. Thus, the current study is designed to delineate the effect of each dose of vaccine on the transcriptional profiles of subjects to determine its efficacy and understand the molecular mechanisms underlying the protection this vaccine provides. Here, we used gene expression profiles of pre and post-vaccination patients after various doses of RTS,S based on samples collected from the Gene Expression Omnibus datasets. Subsequently, differential gene expression analysis using edgeR revealed the significantly (false discovery rate < 0.005) 158 downregulated and 61 upregulated genes between control vs. controlled human malaria infection samples. Further, enrichment analysis of significant genes delineated the involvement of CCL8, CXCL10, CXCL11, XCR1, CSF3, IFNB1, IFNE, IL12B, IL22, IL6, IL27, etc., genes which found to be upregulated after earlier doses but downregulated after the 3rd dose in cytokine-chemokine pathways. Notably, we identified 13 cytokine genes whose expression significantly varied during three doses. Eventually, these findings give insight into the dual role of cytokine responses in malaria pathogenesis. The variations in their expression patterns after various doses of vaccination are linked to the protection as it decreases the severe inflammatory effects in malaria patients. This study will be helpful in designing a better vaccine against malaria and understanding the functions of cytokine response as well.
Collapse
Affiliation(s)
- Supantha Dey
- Department of Genetic Engineering and Biotechnology, University of Dhaka, Dhaka 1000, Bangladesh
- Pine Biotech, New Orleans, LA 70112, USA
- Corresponding author: ,
| | | | | | | |
Collapse
|
18
|
The Variation of Transcriptomic Perturbations is Associated with the Development and Progression of Various Diseases. DISEASE MARKERS 2022; 2022:2148627. [PMID: 36204511 PMCID: PMC9530920 DOI: 10.1155/2022/2148627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Revised: 08/31/2022] [Accepted: 09/07/2022] [Indexed: 11/28/2022]
Abstract
Background Although transcriptomic data have been widely applied to explore various diseases, few studies have investigated the association between transcriptomic perturbations and disease development in a wide variety of diseases. Methods Based on a previously developed algorithm for quantifying intratumor heterogeneity at the transcriptomic level, we defined the variation of transcriptomic perturbations (VTP) of a disease relative to the health status. Based on publicly available transcriptome datasets, we compared VTP values between the disease and health status and analyzed correlations between VTP values and disease progression or severity in various diseases, including neurological disorders, infectious diseases, cardiovascular diseases, respiratory diseases, liver diseases, kidney diseases, digestive diseases, and endocrine diseases. We also identified the genes and pathways whose expression perturbations correlated positively with VTP across diverse diseases. Results VTP values were upregulated in various diseases relative to their normal controls. VTP values were significantly greater in define than in possible or probable Alzheimer's disease. VTP values were significantly larger in intensive care unit (ICU) COVID-19 patients than in non-ICU patients, and in COVID-19 patients requiring mechanical ventilatory support (MVS) than in those not requiring MVS. VTP correlated positively with viral loads in acquired immune deficiency syndrome (AIDS) patients. Moreover, the AIDS patients treated with abacavir or zidovudine had lower VTP values than those without such therapies. In pulmonary tuberculosis (TB) patients, VTP values followed the pattern: active TB > latent TB > normal controls. VTP values were greater in clinically apparent than in presymptomatic malaria. VTP correlated negatively with the cardiac index of left ventricular ejection fraction (LVEF). In chronic obstructive pulmonary disease (COPD), VTP showed a negative correlation with forced expiratory volume in the first second (FEV1). VTP values increased with H. pylori infection and were upregulated in atrophic gastritis caused by H. pylori infection. The genes and pathways whose expression perturbations correlated positively with VTP scores across diseases were mainly involved in the regulation of immune, metabolic, and cellular activities. Conclusions VTP is upregulated in the disease versus health status, and its upregulation is associated with disease progression and severity in various diseases. Thus, VTP has potential clinical implications for disease diagnosis and prognosis.
Collapse
|
19
|
Asiedu E, Larbi A, Adankwah E, Yambah JK, Sakyi SA, Kwarteng EVS, Obiri-Yeboah D, Kwarteng A. Transcriptomic profiling identifies host-derived biomarker panels for assessing cerebral malaria. GENE REPORTS 2022. [DOI: 10.1016/j.genrep.2022.101650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
20
|
An update on cerebral malaria for therapeutic intervention. Mol Biol Rep 2022; 49:10579-10591. [PMID: 35670928 DOI: 10.1007/s11033-022-07625-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Accepted: 05/20/2022] [Indexed: 10/18/2022]
Abstract
BACKGROUND Cerebral malaria is often pronounced as a major life-threatening neurological complication of Plasmodium falciparum infection. The complex pathogenic landscape of the parasite and the associated neurological complications are still not elucidated properly. The growing concerns of drugresistant parasite strains along with the failure of anti-malarial drugs to subdue post-recovery neuro-cognitive dysfunctions in cerebral malaria patients have called for a demand to explore novel biomarkers and therapeutic avenues. Due course of the brain infection journey of the parasite, events such as sequestration of infected RBCs, cytoadherence, inflammation, endothelial activation, and blood-brain barrier disruption are considered critical. METHODS In this review, we briefly summarize the diverse pathogenesis of the brain-invading parasite associated with loss of the blood-brain barrier integrity. In addition, we also discuss proteomics, transcriptomics, and bioinformatics strategies to identify an array of new biomarkers and drug candidates. CONCLUSION A proper understanding of the parasite biology and mechanism of barrier disruption coupled with emerging state-of-art therapeutic approaches could be helpful to tackle cerebral malaria.
Collapse
|
21
|
Sánchez-Arcila JC, Jensen KDC. Forward Genetics in Apicomplexa Biology: The Host Side of the Story. Front Cell Infect Microbiol 2022; 12:878475. [PMID: 35646724 PMCID: PMC9133346 DOI: 10.3389/fcimb.2022.878475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
Forward genetic approaches have been widely used in parasitology and have proven their power to reveal the complexities of host-parasite interactions in an unbiased fashion. Many aspects of the parasite's biology, including the identification of virulence factors, replication determinants, antibiotic resistance genes, and other factors required for parasitic life, have been discovered using such strategies. Forward genetic approaches have also been employed to understand host resistance mechanisms to parasitic infection. Here, we will introduce and review all forward genetic approaches that have been used to identify host factors involved with Apicomplexa infections, which include classical genetic screens and QTL mapping, GWAS, ENU mutagenesis, overexpression, RNAi and CRISPR-Cas9 library screens. Collectively, these screens have improved our understanding of host resistance mechanisms, immune regulation, vaccine and drug designs for Apicomplexa parasites. We will also discuss how recent advances in molecular genetics give present opportunities to further explore host-parasite relationships.
