1
|
Corral-Ruiz GM, Pérez-Vega MJ, Galán-Salinas A, Mancilla-Herrera I, Barrios-Payán J, Fabila-Castillo L, Hernández-Pando R, Sánchez-Torres LE. Thymic atrophy induced by Plasmodium berghei ANKA and Plasmodium yoelii 17XL infection. Immunol Lett 2023; 264:4-16. [PMID: 37875239 DOI: 10.1016/j.imlet.2023.10.006] [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: 04/05/2023] [Revised: 10/06/2023] [Accepted: 10/21/2023] [Indexed: 10/26/2023]
Abstract
The thymus is the anatomical site where T cells undergo a complex process of differentiation, proliferation, selection, and elimination of autorreactive cells which involves molecular signals in different intrathymic environment. However, the immunological functions of the thymus can be compromised upon exposure to different infections, affecting thymocyte populations. In this work, we investigated the impact of malaria parasites on the thymus by using C57BL/6 mice infected with Plasmodium berghei ANKA and Plasmodium yoelii 17XL; these lethal infection models represent the most severe complications, cerebral malaria, and anemia respectively. Data showed a reduction in the thymic weight and cellularity involving different T cell maturation stages, mainly CD4-CD8- and CD4+CD8+ thymocytes, as well as an increased presence of apoptotic cells, leading to significant thymic cortex reduction. Thymus atrophy showed no association with elevated serum cytokines levels, although increased glucocorticoid levels did. The severity of thymic damage in both models reached the same extend although it occurs at different stages of infection, showing that thymic atrophy does not depend on parasitemia level but on the specific host-parasite interaction.
Collapse
Affiliation(s)
- G M Corral-Ruiz
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico; Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - M J Pérez-Vega
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico; Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - A Galán-Salinas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico; Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - I Mancilla-Herrera
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología, Mexico City, Mexico
| | - J Barrios-Payán
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - L Fabila-Castillo
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
| | - R Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - L E Sánchez-Torres
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico.
| |
Collapse
|
2
|
de Jesus MCS, Cerilo-Filho M, Ramirez ADR, Menezes RAO, Gomes MSM, Cassiano GC, Gurgel RQ, Silva JRS, Moura TR, Pratt-Riccio LR, Baptista ARS, Storti-Melo LM, Machado RLD. Influence of trem-1 gene polymorphisms on cytokine levels during malaria by Plasmodium vivax in a frontier area of the Brazilian Amazon. Cytokine 2023; 169:156264. [PMID: 37327529 DOI: 10.1016/j.cyto.2023.156264] [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: 01/29/2023] [Revised: 06/01/2023] [Accepted: 06/04/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND The immunopathology during malaria depends on the level of inflammatory response generated. In this scenario, the TREM-1 has been associated with the severity of infectious diseases and could play an important role in the inflammatory course of malaria. We aimed to describe the allelic and genotypic frequency of four polymorphisms in the trem-1 gene in Plasmodium vivax-infected patients and to verify the association of these polymorphisms with clinical and immunological factors in a frontier area of the Brazilian Amazon. METHODS We included 76 individuals infected with P. vivax and 144 healthy controls living in the municipality of Oiapoque, Amapá, Brazil. The levels of TNF-α, IL-10, IL-2, IL-4, IL-5, and IFN-γ were measured by flow cytometry, while IL-6, sTREM-1, and antibodies against PvMSP-119 were evaluated by ELISA. The SNPs were genotyped by qPCR technique. Polymorphisms analysis, allelic and genotype, frequencies, and HWE calculation were determined by x2 test in R Software. The association between the parasitemia, gametocytes, antibodies, cytokines, and sTREM-1 with the genotypes of malaria and control groups was performed using the Kruskal-Wallis test, these analyzes were conducted in SPSS Software, at 5% significance level. RESULTS All SNPs were successfully genotyped. Allelic and genotypic distribution was in Hardy-Weinberg Equilibrium. Furthermore, several associations were identified between malaria and control groups, with increased levels of IL-5, IL-6, IL-10, TNF-α, and IFN-γ in the infected individuals with rs6910730A, rs2234237T, rs2234246T, rs4711668C alleles compared to the homozygous wild-type and heterozygous genotypes of the controls (p-value < 0.05). No association was found for these SNPs and the levels of IL-2, and sTREM-1. CONCLUSIONS The SNPs on the trem-1 gene are associated with the effector molecules of the innate immunity and may contribute to the identification and effective participation of trem-1 in the modulation of the immune response. This association may be essential for the establishment of immunization strategies against malaria.
Collapse
Affiliation(s)
- Myrela C S de Jesus
- Center for Microorganisms' Investigation, Department of Microbiology and Parasitology, Biomedical Institute, Fluminense Federal University, Niterói 24020-141, Rio de Janeiro, Brazil; Postgraduate Program in Applied Microbiology and Parasitology, Biomedical Institute, Fluminense Federal University, Niterói 24210-130, Rio de Janeiro, Brazil.
