1
|
Charles-Chess NAE, Kurup SP. Regulatory T cell memory: implications for malaria. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025:vkaf067. [PMID: 40267394 DOI: 10.1093/jimmun/vkaf067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/12/2025] [Indexed: 04/25/2025]
Abstract
Regulatory T cells (Tregs) can persist as memory cells (mTregs) in both infectious and non-infectious settings. However, their functional behavior, phenotypic stability, and suppressive properties upon antigen re-exposure remain poorly understood. Emerging evidence suggests that mTregs exhibit enhanced proliferation and suppressive capacity upon re-encountering the same antigen, a feature that may be critical in recurrent infections such as malaria. In malaria, Tregs are known to modulate immune responses and influence acute disease outcomes, suggesting that mTreg recall may play a significant role in long-term immunity. This review explores the biology of Treg memory, with a focus on malaria, and examines the immunological implications of maintaining a suppressive mTreg population in malaria immunity.
Collapse
Affiliation(s)
- Nana Appiah Essel Charles-Chess
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
- Center for Tropical & Emerging Global Diseases, University of Georgia, Athens, GA, United States
| | - Samarchith P Kurup
- Department of Cellular Biology, University of Georgia, Athens, GA, United States
- Center for Tropical & Emerging Global Diseases, University of Georgia, Athens, GA, United States
| |
Collapse
|
2
|
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] [MESH Headings] [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
|
3
|
Segbefia SP, Asandem DA, Amoah LE, Kusi KA. Cytokine gene polymorphisms implicated in the pathogenesis of Plasmodium falciparum infection outcome. Front Immunol 2024; 15:1285411. [PMID: 38404582 PMCID: PMC10884311 DOI: 10.3389/fimmu.2024.1285411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 01/24/2024] [Indexed: 02/27/2024] Open
Abstract
Cytokines play a critical role in the immune mechanisms involved in fighting infections including malaria. Polymorphisms in cytokine genes may affect immune responses during an infection with Plasmodium parasites and immunization outcomes during routine administration of malaria vaccines. These polymorphisms can increase or reduce susceptibility to this deadly infection, and this may affect the physiologically needed balance between anti-inflammatory and pro-inflammatory cytokines. The purpose of this review is to present an overview of the effect of selected cytokine gene polymorphisms on immune responses against malaria.
Collapse
Affiliation(s)
- Selorm Philip Segbefia
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- Department of Molecular Medicine, School of Medicine and Dentistry, College of Science, Kwame Nkrumah University of Science and Technology (KNUST), Kumasi, Ghana
| | - Diana Asema Asandem
- Department of Virology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Linda Eva Amoah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Kwadwo Asamoah Kusi
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
- West African Centre for Cell Biology of Infectious Pathogens, Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
4
|
Dvorak NM, Domingo ND, Tapia CM, Wadsworth PA, Marosi M, Avchalumov Y, Fongsaran C, Koff L, Di Re J, Sampson CM, Baumgartner TJ, Wang P, Villarreal PP, Solomon OD, Stutz SJ, Aditi, Porter J, Gbedande K, Prideaux B, Green TA, Seeley EH, Samir P, Dineley KT, Vargas G, Zhou J, Cisneros I, Stephens R, Laezza F. TNFR1 signaling converging on FGF14 controls neuronal hyperactivity and sickness behavior in experimental cerebral malaria. J Neuroinflammation 2023; 20:306. [PMID: 38115011 PMCID: PMC10729485 DOI: 10.1186/s12974-023-02992-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 12/11/2023] [Indexed: 12/21/2023] Open
Abstract
BACKGROUND Excess tumor necrosis factor (TNF) is implicated in the pathogenesis of hyperinflammatory experimental cerebral malaria (eCM), including gliosis, increased levels of fibrin(ogen) in the brain, behavioral changes, and mortality. However, the role of TNF in eCM within the brain parenchyma, particularly directly on neurons, remains underdefined. Here, we investigate electrophysiological consequences of eCM on neuronal excitability and cell signaling mechanisms that contribute to observed phenotypes. METHODS The split-luciferase complementation assay (LCA) was used to investigate cell signaling mechanisms downstream of tumor necrosis factor receptor 1 (TNFR1) that could contribute to changes in neuronal excitability in eCM. Whole-cell patch-clamp electrophysiology was performed in brain slices from eCM mice to elucidate consequences of infection on CA1 pyramidal neuron excitability and cell signaling mechanisms that contribute to observed phenotypes. Involvement of identified signaling molecules in mediating behavioral changes and sickness behavior observed in eCM were investigated in vivo using genetic silencing. RESULTS Exploring signaling mechanisms that underlie TNF-induced effects on neuronal excitability, we found that the complex assembly of fibroblast growth factor 14 (FGF14) and the voltage-gated Na+ (Nav) channel 1.6 (Nav1.6) is increased upon tumor necrosis factor receptor 1 (TNFR1) stimulation via Janus Kinase 2 (JAK2). On account of the dependency of hyperinflammatory experimental cerebral malaria (eCM) on TNF, we performed patch-clamp studies in slices from eCM mice and showed that Plasmodium chabaudi infection augments Nav1.6 channel conductance of CA1 pyramidal neurons through the TNFR1-JAK2-FGF14-Nav1.6 signaling network, which leads to hyperexcitability. Hyperexcitability of CA1 pyramidal neurons caused by infection was mitigated via an anti-TNF antibody and genetic silencing of FGF14 in CA1. Furthermore, knockdown of FGF14 in CA1 reduced sickness behavior caused by infection. CONCLUSIONS FGF14 may represent a therapeutic target for mitigating consequences of TNF-mediated neuroinflammation.
Collapse
Affiliation(s)
- Nolan M Dvorak
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Nadia D Domingo
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Cynthia M Tapia
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Paul A Wadsworth
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Mate Marosi
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Yosef Avchalumov
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Chanida Fongsaran
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Leandra Koff
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jessica Di Re
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Catherine M Sampson
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Timothy J Baumgartner
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Pingyuan Wang
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Paula P Villarreal
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Clinical Sciences Program, The Institute for Translational Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Olivia D Solomon
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Sonja J Stutz
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Aditi
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jacob Porter
- Department of Chemistry, University of Texas, Austin, TX, 78712, USA
| | - Komi Gbedande
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Center for Immunity and Inflammation and Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, 07301, USA
| | - Brendan Prideaux
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Thomas A Green
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Erin H Seeley
- Department of Chemistry, University of Texas, Austin, TX, 78712, USA
| | - Parimal Samir
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kelley T Dineley
- Center for Addiction Sciences and Therapeutics, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Gracie Vargas
- Department of Neurobiology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Jia Zhou
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Irma Cisneros
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Robin Stephens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
- Center for Immunity and Inflammation and Department of Pharmacology, Physiology and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ, 07301, USA.
| | - Fernanda Laezza
- Department of Pharmacology & Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
5
|
Gupta A, Skjefte M, Muppidi P, Sikka R, Pandey M, Bharti PK, Gupta H. Unravelling the Influence of Host Genetic Factors on Malaria Susceptibility in Asian Populations. Acta Trop 2023; 249:107055. [PMID: 39491156 DOI: 10.1016/j.actatropica.2023.107055] [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: 08/08/2023] [Revised: 10/21/2023] [Accepted: 10/26/2023] [Indexed: 11/05/2024]
Abstract
Malaria is a deadly blood-borne disease caused by a Plasmodium parasite. Infection results in various forms of malaria, including an asymptomatic state, uncomplicated disease, or severe disease. Severe malaria (SM) is particularly prevalent among young children and is a significant cause of mortality. SM is associated with the sequestration of parasitized erythrocytes in the microvasculature of vital host organs, disrupting the normal functioning of the immune system. Although the exact mechanisms of malaria pathogenesis are yet to be fully understood, researchers have been investigating the role of host genetics in determining the severity of the disease and the outcome of infection. The objective of this study is to identify specific host genes that have been examined for their association with malaria in Asian populations and pinpoint those most likely to influence susceptibility. Through an extensive screening process, a total of 982 articles were initially identified, and after careful review, 40 articles discussing 68 genes were included in this review. By constructing a network of protein-protein interactions (PPIs), we identified six key proteins (TNF, IL6, TLR4, IL1β, IL10, and IL8) that exhibited substantial interactions (more than 30 edges), suggesting their potential as significant targets for influencing malaria susceptibility. Notably, these six proteins have been previously identified as crucial components of the immune response, associated with malaria susceptibility, and capable of affecting different clinical forms of the disease. Identifying genes that contribute to malaria susceptibility or resistance holds the promise of enhancing the diagnosis and treatment of this debilitating illness. Such knowledge has the potential to pave the way for more targeted and effective strategies in combating malaria, particularly in Asian populations where controlling Plasmodium vivax is challenging, and India contributes the highest number of cases. By understanding the genetic factors underlying malaria vulnerability, we can develop interventions that are tailored to the specific needs of Asian populations, ultimately leading to better outcomes in the fight against this disease.
Collapse
Affiliation(s)
- Aditi Gupta
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, Uttar Pradesh, India
| | - Malia Skjefte
- Population Services International, Malaria Department, Washington, DC, USA
| | - Pranavi Muppidi
- GKT School of Medical Education, King's College London, London, UK
| | - Ruhi Sikka
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, Uttar Pradesh, India.
| | - Manju Pandey
- Department of Medicine, K. D. Medical College Hospital & Research Center, Mathura, Uttar Pradesh, India
| | - Praveen Kumar Bharti
- ICMR- National Institute of Malaria Research (ICMR-NIMR), Dwarka, New Delhi, India
| | - Himanshu Gupta
- Department of Biotechnology, Institute of Applied Sciences & Humanities, GLA University, Mathura, Uttar Pradesh, India.
| |
Collapse
|
6
|
Fan YX, Chen LR, Gan RX, Yin SJ, Wang P, Meng R, Huang YH, Jiang FF, He GH. A meta-analysis of associations of IL-10 gene polymorphisms with acute leukemia susceptibility. Cytokine 2023; 170:156312. [PMID: 37542945 DOI: 10.1016/j.cyto.2023.156312] [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: 07/07/2023] [Revised: 07/26/2023] [Accepted: 07/28/2023] [Indexed: 08/07/2023]
Abstract
BACKGROUND Recently, increasing evidence has demonstrated that IL-10 single nucleotide polymorphisms (SNPs) are associated with the risk of acute leukemia (AL), but the findings of different articles remain controversial. Thus, we performed a meta-analysis to further investigate the exact roles of IL-10 SNPs in AL susceptibility. METHODS Six common Chinese and English databases were utilized to retrieve eligible studies. The strength of the association was assessed by calculating odds ratios and 95 % confidence intervals. All analyses were carried out using Review Manager (version 5.3) and STATA (version 15.1). The registered number of this research is CRD42022373362. RESULTS A total of 6391 participants were enrolled in this research. The results showed that the AG genotype of rs1800896 increased AL risk in the heterozygous codominant model (AG vs. AA, OR = 1.41, 95 % CI = 1.04-1.92, P = 0.03) and overdominant model (AG vs. AA + GG, OR = 1.32, 95 % CI = 1.04-1.70, P = 0.03). In the subgroup analysis, associations between the G allele, GG genotype, AG genotype, AG + GG genotype of rs1800896 and increased AL risk were also observed in the mixed population based on allelic, homozygote codominant, heterozygous codominant, dominant, and overdominant models. Furthermore, an association between the AC genotype of rs1800872 and increased AL risk was observed in the Caucasian population in the overdominant model. However, the rs1800871, rs3024489 and rs3024493 polymorphisms did not affect AL risk. CONCLUSION IL-10 rs1800896 and rs1800872 affected the susceptibility of AL and therefore may be biomarkers for early screening and risk prediction of AL.
Collapse
Affiliation(s)
- Yu-Xin Fan
- Research Center of Clinical Pharmacology, The First Affiliated Hospital of Yunnan University of Chinese Medicine, Kunming, China
| | - Li-Rong Chen
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
| | - Run-Xin Gan
- Reproductive Medicine Center, Xiangya Hospital, Central South University, Changsha, China
| | - Sun-Jun Yin
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
| | - Ping Wang
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
| | - Rui Meng
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
| | - Yan-Hua Huang
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
| | - Fang-Fang Jiang
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China
| | - Gong-Hao He
- Department of Clinical Pharmacy, 920th Hospital of Joint Logistics Support Force of People's Liberation Army, Kunming, China.
| |
Collapse
|
7
|
Cahill S, Chandola T, Hager R. Genetic Variants Associated With Resilience in Human and Animal Studies. Front Psychiatry 2022; 13:840120. [PMID: 35669264 PMCID: PMC9163442 DOI: 10.3389/fpsyt.2022.840120] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/19/2022] [Indexed: 11/15/2022] Open
Abstract
Resilience is broadly defined as the ability to maintain or regain functioning in the face of adversity and is influenced by both environmental and genetic factors. The identification of specific genetic factors and their biological pathways underpinning resilient functioning can help in the identification of common key factors, but heterogeneities in the operationalisation of resilience have hampered advances. We conducted a systematic review of genetic variants associated with resilience to enable the identification of general resilience mechanisms. We adopted broad inclusion criteria for the definition of resilience to capture both human and animal model studies, which use a wide range of resilience definitions and measure very different outcomes. Analyzing 158 studies, we found 71 candidate genes associated with resilience. OPRM1 (Opioid receptor mu 1), NPY (neuropeptide Y), CACNA1C (calcium voltage-gated channel subunit alpha1 C), DCC (deleted in colorectal carcinoma), and FKBP5 (FKBP prolyl isomerase 5) had both animal and human variants associated with resilience, supporting the idea of shared biological pathways. Further, for OPRM1, OXTR (oxytocin receptor), CRHR1 (corticotropin-releasing hormone receptor 1), COMT (catechol-O-methyltransferase), BDNF (brain-derived neurotrophic factor), APOE (apolipoprotein E), and SLC6A4 (solute carrier family 6 member 4), the same allele was associated with resilience across divergent resilience definitions, which suggests these genes may therefore provide a starting point for further research examining commonality in resilience pathways.
Collapse
Affiliation(s)
- Stephanie Cahill
- Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
- Faculty of Humanities, Cathie Marsh Institute for Social Research, The University of Manchester, Manchester, United Kingdom
| | - Tarani Chandola
- Faculty of Humanities, Cathie Marsh Institute for Social Research, The University of Manchester, Manchester, United Kingdom
- Methods Hub, Department of Sociology, Faculty of Social Sciences, The University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Reinmar Hager
- Evolution, Infection and Genomics, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, The University of Manchester, Manchester, United Kingdom
| |
Collapse
|
8
|
Pratap PD, Raza ST, Zaidi G, Kunwar S, Ahmad S, Charles MR, Eba A, Rajput M. Genetic Variants in Interleukin-10 Gene Association with Susceptibility and Cervical Cancer Development: A Case Control Study. Glob Med Genet 2022; 9:129-140. [PMID: 35707782 PMCID: PMC9192188 DOI: 10.1055/s-0042-1743262] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Accepted: 12/29/2021] [Indexed: 12/24/2022] Open
Abstract
Objectives
Cervical cancer (CC) is one of the most destructive disease caused by persistent HPV infection which affects women worldwide, especially in developing countries. The genetic basis of host immune response especially cytokine function has been shown to influence CC susceptibility. Studies have demonstrated that IL-10 gene polymorphism have been associated with numerous malignancies, but in context to CC results were inconclusive. Though, aim of our study to investigate the association between IL-10 -1082A/G and -819C/T promoter polymorphism and CC susceptibility.