Collapse
Affiliation(s)
- Juan C. Sánchez-Arcila
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
| | - Kirk D. C. Jensen
- Department of Molecular and Cell Biology, University of California Merced, Merced, CA, United States
- Health Science Research Institute, University of California, Merced, Merced, CA, United States
| |
Collapse
|
22
|
Studniberg SI, Ioannidis LJ, Utami RAS, Trianty L, Liao Y, Abeysekera W, Li‐Wai‐Suen CSN, Pietrzak HM, Healer J, Puspitasari AM, Apriyanti D, Coutrier F, Poespoprodjo JR, Kenangalem E, Andries B, Prayoga P, Sariyanti N, Smyth GK, Cowman AF, Price RN, Noviyanti R, Shi W, Garnham AL, Hansen DS. Molecular profiling reveals features of clinical immunity and immunosuppression in asymptomatic P. falciparum malaria. Mol Syst Biol 2022; 18:e10824. [PMID: 35475529 PMCID: PMC9045086 DOI: 10.15252/msb.202110824] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 03/30/2022] [Accepted: 03/31/2022] [Indexed: 01/12/2023] Open
Abstract
Clinical immunity to P. falciparum malaria is non-sterilizing, with adults often experiencing asymptomatic infection. Historically, asymptomatic malaria has been viewed as beneficial and required to help maintain clinical immunity. Emerging views suggest that these infections are detrimental and constitute a parasite reservoir that perpetuates transmission. To define the impact of asymptomatic malaria, we pursued a systems approach integrating antibody responses, mass cytometry, and transcriptional profiling of individuals experiencing symptomatic and asymptomatic P. falciparum infection. Defined populations of classical and atypical memory B cells and a TH2 cell bias were associated with reduced risk of clinical malaria. Despite these protective responses, asymptomatic malaria featured an immunosuppressive transcriptional signature with upregulation of pathways involved in the inhibition of T-cell function, and CTLA-4 as a predicted regulator in these processes. As proof of concept, we demonstrated a role for CTLA-4 in the development of asymptomatic parasitemia in infection models. The results suggest that asymptomatic malaria is not innocuous and might not support the induction of immune processes to fully control parasitemia or efficiently respond to malaria vaccines.
Collapse
Affiliation(s)
- Stephanie I Studniberg
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| | - Lisa J Ioannidis
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| | - Retno A S Utami
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia,Eijkman Institute for Molecular BiologyJakartaIndonesia
| | - Leily Trianty
- Eijkman Institute for Molecular BiologyJakartaIndonesia
| | - Yang Liao
- Olivia Newton‐John Cancer Research InstituteHeidelbergVic.Australia
| | - Waruni Abeysekera
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,School of Mathematics and StatisticsThe University of MelbourneParkvilleVic.Australia
| | - Connie S N Li‐Wai‐Suen
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,School of Mathematics and StatisticsThe University of MelbourneParkvilleVic.Australia
| | - Halina M Pietrzak
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| | - Julie Healer
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| | | | - Dwi Apriyanti
- Eijkman Institute for Molecular BiologyJakartaIndonesia
| | | | | | | | | | - Pak Prayoga
- Papuan Health and Community FoundationPapuaIndonesia
| | | | - Gordon K Smyth
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,School of Mathematics and StatisticsThe University of MelbourneParkvilleVic.Australia
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| | - Ric N Price
- Global and Tropical Health DivisionMenzies School of Health Research and Charles Darwin UniversityDarwinNTAustralia,Centre for Tropical Medicine and Global HealthNuffield Department of MedicineUniversity of OxfordOxfordUK,Mahidol‐Oxford Tropical Medicine Research UnitMahidol UniversityBangkokThailand
| | | | - Wei Shi
- Olivia Newton‐John Cancer Research InstituteHeidelbergVic.Australia
| | - Alexandra L Garnham
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,School of Mathematics and StatisticsThe University of MelbourneParkvilleVic.Australia
| | - Diana S Hansen
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleVic.Australia,Department of Medical BiologyThe University of MelbourneParkvilleVic.Australia
| |
Collapse
|
23
|
Parreira KS, Scarpelli P, Rezende Lima W, Garcia RS. Contribution of Transcriptome to Elucidate the Biology of Plasmodium spp. Curr Top Med Chem 2022; 22:169-187. [PMID: 35021974 DOI: 10.2174/1568026622666220111140803] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 12/22/2021] [Accepted: 12/26/2021] [Indexed: 11/22/2022]
Abstract
In the present review, we discuss some of the new technologies that have been applied to elucidate how Plasmodium spp escape from the immune system and subvert the host physiology to orchestrate the regulation of its biological pathways. Our manuscript describes how techniques such as microarray approaches, RNA-Seq and single-cell RNA sequencing have contributed to the discovery of transcripts and changed the concept of gene expression regulation in closely related malaria parasite species. Moreover, the text highlights the contributions of high-throughput RNA sequencing for the current knowledge of malaria parasite biology, physiology, vaccine target and the revelation of new players in parasite signaling.
Collapse
Affiliation(s)
| | - Pedro Scarpelli
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo - USP, São Paulo, Brazil
| | - Wânia Rezende Lima
- Departamento de Medicina, Instituto de Biotecnologia-Universidade Federal de Catalão
| | - R S Garcia
- Departamento de Análises Clínicas e Toxicológicas, Faculdade de Ciências Farmacêuticas, Universidade de São Paulo - USP, São Paulo, Brazil
| |
Collapse
|
24
|
Georgiadou A, Dunican C, Soro-Barrio P, Lee HJ, Kaforou M, Cunnington AJ. Comparative transcriptomic analysis reveals translationally relevant processes in mouse models of malaria. eLife 2022; 11:e70763. [PMID: 35006075 PMCID: PMC8747512 DOI: 10.7554/elife.70763] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 12/22/2021] [Indexed: 02/07/2023] Open
Abstract
Recent initiatives to improve translation of findings from animal models to human disease have focussed on reproducibility but quantifying the relevance of animal models remains a challenge. Here, we use comparative transcriptomics of blood to evaluate the systemic host response and its concordance between humans with different clinical manifestations of malaria and five commonly used mouse models. Plasmodium yoelii 17XL infection of mice most closely reproduces the profile of gene expression changes seen in the major human severe malaria syndromes, accompanied by high parasite biomass, severe anemia, hyperlactatemia, and cerebral microvascular pathology. However, there is also considerable discordance of changes in gene expression between the different host species and across all models, indicating that the relevance of biological mechanisms of interest in each model should be assessed before conducting experiments. These data will aid the selection of appropriate models for translational malaria research, and the approach is generalizable to other disease models.