| | - Marcelo Cerilo-Filho
- Center for Microorganisms' Investigation, Department of Microbiology and Parasitology, Biomedical Institute, Fluminense Federal University, Niterói 24020-141, Rio de Janeiro, Brazil; Postgraduate Program in Applied Microbiology and Parasitology, Biomedical Institute, Fluminense Federal University, Niterói 24210-130, Rio de Janeiro, Brazil
| | - Aina D R Ramirez
- Center for Microorganisms' Investigation, Department of Microbiology and Parasitology, Biomedical Institute, Fluminense Federal University, Niterói 24020-141, Rio de Janeiro, Brazil; Postgraduate Program in Applied Microbiology and Parasitology, Biomedical Institute, Fluminense Federal University, Niterói 24210-130, Rio de Janeiro, Brazil
| | - Rubens A O Menezes
- Postgraduate Program in Applied Microbiology and Parasitology, Biomedical Institute, Fluminense Federal University, Niterói 24210-130, Rio de Janeiro, Brazil; Postgraduate Program in Health Sciences, Federal University of Amapá (UNIFAP), Macapá 68903-419, Amapá, Brazil
| | - Margarete S M Gomes
- Superintendence of Health Surveillance of the State of Amapá, Macapá 68902-865, Amapá, Brazil
| | | | - Ricardo Q Gurgel
- Postgraduate Program in Parasite Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - José R S Silva
- Postgraduate Program in Parasite Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - Tatiana R Moura
- Postgraduate Program in Parasite Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - Lilian R Pratt-Riccio
- Laboratory for Malaria Research, Oswaldo Cruz Foundation, Oswaldo Cruz Institute, Rio de Janeiro 21040-900, Rio de Janeiro, Brazil
| | - Andrea R S Baptista
- Center for Microorganisms' Investigation, Department of Microbiology and Parasitology, Biomedical Institute, Fluminense Federal University, Niterói 24020-141, Rio de Janeiro, Brazil; Postgraduate Program in Applied Microbiology and Parasitology, Biomedical Institute, Fluminense Federal University, Niterói 24210-130, Rio de Janeiro, Brazil
| | - Luciane M Storti-Melo
- Postgraduate Program in Parasite Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil; Laboratory of Molecular Genetics and Biotechnology, Department of Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| | - Ricardo L D Machado
- Center for Microorganisms' Investigation, Department of Microbiology and Parasitology, Biomedical Institute, Fluminense Federal University, Niterói 24020-141, Rio de Janeiro, Brazil; Postgraduate Program in Applied Microbiology and Parasitology, Biomedical Institute, Fluminense Federal University, Niterói 24210-130, Rio de Janeiro, Brazil; Postgraduate Program in Parasite Biology, Center for Biological and Health Sciences, Federal University of Sergipe, São Cristóvão 49100-000, Sergipe, Brazil
| |
Collapse
|
3
|
Zhang Y, Gao S, Yao S, Weng D, Wang Y, Huang Q, Zhang X, Wang H, Xu W. IL-27 mediates immune response of pneumococcal vaccine SPY1 through Th17 and memory CD4 +T cells. iScience 2023; 26:107464. [PMID: 37588169 PMCID: PMC10425906 DOI: 10.1016/j.isci.2023.107464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Revised: 06/16/2023] [Accepted: 07/19/2023] [Indexed: 08/18/2023] Open
Abstract
Vaccination is an effective means of preventing pneumococcal disease and SPY1 is a live attenuated pneumococcal vaccine we obtained earlier. We found IL-27 and its specific receptor (WSX-1) were increased in SPY1 vaccinated mice. Bacterial clearance and survival rates were decreased in SPY1 vaccinated IL-27Rα-/- mice. The vaccine-induced Th17 cell response and IgA secretion were also suppressed in IL-27Rα-/- mice. STAT3 and NF-κB signaling and expression of the Th17 cell polarization-related cytokines were also decreased in IL-27Rα-/- bone-marrow-derived dendritic cells(BMDC) stimulated with inactivated SPY1. The numbers of memory CD4+T cells were also decreased in SPY1 vaccinated IL-27Rα-/- mice. These results suggested that IL-27 plays a protective role in SPY1 vaccine by promoting Th17 polarization through STAT3 and NF-κB signaling pathways and memory CD4+T cells production in the SPY1 vaccine. In addition, we found that the immune protection of SPY1 vaccine was independent of aerobic glycolysis.
Collapse
Affiliation(s)
- Yanyu Zhang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Song Gao
- Department of Laboratory Medicine, Affiliated Hospital of Zunyi Medical University, School of Laboratory Medicine, Zunyi Medical University, Zunyi, Guizhou, China
| | - Shifei Yao
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Danlin Weng
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yan Wang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Qi Huang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xuemei Zhang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Hong Wang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Wenchun Xu
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| |
Collapse
|
4
|
Amo L, Kole HK, Scott B, Qi CF, Krymskaya L, Wang H, Miller LH, Janse CJ, Bolland S. Plasmodium curtails autoimmune nephritis via lasting bone marrow alterations, independent of hemozoin accumulation. Front Immunol 2023; 14:1192819. [PMID: 37539049 PMCID: PMC10394379 DOI: 10.3389/fimmu.2023.1192819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 06/30/2023] [Indexed: 08/05/2023] Open
Abstract
The host response against infection with Plasmodium commonly raises self-reactivity as a side effect, and antibody deposition in kidney has been cited as a possible cause of kidney injury during severe malaria. In contrast, animal models show that infection with the parasite confers long-term protection from lethal lupus nephritis initiated by autoantibody deposition in kidney. We have limited knowledge of the factors that make parasite infection more likely to induce kidney damage in humans, or the mechanisms underlying protection from autoimmune nephritis in animal models. Our experiments with the autoimmune-prone FcγR2B[KO] mice have shown that a prior infection with P. yoelii 17XNL protects from end-stage nephritis for a year, even when overall autoreactivity and systemic inflammation are maintained at high levels. In this report we evaluate post-infection alterations, such as hemozoin accumulation and compensatory changes in immune cells, and their potential role in the kidney-specific protective effect by Plasmodium. We ruled out the role of pigment accumulation with the use of a hemozoin-restricted P. berghei ANKA parasite, which induced a self-resolved infection that protected from autoimmune nephritis with the same mechanism as parasitic infections that accumulated normal levels of hemozoin. In contrast, adoptive transfer experiments revealed that bone marrow cells were altered by the infection and could transmit the kidney protective effect to a new host. While changes in the frequency of bone marrow cell populations after infection were variable and unique to a particular parasite strain, we detected a sustained bias in cytokine/chemokine expression that suggested lower fibrotic potential and higher Th1 bias likely affecting multiple cell populations. Sustained changes in bone marrow cell activation profile could have repercussions in immune responses long after the infection was cleared.