Material and Methods
This study comprised 192 women with CC and 200 controls. HPV detection was done by RT-PCR and genotyping was assessed through PCR-RFLP method. Serum concentration of IL-10 measured by ELISA.
Results
Women with AG and AG+GG genotypes of IL-10 -1082A/G had two-fold increased risk of CC [OR, 2.35 (95% CI, 1.54–3.58),
p
= 0.005], [OR, 2.03 (95% CI, 1.36–3.04),
p
= 0.0005] compared to controls. Women with G allele of -1082A/G polymorphism had linked with CC susceptibility [OR, 1.39 (95% CI, 1.02–1.88),
p
= 0.036] compared to controls. No significant difference was found between patients and controls in the genotype or allele frequencies of IL–10 -819C/T polymorphism [OR, 1.00 (95% CI, 0.63–1.58),
p
= 0.99]. The level of serum concentration of IL-10 was significantly higher in cases compared to controls.
Conclusion
These findings help to understand that polymorphism of IL-10 -1082A/G gene is associated with increased risk of CC development and can serve as a marker of genetic susceptibility to CC.
Collapse
Affiliation(s)
- Pushpendra D. Pratap
- Central Research Laboratory, Molecular Diagnostic Unit, Department of Biochemistry, ERA's Lucknow Medical College, ERA University, Lucknow, Uttar Pradesh, India
| | - Syed Tasleem Raza
- Central Research Laboratory, Molecular Diagnostic Unit, Department of Biochemistry, ERA's Lucknow Medical College, ERA University, Lucknow, Uttar Pradesh, India
| | - Ghazala Zaidi
- Department of Allied Health Sciences, ERA University, Lucknow, Uttar Pradesh, India
| | - Shipra Kunwar
- Department of Obstetrics & Gynecology, ERA University, Lucknow, Uttar Pradesh, India
| | - Sharique Ahmad
- Department of Pathology ERA's Lucknow Medical College, ERA University, Lucknow, Uttar Pradesh, India
| | - Mark Rector Charles
- Central Research Laboratory, Molecular Diagnostic Unit, Department of Biochemistry, ERA's Lucknow Medical College, ERA University, Lucknow, Uttar Pradesh, India
| | - Ale Eba
- Central Research Laboratory, Molecular Diagnostic Unit, Department of Biochemistry, ERA's Lucknow Medical College, ERA University, Lucknow, Uttar Pradesh, India
| | - Muneshwar Rajput
- Central Research Laboratory, Molecular Diagnostic Unit, Department of Biochemistry, ERA's Lucknow Medical College, ERA University, Lucknow, Uttar Pradesh, India
| |
Collapse
|
9
|
Sirisabhabhorn K, Chaijaroenkul W, Na-Bangchang K. Genetic Diversity of Human Host Genes Involved in Immune Response and the Binding of Malaria Parasite in Patients Residing along the Thai-Myanmar border. Trop Med Infect Dis 2021; 6:tropicalmed6040174. [PMID: 34698295 PMCID: PMC8544681 DOI: 10.3390/tropicalmed6040174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 09/22/2021] [Accepted: 09/22/2021] [Indexed: 12/05/2022] Open
Abstract
Polymorphisms of the genes encoding proteins involved in immune functions and the binding of malaria parasites to human host cells have been the focus of research in recent years, aiming to understand malaria pathogenesis and case severity and to exploit this knowledge to assert control over malaria. This study investigated the genetic diversity of the human host genes encoding proteins that are involved in immune functions and malaria parasite binding, i.e., MCP1 (−2518), TGFβ1 (−509), TNFα (−308), IL4 (VNTR), IL6 (−174), IL10 (−3575), TLR4 (299), CD36 (−188), and ICAM1 (469) in patients with mono-infection of Plasmodium falciparum and Plasmodium vivax infections in the multidrug-resistant areas along the Thai-Myanmar border. The association between gene polymorphisms and parasite density was also investigated. Genomic DNA (gDNA) of P. falciparum and P. vivax were extracted from whole blood and dried blood spot (DBS). Gene amplification and genotyping were performed by PCR and PCR-RFLP analysis, respectively. Of these samples, 178 and 209 samples were, respectively, mono-infection with P. falciparum and P. vivax. The ratio of P. falciparum: P. vivax was 46%:54%. Results showed marked variation in the frequency distribution and patterns of the genotypes and gene alleles of the nine immune response genes or human host genes. The SNPs of TGFβ1, IL10 and ICAM1, were significantly associated with P. falciparum, but not P. vivax parasite density. TGFβ1, IL10 and ICAM1, may play more significant roles in modulating P. falciparum than P. vivax parasitemia. The prevalence of the genotypes and gene alleles of these genes, including their association with parasite density, may vary depending on patient ethnicity and endemic areas. Information obtained from each endemic area is essential for treatment strategies and the development of vaccines for malaria prophylaxis in specific areas.
Collapse
Affiliation(s)
- Kridsada Sirisabhabhorn
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University (Rangsit Campus), Pathumthani 12121, Thailand; (K.S.); (W.C.)
| | - Wanna Chaijaroenkul
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University (Rangsit Campus), Pathumthani 12121, Thailand; (K.S.); (W.C.)
- Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Thammasat University (Rangsit Campus), Pathumthani 12121, Thailand
| | - Kesara Na-Bangchang
- Graduate Program in Bioclinical Sciences, Chulabhorn International College of Medicine, Thammasat University (Rangsit Campus), Pathumthani 12121, Thailand; (K.S.); (W.C.)
- Center of Excellence in Pharmacology and Molecular Biology of Malaria and Cholangiocarcinoma, Thammasat University (Rangsit Campus), Pathumthani 12121, Thailand
- Drug Discovery and Development Center, Office of Advanced Science and Technology, Thammasat University (Rangsit Campus), Pathumthani 12121, Thailand
- Correspondence: or ; Tel.: +662-564-4440-79 (ext. 1803); Fax: +662-564-4398
| |
Collapse
|
10
|
Patel H, Dunican C, Cunnington AJ. Predictors of outcome in childhood Plasmodium falciparum malaria. Virulence 2020; 11:199-221. [PMID: 32063099 PMCID: PMC7051137 DOI: 10.1080/21505594.2020.1726570] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 01/16/2020] [Accepted: 01/20/2020] [Indexed: 12/16/2022] Open
Abstract
Plasmodium falciparum malaria is classified as either uncomplicated or severe, determining clinical management and providing a framework for understanding pathogenesis. Severe malaria in children is defined by the presence of one or more features associated with adverse outcome, but there is wide variation in the predictive value of these features. Here we review the evidence for the usefulness of these features, alone and in combination, to predict death and other adverse outcomes, and we consider the role that molecular biomarkers may play in augmenting this prediction. We also examine whether a more personalized approach to predicting outcome for specific presenting syndromes of severe malaria, particularly cerebral malaria, has the potential to be more accurate. We note a general need for better external validation in studies of outcome predictors and for the demonstration that predictors can be used to guide clinical management in a way that improves survival and long-term health.
Collapse
Affiliation(s)
- Harsita Patel
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| | - Claire Dunican
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| | - Aubrey J. Cunnington
- Section of Paediatric Infectious Disease, Department of Infectious Disease, Imperial College London, London, UK
| |
Collapse
|
11
|
Mbagwu SI, Lannes N, Walch M, Filgueira L, Mantel PY. Human Microglia Respond to Malaria-Induced Extracellular Vesicles. Pathogens 2019; 9:pathogens9010021. [PMID: 31878288 PMCID: PMC7168629 DOI: 10.3390/pathogens9010021] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Revised: 12/16/2019] [Accepted: 12/20/2019] [Indexed: 12/19/2022] Open
Abstract
Microglia are the chief immune cells of the brain and have been reported to be activated in severe malaria. Their activation may drive towards neuroinflammation in cerebral malaria. Malaria-infected red blood cell derived-extracellular vesicles (MiREVs) are produced during the blood stage of malaria infection. They mediate intercellular communication and immune regulation, among other functions. During cerebral malaria, the breakdown of the blood–brain barrier can promote the migration of substances such as MiREVs from the periphery into the brain, targeting cells such as microglia. Microglia and extracellular vesicle interactions in different pathological conditions have been reported to induce neuroinflammation. Unlike in astrocytes, microglia–extracellular vesicle interaction has not yet been described in malaria infection. Therefore, in this study, we aimed to investigate the uptake of MiREVs by human microglia cells and their cytokine response. Human blood monocyte-derived microglia (MoMi) were generated from buffy coats of anonymous healthy donors using Ficoll-Paque density gradient centrifugation. The MiREVs were isolated from the Plasmodium falciparum cultures. They were purified by ultracentrifugation and labeled with PKH67 green fluorescent dye. The internalization of MiREVs by MoMi was observed after 4 h of co-incubation on coverslips placed in a 24-well plate at 37 °C using confocal microscopy. Cytokine-gene expression was investigated using rt-qPCR, following the stimulation of the MoMi cells with supernatants from the parasite cultures at 2, 4, and 24 h, respectively. MiREVs were internalized by the microglia and accumulated in the perinuclear region. MiREVs-treated cells increased gene expression of the inflammatory cytokine TNFα and reduced gene expression of the immune suppressive IL-10. Overall, the results indicate that MiREVs may act on microglia, which would contribute to enhanced inflammation in cerebral malaria.
Collapse
Affiliation(s)
- Smart Ikechukwu Mbagwu
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
- Department of Anatomy, Faculty of Basic Medical Sciences, Nnamdi Azikiwe University, Nnewi Campus, Nnewi 435101, Nigeria
- Correspondence: (S.I.M.); (L.F.)
| | - Nils Lannes
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| | - Michael Walch
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| | - Luis Filgueira
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
- Correspondence: (S.I.M.); (L.F.)
| | - Pierre-Yves Mantel
- Anatomy Unit, Department of Oncology, Microbiology and Immunology, Faculty of Science and Medicine, University of Fribourg, 1700 Fribourg, Switzerland
| |
Collapse
|
12
|
Zhang YH, Xing YQ, Chen Z, Ma XC, Lu Q. Association between interleukin-10 genetic polymorphisms and risk of primary open angle glaucoma in a Chinese Han population: a case-control study. Int J Ophthalmol 2019; 12:1605-1611. [PMID: 31637197 DOI: 10.18240/ijo.2019.10.13] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 07/04/2019] [Indexed: 01/10/2023] Open
Abstract
AIM To investigate the association between interleukin-10 (IL-10) genetic polymorphisms and risk of POAG through a case-control study in a Han population of China. METHODS A total of 210 patients with POAG and 420 normal subjects were recruited during the period from Dec. 2013 to Dec. 2016. The IL-10 -1082A>G (rs1800870), -819T>C (rs1800871) and -592C>A (rs1800872) polymorphisms were determined using iPlex GOLD SNP genotyping analysis (the SequenomMassARRAY® System, Sequenom, San Diego, USA). The association between IL-10 -1082A>G (rs1800870), -819T>C (rs1800871), and -592C>A (rs1800872) polymorphisms and risk of POAG was assessed by singlelogistic regression analysis. RESULTS We observed that those carrying the CC genotype of rs1800871 was associated with an increased risk of POAG when compared with those harboring the TT genotype (OR=1.84, 95%CI=1.01-3.38). Those with AA genotype of rs1800872 had a 10.62 fold risk of POAG in comparison to the CC genotype (OR=10.62, 95%CI, 3.41-33.09). A completely linkage disequilibrium was found between IL-10 rs1800871-rs1800872 (D'=1.00, r 2=0.16). The A-C-A (OR=2.60, 95%CI, 1.48-4.58) and G-T-A (OR=2.34, 95%CI, 1.42-3.86) haplotypes were associated with an increased risk of POAG, while the A-T-C haplotype showed a decreased risk of POAG (OR=0.63, 95%CI, 0.49-0.81). CONCLUSION Our data suggest that IL-10 rs1800871 and rs1800872 can be predictive factors for the pathogenesis of POAG in the Chinese population.
Collapse
Affiliation(s)
- Yi-Hui Zhang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Pronvince, China.,Ophthalmology Department of Inner Mongolia People's Hospital, Hohhot 010017, Inner Mongolia Autonomous Region, China
| | - Yi-Qiao Xing
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Pronvince, China
| | - Zhen Chen
- Department of Ophthalmology, Renmin Hospital of Wuhan University, Wuhan 430060, Hubei Pronvince, China
| | - Xiao-Cheng Ma
- Ophthalmology Department of Inner Mongolia People's Hospital, Hohhot 010017, Inner Mongolia Autonomous Region, China
| | - Qiang Lu
- Ophthalmology Department of Inner Mongolia People's Hospital, Hohhot 010017, Inner Mongolia Autonomous Region, China
| |
Collapse
|
13
|
Kisia LE, Kempaiah P, Anyona SB, Munde EO, Achieng AO, Ong’echa JM, Lambert CG, Chelimo K, Ouma C, Perkins DJ, Raballah E. Genetic variation in interleukin-7 is associated with a reduced erythropoietic response in Kenyan children infected with Plasmodium falciparum. BMC MEDICAL GENETICS 2019; 20:140. [PMID: 31420016 PMCID: PMC6698010 DOI: 10.1186/s12881-019-0866-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Accepted: 07/26/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Severe malarial anemia (SMA) is a leading cause of malaria-related morbidity and mortality in children. The genetic factors that influence development of SMA and inefficient erythropoiesis, a central pathogenic feature of SMA, are only partially understood. METHODS We performed a pilot Genome-wide Association Study (GWAS) on children with Plasmodium falciparum. The GWAS was performed using the Illumina® Infinium® HD Super Assay in conjunction with Illumina's® Human Omni2.5-8v1 BeadChip (with > 2.45 M markers). Data were analyzed using single SNP logistic regression analysis with an additive model of inheritance controlling for covariates. Results from our pilot global genomics study identified that variation in interleukin (IL)-7 was associated with enhanced risk of SMA. To validate this finding, we investigated the relationship between genotypes and/or haplotypes of two single nucleotide polymorphisms (SNPs) in IL7 [72194 T/C and - 2440 A/G] and susceptibility to both SMA and inefficient erythropoiesis [i.e., reticulocyte production index (RPI) < 2.0 in anemic children (Hb < 11.0 g/dL). Children presenting with P. falciparum malaria (< 3 years, n = 883) were stratified into two groups: Uncomplicated malaria (UM, n = 718) and SMA (n = 165). RESULTS Regression modeling, controlling for anemia-related confounders, revealed that carriage of the TC genotype at position 72194 T/C was associated with enhanced susceptibility to inefficient erythropoiesis (OR = 1.90; 95% CI 1.09-3.30; P = 0.02) as was homozygous CC (OR 5.14; 95% CI = 1.20-21.99; P = 0.03). Consistent with this finding, individuals with the CA (72194C/-2440A) haplotype had an increased risk of inefficient erythropoiesis (OR = 1.90; 95% CI = 1.10-3.30; P = 0.02), whereas TA haplotype carriers had marginal protection against inefficient erythropoiesis (OR = 0.24; 95% CI = 0.06-1.21; P = 0.05). These observations were supported by Cochran-Armitage trend test for inefficient erythropoiesis (CA > TA > CG; P < 0.01). Although none of the genotype and/or haplotypic variants were significantly associated with SMA, the direction of the risk profiles were consistent with the erythropoiesis results. CONCLUSION Taken together, variation in IL7 is associated with erythropoietic responses in children with falciparum malaria, a central physiological feature contributing to development of SMA.