Collapse
Affiliation(s)
- Athina Georgiadou
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College LondonLondonUnited Kingdom
- Centre for Paediatrics and Child Health, Imperial College LondonLondonUnited Kingdom
| | - Claire Dunican
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College LondonLondonUnited Kingdom
- Centre for Paediatrics and Child Health, Imperial College LondonLondonUnited Kingdom
| | - Pablo Soro-Barrio
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College LondonLondonUnited Kingdom
| | - Hyun Jae Lee
- Institute for Molecular Bioscience, University of QueenslandBrisbaneAustralia
| | - Myrsini Kaforou
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College LondonLondonUnited Kingdom
- Centre for Paediatrics and Child Health, Imperial College LondonLondonUnited Kingdom
| | - Aubrey J Cunnington
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College LondonLondonUnited Kingdom
- Centre for Paediatrics and Child Health, Imperial College LondonLondonUnited Kingdom
| |
Collapse
|
25
|
Omar M, Marchionni L, Häcker G, Badr MT. Host Blood Gene Signatures Can Detect the Progression to Severe and Cerebral Malaria. Front Cell Infect Microbiol 2021; 11:743616. [PMID: 34746025 PMCID: PMC8569259 DOI: 10.3389/fcimb.2021.743616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/23/2021] [Indexed: 11/16/2022] Open
Abstract
Malaria is a major international public health problem that affects millions of patients worldwide especially in sub-Saharan Africa. Although many tests have been developed to diagnose malaria infections, we still lack reliable diagnostic biomarkers for the identification of disease severity, especially in endemic areas where the diagnosis of cerebral malaria is very difficult and requires the exclusion of all other possible causes. Previous host and pathogen transcriptomic studies have not yielded homogenous results that can be harnessed into a reliable diagnostic tool. Here we utilized a multi-cohort analysis approach using machine-learning algorithms to identify blood gene signatures that can distinguish severe and cerebral malaria from moderate and non-cerebral cases. Using a Regularized Random Forest model, we identified 28-gene and 32-gene signatures that can reliably distinguish severe and cerebral malaria, respectively. We tested the specificity of both signatures against other common infectious diseases to ensure the signatures reliability and suitability as diagnostic markers. The severe and cerebral malaria gene-signatures were further integrated through k-top scoring pairs classifiers into ten and nine gene pairs that could distinguish severe and cerebral malaria, respectively. These signatures have various implications that can be utilized as blood diagnostic tools for malaria severity in endemic countries.
Collapse
Affiliation(s)
- Mohamed Omar
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Luigi Marchionni
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, United States
| | - Georg Häcker
- Institute of Medical Microbiology and Hygiene, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany.,BIOSS Centre for Biological Signaling Studies, University of Freiburg, Freiburg, Germany
| | - Mohamed Tarek Badr
- Institute of Medical Microbiology and Hygiene, Medical Center - University of Freiburg, Faculty of Medicine, Freiburg, Germany.,IMM-PACT-Program, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| |
Collapse
|
26
|
Makinde FL, Tchamga MSS, Jafali J, Fatumo S, Chimusa ER, Mulder N, Mazandu GK. Reviewing and assessing existing meta-analysis models and tools. Brief Bioinform 2021; 22:bbab324. [PMID: 34415019 PMCID: PMC8575034 DOI: 10.1093/bib/bbab324] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Revised: 07/07/2021] [Accepted: 07/25/2021] [Indexed: 01/03/2023] Open
Abstract
Over the past few years, meta-analysis has become popular among biomedical researchers for detecting biomarkers across multiple cohort studies with increased predictive power. Combining datasets from different sources increases sample size, thus overcoming the issue related to limited sample size from each individual study and boosting the predictive power. This leads to an increased likelihood of more accurately predicting differentially expressed genes/proteins or significant biomarkers underlying the biological condition of interest. Currently, several meta-analysis methods and tools exist, each having its own strengths and limitations. In this paper, we survey existing meta-analysis methods, and assess the performance of different methods based on results from different datasets as well as assessment from prior knowledge of each method. This provides a reference summary of meta-analysis models and tools, which helps to guide end-users on the choice of appropriate models or tools for given types of datasets and enables developers to consider current advances when planning the development of new meta-analysis models and more practical integrative tools.
Collapse
Affiliation(s)
- Funmilayo L Makinde
- Computational Biology Division at University of Cape Town in collaboration with the African Institute for Mathematical Sciences (AIMS), South Africa
| | - Milaine S S Tchamga
- Division of Human Genetics at University of Cape in collaboration with the African Institute for Mathematical Sciences (AIMS), South Africa
| | - James Jafali
- Pathogen Biology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Malawi
| | - Segun Fatumo
- London School of Hygiene and Tropical Medicine, University of London, UK
| | - Emile R Chimusa
- Division of Human Genetics, Department of Pathology, University of Cape Town, South Africa
| | - Nicola Mulder
- Computational Biology Division at University of Cape Town, South Africa
| | - Gaston K Mazandu
- Division of Human Genetics, Department of Pathology at University of Cape Town, and Associate Researcher at the African Institute for Mathematical Sciences (AIMS), South Africa
| |
Collapse
|
27
|
Karikari AA, Wruck W, Adjaye J. Transcriptome-based analysis of blood samples reveals elevation of DNA damage response, neutrophil degranulation, cancer and neurodegenerative pathways in Plasmodium falciparum patients. Malar J 2021; 20:383. [PMID: 34565410 PMCID: PMC8474955 DOI: 10.1186/s12936-021-03918-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 09/14/2021] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Malaria caused by Plasmodium falciparum results in severe complications including cerebral malaria (CM) especially in children. While the majority of falciparum malaria survivors make a full recovery, there are reports of some patients ending up with neurological sequelae or cognitive deficit. METHODS An analysis of pooled transcriptome data of whole blood samples derived from two studies involving various P. falciparum infections, comprising mild malaria (MM), non-cerebral severe malaria (NCM) and CM was performed. Pathways and gene ontologies (GOs) elevated in the distinct P. falciparum infections were determined. RESULTS In all, 2876 genes were expressed in common between the 3 forms of falciparum malaria, with CM having the least number of expressed genes. In contrast to other research findings, the analysis from this study showed MM share similar biological processes with cancer and neurodegenerative diseases, NCM is associated with drug resistance and glutathione metabolism and CM is correlated with endocannabinoid signalling and non-alcoholic fatty liver disease (NAFLD). GO revealed the terms biogenesis, DNA damage response and IL-10 production in MM, down-regulation of cytoskeletal organization and amyloid-beta clearance in NCM and aberrant signalling, neutrophil degranulation and gene repression in CM. Differential gene expression analysis between CM and NCM showed the up-regulation of neutrophil activation and response to herbicides, while regulation of axon diameter was down-regulated in CM. CONCLUSIONS Results from this study reveal that P. falciparum-mediated inflammatory and cellular stress mechanisms may impair brain function in MM, NCM and CM. However, the neurological deficits predominantly reported in CM cases could be attributed to the down-regulation of various genes involved in cellular function through transcriptional repression, axonal dysfunction, dysregulation of signalling pathways and neurodegeneration. It is anticipated that the data from this study, might form the basis for future hypothesis-driven malaria research.