Collapse
Affiliation(s)
- Laura Amo
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Hemanta K. Kole
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Bethany Scott
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Chen-Feng Qi
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Ludmila Krymskaya
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Hongsheng Wang
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Louis H. Miller
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| | - Chris J. Janse
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center, Leiden, Netherlands
| | - Silvia Bolland
- Laboratory of Immunogenetics, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, United States
| |
Collapse
|
5
|
Malaria-derived exosomes exacerbate liver injury during blood stage of Plasmodium berghei infection. Acta Trop 2023; 239:106815. [PMID: 36608749 DOI: 10.1016/j.actatropica.2023.106815] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 12/21/2022] [Accepted: 01/01/2023] [Indexed: 01/05/2023]
Abstract
Liver injury is a common clinical feature of Plasmodium spp. infection and contributes to multi-organ failure of severe malaria. Malaria-derived exosomes (MD-Exos) have recently engaged as key mediators in parasite-host interactions, modulating the subsequent pathogenic process. However, the role of MD-Exos in malaria-related liver injury and the underlying mechanisms remain unclear. Herein, exosomes from C57BL/6 mice infected with or without P. berghei ANKA serum (namely inf-Exos or un-Exos) were isolated and characterized by transmission electron microscopy, western blotting, and nanoparticle tracking analysis. The miRNAs profiling between inf-Exos and un-Exos were generated using RNA-seq and qPCR. The functions of inf-Exos on liver injury were investigated after two types of exosomes injected into mice intravenously (i.v.), by examining histopathological and apoptotic changes, macrophage polarization, and pro-inflammatory response. The infected red blood cells-stimulated mouse Raw264.7 macrophage cells targeted by inf-Exos or un-Exos were cultured for further study and verification the potential mechanisms. We found that both inf-Exos and un-Exos displayed a typical cup-shaped structure with a diameter of 60-200 nm, and had a positive expression of exosomal markers (e.g., CD9, CD63, and CD81). Compared with infected control mice, the treatment of inf-Exos but not un-Exos dramatically enhanced peripheral blood parasitemia and ECM incidence, exacerbated liver histopathological damage, elevated numbers of liver apoptotic cells, CD68+and CD86+ macrophages. The CD68+-TREM-1+ macrophages in liver tissues and the mRNA levels of pro-inflammatory cytokines (e.g., iNOS, TNF-α, IL-1β, and IL-6) were increased by inf-Exos treatment in vivo. Meanwhile, the treatment of inf-Exos resulted in a substantial increase of the mRNA levels of CD86, iNOS, TNF-α, IL-1β, and IL-6, but led to a remarkable decrease of Bcl-6 and SOCS-1 in Raw264.7 cells stimulated with iRBC in vitro. Notably, compared to un-Exos, five types of miRNAs (including miR-10a-5p, miR-10b-5p, miR-155-5p, miR-205-5p, and miR-21a-5p), that were previously reported to target Bcl-6 or SOCS-1, present higher abundance on inf-Exos, as demonstrated by RNA-seq and qPCR. Collectively, our data suggest that inf-Exos exacerbate malaria-induced liver pathology via triggering excessive pro-inflammatory response and promoting macrophage M1 polarization. Our findings will provide new insights into the roles of inf-Exos in malaria parasite-host interaction and pathogenesis of liver injury.
Collapse
|
6
|
Kotepui M, Mala W, Kwankaew P, Kotepui KU, Masangkay FR, Wilairatana P. Distinct cytokine profiles in malaria coinfections: A systematic review. PLoS Negl Trop Dis 2023; 17:e0011061. [PMID: 36716305 PMCID: PMC9886258 DOI: 10.1371/journal.pntd.0011061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Accepted: 12/23/2022] [Indexed: 02/01/2023] Open
Abstract
BACKGROUND Few data exist on the distinct cytokine profiles of individuals with malaria coinfections and other diseases. This study focuses on data collation of distinct cytokine profiles between individuals with malaria coinfections and monoinfections to provide evidence for further diagnostic or prognostic studies. METHODS We searched five medical databases, including Embase, MEDLINE, PubMed, Ovid, and Scopus, for articles on cytokines in malaria coinfections published from January 1, 1983 to May 3, 2022, after which the distinct cytokine patterns between malaria coinfection and monoinfection were illustrated in heat maps. RESULTS Preliminary searches identified 2127 articles, of which 34 were included in the systematic review. Distinct cytokine profiles in malaria coinfections with bacteremia; HIV; HBV; dengue; filariasis; intestinal parasites; and schistosomiasis were tumor necrosis factor (TNF), interferon (IFN)-γ, IFN-α, interleukin (IL)-1, IL-1 receptor antagonist (Ra), IL-4, IL-7, IL-12, IL-15, IL-17; TNF, IL-1Ra, IL-4, IL-10, IL-12, IL-18, CCL3, CCL5, CXCL8, CXCL9, CXCL11, granulocyte colony-stimulating factor (G-CSF); TNF, IFN-γ, IL-4, IL-6, IL-10, IL-12, CCL2; IFN-γ, IL-1, IL-4, IL-6, IL-10, IL-12, IL-13, IL-17, CCL2, CCL3, CCL4, G-CSF; IL-1Ra, IL-10, CXCL5, CXCL8, CXCL10; TNF, IL-2, IL-4, IL-6, IL-10; and TNF, IFN-γ, IL-4, IL-5, IL-10, transforming growth factor-β, CXCL8, respectively. CONCLUSION This systematic review provides information on distinct cytokine profiles of malaria coinfections and malaria monoinfections. Further studies should investigate whether specific cytokines for each coinfection type could serve as essential diagnostic or prognostic biomarkers for malaria coinfections.