Collapse
Affiliation(s)
- Lily E. Kisia
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
- University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Kisumu, Kenya
| | - Prakasha Kempaiah
- Department of Internal Medicine, Center for Global Health, University of New Mexico Health Sciences Center, Albuquerque, NM USA
| | - Samuel B. Anyona
- University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Kisumu, Kenya
| | - Elly O. Munde
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
- University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Kisumu, Kenya
| | - Angela O. Achieng
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
- University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Kisumu, Kenya
| | - John M. Ong’echa
- University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Kisumu, Kenya
| | - Christophe G. Lambert
- Department of Internal Medicine, Center for Global Health, University of New Mexico Health Sciences Center, Albuquerque, NM USA
| | - Kiprotich Chelimo
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - Collins Ouma
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
- University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Kisumu, Kenya
| | - Douglas J. Perkins
- University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Kisumu, Kenya
- Department of Internal Medicine, Center for Global Health, University of New Mexico Health Sciences Center, Albuquerque, NM USA
| | - Evans Raballah
- University of New Mexico/KEMRI Laboratories of Parasitic and Viral Diseases, Kisumu, Kenya
- Department of Medical Laboratory Sciences, School of Public Health, Biomedical Sciences and Technology, Masinde Muliro University of Science and Technology, P.O. Box 190-50100, Kakamega, Kenya
| |
Collapse
|
14
|
Achieng AO, Guyah B, Cheng Q, Ong'echa JM, Ouma C, Lambert CG, Perkins DJ. Molecular basis of reduced LAIR1 expression in childhood severe malarial anaemia: Implications for leukocyte inhibitory signalling. EBioMedicine 2019; 45:278-289. [PMID: 31257148 PMCID: PMC6642411 DOI: 10.1016/j.ebiom.2019.06.040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 06/18/2019] [Accepted: 06/20/2019] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Leukocyte-associated immunoglobulin like receptor-1 (LAIR1) is a transmembrane inhibitory receptor that influences susceptibility to a myriad of inflammatory diseases. Our recent investigations of severe malarial anaemia (SMA) pathogenesis in Kenyan children discovered that novel LAIR1 genetic variants which were associated with decreased LAIR1 transcripts enhanced the longitudinal risk of SMA and all-cause mortality. METHODS To characterize the molecular mechanism(s) responsible for altered LAIR1 signalling in severe malaria, we determined LAIR1 transcripts and protein, sLAIR1, sLAIR2, and complement component 1q (C1q) in children with malarial anaemia, followed by a series of in vitro experiments investigating the LAIR1 signalling cascade. FINDINGS Kenyan children with SMA had elevated circulating levels of soluble LAIR1 (sLAIR1) relative to non-SMA (1.69-fold P < .0001). The LAIR1 antagonist, sLAIR2, was also elevated in the circulation of children with SMA (1.59 fold-change, P < .0001). There was a positive correlation between sLAIR1 and sLAIR2 (ρ = 0.741, P < .0001). Conversely, circulating levels of complement component 1q (C1q), a LAIR1 natural ligand, were lower in SMA (-1.21-fold P = .048). These in vivo findings suggest that reduced membrane-bound LAIR1 expression in SMA is associated with elevated production of sLAIR1, sLAIR2 (antagonist), and limited C1q (agonist) availability. Since reduced LAIR1 transcripts in SMA were associated with increased acquisition of haemozoin (PfHz) by monocytes (P = .028), we explored the relationship between acquisition of intraleukocytic PfHz, LAIR1 expression, and subsequent impacts on leukocyte signalling in cultured PBMCs from malaria-naïve donors stimulated with physiological concentrations of PfHz (10 μg/mL). Phagocytosis of PfHz reduced LAIR1 transcript and protein expression in a time-dependent manner (P < .050), and inhibited LAIR1 signalling through decreased phosphorylation of LAIR1 (P < .0001) and SH2-domain containing phosphatase-1 (SHP-1) (P < .001). This process was associated with NF-κB activation (P < .0001) and enhanced production of IL-6, IL-1β, and TNF-α (all P < .0001). INTERPRETATION Collectively, these findings demonstrate that SMA is characterized by reduced LAIR1 transmembrane expression, reduced C1q, and enhanced production of sLAIR1 and sLAIR2, molecular events which can promote enhanced production of cytokines that contribute to the pathogenesis of SMA. These investigations are important for discovering immune checkpoints that could be future targets of immunotherapy to improve disease outcomes.
Collapse
Affiliation(s)
- Angela O Achieng
- University of New Mexico-Kenya Global Health Programs, Kisumu and Siaya, Kenya; Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - Bernard Guyah
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - Qiuying Cheng
- University of New Mexico, Center for Global Health, Department of Internal Medicine, NM, USA
| | - John M Ong'echa
- Kenya Medical Research Institute, Centre for Global Health Research, Kisumu, Kenya
| | - Collins Ouma
- University of New Mexico-Kenya Global Health Programs, Kisumu and Siaya, Kenya; Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | - Christophe G Lambert
- University of New Mexico, Center for Global Health, Department of Internal Medicine, NM, USA
| | - Douglas J Perkins
- University of New Mexico-Kenya Global Health Programs, Kisumu and Siaya, Kenya; University of New Mexico, Center for Global Health, Department of Internal Medicine, NM, USA.
| |
Collapse
|
15
|
Mohanty S, Singh US, Mohanty S, Mohanty AK, Pande V, Das A. Evolutionary interplay of single nucleotide polymorphisms at the promoter region of TNF-α gene in different clinical outcomes of malaria in India. INFECTION GENETICS AND EVOLUTION 2019; 69:107-116. [PMID: 30677532 DOI: 10.1016/j.meegid.2019.01.025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2018] [Revised: 01/16/2019] [Accepted: 01/21/2019] [Indexed: 01/17/2023]
Abstract
Host genetic factors are frequently ascribed to differential malaria outcomes as a by-product of evolutionary adaptation. To this respect, Tumor Necrosis factor alpha (TNF-α), a human cytokine, is known to be associated with malaria through its differential regulation in diverse malaria manifestations. Since diversity in differential malaria outcome is uncommon in every endemic settings, possible association of TNF-α and malaria is not commonly established. In order to check for association between the occurrence of Single Nucleotide Polymorphisms (SNPs) in the TNF-α gene with different malaria manifestations, we have sequenced a 4011 bp region constituting the promoter and the whole gene of human TNF-α in 61 patients [(16 cerebral plus severe (SCM), 21 severe (SM) and 24 uncomplicated (UM)] samples in a highly malaria endemic state (Odisha) of India. Multiple sequence alignment revealed presence of six SNPs (-1031 T > C, -863C > A, -857C > T, -308G > A, -806C > T, +787C > A), out of which the -806C > T and +787C > A are novel in malaria patients in general and the +787C > A was detected for the first time in humans. Although alleles due to six different SNPs segregate differentially in the three groups of malaria (SCM, SM and UM) in the present study, interestingly, for the -1031 T > C position, the frequency of individuals possessing the homozygous rare allele was higher in the SCM group with a higher number of heterozygotes in the UM group. The Tajima's D values considering all the SNPs in a defined group were positive and statistically insignificant conforming no evolutionary constraint. However, statistically significant deviation from expectation under Hardy-Weinberg equilibrium for -1031 T > C SNP in the UM group points towards the probable role of natural selection providing some kind of protection to malaria in Odisha, India.
Collapse
Affiliation(s)
- Stuti Mohanty
- Division of Vector Borne Diseases, ICMR-National Institute of Research in Tribal Health, Garha, Jabalpur, Madhya Pradesh, India
| | - Upasana Shyamsunder Singh
- Division of Vector Borne Diseases, ICMR-National Institute of Research in Tribal Health, Garha, Jabalpur, Madhya Pradesh, India; School of Earth and Environmental Sciences, The University of Manchester, Manchester M139PL, United Kingdom
| | - Sanjib Mohanty
- Community Welfare Society Hospital, Rourkela, Odisha, India
| | | | - Veena Pande
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, India
| | - Aparup Das
- Division of Vector Borne Diseases, ICMR-National Institute of Research in Tribal Health, Garha, Jabalpur, Madhya Pradesh, India.
| |
Collapse
|
16
|
Martínez-Laperche C, Buces E, Aguilera-Morillo MC, Picornell A, González-Rivera M, Lillo R, Santos N, Martín-Antonio B, Guillem V, Nieto JB, González M, de la Cámara R, Brunet S, Jiménez-Velasco A, Espigado I, Vallejo C, Sampol A, Bellón JM, Serrano D, Kwon M, Gayoso J, Balsalobre P, Urbano-Izpizua Á, Solano C, Gallardo D, Díez-Martín JL, Romo J, Buño I. A novel predictive approach for GVHD after allogeneic SCT based on clinical variables and cytokine gene polymorphisms. Blood Adv 2018; 2:1719-1737. [PMID: 30030270 PMCID: PMC6058238 DOI: 10.1182/bloodadvances.2017011502] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Accepted: 05/11/2018] [Indexed: 02/07/2023] Open
Abstract
Despite considerable advances in our understanding of the pathophysiology of graft-versus-host disease (GVHD), its prediction remains unresolved and depends mainly on clinical data. The aim of this study is to build a predictive model based on clinical variables and cytokine gene polymorphism for predicting acute GVHD (aGVHD) and chronic GVHD (cGVHD) from the analysis of a large cohort of HLA-identical sibling donor allogeneic stem cell transplant (allo-SCT) patients. A total of 25 SNPs in 12 cytokine genes were evaluated in 509 patients. Data were analyzed using a linear regression model and the least absolute shrinkage and selection operator (LASSO). The statistical model was constructed by randomly selecting 85% of cases (training set), and the predictive ability was confirmed based on the remaining 15% of cases (test set). Models including clinical and genetic variables (CG-M) predicted severe aGVHD significantly better than models including only clinical variables (C-M) or only genetic variables (G-M). For grades 3-4 aGVHD, the correct classification rates (CCR1) were: 100% for CG-M, 88% for G-M, and 50% for C-M. On the other hand, CG-M and G-M predicted extensive cGVHD better than C-M (CCR1: 80% vs. 66.7%, respectively). A risk score was calculated based on LASSO multivariate analyses. It was able to correctly stratify patients who developed grades 3-4 aGVHD (P < .001) and extensive cGVHD (P < .001). The novel predictive models proposed here improve the prediction of severe GVHD after allo-SCT. This approach could facilitate personalized risk-adapted clinical management of patients undergoing allo-SCT.
Collapse
Affiliation(s)
- Carolina Martínez-Laperche
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Elena Buces
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | | | - Antoni Picornell
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Oncology and
| | - Milagros González-Rivera
- Department of Oncology and
- DNA Sequencing and Genotyping Core Facility, H.G.U. Gregorio Marañón, Madrid, Spain
| | - Rosa Lillo
- Department of Statistics, Universidad Carlos III de Madrid, Madrid, Spain
| | - Nazly Santos
- Department of Hematology, Instituto Catalán de Oncología Hospital Josep Trueta, Girona, Spain
| | - Beatriz Martín-Antonio
- Department of Hematology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Vicent Guillem
- Department of Hematology, Hospital Clínico de Valencia, Valencia, Spain
| | - José B Nieto
- Department of Hematology, Hospital Universitario Morales Meseguer, Murcia, Spain
| | - Marcos González
- Department of Hematology, Hospital de Salamanca, Salamanca, Spain
| | - Rafael de la Cámara
- Department of Hematology, Hospital Universitario de La Princesa, Madrid, Spain
| | - Salut Brunet
- Department of Haematology, Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | | | - Ildefonso Espigado
- Department of Hematology, Hospital Universitario Virgen del Rocío, Seville, Spain
| | - Carlos Vallejo
- Department of Hematology, Hospital Central de Asturias, Oviedo, Spain
| | - Antonia Sampol
- Department of Hematology, Hospital Universitario Son Espases, Palma de Mallorca, Spain
| | - José María Bellón
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - David Serrano
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Mi Kwon
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Jorge Gayoso
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Pascual Balsalobre
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
| | - Álvaro Urbano-Izpizua
- Department of Hematology, Hospital Clínic, Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Carlos Solano
- Department of Hematology, Hospital Clínico de Valencia, Valencia, Spain
| | - David Gallardo
- Department of Hematology, Instituto Catalán de Oncología Hospital Josep Trueta, Girona, Spain
| | - José Luis Díez-Martín
- Department of Hematology, Hospital General Universitario (H.G.U.) Gregorio Marañón, Madrid, Spain
- Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain
- Department of Medicine, Facultad de Medicina, Universidad Complutense de Madrid, Madrid, Spain; and
| | - Juan Romo
- Department of Statistics, Universidad Carlos III de Madrid, Madrid, Spain
| | | |
Collapse
|
17
|
Wilson KD, Ochoa LF, Solomon OD, Pal R, Cardona SM, Carpio VH, Keiser PH, Cardona AE, Vargas G, Stephens R. Elimination of intravascular thrombi prevents early mortality and reduces gliosis in hyper-inflammatory experimental cerebral malaria. J Neuroinflammation 2018; 15:173. [PMID: 29866139 PMCID: PMC5987620 DOI: 10.1186/s12974-018-1207-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Accepted: 05/17/2018] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Cerebral malaria (CM) is the most lethal outcome of Plasmodium infection. There are clear correlations between expression of inflammatory cytokines, severe coagulopathies, and mortality in human CM. However, the mechanisms intertwining the coagulation and inflammation pathways, and their roles in CM, are only beginning to be understood. In mice with T cells deficient in the regulatory cytokine IL-10 (IL-10 KO), infection with Plasmodium chabaudi leads to a hyper-inflammatory response and lethal outcome that can be prevented by anti-TNF treatment. However, inflammatory T cells are adherent within the vasculature and not present in the brain parenchyma, suggesting a novel form of cerebral inflammation. We have previously documented behavioral dysfunction and microglial activation in infected IL-10 KO animals suggestive of neurological involvement driven by inflammation. In order to understand the relationship of intravascular inflammation to parenchymal dysfunction, we studied the congestion of vessels with leukocytes and fibrin(ogen) and the relationship of glial cell activation to congested vessels in the brains of P. chabaudi-infected IL-10 KO mice. METHODS Using immunofluorescence microscopy, we describe severe thrombotic congestion in these animals. We stained for immune cell surface markers (CD45, CD11b, CD4), fibrin(ogen), microglia (Iba-1), and astrocytes (GFAP) in the brain at the peak of behavioral symptoms. Finally, we investigated the roles of inflammatory cytokine tumor necrosis factor (TNF) and coagulation on the pathology observed using neutralizing antibodies and low-molecular weight heparin to inhibit both inflammation and coagulation, respectively. RESULTS Many blood vessels in the brain were congested with thrombi containing adherent leukocytes, including CD4 T cells and monocytes. Despite containment of the pathogen and leukocytes within the vasculature, activated microglia and astrocytes were prevalent in the parenchyma, particularly clustered near vessels with thrombi. Neutralization of TNF, or the coagulation cascade, significantly reduced both thrombus formation and gliosis in P. chabaudi-infected IL-10 KO mice. CONCLUSIONS These findings support the contribution of cytokines, coagulation, and leukocytes within the brain vasculature to neuropathology in malaria infection. Strikingly, localization of inflammatory leukocytes within intravascular clots suggests a mechanism for interaction between the two cascades by which cytokines drive local inflammation without considerable cellular infiltration into the brain parenchyma.