Collapse
Affiliation(s)
- Akua A. Karikari
- Department of Biomedical Sciences, College of Health and Allied Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Wasco Wruck
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany
| | - James Adjaye
- Institute for Stem Cell Research and Regenerative Medicine, Medical Faculty, Heinrich-Heine University, 40225 Düsseldorf, Germany
| |
Collapse
|
28
|
Olupot-Olupot P, Okiror W, Mnjalla H, Muhindo R, Uyoga S, Mpoya A, Williams TN, terHeine R, Burger DM, Urban B, Connon R, George EC, Gibb DM, Walker AS, Maitland K. Pharmacokinetics and pharmacodynamics of azithromycin in severe malaria bacterial co-infection in African children (TABS-PKPD): a protocol for a Phase II randomised controlled trial. Wellcome Open Res 2021. [DOI: 10.12688/wellcomeopenres.16968.1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: African children with severe malaria are susceptible to Gram-negative bacterial co-infection, largely non-typhoidal Salmonellae, leading to a substantially higher rates of in-hospital and post-discharge mortality than those without bacteraemia. Current evidence for treating co-infection is lacking, and there is no consensus on the dosage or length of treatment required. We therefore aimed to establish the appropriate dose of oral dispersible azithromycin as an antimicrobial treatment for children with severe malaria and to investigate whether antibiotics can be targeted to those at greatest risk of bacterial co-infection using clinical criteria alone or in combination with rapid diagnostic biomarker tests. Methods: A Phase I/II open-label trial comparing three doses of azithromycin: 10, 15 and 20 mg/kg spanning the lowest to highest mg/kg doses previously demonstrated to be equally effective as parenteral treatment for other salmonellae infection. Children with the highest risk of bacterial infection will receive five days of azithromycin and followed for 90 days. We will generate relevant pharmacokinetic data by sparse sampling during dosing intervals. We will use population pharmacokinetic modelling to determine the optimal azithromycin dose in severe malaria and investigate azithromycin exposure to change in C-reactive protein, a putative marker of sepsis at 72 hours, and microbiological cure (seven-day), alone and as a composite with seven-day survival. We will also evaluate whether a combination of clinical, point-of-care diagnostic tests, and/or biomarkers can accurately identify the sub-group of severe malaria with culture-proven bacteraemia by comparison with a control cohort of children hospitalized with severe malaria at low risk of bacterial co-infection. Discussion: We plan to study azithromycin because of its favourable microbiological spectrum, its inherent antimalarial and immunomodulatory properties and dosing and safety profile. This study will generate new data to inform the design and sample size for definitive Phase III trial evaluation. Registration: ISRCTN49726849 (27th October 2017).
Collapse
|
29
|
Li K, Wang H, Zhang HF, Zhao XX, Lai YJ, Liu FF. Genomic analysis of host gene responses to cerebral Plasmodium falciparum malaria. IMMUNITY INFLAMMATION AND DISEASE 2021; 9:819-826. [PMID: 33942992 PMCID: PMC8342194 DOI: 10.1002/iid3.436] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Revised: 03/16/2021] [Accepted: 03/31/2021] [Indexed: 11/09/2022]
Abstract
Introduction A vaccine for malaria is urgently required but no vaccine has yet shown satisfactory protective efficacy especially for Plasmodium falciparum. P. falciparum infection can progress to cerebral malaria (CM), a neurological syndrome with exceedingly high mortality. Designing effective P. falciparum vaccines require more understanding of the protective immune response while the host immune response to CM and the mechanisms are still elusive. Here, we aim to identify host gene responses to CM and host gene networks associated with CM pathogenesis. Methods An innovative genomic analysis strategy, the weighted gene coexpression network analysis (WGCNA) combined with differential gene expression analysis, was used in this study. Data for analysis contain 93 whole blood samples, derived from two previous public transcriptome datasets. Results This approach led to the identification of numerous differentially expressed human transcripts and dozens of coexpression gene modules. We further identified nine key genes, including MBP, SAMSN1, PSMF1, SLC39A8, EIF3B, SMPDL3A, FABP5, SPSB3, and SHARPIN, of which the last four genes were first identified to be related to CM in the present study. Conclusion The results provided a comprehensive characterization of host gene expression profiles in CM and offered some new insight into malaria vaccine design. These identified key genes could be potential targets or immune modulators for novel therapeutic interventions of CM.