Collapse
Affiliation(s)
- Manas Kotepui
- Medical Technology Program, School of Allied Health Sciences, Walailak University, Tha Sala, Nakhon Si Thammarat, Thailand
- * E-mail: (MK); (PW)
| | - Wanida Mala
- Medical Technology Program, School of Allied Health Sciences, Walailak University, Tha Sala, Nakhon Si Thammarat, Thailand
| | - Pattamaporn Kwankaew
- Medical Technology Program, School of Allied Health Sciences, Walailak University, Tha Sala, Nakhon Si Thammarat, Thailand
| | - Kwuntida Uthaisar Kotepui
- Medical Technology Program, School of Allied Health Sciences, Walailak University, Tha Sala, Nakhon Si Thammarat, Thailand
| | | | - Polrat Wilairatana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
- * E-mail: (MK); (PW)
| |
Collapse
|
7
|
Zhang D, Yu Y, Duan T, Zhou Q. The role of macrophages in reproductive-related diseases. Heliyon 2022; 8:e11686. [DOI: 10.1016/j.heliyon.2022.e11686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/03/2022] [Accepted: 11/10/2022] [Indexed: 11/23/2022] Open
|
8
|
Oliveira FMS, Cruz RE, Pinheiro GRG, Caliari MV. Comorbidities involving parasitic diseases: A look at the benefits and complications. Exp Biol Med (Maywood) 2022; 247:1819-1826. [PMID: 35876147 PMCID: PMC9679356 DOI: 10.1177/15353702221108387] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Parasitic infections acquired by the population cause substantial morbidity worldwide, with individuals from developing countries being most affected. Some parasites remain in the host for long periods, settling in different organs, manipulating the flow of nutrients and metabolites, and influencing the immune response, favoring their adaptation. The host attempts to counteract the metabolic and immunological alterations and the possible damage caused by infection. These metabolic and immunological changes experienced by the host can influence the progression of other existing morbidities or those that will be acquired in the future. Cancer and metabolic diseases are also frequent causes of morbidity in the world population. The large numbers of individuals affected by cancer and metabolic diseases and the high prevalence of morbidity caused by parasitic diseases favor the development of comorbidity involving these pathologies. This review provides an overview of major advances in research on cancer and metabolic diseases associated with parasitic infections. Information about hosts and parasites such as alterations of the immune response, metabolism and adaptation mechanisms of the parasites, and parasitic molecules with therapeutic potential is provided, as well as the beneficial results or complications related to the comorbidities discussed herein. We emphasize the need to conduct additional studies addressing comorbidities associated with parasitic infections to improve the understanding of the impact of this association on the progression of morbidities, as well as the possibility of the therapeutic use of and therapeutic approaches involving parasites.
Collapse
Affiliation(s)
- Fabrício Marcus Silva Oliveira
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Ruth Elizabeth Cruz
- Department of Parasitology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Guilherme Rafael Gomide Pinheiro
- Department of Preventive Veterinary Medicine, Veterinary School, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil
| | - Marcelo Vidigal Caliari
- Department of General Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31270-901, Minas Gerais, Brazil,Marcelo Vidigal Caliari.
| |
Collapse
|
9
|
Saini I, Joshi J, Kaur S. Unwelcome prevalence of leishmaniasis with several other infectious diseases. Int Immunopharmacol 2022; 110:109059. [DOI: 10.1016/j.intimp.2022.109059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 11/17/2022]
|
10
|
IL-10 Producing Regulatory B Cells Mediated Protection against Murine Malaria Pathogenesis. BIOLOGY 2022; 11:biology11050669. [PMID: 35625397 PMCID: PMC9138363 DOI: 10.3390/biology11050669] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Revised: 03/05/2022] [Accepted: 03/06/2022] [Indexed: 02/06/2023]
Abstract
Simple Summary The immunomodulatory role of B cell subset called regulatory B cells was evaluated during Plasmodium infection to study their role in susceptibility or resistance during infection. The expansion of regulatory B cells during Plasmodium infection indicated their important role in regulating the immune response. Adoptive transfer of regulatory B cells following infection with a lethal parasite resulted in enhanced survival of mice and inhibited growth of the Plasmodium parasite. Moreover, by inhibiting the production of the pro-inflammatory cytokine, IFN-γ, and stimulating anti-inflammatory IL-10 production, regulatory B cells may serve as an important contributor to protective immune response. Abstract Various immune cells are known to participate in combating infection. Regulatory B cells represent a subset of B cells that take part in immunomodulation and control inflammation. The immunoregulatory function of regulatory B cells has been shown in various murine models of several disorders. In this study, a comparable IL-10 competent B-10 cell subset (regulatory B cells) was characterized during lethal and non-lethal infection with malaria parasites using the mouse model. We observed that infection of Balb/c mice with P. yoelii I 7XL was lethal, and a rapid increase in dynamics of IL-10 producing B220+CD5+CD1d+ regulatory B cells over the course of infection was observed. However, animals infected with a less virulent strain of the parasite P. yoelii I7XNL attained complete resistance. It was observed that there is an increase in the population of regulatory B cells with an increase of parasitemia; however, a sudden drop in the frequency of these cells was observed with parasite clearance. Adoptive transfer of regulatory B cells to naïve mice followed by infection results in slow parasite growth and enhancement of survival in P. yoelii 17XL (lethal) infected animals. Adoptively transferred regulatory B cells also resulted in decreased production of pro-inflammatory cytokine (IFN-γ) and enhanced production of anti-inflammatory cytokine (IL-10). It infers that these regulatory B cells may contribute in immune protection by preventing the inflammation associated with disease and inhibiting the parasite growth.