Collapse
Affiliation(s)
- Kyle D Wilson
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Lorenzo F Ochoa
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Olivia D Solomon
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Rahul Pal
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Sandra M Cardona
- Department of Biology, One UTSA Circle, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Victor H Carpio
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Philip H Keiser
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555-0435, USA
| | - Astrid E Cardona
- Department of Biology, One UTSA Circle, University of Texas at San Antonio, San Antonio, TX, 78249, USA
| | - Gracie Vargas
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA.,Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA
| | - Robin Stephens
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA. .,Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555-0435, USA. .,Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Boulevard, Galveston, TX, 77555, USA.
| |
Collapse
|
18
|
Potential Role for Regulatory B Cells as a Major Source of Interleukin-10 in Spleen from Plasmodium chabaudi-Infected Mice. Infect Immun 2018. [PMID: 29531131 DOI: 10.1128/iai.00016-18] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Interleukin-10 (IL-10)-producing regulatory B (Breg) cells were found to be induced in a variety of infectious diseases. However, its importance in the regulation of immune response to malaria is still unclear. Here, we investigated the dynamics, phenotype, and function of Breg cells using Plasmodium chabaudi chabaudi AS-infected C57BL/6 and BALB/c mice. BALB/c mice were more susceptible to infection and had a stronger IL-10 response in spleen than C57BL/6 mice. Analysis of the surface markers of IL-10-producing cells with flow cytometry showed that CD19+ B cells were one of the primary IL-10-producing populations in P. c. chabaudi AS-infected C57BL/6 and BALB/c mice, especially in the latter one. The Breg cells had a heterogeneous phenotype which shifted during infection. The well-established Breg subset, CD19+ CD5+ CD1dhi cells, accounted for less than 20% of IL-10-producing B cells in both strains during the course of infection. Most Breg cells were IgG+ and CD138- from day 0 to day 8 postinfection. Adoptive transfer of Breg cells to C57BL/6 mice infected with P. c. chabaudi AS led to a transient increase of parasitemia without an impact on survival rate. Our finding reveals that B cells play an active and important regulatory role in addition to mediating humoral immunity in immune response against malaria, which should be paid more attention in developing therapeutic or vaccine strategies against malaria involving stimulation of B cells.
Collapse
|
19
|
Guo C, Wen L, Song JK, Zeng WJ, Dan C, Niu YM, Shen M. Significant association between interleukin-10 gene polymorphisms and cervical cancer risk: a meta-analysis. Oncotarget 2018; 9:12365-12375. [PMID: 29552317 PMCID: PMC5844753 DOI: 10.18632/oncotarget.24193] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 12/04/2017] [Indexed: 12/18/2022] Open
Abstract
Previous studies have suggested that interleukin-10 (IL-10) polymorphisms may be associated with an increased risk of developing cervical cancer. However, the published results on this subject matter are controversial. The aim of this study was to conduct a meta-analysis of published reports to more precisely investigate the relationship between IL-10 polymorphisms and cervical cancer risk. Five online databases (PubMed, Embase, Web of SCI, CNKI and Wanfang) were searched, and seventeen articles with sufficient quantitative information were included in our meta-analysis. The odds ratios (ORs) and 95% confidence intervals (CIs) were calculated to assess the association between IL-10 polymorphisms and cervical cancer risk. Publication bias, sensitivity and cumulative analyses were also performed to support our findings. Overall, there was a significant association between the IL-10 -1082A > G polymorphism and cervical cancer risk observed in the total population (G vs. A: OR = 1.60, 95% CI = 1.12–2.29, P = 0.01, I2 = 92.3%; AG vs. AA: OR = 1.34, 95% CI = 1.04-1.74, P = 0.03, I2 = 65.9%; AG + GG vs. AA: OR = 1.58, 95% CI = 1.11–2.25, P = 0.01, I2 = 84.4%), and the same results were obtained in the subgroup analysis. Moreover, the IL-10 -819 T > C polymorphism exhibited a significant, protective effect against cervical cancer. In summary, our meta-analysis suggests that IL-10 polymorphisms may play a variety of roles in regard to cervical cancer risk, especially in Asians.
Collapse
Affiliation(s)
- Chong Guo
- Center for Evidence-Based Medicine and Clinical Research, Department of Gynecology and Obstetrics, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Li Wen
- Department of Dermatology, Suizhou Central Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Ju-Kun Song
- Department of Oral and Maxillary Surgery, Guizhou Provincial People's Hospital, Guiyang 550002, China
| | - Weng-Jing Zeng
- Department of Anesthesiology, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Chao Dan
- Department of Urinary Surgery, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Yu-Ming Niu
- Center for Evidence-Based Medicine and Clinical Research, Department of Gynecology and Obstetrics, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China.,Center for Evidence-Based Medicine and Clinical Research, Taihe Hospital, Hubei University of Medicine, Shiyan 442000, China
| | - Ming Shen
- Jiangsu Key Laboratory of Oral Diseases, Department of Dental Implant, Affiliated Hospital of Stomatology, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
20
|
Adedoja A, Hoan NX, van Tong H, Adukpo S, Tijani DB, Akanbi AA, Meyer CG, Ojurongbe O, Velavan TP. Differential contribution of interleukin-10 promoter variants in malaria and schistosomiasis mono- and co-infections among Nigerian children. Trop Med Int Health 2017; 23:45-52. [PMID: 29131459 DOI: 10.1111/tmi.13007] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
OBJECTIVE Interleukin-10 (IL-10) is an anti-inflammatory cytokine produced by Th1 cells and macrophages. The rationale of this study was to examine and validate possible contributions of IL-10 promoter polymorphisms in sub-Saharan Africa in children infected with either Plasmodium falciparum or Schistosoma haematobium and in children co-infected with both parasites. MATERIALS AND METHODS A total of 309 Nigerian children aged 4-15 years were recruited. The study group consisted of individuals infected either with P. falciparum (n = 76) or S. haematobium (n = 94) in mono-infections, a group of children co-infected with both P. falciparum and S. haematobium (n = 62) and matched healthy controls (n = 77). The IL-10 promoter polymorphisms -1082G/A, -819C/T and -592C/A were genotyped by direct sequencing. RESULTS The frequencies of the IL-10 -1082GG genotype, the -1082G allele and haplotype GCC (positions -1082, -819 and -592) were higher in children infected with P. falciparum than in healthy controls, indicating that the -1082GG genotype and the -1082G allele and the GCC haplotype are associated with increased susceptibility to malaria infection (OR = 3.4, 95% CI = 1.2-10.8, P = 0.02; OR = 2.5, 95% CI = 1.1-3.4, P = 0.02; OR = 3.8, 95% CI = 2.0-7.2, P = 0.0001, respectively). Children with the -1082GG genotype had a higher parasitaemia than children with the -1082AA or -1082AG genotypes (P = 0.0017). Haplotype GCC occurred more frequently in children infected with S. haematobium, while haplotype GTA was less frequent than in controls (OR = 2.2, 95% CI = 1.2-4.4, P = 0.017 and OR = 0.1, 95% CI = 0.02-0.5, P = 0.0004, respectively). No differences in the frequencies of IL-10 promoter polymorphisms were observed between children with P. falciparum-S. haematobium co-infections and healthy controls. CONCLUSION Although IL-10 promoter polymorphisms are not associated with P. falciparum and S. haematobium co-infection, variant -1082G/A and haplotype GCC are associated with malaria, whereas the IL-10 haplotypes GCC and GTA are associated with schistosomiasis.
Collapse
Affiliation(s)
- Ayodele Adedoja
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,Department of Medical Microbiology and Parasitology, Ladoke Akintola University of Technology, Osogbo, Nigeria.,Department of Medical Microbiology and Parasitology, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Nghiem Xuan Hoan
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Hoang van Tong
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Selorme Adukpo
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany
| | - Deborah B Tijani
- Department of Medical Microbiology and Parasitology, Ladoke Akintola University of Technology, Osogbo, Nigeria.,Department of Medical Microbiology and Parasitology, University of Ilorin Teaching Hospital, Ilorin, Nigeria
| | - Ajibola A Akanbi
- Department of Medical Microbiology and Parasitology, University of Ilorin, Ilorin, Nigeria
| | - Christian G Meyer
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,Duy Tan University, Da Nang, Vietnam.,Vietnamese-German Centre for Excellence in Medical Research, Hanoi, Vietnam
| | - Olusola Ojurongbe
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,Department of Medical Microbiology and Parasitology, Ladoke Akintola University of Technology, Osogbo, Nigeria
| | - Thirumalaisamy P Velavan
- Institute of Tropical Medicine, University of Tübingen, Tübingen, Germany.,Duy Tan University, Da Nang, Vietnam.,Vietnamese-German Centre for Excellence in Medical Research, Hanoi, Vietnam
| |
Collapse
|
21
|
How gene polymorphisms can influence clinical response and toxicity following R-CHOP therapy in patients with diffuse large B cell lymphoma. Blood Rev 2017; 31:235-249. [DOI: 10.1016/j.blre.2017.02.005] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2016] [Revised: 12/07/2016] [Accepted: 02/03/2017] [Indexed: 12/20/2022]
|
22
|
Winchester D, Till C, Goodman PJ, Tangen CM, Santella RM, Johnson-Pais TL, Leach RJ, Xu J, Zheng SL, Thompson IM, Lucia MS, Lippman SM, Parnes HL, Isaacs WB, De Marzo AM, Drake CG, Platz EA. Association between variants in genes involved in the immune response and prostate cancer risk in men randomized to the finasteride arm in the Prostate Cancer Prevention Trial. Prostate 2017; 77:908-919. [PMID: 28317149 PMCID: PMC5400704 DOI: 10.1002/pros.23346] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Accepted: 02/22/2017] [Indexed: 11/07/2022]
Abstract
BACKGROUND We reported that some, but not all single nucleotide polymorphisms (SNPs) in select immune response genes are associated with prostate cancer, but not individually with the prevalence of intraprostatic inflammation in the Prostate Cancer Prevention Trial (PCPT) placebo arm. Here, we investigated whether these same SNPs are associated with risk of lower- and higher-grade prostate cancer in men randomized to finasteride, and with prevalence of intraprostatic inflammation among controls. Methods A total of 16 candidate SNPs in IL1β, IL2, IL4, IL6, IL8, IL10, IL12(p40), IFNG, MSR1, RNASEL, TLR4, and TNFA and 7 tagSNPs in IL10 were genotyped in 625 white prostate cancer cases, and 532 white controls negative for cancer on an end-of-study biopsy nested in the PCPT finasteride arm. We used logistic regression to estimate log-additive odds ratios (OR) and 95% confidence intervals (CI) adjusting for age and family history. RESULTS Minor alleles of rs2243250 (T) in IL4 (OR = 1.46, 95% CI 1.03-2.08, P-trend = 0.03), rs1800896 (G) in IL10 (OR = 0.77, 95% CI 0.61-0.96, P-trend = 0.02), rs2430561 (A) in IFNG (OR = 1.33, 95% CI 1.02-1.74; P-trend = 0.04), rs3747531 (C) in MSR1 (OR = 0.55, 95% CI 0.32-0.95; P-trend = 0.03), and possibly rs4073 (A) in IL8 (OR = 0.81, 95% CI 0.64-1.01, P-trend = 0.06) were associated with higher- (Gleason 7-10; N = 222), but not lower- (Gleason 2-6; N = 380) grade prostate cancer. In men with low PSA (<2 ng/mL), these higher-grade disease associations were attenuated and/or no longer significant, whereas associations with higher-grade disease were apparent for minor alleles of rs1800795 (C: OR = 0.70, 95% CI 0.51-0.94, P-trend = 0.02) and rs1800797 (A: OR = 0.72, 95% CI 0.53-0.98, P-trend = 0.04) in IL6. While some IL10 tagSNPs were associated with lower- and higher-grade prostate cancer, distributions of IL10 haplotypes did not differ, except possibly between higher-grade cases and controls among those with low PSA (P = 0.07). We did not observe an association between the studied SNPs and intraprostatic inflammation in the controls. CONCLUSION In the PCPT finasteride arm, variation in genes involved in the immune response, including possibly IL8 and IL10 as in the placebo arm, may be associated with prostate cancer, especially higher-grade disease, but not with intraprostatic inflammation. We cannot rule out PSA-associated detection bias or chance due to multiple testing.
Collapse
Affiliation(s)
- Danyelle Winchester
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
| | - Cathee Till
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Phyllis J. Goodman
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Catherine M. Tangen
- SWOG Statistical Center, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Regina M. Santella
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY
| | - Teresa L. Johnson-Pais
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Robin J. Leach
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - Jianfeng Xu
- Program for Personalized Cancer Care and Department of Surgery, NorthShore University Health System, Evanston, IL
| | - S. Lilly Zheng
- Program for Personalized Cancer Care and Department of Surgery, NorthShore University Health System, Evanston, IL
- Center for Cancer Genomics, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Ian M. Thompson
- Department of Urology, University of Texas Health Science Center at San Antonio, San Antonio, TX
| | - M. Scott Lucia
- Department of Pathology, University of Colorado Denver School of Medicine, Aurora, CO
| | - Scott M. Lippman
- Moores Cancer Center, University of California San Diego, La Jolla, CA
| | - Howard L. Parnes
- Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Department of Health and Human Services, Bethesda, MD
| | - William B. Isaacs
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| | - Angelo M. De Marzo
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Charles G. Drake
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
- Department of Immunology, Johns Hopkins University School of Medicine, Baltimore, MD
- Department of Oncology, Columbia University, New York, NY
| | - Elizabeth A. Platz
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD
- James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD
| |
Collapse
|
23
|
Mijac D, Petrovic IV, Djuranovic S, Perovic V, Bojic D, Culafic D, Popovic D, Krstic M, Jankovic G, Djoric M, Pravica V, Markovic M. The Polymorphism rs3024505 (C/T) Downstream of the IL10 Gene Is Associated with Crohn's Disease in Serbian Patients with Inflammatory Bowel Disease. TOHOKU J EXP MED 2017; 240:15-24. [PMID: 27558476 DOI: 10.1620/tjem.240.15] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2022]
Abstract
Inflammatory bowel disease (IBD), manifesting as Crohn's disease (CD) and ulcerative colitis (UC), is characterized by recurring episodes of inflammation in gastrointestinal tract, in which aberrant production of regulatory cytokine interleukin-10 (IL-10) presumably plays important role. Single nucleotide polymorphisms (SNPs) that affect IL-10 production, such as rs1800896 (G/A) at position -1082 and rs1800871 (C/T) at position -819 in the promoter region of the IL10 gene, have been associated with CD and/or UC, but the results were inconsistent. Another SNP that may alter IL-10 production, rs3024505 (C/T) located immediately downstream of the IL10 gene has been recently identified. T allele of rs3024505 was associated with both UC and CD in Western populations, but the studies from East European countries are lacking. Therefore, our aim was to assess the association of rs3024505, rs1800896 and rs1800871 with Serbian IBD patients. To this end, 107 CD and 99 UC patients and 255 healthy controls were genotyped. As a result, T allele of rs3024505 was associated with CD at allelic, genotypic (GT genotype) and haplotypic (GCCT haplotype) level, suggesting potential role of this variant in susceptibility to CD. In contrast, CD patients carrying C allele of rs3024505 had significantly increased risk of anemia and stricturing/penetrating behavior. No association was observed between rs3024505 and UC or SNPs in IL10 promoter region and any form of IBD. In conclusion, rs3024505 SNP flanking the IL10 gene is associated with susceptibility and severity of disease in Serbian CD patients, further validating its role as a potential biomarker in IBD.