Collapse
Affiliation(s)
- Ke Li
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Wang
- Department of Pharmacy, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Hong-Feng Zhang
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiao-Xiao Zhao
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yong-Ji Lai
- Department of Pharmacy, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fang-Fang Liu
- Department of Pathology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
30
|
Romero-Cordero S, Kirwan R, Noguera-Julian A, Cardellach F, Fortuny C, Morén C. A Mitocentric View of the Main Bacterial and Parasitic Infectious Diseases in the Pediatric Population. Int J Mol Sci 2021; 22:3272. [PMID: 33806981 PMCID: PMC8004694 DOI: 10.3390/ijms22063272] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 03/13/2021] [Accepted: 03/16/2021] [Indexed: 01/04/2023] Open
Abstract
Infectious diseases occur worldwide with great frequency in both adults and children. Both infections and their treatments trigger mitochondrial interactions at multiple levels: (i) incorporation of damaged or mutated proteins to the complexes of the electron transport chain, (ii) mitochondrial genome (depletion, deletions, and point mutations) and mitochondrial dynamics (fusion and fission), (iii) membrane potential, (iv) apoptotic regulation, (v) generation of reactive oxygen species, among others. Such alterations may result in serious adverse clinical events with great impact on children's quality of life, even resulting in death. As such, bacterial agents are frequently associated with loss of mitochondrial membrane potential and cytochrome c release, ultimately leading to mitochondrial apoptosis by activation of caspases-3 and -9. Using Rayyan QCRI software for systematic reviews, we explore the association between mitochondrial alterations and pediatric infections including (i) bacterial: M. tuberculosis, E. cloacae, P. mirabilis, E. coli, S. enterica, S. aureus, S. pneumoniae, N. meningitidis and (ii) parasitic: P. falciparum. We analyze how these pediatric infections and their treatments may lead to mitochondrial deterioration in this especially vulnerable population, with the intention of improving both the understanding of these diseases and their management in clinical practice.
Collapse
Affiliation(s)
- Sonia Romero-Cordero
- Faculty of Medicine, Pompeu Fabra University and Universitat Autònoma de Barcelona, 08002 Barcelona, Spain;
| | - Richard Kirwan
- School of Biological and Environmental Sciences, Liverpool John Moores University, Liverpool L2 2QP, UK
| | - Antoni Noguera-Julian
- Malalties Infeccioses i Resposta Inflamatòria Sistèmica en Pediatria, Unitat d’Infeccions, Servei de Pediatria, Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (A.N.-J.); (C.F.)
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Red de Investigación Translacional en Infectología Pediátrica (RITIP), 28029 Madrid, Spain
| | - Francesc Cardellach
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain;
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (ISCIII), 28029 Madrid, Spain
- Internal Medicine Department-Hospital Clínic of Barcelona (HCB), 08036 Barcelona, Spain
| | - Clàudia Fortuny
- Malalties Infeccioses i Resposta Inflamatòria Sistèmica en Pediatria, Unitat d’Infeccions, Servei de Pediatria, Institut de Recerca Pediàtrica Hospital Sant Joan de Déu, 08950 Barcelona, Spain; (A.N.-J.); (C.F.)
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain;
- Centro de Investigación Biomédica en Red de Epidemiología y Salud Pública (CIBERESP), 28029 Madrid, Spain
- Red de Investigación Translacional en Infectología Pediátrica (RITIP), 28029 Madrid, Spain
| | - Constanza Morén
- Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain;
- Muscle Research and Mitochondrial Function Laboratory, Cellex-IDIBAPS, Faculty of Medicine and Health Sciences, University of Barcelona, 08036 Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) (ISCIII), 28029 Madrid, Spain
- Internal Medicine Department-Hospital Clínic of Barcelona (HCB), 08036 Barcelona, Spain
| |
Collapse
|
31
|
Nambou K, Nie X, Tong Y, Anakpa M. Weighted gene co-expression network analysis and drug-gene interaction bioinformatics uncover key genes associated with various presentations of malaria infection in African children and major drug candidates. INFECTION GENETICS AND EVOLUTION 2021; 89:104723. [PMID: 33444859 DOI: 10.1016/j.meegid.2021.104723] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 01/04/2021] [Accepted: 01/08/2021] [Indexed: 01/06/2023]
Abstract
Malaria is a fatal parasitic disease with unelucidated pathogenetic mechanism. Herein, we aimed to uncover genes associated with different clinical aspects of malaria based on the GSE1124 dataset that is publicly accessible by using WGCNA. We obtained 16 co-expression modules and their correlations with clinical features. Using the MCODE tool, we identified THEM4, STYX, VPS36, LCOR, KIAA1143, EEA1, RAPGEF6, LOC439994, ZBTB33, PTPN22, ESCO1, and KLF3 as hub genes positively associated with Plasmodium falciparum infection (ASPF). These hub genes were involved in the biological processes of endosomal transport, regulation of natural killer cell proliferation, and KEGG pathways of endocytosis and fatty acid elongation. For the purple module negatively correlated with ASPF, we identified 19 hub genes that were involved in the biological processes of positive regulation of cellular protein catabolic process and KEGG pathways of other glycan degradation. For the salmon module positively correlated with severe malaria anemia (SMA), we identified 17 hub genes that were among those driving the biological processes of positive regulation of erythrocyte differentiation. For the brown module positively correlated with cerebral malaria (CM), we identified eight hub genes and these genes participated in phagolysosome assembly and positive regulation of exosomal secretion, and animal mitophagy pathway. For the tan module negatively correlated with CM, we identified four hub genes that were involved in CD8-positive, alpha-beta T cell differentiation and notching signaling pathway. These findings may provide new insights into the pathogenesis of malaria and help define new diagnostic and therapeutic approaches for malaria patients.
Collapse
Affiliation(s)
- Komi Nambou
- Shenzhen Nambou1 Biotech Company Limited, West Silicon Valley, No. 5010 Bao'an Avenue, Shenzhen 518000, Guangdong Province, China.