Collapse
|
11
|
Chauhan R, Awasthi V, Thakur RS, Pande V, Chattopadhyay D, Das J. CD4 +ICOS +Foxp3 +: a sub-population of regulatory T cells contribute to malaria pathogenesis. Malar J 2022; 21:32. [PMID: 35109868 PMCID: PMC8812217 DOI: 10.1186/s12936-022-04055-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 01/19/2022] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Regulatory T cells are known to play a key role to counter balance the protective immune response and immune mediated pathology. However, the role of naturally occurring regulatory cells CD4+CD25+Foxp3+ in malaria infection during the disease pathogenesis is controversial. Beside this, ICOS molecule has been shown to be involved in the development and function of regulatory T cell enhance IL-10 production. Therefore, possible involvement of the ICOS dependent regulatory CD4+ICOS+Foxp3+ T cells in resistance/susceptibility during malaria parasite is explored in this study. METHODS 5 × 105 red blood cells infected with non-lethal and lethal parasites were inoculated in female Balb/c mice by intra-peritoneal injection. Infected or uninfected mice were sacrificed at early (3rd day post infection) and later stage (10th day post infection) of infection. Harvested cells were analysed by using flow cytometer and serum cytokine by Bioplex assay. RESULTS Thin blood films show that percentages of parasitaemia increases with disease progression in infections with the lethal malaria parasite and mice eventually die by day 14th post-infection. Whereas in case of non-lethal malaria parasite, parasitaemia goes down by 7th day post infection and gets cleared within 13th day. The number of CD4+ ICOS+ T cells increases in lethal infection with disease progression. Surprisingly, in non-lethal parasite, ICOS expression decreases after day 7th post infection as parasitaemia goes down. The frequency of CD4+ICOS+FoxP3+ Tregs was significantly higher in lethal parasitic infection as compared to the non-lethal parasite. The level of IL-12 cytokine was remarkably higher in non-lethal infection compared to the lethal infection. In contrast, the level of IL-10 cytokines was higher in lethal parasite infection compared to the non-lethal parasite. CONCLUSION Taken together, these data suggest that lethal parasite induce immunosuppressive environment, protecting from host immune responses and help the parasite to survive whereas non-lethal parasite leads to low frequencies of Treg cells seldom impede immune response that allow the parasite to get self-resolved.
Collapse
Affiliation(s)
- Rubika Chauhan
- Parasite-Host Biology, National Institute of Malaria Research, Sector-8, Dwarka, New Delhi, 110077, India
| | - Vikky Awasthi
- Parasite-Host Biology, National Institute of Malaria Research, Sector-8, Dwarka, New Delhi, 110077, India
| | - Reva Sharan Thakur
- Parasite-Host Biology, National Institute of Malaria Research, Sector-8, Dwarka, New Delhi, 110077, India
| | - Veena Pande
- Biotechnology Department, Kumaun University, Nainital, India
| | - Debprasad Chattopadhyay
- ICMR Virus Unit, ID and BG Hospital, Kolkata, 700010, India.,ICMR-National Institute of Traditional Medicine (NITM), Belagavi, 590010, India
| | - Jyoti Das
- Parasite-Host Biology, National Institute of Malaria Research, Sector-8, Dwarka, New Delhi, 110077, India.
| |
Collapse
|
12
|
Alves KCS, Guimarães JM, Almeida MEMD, Mariúba LAM. Plasmodium falciparum merozoite surface protein 3 as a vaccine candidate: a brief review. Rev Inst Med Trop Sao Paulo 2022; 64:e23. [PMID: 35293561 PMCID: PMC8916589 DOI: 10.1590/s1678-9946202264023] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/24/2022] [Indexed: 11/22/2022] Open
Abstract
Despite the many efforts of researchers around the world, there is currently no effective vaccine for malaria. Numerous studies have been developed to find vaccine antigens that are immunogenic and safe. Among antigen candidates, Plasmodium falciparum merozoite surface protein 3 (MSP3) has stood out in a number of these studies for its ability to induce a consistent and protective immune response, also being safe for use in humans. This review presents the main studies that explored MSP3 as a vaccine candidate over the last few decades. MSP3 formulations were tested in animals and humans and the most advanced candidate formulations are MSP3-LSP, a combination of MSP3 and LSP1, and GMZ2 (a vaccine based on the recombinant protein fusion GLURP and MSP3) which is currently being tested in phase II clinical studies. This brief review highlights the history and the main formulations of MSP3-based vaccines approaches against P. falciparum .