Collapse
Affiliation(s)
- Dragana Mijac
- Clinic for Gastroenterology and Hepatology, Clinical Center of Serbia, School of Medicine, University of Belgrade
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Frequency of TNFA, INFG, and IL10 Gene Polymorphisms and Their Association with Malaria Vivax and Genomic Ancestry. Mediators Inflamm 2016; 2016:5168363. [PMID: 27999453 PMCID: PMC5143728 DOI: 10.1155/2016/5168363] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 07/14/2016] [Accepted: 09/27/2016] [Indexed: 02/08/2023] Open
Abstract
Polymorphisms in cytokine genes can alter the production of these proteins and consequently affect the immune response. The trihybrid heterogeneity of the Brazilian population is characterized as a condition for the use of ancestry informative markers. The objective of this study was to evaluate the frequency of -1031T>C, -308G>A and -238G>A TNFA, +874 A>T IFNG and -819C>T, and -592C>A IL10 gene polymorphisms and their association with malaria vivax and genomic ancestry. Samples from 90 vivax malaria-infected individuals and 51 noninfected individuals from northern Brazil were evaluated. Genotyping was carried out by using ASO-PCR or PCR/RFLP. The genomic ancestry of the individuals was classified using 48 insertion/deletion polymorphism biallelic markers. There were no differences in the proportions of African, European, and Native American ancestry between men and women. No significant association was observed for the allele and genotype frequencies of the 6 SNPs between malaria-infected and noninfected individuals. However, there was a trend toward decreasing the frequency of individuals carrying the TNF-308A allele with the increasing proportion of European ancestry. No ethnic-specific SNPs were identified, and there was no allelic or genotype association with susceptibility or resistance to vivax malaria. Understanding the genomic mechanisms by which ancestry influences this association is critical and requires further study.
Collapse
|
25
|
Wilson KD, Stutz SJ, Ochoa LF, Valbuena GA, Cravens PD, Dineley KT, Vargas G, Stephens R. Behavioural and neurological symptoms accompanied by cellular neuroinflammation in IL-10-deficient mice infected with Plasmodium chabaudi. Malar J 2016; 15:428. [PMID: 27557867 PMCID: PMC4995805 DOI: 10.1186/s12936-016-1477-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 08/10/2016] [Indexed: 11/10/2022] Open
Abstract
Background Cerebral malaria is one of the most severe complications of Plasmodium falciparum infection and occurs mostly in young African children. This syndrome results from a combination of high levels of parasitaemia and inflammation. Although parasite sequestration in the brain is a feature of the human syndrome, sequestering strains do not uniformly cause severe malaria, suggesting interplay with other factors. Host genetic factors such as mutations in the promoters of the cytokines IL-10 and TNF are also clearly linked to severe disease. Plasmodium chabaudi, a rodent malaria parasite, leads to mild illness in wildtype animals. However, IL-10−/− mice respond to parasite with increased levels of pro-inflammatory cytokines IFN-γ and TNF, leading to lethal disease in the absence of sequestration in the brain. These mice also exhibit cerebral symptoms including gross cerebral oedema and haemorrhage, allowing study of these critical features of disease without the influence of sequestration. Methods The neurological consequences of P. chabaudi infection were investigated by performing a general behavioural screen (SHIRPA). The immune cell populations found in the brain during infection were also analysed using flow cytometry and confocal microscopy. Results IL-10−/− mice suffer significant declines in behavioural and physical capacities during infection compared to wildtype. In addition, grip strength and pain sensitivity were affected, suggestive of neurological involvement. Several immune cell populations were identified in the perfused brain on day 7 post-infection, suggesting that they are tightly adherent to the vascular endothelium, or potentially located within the brain parenchyma. There was an increase in both inflammatory monocyte and resident macrophage (CD11bhi, CD45+, MHCII+, Ly6C+/−) numbers in IL-10−/− compared to wildtype animals. In addition, the activation state of all monocytes and microglia (CD11bint, CD45−, MHC-II+) were increased. T cells making IFN-γ were also identified in the brain, but were localized within the vasculature, and not the parenchyma. Conclusions These studies demonstrate exacerbated neuroinflammation concurrent with development of behavioural symptoms in P. chabaudi infection of IL-10−/− animals. Electronic supplementary material The online version of this article (doi:10.1186/s12936-016-1477-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Kyle D Wilson
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Sonja J Stutz
- Mitchell Center for Neurodegenerative Diseases, Center for Addiction Research, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Lorenzo F Ochoa
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Gustavo A Valbuena
- Department of Pathology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Petra D Cravens
- Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Kelly T Dineley
- Mitchell Center for Neurodegenerative Diseases, Center for Addiction Research, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.,Department of Neurology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Gracie Vargas
- Center for Biomedical Engineering, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.,Department of Neuroscience and Cell Biology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.,Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA
| | - Robin Stephens
- Department of Microbiology and Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA. .,Department of Internal Medicine, Division of Infectious Diseases, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA. .,Institute for Human Infections and Immunity, University of Texas Medical Branch, 301 University Blvd., Galveston, TX, 77555, USA.
| |
Collapse
|
26
|
Furini AAC, Capobianco MP, Storti-Melo LM, Cunha MG, Cassiano GC, Machado RLD. Cytokine gene polymorphisms are not associated with anti-PvDBP, anti-PvAMA-1 or anti-PvMSP-119 IgG antibody levels in a malaria-endemic area of the Brazilian Amazon. Malar J 2016; 15:374. [PMID: 27435973 PMCID: PMC4952271 DOI: 10.1186/s12936-016-1414-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2016] [Accepted: 07/04/2016] [Indexed: 11/18/2022] Open
Abstract
Background The immune response against Plasmodium vivax immunogenic epitopes is regulated by pro- and anti-inflammatory cytokines that determine antibody levels and class switching. Cytokine gene polymorphisms may be responsible for changes in the humoral immune response against malaria. The aim of this study was to evaluate whether polymorphisms in the TNFA, IFNG and IL10 genes would alter the levels of anti-PvAMA1, PvDBP and -PvMSP-119 IgG antibodies in patients with vivax malaria. Methods Samples from 90 vivax malaria-infected and 51 uninfected subjects from an endemic area of the Brazilian Amazon were genotyped using polymerase chain reaction-restriction fragment length polymorphism (PCR–RFLP) to identify polymorphisms of the genes TNFA (−1031T > C, −308G > A, −238G > A), IFNG (+874T > A) and IL10 (−819C > T, −592C > A). The levels of total IgG against PvAMA1, PvDBP and PvMSP-119 were determined using an enzyme-linked immunosorbent assay (ELISA). Associations between the polymorphisms and the antibody response were assessed by means of logistic regression models. Results No significant differences were found in the levels of IgG antibodies against the PvAMA-1, PvDBP or PvMSP-119 proteins in relation to the studied polymorphisms. Conclusions Although no associations were found among the evaluated genotypes and alleles and anti-merozoite IgG class P. vivax antibody levels, this study helps elucidate the immunogenic profile involved in the humoral immune response in malaria. Electronic supplementary material The online version of this article (doi:10.1186/s12936-016-1414-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Adriana A C Furini
- Department of Dermatology, Infectious and Parasitic Diseases, São José do Rio Preto Medical School, São José do Rio Preto, São Paulo, Brazil.
| | - Marcela P Capobianco
- Department of Biology, São Paulo State University, São José do Rio Preto, São Paulo, Brazil
| | - Luciane M Storti-Melo
- Laboratory of Molecular Genetics and Biotechnology, Department of Biology, Federal University of Sergipe, São Cristóvão, SE, Brazil
| | - Maristela G Cunha
- Laboratory of Microbiology and Immunology, Institute of Biological Sciences, Federal University of Pará (Universidade Federal do Pará-UFPA), Belém, State of Pará (PA), Brazil
| | - Gustavo C Cassiano
- Laboratory of Tropical Diseases-Department of Genetics, Evolution and Bioagents, Universidade de Campinas, Campinas, SP, Brazil
| | - Ricardo Luiz D Machado
- Department of Dermatology, Infectious and Parasitic Diseases, São José do Rio Preto Medical School, São José do Rio Preto, São Paulo, Brazil.,Department of Biology, São Paulo State University, São José do Rio Preto, São Paulo, Brazil.,Laboratory of Basic Research in Malaria, Section of Parasitology, Evandro Chagas Institute, Belém, PA, Brazil
| |
Collapse
|
27
|
Reduced Parasite Burden in Children with Falciparum Malaria and Bacteremia Coinfections: Role of Mediators of Inflammation. Mediators Inflamm 2016; 2016:4286576. [PMID: 27418744 PMCID: PMC4933845 DOI: 10.1155/2016/4286576] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 04/28/2016] [Indexed: 01/07/2023] Open
Abstract
Bacteremia and malaria coinfection is a common and life-threatening condition in children residing in sub-Saharan Africa. We previously showed that coinfection with Gram negative (G[-]) enteric Bacilli and Plasmodium falciparum (Pf[+]) was associated with reduced high-density parasitemia (HDP, >10,000 parasites/μL), enhanced respiratory distress, and severe anemia. Since inflammatory mediators are largely unexplored in such coinfections, circulating cytokines were determined in four groups of children (n = 206, aged <3 yrs): healthy; Pf[+] alone; G[-] coinfected; and G[+] coinfected. Staphylococcus aureus and non-Typhi Salmonella were the most frequently isolated G[+] and G[-] organisms, respectively. Coinfected children, particularly those with G[-] pathogens, had lower parasite burden (peripheral and geometric mean parasitemia and HDP). In addition, both coinfected groups had increased IL-4, IL-5, IL-7, IL-12, IL-15, IL-17, IFN-γ, and IFN-α and decreased TNF-α relative to malaria alone. Children with G[-] coinfection had higher IL-1β and IL-1Ra and lower IL-10 than the Pf[+] group and higher IFN-γ than the G[+] group. To determine how the immune response to malaria regulates parasitemia, cytokine production was investigated with a multiple mediation model. Cytokines with the greatest mediational impact on parasitemia were IL-4, IL-10, IL-12, and IFN-γ. Results here suggest that enhanced immune activation, especially in G[-] coinfected children, acts to reduce malaria parasite burden.
Collapse
|
28
|
Lima-Junior JDC, Pratt-Riccio LR. Major Histocompatibility Complex and Malaria: Focus on Plasmodium vivax Infection. Front Immunol 2016; 7:13. [PMID: 26858717 PMCID: PMC4728299 DOI: 10.3389/fimmu.2016.00013] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 01/12/2016] [Indexed: 01/13/2023] Open
Abstract
The importance of host and parasite genetic factors in malaria resistance or susceptibility has been investigated since the middle of the last century. Nowadays, of all diseases that affect man, malaria still plays one of the highest levels of selective pressure on human genome. Susceptibility to malaria depends on exposure profile, epidemiological characteristics, and several components of the innate and adaptive immune system that influences the quality of the immune response generated during the Plasmodium lifecycle in the vertebrate host. But it is well known that the parasite's enormous capacity of genetic variation in conjunction with the host genetics polymorphism is also associated with a wide spectrum of susceptibility degrees to complicated or severe forms of the disease. In this scenario, variations in genes of the major histocompatibility complex (MHC) associated with host resistance or susceptibility to malaria have been identified and used as markers in host-pathogen interaction studies, mainly those evaluating the impact on the immune response, acquisition of resistance, or increased susceptibility to infection or vulnerability to disease. However, due to the intense selective pressure, number of cases, and mortality rates, the majority of the reported associations reported concerned Plasmodium falciparum malaria. Studies on the MHC polymorphism and its association with Plasmodium vivax, which is the most widespread Plasmodium and the most prevalent species outside the African continent, are less frequent but equally important. Despite punctual contributions, there are accumulated evidences of human genetic control in P. vivax infection and disease. Herein, we review the current knowledge in the field of MHC and derived molecules (HLA Class I, Class II, TNF-α, LTA, BAT1, and CTL4) regarding P. vivax malaria. We discuss particularly the results of P. vivax studies on HLA class I and II polymorphisms in relation to host susceptibility, naturally acquired immune response against specific antigens and the implication of this knowledge to overcome the parasite immune evasion. Finally, the potential impact of such polymorphisms on the development of vaccine candidate antigens against P. vivax will be studied.
Collapse
|
29
|
Xu X, Yang J, Li N, Wu R, Tian H, Song H, Wang H. Role of Endothelial Progenitor Cell Transplantation in Rats With Sepsis. Transplant Proc 2015; 47:2991-3001. [DOI: 10.1016/j.transproceed.2015.10.024] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Accepted: 10/07/2015] [Indexed: 12/11/2022]
|
30
|
Parasite-Specific CD4+ IFN-γ+ IL-10+ T Cells Distribute within Both Lymphoid and Nonlymphoid Compartments and Are Controlled Systemically by Interleukin-27 and ICOS during Blood-Stage Malaria Infection. Infect Immun 2015; 84:34-46. [PMID: 26459508 PMCID: PMC4693994 DOI: 10.1128/iai.01100-15] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/04/2015] [Indexed: 01/18/2023] Open
Abstract
Immune-mediated pathology in interleukin-10 (IL-10)-deficient mice during blood-stage malaria infection typically manifests in nonlymphoid organs, such as the liver and lung. Thus, it is critical to define the cellular sources of IL-10 in these sensitive nonlymphoid compartments during infection. Moreover, it is important to determine if IL-10 production is controlled through conserved or disparate molecular programs in distinct anatomical locations during malaria infection, as this may enable spatiotemporal tuning of the regulatory immune response. In this study, using dual gamma interferon (IFN-γ)–yellow fluorescent protein (YFP) and IL-10–green fluorescent protein (GFP) reporter mice, we show that CD4+ YFP+ T cells are the major source of IL-10 in both lymphoid and nonlymphoid compartments throughout the course of blood-stage Plasmodium yoelii infection. Mature splenic CD4+ YFP+ GFP+ T cells, which preferentially expressed high levels of CCR5, were capable of migrating to and seeding the nonlymphoid tissues, indicating that the systemically distributed host-protective cells have a common developmental history. Despite exhibiting comparable phenotypes, CD4+ YFP+ GFP+ T cells from the liver and lung produced significantly larger quantities of IL-10 than their splenic counterparts, showing that the CD4+ YFP+ GFP+ T cells exert graded functions in distinct tissue locations during infection. Unexpectedly, given the unique environmental conditions within discrete nonlymphoid and lymphoid organs, we show that IL-10 production by CD4+ YFP+ T cells is controlled systemically during malaria infection through IL-27 receptor signaling that is supported after CD4+ T cell priming by ICOS signaling. The results in this study substantially improve our understanding of the systemic IL-10 response to malaria infection, particularly within sensitive nonlymphoid organs.
Collapse
|
31
|
RAHMAH Z, SASMITO SD, SISWANTO B, SARDJONO TW, FITRI LE. Parasitemia Induces High Plasma Levels of Interleukin-17 (IL-17) and Low Levels of Interleukin-10 (IL-10) and Transforming Growth Factor-ß (TGF-ß) in Pregnant Mice Infected with Malaria. Malays J Med Sci 2015; 22:25-32. [PMID: 26715893 PMCID: PMC4681718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 04/11/2015] [Indexed: 06/05/2023] Open
Abstract
BACKGROUND During pregnancy, the balanced dominance of the T helper17 response shifts to a Th2 response that is characterised by the production of IL-10, following the completion of the implantation process. Transforming growth factor-β (TGF-β) expression is associated with the completion of trophoblast invasion and placental growth. This study assessed the effect of malaria infection on the levels of IL-17, IL-10, and TGF-β in the plasma of pregnant mice with malaria. METHODS Seventeen pregnant BALB/C mice were divided into two groups: mice infected with Plasmodium berghei (treatment group) and uninfected mice (control group). The mice were sacrificed on day 18 post-mating. Parasitemia was measured by Giemsa staining. The levels of IL-17, IL-10, and TGF-β were measured by ELISA. RESULTS Using independent t test, the IL-17 levels in the treatment group were higher than those in the control group (= = 0.040). The IL-10 levels in the treatment group were lower than those in the control group (= = 0.00). There was no significant difference in the TGF-β levels (= = 0.055) between two groups. However, using SEM analysis the degree of parasitemia decreased the plasma TGF-β levels (tcount = 5.148; ≥ ttable = 1.96). SEM analysis showed that a high degree of parasitemia increased the IL-17 levels and decreased the IL-10 and TGF-β levels. CONCLUSION Malaria infection during pregnancy interferes with the systemic balance by increasing the IL-17 levels and decreasing the IL-10 and TGF-β levels.