| | - Xiaoling Nie
- Shenzhen Nambou1 Biotech Company Limited, West Silicon Valley, No. 5010 Bao'an Avenue, Shenzhen 518000, Guangdong Province, China
| | - Yin Tong
- Shenzhen Nambou1 Biotech Company Limited, West Silicon Valley, No. 5010 Bao'an Avenue, Shenzhen 518000, Guangdong Province, China
| | - Manawa Anakpa
- Key Laboratory of Trustworthy Distributed Computing and Service, School of Computer Science (National Pilot Software Engineering School), Beijing University of Posts and Telecommunications, Ministry of Education, Beijing 100876, China
| |
Collapse
|
32
|
Mario-Vásquez JE, Naranjo-González CA, Montiel J, Zuluaga LM, Vásquez AM, Tobón-Castaño A, Bedoya G, Segura C. Association of variants in IL1B, TLR9, TREM1, IL10RA, and CD3G and Native American ancestry on malaria susceptibility in Colombian populations. INFECTION GENETICS AND EVOLUTION 2020; 87:104675. [PMID: 33316430 DOI: 10.1016/j.meegid.2020.104675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2020] [Revised: 11/19/2020] [Accepted: 12/09/2020] [Indexed: 12/24/2022]
Abstract
Host genetics is an influencing factor in the manifestation of infectious diseases. In this study, the association of mild malaria with 28 variants in 16 genes previously reported in other populations and/or close to ancestry-informative markers (AIMs) selected was evaluated in an admixed 736 Colombian population sample. Additionally, the effect of genetic ancestry on phenotype expression was explored. For this purpose, the ancestral genetic composition of Turbo and El Bagre was determined. A higher Native American ancestry trend was found in the population with lower malaria susceptibility [odds ratio (OR) = 0.416, 95% confidence interval (95% CI) = 0.234-0.740, P = 0.003]. Three AIMs presented significant associations with the disease phenotype (MID1752, MID921, and MID1586). The first two were associated with greater malaria susceptibility (D/D, OR = 2.23, 95% CI = 1.06-4.69, P = 0.032 and I/D-I/I, OR = 2.14, 95% CI = 1.18-3.87, P = 0.011, respectively), and the latter has a protective effect on the appearance of malaria (I/I, OR = 0.18, 95% CI = 0.08-0.40, P < 0.0001). After adjustment by age, sex, municipality, and genetic ancestry, genotype association analysis showed evidence of association with malaria susceptibility for variants in or near IL1B, TLR9, TREM1, IL10RA, and CD3G genes: rs1143629-IL1B (G/A-A/A, OR = 0.41, 95% CI = 0.21-0.78, P = 0.0051), rs352139-TLR9 (T/T, OR = 0.28, 95% CI = 0.11-0.72, P = 0.0053), rs352140-TLR9 (C/C, OR = 0.41, 95% CI = 0.20-0.87, P = 0.019), rs2234237-TREM1 (T/A-A/A, OR = 0.43, 95% CI = 0.23-0.79, P = 0.0056), rs4252246-IL10RA (C/A-A/A, OR = 2.11, 95% CI = 1.18-3.75, P = 0.01), and rs1561966-CD3G (A/A, OR = 0.20, 95% CI = 0.06-0.69, P = 0.0058). The results showed the participation of genes involved in immunological processes and suggested an effect of ancestral genetic composition over the traits analyzed. Compared to the paisa population (Antioquia), Turbo and El Bagre showed a strong decrease in European ancestry and an increase in African and Native American ancestries. Also, a novel association of two single nucleotide polymorphisms with malaria susceptibility was identified in this study.
Collapse
Affiliation(s)
- Jorge Eliécer Mario-Vásquez
- Grupo Genética Molecular (GENMOL), Universidad de Antioquia, Carrera 53 No. 61-30, Lab 430. Medellín, Colombia
| | | | - Jehidys Montiel
- Grupo Malaria-Facultad de Medicina, Universidad de Antioquia, Carrera 53 No. 61-30, Lab 610, Medellín, Colombia
| | - Lina M Zuluaga
- Grupo Malaria-Facultad de Medicina, Universidad de Antioquia, Carrera 53 No. 61-30, Lab 610, Medellín, Colombia
| | - Ana M Vásquez
- Grupo Malaria-Facultad de Medicina, Universidad de Antioquia, Carrera 53 No. 61-30, Lab 610, Medellín, Colombia
| | - Alberto Tobón-Castaño
- Grupo Malaria-Facultad de Medicina, Universidad de Antioquia, Carrera 53 No. 61-30, Lab 610, Medellín, Colombia
| | - Gabriel Bedoya
- Grupo Genética Molecular (GENMOL), Universidad de Antioquia, Carrera 53 No. 61-30, Lab 430. Medellín, Colombia
| | - Cesar Segura
- Grupo Malaria-Facultad de Medicina, Universidad de Antioquia, Carrera 53 No. 61-30, Lab 610, Medellín, Colombia.
| |
Collapse
|
33
|
Zhao Y, Hosking C, Cunningham D, Langhorne J, Lin JW. Transcriptome analysis of blood and spleen in virulent and avirulent mouse malaria infection. Sci Data 2020; 7:253. [PMID: 32753619 PMCID: PMC7403358 DOI: 10.1038/s41597-020-00592-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Accepted: 07/03/2020] [Indexed: 01/21/2023] Open
Abstract
Malaria is a devastating infectious disease and the immune response is complex and dynamic during a course of a malarial infection. Rodent malaria models allow detailed time-series studies of the host response in multiple organs. Here, we describe two comprehensive datasets containing host transcriptomic data from both the blood and spleen throughout an acute blood stage infection of virulent or avirulent Plasmodium chabaudi infection in C57BL/6 mice. The mRNA expression profiles were generated using Illumina BeadChip microarray. These datasets provide a groundwork for comprehensive and comparative studies on host gene expression in early, acute and recovering phases of a blood stage infection in both the blood and spleen, to explore the interaction between the two, and importantly to investigate whether these responses differ in virulent and avirulent infections. Measurement(s) | transcriptome • gene expression • malaria | Technology Type(s) | Microarray | Factor Type(s) | blood versus spleen • virulent versus avirulent malaria infection | Sample Characteristic - Organism | Mus musculus |
Machine-accessible metadata file describing the reported data: 10.6084/m9.figshare.12619232
Collapse
Affiliation(s)
- Yuancun Zhao
- Division of Pediatric Infectious Diseases and Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Caroline Hosking
- Malaria Immunology laboratory, The Francis Crick Institute, London, NW1 1AT, United Kingdom
| | - Deirdre Cunningham
- Malaria Immunology laboratory, The Francis Crick Institute, London, NW1 1AT, United Kingdom
| | - Jean Langhorne
- Malaria Immunology laboratory, The Francis Crick Institute, London, NW1 1AT, United Kingdom.
| | - Jing-Wen Lin
- Division of Pediatric Infectious Diseases and Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, State Key Laboratory of Biotherapy, West China Second University Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China.