Collapse
Affiliation(s)
| | | | | | - Luís André Morais Mariúba
- Instituto Leônidas e Maria Deane, Brazil; Universidade Federal do Amazonas, Brazil; Instituto Oswaldo Cruz, Brazil; Universidade Federal do Amazonas, Brazil
| |
Collapse
|
13
|
Vinhaes CL, Carmo TA, Queiroz ATL, Fukutani KF, Araújo-Pereira M, Arriaga MB, Lacerda MVG, Barral-Netto M, Andrade BB. Dissecting disease tolerance in Plasmodium vivax malaria using the systemic degree of inflammatory perturbation. PLoS Negl Trop Dis 2021; 15:e0009886. [PMID: 34727121 PMCID: PMC8589215 DOI: 10.1371/journal.pntd.0009886] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 11/12/2021] [Accepted: 10/08/2021] [Indexed: 01/18/2023] Open
Abstract
Homeostatic perturbation caused by infection fosters two major defense strategies, resistance and tolerance, which promote the host's survival. Resistance relates to the ability of the host to restrict the pathogen load. Tolerance minimizes collateral tissue damage without directly affecting pathogen fitness. These concepts have been explored mechanistically in murine models of malaria but only superficially in human disease. Indeed, individuals infected with Plasmodium vivax may present with asymptomatic malaria, only mild symptoms, or be severely ill. We and others have reported a diverse repertoire of immunopathological events that potentially underly susceptibility to disease severity in vivax malaria. Nevertheless, the combined epidemiologic, clinical, parasitological, and immunologic features associated with defining the disease outcomes are still not fully understood. In the present study, we perform an extensive outlining of cytokines and inflammatory proteins in plasma samples from a cohort of individuals from the Brazilian Amazon infected with P. vivax and presenting with asymptomatic (n = 108) or symptomatic (n = 134) disease (106 with mild presentation and 28 with severe malaria), as well as from uninfected endemic controls (n = 128) to elucidate these gaps further. We employ highly multidimensional Systems Immunology analyses using the molecular degree of perturbation to reveal nuances of a unique profile of systemic inflammation and imbalanced immune activation directly linked to disease severity as well as with other clinical and epidemiologic characteristics. Additionally, our findings reveal that the main factor associated with severe cases of P. vivax infection was the number of symptoms, despite of a lower global inflammatory perturbation and parasitemia. In these participants, the number of symptoms directly correlated with perturbation of markers of inflammation and tissue damage. On the other hand, the main factor associated with non-severe infections was the parasitemia values, that correlated only with perturbation of inflammatory markers, such as IL-4 and IL-1β, with a relatively lower number of symptoms. These observations suggest that some persons present severe vivax regardless of pathogen burden and global inflammatory perturbation. Such patients are thus little tolerant to P. vivax infection and show higher susceptibility to disrupt homeostasis and consequently exhibit more clinical manifestations. Other persons are capable to tolerate higher parasitemia with lower inflammatory perturbation and fewer symptoms, developing non-severe malaria. The analytical approach presented here has capability to define in more details the determinants of disease tolerance in vivax malaria.
Collapse
Affiliation(s)
- Caian L. Vinhaes
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
| | - Thomas A. Carmo
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Curso de Medicina, Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Brazil
| | - Artur T. L. Queiroz
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
| | - Kiyoshi F. Fukutani
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Curso de Medicina, Centro Universitário Facultade de Tecnologia e Ciências (UniFTC), Salvador, Brazil
| | - Mariana Araújo-Pereira
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Faculdade de Medicina, Salvador, Brazil
| | - María B. Arriaga
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Faculdade de Medicina, Salvador, Brazil
| | - Marcus V. G. Lacerda
- Instituto de Pesquisa Clínica Carlos Borborema, Fundação de Medicina Tropical Dr Heitor Vieira Dourado, Manaus, Brazil
- Instituto Leônidas & Maria Deane, Fundação Oswaldo Cruz, Manaus, Brazil
| | - Manoel Barral-Netto
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Faculdade de Medicina, Salvador, Brazil
- Instituto Nacional de Ciência e Tecnologia, Instituto de Investigação em Imunologia, São Paulo, Brazil
| | - Bruno B. Andrade
- Instituto Gonçalo Moniz, Fundação Oswaldo Cruz (FIOCRUZ), Salvador, Brazil
- Multinational Organization Network Sponsoring Translational and Epidemiological Research (MONSTER) Initiative, Salvador, Brazil
- Escola Bahiana de Medicina e Saúde Pública (EBMSP), Salvador, Brazil
- Curso de Medicina, Universidade Salvador (UNIFACS), Laureate Universities, Salvador, Brazil
- Curso de Medicina, Centro Universitário Facultade de Tecnologia e Ciências (UniFTC), Salvador, Brazil
- Faculdade de Medicina, Universidade Federal da Bahia, Faculdade de Medicina, Salvador, Brazil
- * E-mail:
| |
Collapse
|
14
|
Immunosuppression in Malaria: Do Plasmodium falciparum Parasites Hijack the Host? Pathogens 2021; 10:pathogens10101277. [PMID: 34684226 PMCID: PMC8536967 DOI: 10.3390/pathogens10101277] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/27/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Malaria reflects not only a state of immune activation, but also a state of general immune defect or immunosuppression, of complex etiology that can last longer than the actual episode. Inhabitants of malaria-endemic regions with lifelong exposure to the parasite show an exhausted or immune regulatory profile compared to non- or minimally exposed subjects. Several studies and experiments to identify and characterize the cause of this malaria-related immunosuppression have shown that malaria suppresses humoral and cellular responses to both homologous (Plasmodium) and heterologous antigens (e.g., vaccines). However, neither the underlying mechanisms nor the relative involvement of different types of immune cells in immunosuppression during malaria is well understood. Moreover, the implication of the parasite during the different stages of the modulation of immunity has not been addressed in detail. There is growing evidence of a role of immune regulators and cellular components in malaria that may lead to immunosuppression that needs further research. In this review, we summarize the current evidence on how malaria parasites may directly and indirectly induce immunosuppression and investigate the potential role of specific cell types, effector molecules and other immunoregulatory factors.
Collapse
|
15
|
Sabourin KR, Daud I, Ogolla S, Labo N, Miley W, Lamb M, Newton R, Whitby D, Rochford R. Malaria Is Associated With Kaposi Sarcoma-Associated Herpesvirus Seroconversion in a Cohort of Western Kenyan Children. J Infect Dis 2021; 224:303-311. [PMID: 33249494 PMCID: PMC8280487 DOI: 10.1093/infdis/jiaa740] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 11/25/2020] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND We aimed to determine whether Plasmodium falciparum infection affects age of Kaposi sarcoma-associated herpesvirus (KSHV) seroconversion in Kenyan children. METHODS Kenyan children (n = 144) enrolled at age 1 month, from 2 sites with different levels of malaria transmission (stable/high vs unstable/low) were followed to age 24 months. Plasma was tested for KSHV antibodies using enzyme-linked immunosorbent assay (ELISA; K8.1 and LANA) and a multiplex bead-based assay (K8.1, K10.5, ORF38, ORF50, and LANA) and whole blood tested for P. falciparum DNA using quantitative PCR. Cox proportional hazards models were used to assess associations between P. falciparum DNA detection, malaria annualized rate (P. falciparum detections/person-years), and enrollment site (malaria-high vs malaria-low) with time to KSHV seroconversion. RESULTS KSHV seroprevalence was 63% by age 2 years when assessed by multiplex assay. Children with P. falciparum were at increased hazards of earlier KSHV seroconversion and, among children with malaria, the hazard of becoming KSHV seropositive increased significantly with increasing malaria annualized rate. Children from the malaria-high transmission region had no significant difference in hazards of KSHV seroconversion at 12 months but were more likely to become KSHV seropositive by age 24 months. DISCUSSION Malaria exposure increases the risk for KSHV seroconversion early in life.