Collapse
Affiliation(s)
- Zainabur RAHMAH
- Doctoral Program in Medical Science, Faculty of Medicine, Universitas Brawijaya, Jalan Veteran Malang, East Java 65145, Indonesia
| | - Sujarot Dwi SASMITO
- Master Program in Biomedical Sciences, Faculty of Medicine, Universitas Brawijaya, Jalan Veteran Malang, East Java 65145, Indonesia
| | - Budi SISWANTO
- Department of Obstetric and Gynecology, Faculty of Medicine Universitas Brawijaya, Jalan Jaksa Agung Suprapto, No. 2 Malang, East Java, 65122, Indonesia
| | - Teguh Wahju SARDJONO
- Department of Parasitology Faculty of Medicine Universitas Brawijaya, Jalan Veteran Malang, East Java, 65145, Indonesia
| | - Loeki Enggar FITRI
- Department of Parasitology Faculty of Medicine Universitas Brawijaya, Jalan Veteran Malang, East Java, 65145, Indonesia
| |
Collapse
|
32
|
Pereira VA, Sánchez-Arcila JC, Teva A, Perce-da-Silva DS, Vasconcelos MPA, Lima CAM, Aprígio CJL, Rodrigues-da-Silva RN, Santos DO, Banic DM, Bonecini-Almeida MG, Lima-Júnior JC, Oliveira-Ferreira J. IL10A genotypic association with decreased IL-10 circulating levels in malaria infected individuals from endemic area of the Brazilian Amazon. Malar J 2015; 14:30. [PMID: 25627396 PMCID: PMC4334410 DOI: 10.1186/s12936-015-0548-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2014] [Accepted: 01/06/2015] [Indexed: 12/25/2022] Open
Abstract
Background Cytokines play an important role in human immune responses to malaria and variation in their production may influence the course of infection and determine the outcome of the disease. The differential production of cytokines has been linked to single nucleotide polymorphisms in gene promoter regions, signal sequences, and gene introns. Although some polymorphisms play significant roles in susceptibility to malaria, gene polymorphism studies in Brazil are scarce. Methods A population of 267 individuals from Brazilian Amazon exposed to malaria was genotyped for five single nucleotide polymorphisms (SNPs), IFNG + 874 T/A, IL10A-1082G/A, IL10A-592A/C, IL10A-819 T/C and NOS2A-954G/C. Specific DNA fragments were amplified by polymerase chain reaction, allowing the detection of the polymorphism genotypes. The polymorphisms IL10A-592A/C and IL10A-819 T/C were estimated by a single analysis due to the complete linkage disequilibrium between the two SNPs with D’ = 0.99. Plasma was used to measure the levels of IFN-γ and IL-10 cytokines by Luminex and nitrogen radicals by Griess reaction. Results No differences were observed in genotype and allelic frequency of IFNG + 874 T/A and NOS2A-954G/C between positive and negative subjects for malaria infection. Interesting, the genotype NOS2A-954C/C was not identified in the study population. Significant differences were found in IL10A-592A/C and IL10A-819 T/C genotypes distribution, carriers of IL10A -592A/-819 T alleles (genotypes AA/TT + AC/TC) were more frequent among subjects with malaria than in negative subjects that presented a higher frequency of the variant C allele (p < 0.0001). The presence of the allele C was associated with low producer of IL-10 and low parasitaemia. In addition, the GTA haplotypes formed from combinations of investigated polymorphisms in IL10A were significantly associated with malaria (+) and the CCA haplotype with malaria (−) groups. The IL10A-1082G/A polymorphism showed high frequency of heterozygous AG genotype in the population, but it was not possible to infer any association of the polymorphism because their distribution was not in Hardy Weinberg equilibrium. Conclusion This study shows that the IL10A-592A/C and IL10A-819 T/C polymorphisms were associated with malaria and decreased IL-10 levels and low parasite density suggesting that this polymorphism influence IL-10 levels and may influence in the susceptibility to clinical malaria.
Collapse
Affiliation(s)
- Virginia A Pereira
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz/Fiocruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro, Brazil.
| | - Juan C Sánchez-Arcila
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz/Fiocruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro, Brazil.
| | - Antonio Teva
- Laboratório de Imunodiagnóstico /Departamento de Ciências Biológicas, Escola Nacional de Saúde Pública/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Daiana S Perce-da-Silva
- Laboratório de Simulídeos e Oncocercose, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Brazil.
| | | | - Cleoni A M Lima
- Centro Interdepartamental de Biologia Experimental e Biotecnologia, Universidade Federal de Rondônia, Porto Velho, Rondônia, Brazil.
| | - Cesarino J L Aprígio
- Laboratório de Quimioterapia/Fiocruz, Porto Velho, Rondônia, Brazil and Universidade Federal de Rondônia, Porto Velho, Rondônia, Brazil.
| | - Rodrigo N Rodrigues-da-Silva
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz/Fiocruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro, Brazil.
| | - Davi O Santos
- Laboratório de Imunologia e Imunogenética, Instituto de Pesquisa Clínica Evandro Chagas (IPEC)/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Dalma M Banic
- Laboratório de Simulídeos e Oncocercose, Instituto Oswaldo Cruz/Fiocruz, Rio de Janeiro, Brazil.
| | - Maria G Bonecini-Almeida
- Laboratório de Imunologia e Imunogenética, Instituto de Pesquisa Clínica Evandro Chagas (IPEC)/Fiocruz, Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Josué C Lima-Júnior
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz/Fiocruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro, Brazil.
| | - Joseli Oliveira-Ferreira
- Laboratório de Imunoparasitologia, Instituto Oswaldo Cruz/Fiocruz, Av. Brasil 4365, Manguinhos, Rio de Janeiro, Brazil.
| |
Collapse
|
33
|
Oduor CI, Chelimo K, Ouma C, Mulama DH, Foley J, Vulule J, Bailey JA, Moormann AM. Interleukin-6 and interleukin-10 gene promoter polymorphisms and risk of endemic Burkitt lymphoma. Am J Trop Med Hyg 2014; 91:649-54. [PMID: 25071000 DOI: 10.4269/ajtmh.13-0616] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Overexpression of interleukin-6 (IL-6) and IL-10 in endemic Burkitt lymphoma (eBL) may facilitate tumorigenesis by providing a permissive cytokine milieu. Promoter polymorphisms influence interindividual differences in cytokine production. We hypothesized that children genetically predisposed for elevated cytokine levels may be more susceptible to eBL. Using case-control samples from western Kenya consisting of 117 eBL cases and 88 ethnically matched healthy controls, we tested for the association between eBL risk and IL-10 (rs1800896, rs1800871, and rs1800872) and IL-6 (rs1800795) promoter single nucleotide polymorphisms (SNPs) as well as IL-10 promoter haplotypes. In addition, the association between these variants and Epstein Barr Virus (EBV) load was examined. Results showed that selected IL-10 and IL-6 promoter SNPs and IL-10 promoter haplotypes were not associated with risk eBL or EBV levels in EBV-seropositive children. Findings from this study reveal that common variants within the IL-10 and IL-6 promoters do not independently increase eBL risk in this vulnerable population.
Collapse
Affiliation(s)
- Cliff I Oduor
- Department of Biomedical Sciences and Technology, Maseno University, Maseno, Kenya; Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Medicine, Division of Transfusion, and Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Kiprotich Chelimo
- Department of Biomedical Sciences and Technology, Maseno University, Maseno, Kenya; Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Medicine, Division of Transfusion, and Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Collins Ouma
- Department of Biomedical Sciences and Technology, Maseno University, Maseno, Kenya; Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Medicine, Division of Transfusion, and Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts
| | - David H Mulama
- Department of Biomedical Sciences and Technology, Maseno University, Maseno, Kenya; Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Medicine, Division of Transfusion, and Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Joslyn Foley
- Department of Biomedical Sciences and Technology, Maseno University, Maseno, Kenya; Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Medicine, Division of Transfusion, and Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts
| | - John Vulule
- Department of Biomedical Sciences and Technology, Maseno University, Maseno, Kenya; Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Medicine, Division of Transfusion, and Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Jeffrey A Bailey
- Department of Biomedical Sciences and Technology, Maseno University, Maseno, Kenya; Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Medicine, Division of Transfusion, and Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Ann M Moormann
- Department of Biomedical Sciences and Technology, Maseno University, Maseno, Kenya; Center for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya; Department of Pediatrics, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Medicine, Division of Transfusion, and Program in Bioinformatics and Integrative Biology, University of Massachusetts Medical School, Worcester, Massachusetts; Department of Quantitative Health Sciences, University of Massachusetts Medical School, Worcester, Massachusetts
| |
Collapse
|
34
|
Apinjoh TO, Anchang-Kimbi JK, Njua-Yafi C, Ngwai AN, Mugri RN, Clark TG, Rockett KA, Kwiatkowski DP, Achidi EA. Association of candidate gene polymorphisms and TGF-beta/IL-10 levels with malaria in three regions of Cameroon: a case-control study. Malar J 2014; 13:236. [PMID: 24934404 PMCID: PMC4077225 DOI: 10.1186/1475-2875-13-236] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2014] [Accepted: 06/07/2014] [Indexed: 11/11/2022] Open
Abstract
BACKGROUND Plasmodium falciparum malaria is one of the most widespread and deadliest infectious diseases in children under five years in endemic areas. The disease has been a strong force for evolutionary selection in the human genome, and uncovering the critical host genetic factors that confer resistance to the disease would provide clues to the molecular basis of protective immunity and improve vaccine development initiatives. METHODS The effect of single nucleotide polymorphisms (SNPs) and plasma transforming growth factor beta (TGF-β) and interleukin 10 (IL-10) levels on malaria pathology was investigated in a case-control study of 1862 individuals from two major ethnic groups in three regions with intense perennial P. falciparum transmission in Cameroon. Thirty-four malaria candidate polymorphisms, including the sickle cell trait (HbS), were assayed on the Sequenom iPLEX platform while plasma TGF-β and IL-10 levels were measured by sandwich ELISA. RESULTS The study confirms the known protective effect of HbS against severe malaria and also reveals a protective effect of SNPs in the nitrogen oxide synthase 2 (NOS2) gene against malaria infection, anaemia and uncomplicated malaria. Furthermore, ADCY9 rs10775349 (additive G) and ABO rs8176746 AC individuals were associated with protection from hyperpyrexia and hyperparasitaemia, respectively. Meanwhile, individuals with the EMR1 rs373533 GT, EMR1 rs461645 CT and RTN3 rs542998 (additive C) genotypes were more susceptible to hyperpyrexia while both females and males with the rs1050828 and rs1050829 SNPs of G6PD, respectively, were more vulnerable to anaemia. Plasma TGF-β levels were strongly correlated with heterozygosity for the ADCY9 rs2230739 and HBB rs334 SNPs while individuals with the ABO rs8176746 AC genotype had lower IL-10 levels. CONCLUSION Taken together, this study suggests that some rare polymorphisms in candidate genes may have important implications for the susceptibility of Cameroonians to severe malaria. Moreover using the uncomplicated malaria phenotype may permit the identification of novel pathways in the early development of the disease.
Collapse
Affiliation(s)
- Tobias O Apinjoh
- Department of Biochemistry and Molecular Biology, University of Buea, Buea, Cameroon
| | | | - Clarisse Njua-Yafi
- Department of Animal Biology and Physiology, University of Yaounde I, Yaounde, Cameroon
| | - André N Ngwai
- Department of Medical Laboratory Science, University of Buea, Buea, Cameroon
| | - Regina N Mugri
- Department of Medical Laboratory Science, University of Buea, Buea, Cameroon
| | - Taane G Clark
- London School of Hygiene and Tropical Medicine, London, UK
| | - Kirk A Rockett
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Eric A Achidi
- Department of Biochemistry and Molecular Biology, University of Buea, Buea, Cameroon
- Department of Medical Laboratory Science, University of Buea, Buea, Cameroon
| |
Collapse
|
35
|
Gonçalves RM, Lima NF, Ferreira MU. Parasite virulence, co-infections and cytokine balance in malaria. Pathog Glob Health 2014; 108:173-8. [PMID: 24854175 DOI: 10.1179/2047773214y.0000000139] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Strong early inflammatory responses followed by a timely production of regulatory cytokines are required to control malaria parasite multiplication without inducing major host pathology. Here, we briefly examine the homeostasis of inflammatory responses to malaria parasite species with varying virulence levels and discuss how co-infections with bacteria, viruses, and helminths can modulate inflammation, either aggravating or alleviating malaria-related morbidity.
Collapse
|
36
|
Qaddourah RH, Magdoud K, Saldanha FL, Mahmood N, Mustafa FE, Mahjoub T, Almawi WY. IL-10 gene promoter and intron polymorphisms and changes in IL-10 secretion in women with idiopathic recurrent miscarriage. Hum Reprod 2014; 29:1025-34. [PMID: 24626804 DOI: 10.1093/humrep/deu043] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
STUDY QUESTION Is recurrent pregnancy loss (RPL) associated with polymorphisms in the promoter and intron regions of the interleukin-10 (IL-10) gene? SUMMARY ANSWER IL-10 rs1518111 was found to be associated with RPL but the commonly studied promoter variants rs1800872, rs1800871 and 1800896 were not. WHAT IS KNOWN ALREADY Reduced expression of IL-10 is implicated in RPL, due to defective maternal immune tolerance (causing early miscarriages) or placental vascular insufficiency (causing late losses). IL-10 production is in part inherited, and IL-10 gene variants associated with reduced IL-10 expression have been analyzed for their association with RPL, often with inconclusive results. STUDY DESIGN, SIZE, DURATION A retrospective case-control study was performed between January 2011 and April 2012. The subjects comprised 296 RPL cases and 305 control women. PARTICIPANTS/MATERIALS, SETTING, METHODS Genotyping of the IL-10 intron (rs1878672, rs3024492, rs1554286, rs1518111, rs3024491, rs3024490) and promoter (rs1800872, rs1800871, rs1800896) variants was done by real-time PCR, with defined clusters. MAIN RESULTS AND THE ROLE OF CHANCE A higher minor allele frequency (MAF) of rs1518111 (P = 0.03) was in seen RPL cases; but the MAFs of the remaining SNPs were comparable between cases and controls. Setting the homozygous major allele genotype (1/1) as the reference, significantly higher frequencies of heterozygous rs1554286 and rs1800872, and homozygous rs1800896 genotype carriers, and a reduced frequency of homozygous rs1518111 genotype carriers, were seen in RPL cases, while the distribution of the remaining genotypes were comparable between cases and controls. Serum IL-10 levels were significantly reduced in RPL cases compared with control women (P = 0.002), and this correlated with rs1518111 and rs1800871 genotypes in both groups, and with the rs1800872 genotype among control women. A nine-locus (rs1878672, rs3024492, rs1554286, rs1518111, rs3024491, rs3024490, rs1800872, rs1800871 and rs1800896) haploview analysis demonstrated an increased frequency of haplotype 112112121 in RPL cases, thus conferring a disease susceptibility nature to this haplotype. LIMITATIONS, REASONS FOR CAUTION The main limitation of this study was that it was limited to Bahraini Arabs, thereby necessitating parallel studies of other ethnic groups. Another limitation is the study design, which prompts speculation on whether it is a cause-effect relationship. WIDER IMPLICATIONS OF THE FINDINGS While the lack of association of the various IL-10 promoter variants with RPL was in agreement with reports from varied ethnic groups, this is the first study to confirm the association between IL-10 rs151811 intronic variant and RPL. STUDY FUNDING/COMPETING INTEREST(S) The study was funded by grants from the Arabian Gulf University Research Fund. None of the authors report any competing interests.