| |
Collapse
|
34
|
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: 111] [Impact Index Per Article: 27.8] [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
|
35
|
Hodgson SH, Muller J, Lockstone HE, Hill AVS, Marsh K, Draper SJ, Knight JC. Use of gene expression studies to investigate the human immunological response to malaria infection. Malar J 2019; 18:418. [PMID: 31835999 PMCID: PMC6911278 DOI: 10.1186/s12936-019-3035-0] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Accepted: 11/26/2019] [Indexed: 01/02/2023] Open
Abstract
Background Transcriptional profiling of the human immune response to malaria has been used to identify diagnostic markers, understand the pathogenicity of severe disease and dissect the mechanisms of naturally acquired immunity (NAI). However, interpreting this body of work is difficult given considerable variation in study design, definition of disease, patient selection and methodology employed. This work details a comprehensive review of gene expression profiling (GEP) of the human immune response to malaria to determine how this technology has been applied to date, instances where this has advanced understanding of NAI and the extent of variability in methodology between studies to allow informed comparison of data and interpretation of results. Methods Datasets from the gene expression omnibus (GEO) including the search terms; ‘plasmodium’ or ‘malaria’ or ‘sporozoite’ or ‘merozoite’ or ‘gametocyte’ and ‘Homo sapiens’ were identified and publications analysed. Datasets of gene expression changes in relation to malaria vaccines were excluded. Results Twenty-three GEO datasets and 25 related publications were included in the final review. All datasets related to Plasmodium falciparum infection, except two that related to Plasmodium vivax infection. The majority of datasets included samples from individuals infected with malaria ‘naturally’ in the field (n = 13, 57%), however some related to controlled human malaria infection (CHMI) studies (n = 6, 26%), or cells stimulated with Plasmodium in vitro (n = 6, 26%). The majority of studies examined gene expression changes relating to the blood stage of the parasite. Significant heterogeneity between datasets was identified in terms of study design, sample type, platform used and method of analysis. Seven datasets specifically investigated transcriptional changes associated with NAI to malaria, with evidence supporting suppression of the innate pro-inflammatory response as an important mechanism for this in the majority of these studies. However, further interpretation of this body of work was limited by heterogeneity between studies and small sample sizes. Conclusions GEP in malaria is a potentially powerful tool, but to date studies have been hypothesis generating with small sample sizes and widely varying methodology. As CHMI studies are increasingly performed in endemic settings, there will be growing opportunity to use GEP to understand detailed time-course changes in host response and understand in greater detail the mechanisms of NAI.
Collapse
Affiliation(s)
- Susanne H Hodgson
- The Jenner Institute, University of Oxford, Old Road Campus Road Building, Off Roosevelt Drive, Oxford, OX3 7DQ, UK. .,Department of Infectious Diseases & Microbiology, Oxford University Hospitals Trust, Oxford, UK.
| | - Julius Muller
- The Jenner Institute, University of Oxford, Old Road Campus Road Building, Off Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Helen E Lockstone
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Adrian V S Hill
- The Jenner Institute, University of Oxford, Old Road Campus Road Building, Off Roosevelt Drive, Oxford, OX3 7DQ, UK.,Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Kevin Marsh
- Department of Tropical Medicine, University of Oxford, Oxford, UK
| | - Simon J Draper
- The Jenner Institute, University of Oxford, Old Road Campus Road Building, Off Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Julian C Knight
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| |
Collapse
|
36
|
Tran TM, Crompton PD. Decoding the complexities of human malaria through systems immunology. Immunol Rev 2019; 293:144-162. [PMID: 31680289 DOI: 10.1111/imr.12817] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 09/27/2019] [Accepted: 09/30/2019] [Indexed: 12/18/2022]
Abstract
The complexity of the Plasmodium parasite and its life cycle poses a challenge to our understanding of the host immune response against malaria. Studying human immune responses during natural and experimental Plasmodium infections can enhance our understanding of malaria-protective immunity and inform the design of disease-modifying adjunctive therapies and next-generation malaria vaccines. Systems immunology can complement conventional approaches to facilitate our understanding of the complex immune response to the highly dynamic malaria parasite. In this review, recent studies that used systems-based approaches to evaluate human immune responses during natural and experimental Plasmodium falciparum and Plasmodium vivax infections as well as during immunization with candidate malaria vaccines are summarized and related to each other. The potential for next-generation technologies to address the current limitations of systems-based studies of human malaria are discussed.
Collapse
Affiliation(s)
- Tuan M Tran
- Division of Infectious Diseases, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA.,Ryan White Center for Pediatric Infectious Disease and Global Health, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.,Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| |
Collapse
|
37
|
Talavera-López C, Bediako Y, Lin JW, Joseph Valletta J, Recker M, Langhorne J. Comparison of whole blood and spleen transcriptional signatures over the course of an experimental malaria infection. Sci Rep 2019; 9:15853. [PMID: 31676877 PMCID: PMC6825210 DOI: 10.1038/s41598-019-52388-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2019] [Accepted: 10/16/2019] [Indexed: 12/13/2022] Open
Abstract
Although the spleen is broadly accepted as the major lymphoid organ involved in generating immune responses to the erythrocytic stages of the malaria parasite, Plasmodium, human splenic tissue is not readily available in most cases. As a result, most studies of malaria in humans rely on peripheral blood to assess cellular immune responses to malaria. The suitability of peripheral blood as a proxy for splenic immune responses is however unknown. Here, we have simultaneously analysed the transcriptomes of whole blood and spleen over 12 days of erythrocytic stage Plasmodium chabaudi infection in C57BL/6 mice. Using both unsupervised and directed approaches, we compared gene expression between blood and spleen over the course of infection. Taking advantage of publicly available datasets, we used machine learning approaches to infer cell proportions and cell-specific gene expression signatures from our whole tissue transcriptome data. Our findings demonstrate that spleen and blood are quite dissimilar, sharing only a small amount of transcriptional information between them, with transcriptional differences in both cellular composition and transcriptional activity. These results suggest that while blood transcriptome data may be useful in defining surrogate markers of protection and pathology, they should not be used to predict specific immune responses occurring in lymphoid organs.