Collapse
Affiliation(s)
- Katherine R Sabourin
- Department of Epidemiology, Colorado School of Public Health, Denver, Colorado, USA
- Department of Immunology and Microbiology, University of Colorado, Denver, Colorado, USA
| | - Ibrahim Daud
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Sidney Ogolla
- Centre for Global Health Research, Kenya Medical Research Institute, Kisumu, Kenya
| | - Nazzarena Labo
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, USA
| | - Wendell Miley
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, USA
| | - Molly Lamb
- Department of Epidemiology, Colorado School of Public Health, Denver, Colorado, USA
| | - Robert Newton
- Department of Health Sciences, University of York, York, United Kingdom
| | - Denise Whitby
- Frederick National Laboratory for Cancer Research, Leidos Biomedical Research, Frederick, Maryland, USA
| | - Rosemary Rochford
- Department of Immunology and Microbiology, University of Colorado, Denver, Colorado, USA
| |
Collapse
|
16
|
Naing C, Wong ST, Aung HH. Toll-like receptor 9 and 4 gene polymorphisms in susceptibility and severity of malaria: a meta-analysis of genetic association studies. Malar J 2021; 20:302. [PMID: 34217314 PMCID: PMC8255014 DOI: 10.1186/s12936-021-03836-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2020] [Accepted: 06/27/2021] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Malaria is still a major public health problem in sub-Saharan Africa and South-east Asia. The clinical presentations of malaria infection vary from a mild febrile illness to life-threatening severe malaria. Toll like receptors (TLRs) are postulated to be involved in the innate immune responses to malaria. Individual studies showed inconclusive findings. This study aimed to assess the role of TLR4 (D299G, T399I) and TLR9 (T1237C, T1486C) in severity or susceptibility of malaria by meta-analysis of data from eligible studies. METHODS Relevant case-control studies that assessed the association between TLR 4/9 and malaria either in susceptibility or progression were searched in health-related electronic databases. Quality of included studies was evaluated with Newcastle-Ottawa scale. Pooled analyses for specific genetic polymorphisms were done under five genetic models. Stratified analysis was done by age and geographical region (Asian countries vs non-Asian countries). RESULTS Eleven studies (2716 cases and 2376 controls) from nine endemic countries were identified. Five studies (45.4%) obtained high score in quality assessment. Overall, a significant association between TLR9 (T1486C) and severity of malaria is observed in allele model (OR: 1.26, 95% CI: 1.08-1.48, I2 = 0%) or homozygous model (OR: 1.55, 95% CI: 1.08-2.28, I2 = 0%). For TLR9 (T1237C), a significant association with severity of malaria is observed in in heterozygous model (OR:1.89, 95% CI: 1.11-3.22, I2 = 75%). On stratifications, TLR9 (T1486C) is only significantly associated with a subgroup of children of non-Asian countries under allele model (OR: 1.25, 95% CI: 1.02-1.38), while 1237 is with a subgroup of adults from Asian countries under heterozygous model (OR: 2.0, 95% CI: 1.09-3.64, I2 = 39%). Regarding the susceptibility to malaria, TLR9 (T1237C) is significantly associated only with the children group under recessive model (OR: 2.21, 95% CI: 1.06-4.57, I2=85%) and homozygous model (OR: 1.49, 95% CI: 1.09-2.0, I2 = 0%). For TLR4 (D299G, T399I), none is significantly associated with either severity of malaria or susceptibility to malaria under any genetic models. CONCLUSIONS The findings suggest that TLR 9 (T1486C and T1237C) seems to influence the progression of malaria, under certain genetic models and in specific age group of people from specific geographical region. TLR 9 (T1237C) also plays a role in susceptibility to malaria under certain genetic models and only with children of non-Asian countries. To substantiate these, future well designed studies with larger samples across endemic countries are needed.
Collapse
Affiliation(s)
- Cho Naing
- Institute for Research, Development and Innovation (IRDI), International Medical University, 5700, Kuala Lumpur, Malaysia. .,Faculty of Tropical Heath and Medicine, James Cook University, Queensland, Australia.