Collapse
Affiliation(s)
- R H Qaddourah
- Department of Medical Biochemistry, College of Medicine and Medical Sciences, Arabian Gulf University, Manama, Bahrain
| | | | | | | | | | | | | |
Collapse
|
37
|
Borges QI, Fontes CJF, Damazo AS. Analysis of lymphocytes in patients with Plasmodium vivax malaria and its relation to the annexin-A1 and IL-10. Malar J 2013; 12:455. [PMID: 24359168 PMCID: PMC3878186 DOI: 10.1186/1475-2875-12-455] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2013] [Accepted: 11/22/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Malaria is the most prevalent parasitic disease in the world. In Brazil, the largest number of malaria cases (98%) is within the Legal Amazon region, where Plasmodium vivax is responsible for over 80% of diagnosed cases. The aim of this study was to investigate the annexin-A1 expression in CD4+, CD8+ T cells, regulatory T cells (Treg) and cytokine IL-10 quantification in plasma from patients with malaria caused by P. vivax. METHODS The quantification of the cytokine IL-10 of patients infected with P. vivax and healthy controls were evaluated by enzyme-linked immunosorbent assay (ELISA). The determination of the expression of annexin-A1 in lymphocytes from patients and healthy controls was determined by immunofluorescence staining. All results were correlated with the parasitaemia and the number of previous episodes of malaria. RESULTS The cytokine IL-10 plasma levels showed a significant increase in both patients with low (650.4 ± 59.3 pg/mL) and high (2870 ± 185.3 pg/mL) parasitaemia compared to the control (326.1 ± 40.1 pg/mL). In addition, there was an increase of this cytokine in an episode dependent manner (individuals with no previous episodes of malaria--primoinfected: 363.9 ± 31.1 pg/mL; individuals with prior exposure: 659.9 ± 49.4 pg/mL). The quantification of annexin-A1 expression indicated a decrease in CD4+ and CD8+ T cells and an increase in Treg in comparison with the control group. When annexin-A1 expression was compared according to the number of previous episodes of malaria, patients who have been exposed more than once to the parasite was found to have higher levels of CD4+ T cells (96.0 ± 2.5 A.U) compared to primoinfected (50.3 ± 1.7). However, this endogenous protein had higher levels in CD8+ (108.5 ± 3.1) and Treg (87.5 ± 2.5) from patients primoinfected. CONCLUSION This study demonstrates that in the patients infected with P. vivax the release of immunoregulatory molecules can be influenced by the parasitaemia level and the number of previous episodes of malaria. annexin-A1 is expressed differently in lymphocyte sub-populations and may have a role in cell proliferation. Furthermore, annexin-A1 may be contributing to IL-10 release in plasma of patients with vivax malaria.
Collapse
Affiliation(s)
| | | | - Amílcar S Damazo
- Post-graduation in Health Science, Faculty of Medicine (FM), Federal University of Mato Grosso (UFMT), Cuiabá, Mato Grosso 78060-900, Brazil.
| |
Collapse
|
38
|
The association of IL-8-251T/A polymorphism with complicated malaria in Karbi Anglong district of Assam. Cytokine 2013; 65:210-6. [PMID: 24290435 DOI: 10.1016/j.cyto.2013.11.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 10/09/2013] [Accepted: 11/01/2013] [Indexed: 01/21/2023]
Abstract
Amongst host genetic factors, cytokine gene polymorphism can be anticipated to be an important factor as qualitative, quantitative and time of secretion play an important role in disease outcome. We have investigated association of cytokine promoter SNPs with risk of Plasmodium falciparum malaria and disease severity in a case control study in malaria endemic Karbi Anglong district of Assam, India. Frequency of IL-8-251T/A (p=0.03 and p=0.01) and TGF-β1-509C/T (p=0.02 and p=0.03) was higher in malaria in comparison to control participants and non-malarial fever controls. Interestingly, a higher frequency of mutant allele of IL-10-819T/C was observed in non-malarial fever controls compared to malaria thus suggesting its role as a distinguishing marker of the two disease groups. Higher IL-8 expression and increased frequency of IL-8-251T/A in complicated malaria (p=0.002) was reported indicating its role in susceptibility to complicated malaria. In conclusion, our study suggests the role of mutant genotype of IL-8-251T/A as a marker of complicated malaria in our population. Surprisingly, decreased expression of TGF-β1 in uncomplicated malaria even in presence of high expressing mutant genotype was observed and needs to be investigated in context of the pool of activated cells producing the cytokine.
Collapse
|
39
|
Apinjoh TO, Anchang-Kimbi JK, Njua-Yafi C, Mugri RN, Ngwai AN, Rockett KA, Mbunwe E, Besingi RN, Clark TG, Kwiatkowski DP, Achidi EA, in collaboration with The MalariaGEN Consortium. Association of cytokine and Toll-like receptor gene polymorphisms with severe malaria in three regions of Cameroon. PLoS One 2013; 8:e81071. [PMID: 24312262 PMCID: PMC3842328 DOI: 10.1371/journal.pone.0081071] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 10/08/2013] [Indexed: 11/23/2022] Open
Abstract
P. falciparum malaria is one of the most widespread and deadliest infectious diseases in children under five years in endemic areas. The disease has been a strong force for evolutionary selection in the human genome, and uncovering the critical human genetic factors that confer resistance to the disease would provide clues to the molecular basis of protective immunity that would be invaluable for vaccine development. We investigated the effect of single nucleotide polymorphisms (SNPs) on malaria pathology in a case- control study of 1862 individuals from two major ethnic groups in three regions with intense perennial P. falciparum transmission in Cameroon. Twenty nine polymorphisms in cytokine and toll-like receptor (TLR) genes as well as the sickle cell trait (HbS) were assayed on the Sequenom iPLEX platform. Our results confirm the known protective effect of HbS against severe malaria and also reveal a protective effect of SNPs in interleukin-10 (IL10) cerebral malaria and hyperpyrexia. Furthermore, IL17RE rs708567 GA and hHbS rs334 AT individuals were associated with protection from uncomplicated malaria and anaemia respectively in this study. Meanwhile, individuals with the hHbS rs334 TT, IL10 rs3024500 AA, and IL17RD rs6780995 GA genotypes were more susceptible to severe malarial anaemia, cerebral malaria, and hyperpyrexia respectively. Taken together, our results suggest that polymorphisms in some immune response genes may have important implications for the susceptibility to severe malaria in Cameroonians. Moreover using uncomplicated malaria may allow us to identify novel pathways in the early development of the disease.
Collapse
Affiliation(s)
- Tobias O. Apinjoh
- Department of Biochemistry and Molecular Biology, University of Buea, Buea, Cameroon
- * E-mail:
| | | | - Clarisse Njua-Yafi
- Department of Animal Biology and Physiology, University of Yaounde I, Yaounde, Cameroon
| | - Regina N. Mugri
- Department of Medical Laboratory Sciences, University of Buea, Buea, Cameroon
| | - Andre N. Ngwai
- Department of Medical Laboratory Sciences, University of Buea, Buea, Cameroon
| | - Kirk A. Rockett
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Eric Mbunwe
- Department of Medical Laboratory Sciences, University of Buea, Buea, Cameroon
- Diabetes Research Center, Brussels Free University, Brussels, Belgium
| | - Richard N. Besingi
- Department of Biochemistry and Molecular Biology, University of Buea, Buea, Cameroon
- Department of Oral Biology, University of Florida, Gainesville, Florida, United States of America
| | - Taane G. Clark
- London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Dominic P. Kwiatkowski
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
- Wellcome Trust Sanger Institute, Hinxton, United Kingdom
| | - Eric A. Achidi
- Department of Biochemistry and Molecular Biology, University of Buea, Buea, Cameroon
- Department of Medical Laboratory Sciences, University of Buea, Buea, Cameroon
| | | |
Collapse
|
40
|
Butler NS, Harris TH, Blader IJ. Regulation of immunopathogenesis during Plasmodium and Toxoplasma infections: more parallels than distinctions? Trends Parasitol 2013; 29:593-602. [PMID: 24184186 DOI: 10.1016/j.pt.2013.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2013] [Revised: 10/03/2013] [Accepted: 10/04/2013] [Indexed: 01/08/2023]
Abstract
Toxoplasma and Plasmodium parasites exact a significant toll on public health. Host immunity required for efficient control of infection by these Apicomplexans involves the induction of potent T cell responses, which sometimes results in immunopathological damage. Thus, protective immune responses must be balanced by regulatory networks that limit immunopathology. We review several key cellular and molecular immunoregulatory networks operational during Toxoplasma and Plasmodium infections. Accumulating data show that despite differences in how the immune response controls these parasites, many host immunoregulatory pathways and cellular networks are common to both. Thus, understanding the cellular and molecular circuits that prevent or regulate immunopathological responses against one parasite is likely to inform our understanding of the host response to the other parasite.
Collapse
Affiliation(s)
- Noah S Butler
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA.
| | | | | |
Collapse
|
41
|
Lokossou AG, Dechavanne C, Bouraïma A, Courtin D, Le Port A, Ladékpo R, Noukpo J, Bonou D, Ahouangninou C, Sabbagh A, Fayomi B, Massougbodji A, Garcia A, Migot-Nabias F. Association of IL-4 and IL-10 maternal haplotypes with immune responses to P. falciparum in mothers and newborns. BMC Infect Dis 2013; 13:215. [PMID: 23668806 PMCID: PMC3679728 DOI: 10.1186/1471-2334-13-215] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 05/03/2013] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Particular cytokine gene polymorphisms are involved in the regulation of the antibody production. The consequences of already described IL-4, IL-10 and IL-13 gene polymorphisms on biological parameters and antibody levels were investigated among 576 mothers at delivery and their newborns in the context of P. falciparum placental malaria infection. METHODS The study took place in the semi-rural area of Tori-Bossito, in south-west Benin, where malaria is meso-endemic. Six biallelic polymorphisms were determined by quantitative PCR using TaqMan® Pre-Designed SNP Genotyping Assays, in IL-4 (rs2243250, rs2070874), IL-10 (rs1800896, rs1800871, rs1800872) and IL-13 (rs1800925) genes. Antibody responses directed to P. falciparum MSP-1, MSP-2, MSP-3, GLURP-R0, GLURP-R2 and AMA-1 recombinant proteins were determined by ELISA. RESULTS The maternal IL-4(-590)*T/IL-4(+33)*T haplotype (one or two copies) was associated with favorable maternal condition at delivery (high haemoglobin levels, absence of placental parasites) and one of its component, the IL-4(-590)TT genotype, was related to low IgG levels to MSP-1, MSP-2/3D7 and MSP-2/FC27. Inversely, the maternal IL-10(-1082)AA was positively associated with P. falciparum placenta infection at delivery. As a consequence, the IL-10(-819)*T allele (in CT and TT genotypes) as well as the IL-10(-1082)*A/IL-10(-819)*T/IL-10(-592)*A haplotype (one or two copies) in which it is included, were related to an increased risk for anaemia in newborns. The maternal IL-10(-1082)AA genotype was related to high IgG levels to MSP-2/3D7 and AMA-1 in mothers and newborns, respectively. The IL-13 gene polymorphism was only involved in the newborn's antibody response to AMA-1. CONCLUSION These data revealed that IL-4 and IL-10 maternal gene polymorphisms are likely to play a role in the regulation of biological parameters in pregnant women at delivery (anaemia, P. falciparum placenta infection) and in newborns (anaemia). Moreover, IL-4, IL-10 and IL-13 maternal gene polymorphisms were related to IgG responses to MSP-1, MSP-2/3D7 and MSP-2/FC27 in mothers as well as to AMA-1 in newborns.
Collapse
Affiliation(s)
- Adjimon Gatien Lokossou
- Institut de Recherche pour le Développement, UMR 216 Mère et enfant face aux infections tropicales, Paris, France.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Okeyo WA, Munde EO, Okumu W, Raballah E, Anyona SB, Vulule JM, Ong'echa JM, Perkins DJ, Ouma C. Interleukin (IL)-13 promoter polymorphisms (-7402 T/G and -4729G/A) condition susceptibility to pediatric severe malarial anemia but not circulating IL-13 levels. BMC Immunol 2013; 14:15. [PMID: 23521898 PMCID: PMC3618200 DOI: 10.1186/1471-2172-14-15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 03/18/2013] [Indexed: 02/07/2023] Open
Abstract
In holoendemic Plasmodium falciparum transmission areas such as western Kenya, severe malarial anemia [SMA, hemoglobin (Hb) < 6.0 g/dL, with any density parasitemia] is the most common clinical manifestation of severe malaria resulting in high rates of pediatric morbidity and mortality in these regions. Previous studies associated interleukin (IL)-13 with pathogenesis of different infectious diseases, including P. falciparum malaria. However, the functional roles of polymorphic variants within the IL-13 promoter in conditioning susceptibility to SMA remain largely unexplored. As such, the association between the IL-13 variants -7402 T/G (rs7719175) and -4729G/A (rs3091307) and susceptibility to SMA was determined in children (n = 387) presenting with clinical symptoms of falciparum malaria and resident in a holoendemic transmission region in western Kenya. Our results indicated no difference in the proportions of individual genotypes among children presenting with non-SMA (n = 222) versus SMA (n = 165). Similarly, there was no associations between the individual genotypes (-7402 T/G and -4729G/A) and SMA. Additional analyses, however, revealed that proportions of individuals with -7402 T/-4729A (TA) haplotype was significantly higher in children presenting with SMA than non-SMA group (P = 0.043). A further multivariate logistic regression analyses, controlling for confounding factors, demonstrated that carriage of the TA haplotype was associated with increased susceptibility to SMA (OR; 1.564, 95% CI; 1.023-2.389, P = 0.039). In addition, circulating levels of IL-13 were comparable between the clinical groups as well as across genotypes and haplotypes. Collectively, findings presented here suggest that haplotypes within the IL-13 promoter at -7402 T/G and -4729G/A may modulate SMA pathogenesis, but do not affect circulating IL-13 levels.