Collapse
Affiliation(s)
| | - Yaw Bediako
- The Francis Crick Institute, London, United Kingdom.,West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Accra, Ghana
| | - Jing-Wen Lin
- The Francis Crick Institute, London, United Kingdom.,Division of Pediatric Infectious Diseases, State Key Laboratory of Biotherapy, West China Second Hospital, Sichuan University and Collaboration Innovation Centre, Chengdu, 610041, China
| | - John Joseph Valletta
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, United Kingdom
| | - Mario Recker
- College of Engineering, Mathematics and Physical Sciences, University of Exeter, Exeter, United Kingdom
| | | |
Collapse
|
38
|
Kimenyi KM, Wamae K, Ochola-Oyier LI. Understanding P. falciparum Asymptomatic Infections: A Proposition for a Transcriptomic Approach. Front Immunol 2019; 10:2398. [PMID: 31681289 PMCID: PMC6803459 DOI: 10.3389/fimmu.2019.02398] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Accepted: 09/25/2019] [Indexed: 12/11/2022] Open
Abstract
Malaria is still a significant public health burden in the tropics. Infection with malaria causing parasites results in a wide range of clinical disease presentations, from severe to uncomplicated or mild, and in the poorly understood asymptomatic infections. The complexity of asymptomatic infections is due to the intricate interplay between factors derived from the human host, parasite, and environment. Asymptomatic infections often go undetected and provide a silent natural reservoir that sustains malaria transmission. This creates a major obstacle for malaria control and elimination efforts. Numerous studies have tried to characterize asymptomatic infections, unanimously revealing that host immunity is the underlying factor in the maintenance of these infections and in the risk of developing febrile malaria infections. An in-depth understanding of how host immunity and parasite factors interact to cause malaria disease tolerance is thus required. This review primarily focuses on understanding anti-inflammatory and pro-inflammatory responses to asymptomatic infections in malaria endemic areas, to present the view that it is potentially the shift in host immunity toward an anti-inflammatory profile that maintains asymptomatic infections after multiple exposures to malaria. Conversely, symptomatic infections are skewed toward a pro-inflammatory immune profile. Moreover, we propose that these infections can be better interrogated using next generation sequencing technologies, in particular RNA sequencing (RNA-seq), to investigate the immune system using the transcriptome sampled during a clearly defined asymptomatic infection.
Collapse
Affiliation(s)
- Kelvin M Kimenyi
- KEMRI-Wellcome Trust Research Programme, CGMRC, Kilifi, Kenya.,Centre for Biotechnology and Bioinformatics, University of Nairobi, Nairobi, Kenya
| | - Kevin Wamae
- KEMRI-Wellcome Trust Research Programme, CGMRC, Kilifi, Kenya
| | - Lynette Isabella Ochola-Oyier
- KEMRI-Wellcome Trust Research Programme, CGMRC, Kilifi, Kenya.,Pwani University Bioscience Research Centre, Pwani University, Kilifi, Kenya
| |
Collapse
|
39
|
Perturbations in neuroinflammatory pathways are associated with paclitaxel-induced peripheral neuropathy in breast cancer survivors. J Neuroimmunol 2019; 335:577019. [PMID: 31401418 PMCID: PMC6788784 DOI: 10.1016/j.jneuroim.2019.577019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 07/18/2019] [Accepted: 08/01/2019] [Indexed: 01/11/2023]
Abstract
Paclitaxel is a common chemotherapy drug associated with the development of chronic paclitaxel-induced peripheral neuropathy (PIPN). PIPN is associated with neuroinflammatory mechanisms in pre-clinical studies. Here, we evaluated for differential gene expression (DGE) in peripheral blood between breast cancer survivors with and without PIPN and for neuroinflammatory (NI) related signaling pathways and whole-transcriptome profiles from other experiments. Pathway impact analysis identified 8 perturbed NI related pathways. Expression profile analysis found 15 experiments having similar whole-transcriptome profiles of DGE related to neuroinflammation and PIPN. These findings suggest that perturbations in pathways associated with neuroinflammation are found in cancer survivors with PIPN. Paclitaxel-induced peripheral neuropathy (PIPN) is associated with Paclitaxel treatment Differential gene expression was associated with PIPN in breast cancer survivors. Perturbations of neuroinflammatory-related pathways were identified between survivors. Transcriptome profile was similar to other pre-clinical and clinical studies.
Collapse
|
40
|
Fakiola M, Singh OP, Syn G, Singh T, Singh B, Chakravarty J, Sundar S, Blackwell JM. Transcriptional blood signatures for active and amphotericin B treated visceral leishmaniasis in India. PLoS Negl Trop Dis 2019; 13:e0007673. [PMID: 31419223 PMCID: PMC6713396 DOI: 10.1371/journal.pntd.0007673] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 08/28/2019] [Accepted: 07/30/2019] [Indexed: 12/13/2022] Open
Abstract
Amphotericin B provides improved therapy for visceral leishmaniasis (VL) caused by Leishmania donovani, with single dose liposomal-encapsulated Ambisome providing the best cure rates. The VL elimination program aims to reduce the incidence rate in the Indian subcontinent to <1/10,000 population/year. Ability to predict which asymptomatic individuals (e.g. anti-leishmanial IgG and/or Leishmania-specific modified Quantiferon positive) will progress to clinical VL would help in monitoring disease outbreaks. Here we examined whole blood transcriptional profiles associated with asymptomatic infection, active disease, and in treated cases. Two independent microarray experiments were performed, with analysis focussed primarily on differentially expressed genes (DEGs) concordant across both experiments. No DEGs were identified for IgG or Quantiferon positive asymptomatic groups compared to negative healthy endemic controls. We therefore concentrated on comparing concordant DEGs from active cases with all healthy controls, and in examining differences in the transcriptome following different regimens of drug treatment. In these comparisons 6 major themes emerged: (i) expression of genes and enrichment of gene sets associated with erythrocyte function in active cases; (ii) strong evidence for enrichment of gene sets involved in cell cycle in comparing active cases with healthy controls; (iii) identification of IFNG encoding interferon-γ as the major hub gene in concordant gene expression patterns across experiments comparing active cases with healthy controls or with treated cases; (iv) enrichment for interleukin signalling (IL-1/3/4/6/7/8) and a prominent role for CXCL10/9/11 and chemokine signalling pathways in comparing active cases with treated cases; (v) the novel identification of Aryl Hydrocarbon Receptor signalling as a significant canonical pathway when comparing active cases with healthy controls or with treated cases; and (vi) global expression profiling support for more effective cure at day 30 post-treatment with a single dose of liposomal encapsulated amphotericin B compared to multi-dose non-liposomal amphotericin B treatment over 30 days. (296 words; 300 words allowed).
Collapse
Affiliation(s)
- Michaela Fakiola
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- INGM-National Institute of Molecular Genetics "Romeo ed Enrica Invernizzi" Milan, Milan, Italy
| | - Om Prakash Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Genevieve Syn
- Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Toolika Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Bhawana Singh
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Jaya Chakravarty
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Jenefer M. Blackwell
- Department of Pathology, University of Cambridge, Cambridge, United Kingdom
- Telethon Kids Institute, The University of Western Australia, Nedlands, Western Australia, Australia
| |
Collapse
|