Collapse
Affiliation(s)
- Winnie A Okeyo
- Department of Biomedical Sciences and Technology, School of Public Health and Community Development, Maseno University, Maseno, Kenya
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Zakeri S, Mashhadi R, Mehrizi AA, Djadid ND. Analysis of Fcgamma receptor IIa (cd32) gene polymorphism and anti-malarial IgG subclass antibodies to asexual blood-stage antigen of Plasmodium falciparum in an unstable malaria endemic area of Iran. Exp Parasitol 2013; 134:115-21. [PMID: 23458236 DOI: 10.1016/j.exppara.2013.02.012] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2012] [Revised: 02/09/2013] [Accepted: 02/19/2013] [Indexed: 11/19/2022]
Abstract
One of the main host genetic factors involved in inflammation, immune responses and pathogenesis of malaria is FcγRIIa (cd32) gene. A single point mutation at position 131 replace an arginine (R) with a histidine (H) that can affect the affinity of the receptor for human IgG subclasses. This investigation was designed to explore the polymorphisms at FcγRIIa gene in association with both anti-malarial total IgG antibody and IgG subclass profiles to C-terminal region of Plasmodium falciparum merozoite surface protein 1 (PfMSP-1(19)). In this study, 166 infected patients with P. falciparum who are living in a malaria endemic area of Iran were studied using PCR-RFLP and ELISA methods. The results showed that the frequency of FcγRIIa-R/R131, -R/H131 and -H/H131 genotypes was 9.6%, 42.8% and 47.6%, respectively. Level of total IgG to recombinant PfMSP-1(19) antigen showed that there was no difference among the FcγRIIa-R/R131, -R/H131 and -H/H131 groups. With regards to the IgG subclasses, the anti-malarial IgG1 antibodies predominated. Also, there was a significant difference between the frequency of positive responders for anti-PfMSP-1(19) IgG and IgG1 antibodies in P. falciparum-infected individuals with FcγRIIa-R/R131, -R/H131 or -H/H131 genotypes (P<0.05, X(2) test). Regarding to IgG2-PfMSP-1(19) antibody, 27.27% (FcγRIIa-R/R131), 25.71% (FcγRIIa-R/H131) and 22.2% (FcγRIIa-H/H131) of IgG responders showed positive antibody response. Taken together, this study is the first report that exhibits the high frequency of both FcγRIIa-H131H genotypes and H131 allele in the Baluchi ethnic group, which was similar to the Fulani ethnic group. The present results provide additional data to understand the role of FcγRIIa-131 genotypes in the pathogenesis of malaria.
Collapse
Affiliation(s)
- Sedigheh Zakeri
- Malaria and Vector Research Group, Biotechnology Research Center, Pasteur Institute of Iran, P.O. Box 1316943551, Tehran, Iran.
| | | | | | | |
Collapse
|
44
|
The host genetic diversity in malaria infection. J Trop Med 2012; 2012:940616. [PMID: 23316245 PMCID: PMC3532872 DOI: 10.1155/2012/940616] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Revised: 11/06/2012] [Accepted: 11/19/2012] [Indexed: 02/06/2023] Open
Abstract
Populations exposed to Plasmodium infection develop genetic mechanisms of protection against severe disease. The clinical manifestation of malaria results primarily from the lysis of infected erythrocytes and subsequent immune and inflammatory responses. Herein, we review the genetic alterations associated with erythrocytes or mediators of the immune system, which might influence malaria outcome. Moreover, polymorphisms in genes related to molecules involved in mechanisms of cytoadherence and their influence on malaria pathology are also discussed. The results of some studies have suggested that the combinatorial effects of a set of genetic factors in the erythrocyte-immunology pathway might be relevant to host resistance or susceptibility against Plasmodium infection. However, these results must be interpreted with caution because of the differences observed in the functionality and frequency of polymorphisms within different populations. With the recent advances in molecular biology techniques, more robust studies with reliable data have been reported, and the results of these studies have identified individual genetic factors for consideration in preventing severe disease and the individual response to treatment.
Collapse
|
45
|
Manjurano A, Clark TG, Nadjm B, Mtove G, Wangai H, Sepulveda N, Campino SG, Maxwell C, Olomi R, Rockett KR, Jeffreys A, MalariaGen Consortium, Riley EM, Reyburn H, Drakeley C. Candidate human genetic polymorphisms and severe malaria in a Tanzanian population. PLoS One 2012; 7:e47463. [PMID: 23144702 PMCID: PMC3483265 DOI: 10.1371/journal.pone.0047463] [Citation(s) in RCA: 34] [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: 07/23/2012] [Accepted: 09/12/2012] [Indexed: 11/18/2022] Open
Abstract
Human genetic background strongly influences susceptibility to malaria infection and progression to severe disease and death. Classical genetic studies identified haemoglobinopathies and erythrocyte-associated polymorphisms, as protective against severe disease. High throughput genotyping by mass spectrometry allows multiple single nucleotide polymorphisms (SNPs) to be examined simultaneously. We compared the prevalence of 65 human SNP's, previously associated with altered risk of malaria, between Tanzanian children with and without severe malaria. Five hundred children, aged 1–10 years, with severe malaria were recruited from those admitted to hospital in Muheza, Tanzania and compared with matched controls. Genotyping was performed by Sequenom MassArray, and conventional PCR was used to detect deletions in the alpha-thalassaemia gene. SNPs in two X-linked genes were associated with altered risk of severe malaria in females but not in males: heterozygosity for one or other of two SNPs in the G6PD gene was associated with protection from all forms of severe disease whilst two SNPs in the gene encoding CD40L were associated with respiratory distress. A SNP in the adenyl cyclase 9 (ADCY9) gene was associated with protection from acidosis whilst a polymorphism in the IL-1α gene (IL1A) was associated with an increased risk of acidosis. SNPs in the genes encoding IL-13 and reticulon-3 (RTN3) were associated with increased risk of cerebral malaria. This study confirms previously known genetic associations with protection from severe malaria (HbS, G6PD). It identifies two X-linked genes associated with altered risk of severe malaria in females, identifies mutations in ADCY9, IL1A and CD40L as being associated with altered risk of severe respiratory distress and acidosis, both of which are characterised by high serum lactate levels, and also identifies novel genetic associations with severe malaria (TRIM5) and cerebral malaria(IL-13 and RTN3). Further studies are required to test the generality of these associations and to understand their functional consequences.
Collapse
Affiliation(s)
- Alphaxard Manjurano
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Taane G. Clark
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Behzad Nadjm
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - George Mtove
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Hannah Wangai
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Nuno Sepulveda
- Faculty of Epidemiology and Population Health, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | | | - Caroline Maxwell
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Raimos Olomi
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
| | - Kirk R. Rockett
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | - Anna Jeffreys
- Wellcome Trust Centre for Human Genetics, University of Oxford, Oxford, United Kingdom
| | | | - Eleanor M. Riley
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Hugh Reyburn
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Christopher Drakeley
- Joint Malaria Programme, Kilimanjaro Christian Medical Centre, Moshi, Tanzania
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
- * E-mail:
| |
Collapse
|
46
|
Zhu H, Lei X, Liu Q, Wang Y. Interleukin-10-1082A/G polymorphism and inflammatory bowel disease susceptibility: a meta-analysis based on 17,585 subjects. Cytokine 2012; 61:146-53. [PMID: 23046617 DOI: 10.1016/j.cyto.2012.09.009] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2012] [Revised: 09/06/2012] [Accepted: 09/17/2012] [Indexed: 12/19/2022]
Abstract
A large number of studies have shown that the interleukin-10 (IL-10)-1082A/G polymorphism is implicated in susceptibility to inflammatory bowel disease (IBD). However, the results are inconsistent. We performed this meta-analysis to estimate the association between -1082A/G polymorphism in the IL-10 gene and IBD susceptibility. A total number of 18 case-control studies including 17,585 subjects were identified. No association was found between -1082A/G polymorphism and ulcerative colitis (UC) susceptibility. However, increased risk of Crohn's disease (CD) was associated with -1082A/G polymorphism in the dominant genetic model (GG+GA vs. AA: OR=1.22, 95% CI: 1.02-1.46, P=0.028) and the heterozygote comparison (GA vs. AA: OR=1.28, 95% CI: 1.05-1.55, P=0.015). The results of this meta-analysis provide evidence for the association between IL-10-1082A/G polymorphism and susceptibility of CD. Due to several limitations in the present study, well-designed epidemiological studies with large sample size among different ethnicities should be performed in the future.
Collapse
Affiliation(s)
- Hang Zhu
- Maternal and Child Hygiene Department, School of Public Health and Management, Chongqing Medical University, China
| | | | | | | |
Collapse
|
47
|
Davenport GC, Hittner JB, Were T, Ong'echa JM, Perkins DJ. Relationship between inflammatory mediator patterns and anemia in HIV-1 positive and exposed children with Plasmodium falciparum malaria. Am J Hematol 2012; 87:652-8. [PMID: 22570198 DOI: 10.1002/ajh.23200] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 03/01/2012] [Accepted: 03/06/2012] [Indexed: 12/27/2022]
Abstract
Anemia is the primary hematological manifestation of both Plasmodium falciparum malaria and HIV-1 in pediatric populations in sub-Saharan Africa. We have previously shown that HIV-1 positive and exposed children have greater risk of developing severe anemia (hemoglobin, Hb <6.0 g dL⁻¹) during acute malaria. However, enhanced severity of anemia was unrelated to either erythropoietic suppression or parasite-driven red blood cell hemolysis. To further explore mechanisms of anemia, circulating inflammatory mediators (IMs) were determined using a 25-plex bead array in P. falciparum-infected (Pf[+]) children (3-36 month, n = 194) stratified into three groups: HIV-1 negative (HIV-1[-]/Pf[+]); HIV-1 exposed (HIV-1[exp]/Pf[+]); and HIV-1 infected (HIV-1[+]/Pf[+]). IL-12, MIG/CXCL9, eotaxin/CCL11, and GM-CSF differed significantly and progressively increased across the groups (HIV-1[-]→HIV-1[exp]→HIV-1[+]). To further explore the relationship between the inflammatory milieu (i.e., cytokines, chemokines, and growth factors) and HIV-1 status, the large panel of IMs was reduced into discrete groups by principal component factor analysis. Of the six principal components that emerged, three components were significantly higher in the HIV-1 [+]/pf[+] and HIV[exp]/Pf[+] groups, demonstrating that inflammatory profiles differ according to HIV-1 status. Additional analyses exploring the relationship between the components and anemia revealed significant positive correlations between Hb and Component 3 (IL-1Ra, IL-7, IL-17, IFN-α, IFN-γ, MIG/CXCL9) in the HIV-1[-]/Pf[+] group, and Component 4 (IL-4, IL-5, IL-12, Eotaxin/CCL11) in HIV-1[+]/Pf[+] children. Further analyses of the HIV-1[+]/Pf[+] group revealed that IL-12 had the strongest association with anemia. Results presented here demonstrate that there are unique relationships between the inflammatory environment and anemia in HIV-1 positive and exposed children with malaria.
Collapse
Affiliation(s)
- Gregory C Davenport
- Center for Global Health, Department of Internal Medicine, University of New Mexico, Albuquerque, NM 87131-0001, USA
| | | | | | | | | |
Collapse
|
48
|
Investigation of host candidate malaria-associated risk/protective SNPs in a Brazilian Amazonian population. PLoS One 2012; 7:e36692. [PMID: 22615793 PMCID: PMC3353949 DOI: 10.1371/journal.pone.0036692] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2011] [Accepted: 04/05/2012] [Indexed: 02/02/2023] Open
Abstract
The Brazilian Amazon is a hypo-endemic malaria region with nearly 300,000 cases each year. A variety of genetic polymorphisms, particularly in erythrocyte receptors and immune response related genes, have been described to be associated with susceptibility and resistance to malaria. In order to identify polymorphisms that might be associated with malaria clinical outcomes in a Brazilian Amazonian population, sixty-four human single nucleotide polymorphisms in 37 genes were analyzed using a Sequenom massARRAY iPLEX platform. A total of 648 individuals from two malaria endemic areas were studied, including 535 malaria cases (113 individuals with clinical mild malaria, 122 individuals with asymptomatic infection and 300 individuals with history of previous mild malaria) and 113 health controls with no history of malaria. The data revealed significant associations (p<0.003) between one SNP in the IL10 gene (rs1800896) and one SNP in the TLR4 gene (rs4986790) with reduced risk for clinical malaria, one SNP in the IRF1 gene (rs2706384) with increased risk for clinical malaria, one SNP in the LTA gene (rs909253) with protection from clinical malaria and one SNP in the TNF gene (RS1800750) associated with susceptibility to clinical malaria. Also, a new association was found between a SNP in the CTL4 gene (rs2242665), located at the major histocompatibility complex III region, and reduced risk for clinical malaria. This study represents the first association study from an Amazonian population involving a large number of host genetic polymorphisms with susceptibility or resistance to Plasmodium infection and malaria outcomes. Further studies should include a larger number of individuals, refined parameters and a fine-scale map obtained through DNA sequencing to increase the knowledge of the Amazonian population genetic diversity.
Collapse
|
49
|
Interleukin-10 (IL-10) polymorphisms are associated with IL-10 production and clinical malaria in young children. Infect Immun 2012; 80:2316-22. [PMID: 22566507 DOI: 10.1128/iai.00261-12] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
The role of interleukin-10 (IL-10) in malaria remains poorly characterized. The aims of this study were to investigate (i) whether genetic variants of the IL-10 gene influence IL-10 production and (ii) whether IL-10 production as well as the genotypes and haplotypes of the IL-10 gene in young children and their mothers are associated with the incidence of clinical malaria in young children. We genotyped three IL-10 single nucleotide polymorphisms in 240 children and their mothers from a longitudinal prospective cohort and assessed the IL-10 production by maternal peripheral blood mononuclear cells (PBMCs) and cord blood mononuclear cells (CBMCs). Clinical episodes of Plasmodium falciparum malaria in the children were documented until the second year of life. The polymorphism IL-10 A-1082G (GCC haplotype of three SNPs in IL-10) in children was associated with IL-10 production levels by CBMC cultured with P. falciparum-infected erythrocytes (P = 0.043), with the G allele linked to low IL-10 production capacity. The G allele in children was also significantly associated with a decreased risk for clinical malaria infection in their second year of life (P = 0.016). Furthermore, IL-10 levels measured in maternal PBMCs cultured with infected erythrocytes were associated with increased risk of malaria infection in young children (P < 0.001). In conclusion, IL-10 polymorphisms and IL-10 production capacity were associated with clinical malaria infections in young children. High IL-10 production capacity inherited from parents may diminish immunological protection against P. falciparum infection, thereby being a risk for increased malaria morbidity.
Collapse
|
50
|
Freitas do Rosario AP, Langhorne J. T cell-derived IL-10 and its impact on the regulation of host responses during malaria. Int J Parasitol 2012; 42:549-55. [PMID: 22549022 DOI: 10.1016/j.ijpara.2012.03.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2011] [Revised: 03/02/2012] [Accepted: 03/24/2012] [Indexed: 02/04/2023]
Abstract
Despite intense research, malaria still is the one of the most devastating diseases killing more people than any other parasitic infection. In an attempt to control the infection, the host immune system produces a potent pro-inflammatory response. However, this response is also associated with complications, such as severe anaemia, hypoglycaemia and cerebral malaria. This pronounced production of pro-inflammatory cytokines response is a common feature of malaria caused by parasites infecting humans as well as rodents and primates. A balance between pro- and anti-inflammatory responses may be fundamental to the elimination of the parasite without inducing excessive host pathology. IL-10 is a key cytokine that has been shown to have an important regulatory function in establishing this balance in malaria. Here we discuss which cells can produce IL-10 during infection, and present an overview of the evidence showing that T-cell derived IL-10 plays an important role in regulating malaria pathology. Many different subsets of T cells can produce IL-10, however, evidence is accumulating that it is effector Th1 CD4(+) T cells which provide the crucial source that down-regulates inflammatory pathology during blood-stage malaria infections.
Collapse
|