| | - Siew Tung Wong
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| | - Htar Htar Aung
- School of Medicine, International Medical University, Kuala Lumpur, Malaysia
| |
Collapse
|
17
|
Metwally DM, Alajmi RA, El-Khadragy MF, Al-Quraishy S. Silver Nanoparticles Biosynthesized With Salvia officinalis Leaf Exert Protective Effect on Hepatic Tissue Injury Induced by Plasmodium chabaudi. Front Vet Sci 2021; 7:620665. [PMID: 33614756 PMCID: PMC7889953 DOI: 10.3389/fvets.2020.620665] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/29/2020] [Indexed: 01/24/2023] Open
Abstract
Malaria is an important health problem in subtropical and tropical areas around the world. Infection with protozoan parasites of the Plasmodium genus, which grow inside host erythrocytes, causes malaria and may lead to morbidity and mortality. Liver tissue plays an important role in the pathogenesis of malaria and is closely involved in parasitic pre-erythrocytic development. Numerous published studies have demonstrated that the liver is not only the source of Plasmodium parasites prior to erythrocytic growth but is also a primary immune effector toward the blood stage of the malaria life cycle. Despite efforts to improve antimalarial drugs and vaccines, Plasmodium species that cause severe malaria are being detected increasingly frequently in endemic regions. In this study, Salvia officinalis (S. officinalis) leaf extract was employed to synthesize silver nanoparticles (Ag-NPs). This method is eco-friendly and represents a single-step technique for the biosynthetic process; therefore, it has attracted considerable attention. Accordingly, we biosynthesized Ag-NPs with extract of the S. officinalis leaf and examined the antimalarial activity of these nanoparticles in a murine model of Plasmodium chabaudi malaria (P. chabaudi malaria). Forty mice were chosen and classified into four types: infected group, healthy control, pretreated mice infected after treatment with 50 mg/kg of S. officinalis leaf extract-biosynthesized Ag-NPs for two weeks, and post-treated mice infected before treatment with 50 mg/kg of S. officinalis leaf extract-biosynthesized Ag-NPs (administered daily for 7 d). In this study, both pre-treatment and post-treatment with Ag-NPs produced a substantial reduction in parasitemia relative to the infected group. We investigated the antiplasmodial and hepatoprotective effects of S. officinalis leaf extract-biosynthesized Ag-NPs on P. chabaudi-induced inflammation and hepatic oxidative stress markers.
Collapse
Affiliation(s)
- Dina M Metwally
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia.,Department of Parasitology, Faculty of Veterinary Medicine, Zagazig University, Zagazig, Egypt
| | - Reem A Alajmi
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Manal F El-Khadragy
- Department of Biology, Faculty of Science, Princess Nourah Bint Abdelrahman University, Riyadh, Saudi Arabia.,Department of Zoology and Entomology, Faculty of Science, Helwan University, Cairo, Egypt
| | - Saleh Al-Quraishy
- Department of Zoology, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
18
|
Wu X, Thylur RP, Dayanand KK, Punnath K, Norbury CC, Gowda DC. IL-4 Treatment Mitigates Experimental Cerebral Malaria by Reducing Parasitemia, Dampening Inflammation, and Lessening the Cytotoxicity of T Cells. THE JOURNAL OF IMMUNOLOGY 2020; 206:118-131. [PMID: 33239419 DOI: 10.4049/jimmunol.2000779] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Accepted: 10/28/2020] [Indexed: 12/22/2022]
Abstract
Cytokine responses to malaria play important roles in both protective immunity development and pathogenesis. Although the roles of cytokines such as TNF-α, IL-12, IFN-γ, and IL-10 in immunity and pathogenesis to the blood stage malaria are largely known, the role of IL-4 remains less understood. IL-4 targets many cell types and induces multiple effects, including cell proliferation, gene expression, protection from apoptosis, and immune regulation. Accordingly, IL-4 has been exploited as a therapeutic for several inflammatory diseases. Malaria caused by Plasmodium falciparum manifests in many organ-specific fatal pathologies, including cerebral malaria (CM), driven by a high parasite load, leading to parasite sequestration in organs and consequent excessive inflammatory responses and endothelial damage. We investigated the therapeutic potential of IL-4 against fatal malaria in Plasmodium berghei ANKA-infected C57BL/6J mice, an experimental CM model. IL-4 treatment significantly reduced parasitemia, CM pathology, and mortality. The therapeutic effect of IL-4 is mediated through multiple mechanisms, including enhanced parasite clearance mediated by upregulation of phagocytic receptors and increased IgM production, and decreased brain inflammatory responses, including reduced chemokine (CXCL10) production, reduced chemokine receptor (CXCR3) and adhesion molecule (LFA-1) expression by T cells, and downregulation of cytotoxic T cell lytic potential. IL-4 treatment markedly reduced the infiltration of CD8+ T cells and brain pathology. STAT6, PI3K-Akt-NF-κB, and Src signaling mediated the cellular and molecular events that contributed to the IL-4-dependent decrease in parasitemia. Overall, our results provide mechanistic insights into how IL-4 treatment mitigates experimental CM and have implications in developing treatment strategies for organ-specific fatal malaria.
Collapse
Affiliation(s)
- Xianzhu Wu
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Ramesh P Thylur
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Kiran K Dayanand
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Kishore Punnath
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| | - Christopher C Norbury
- Department of Microbiology and Immunology, The Pennsylvania State University College of Medicine, Hershey, PA 17033
| | - D Channe Gowda
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA 17033; and
| |
Collapse
|
19
|
Frimpong A, Amponsah J, Adjokatseh AS, Agyemang D, Bentum-Ennin L, Ofori EA, Kyei-Baafour E, Akyea-Mensah K, Adu B, Mensah GI, Amoah LE, Kusi KA. Asymptomatic Malaria Infection Is Maintained by a Balanced Pro- and Anti-inflammatory Response. Front Microbiol 2020; 11:559255. [PMID: 33281757 PMCID: PMC7705202 DOI: 10.3389/fmicb.2020.559255] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 10/19/2020] [Indexed: 01/12/2023] Open
Abstract
Background Pro- and anti-inflammatory cytokines are important mediators of immunity and are associated with malaria disease outcomes. However, their role in the establishment of asymptomatic infections, which may precede the development of clinical symptoms, is not as well-understood. Methods We determined the association of pro and anti-inflammatory cytokines and other immune effector molecules with the development of asymptomatic malaria. We measured and compared the plasma levels of pro-inflammatory mediators including tumor necrosis factor-alpha (TNF-α), interferon-gamma (IFN-γ), interleukin (IL)-6, IL-12p70, IL-17A, and granzyme B, the anti-inflammatory cytokine IL-4 and the regulatory cytokine IL-10 from children with asymptomatic malaria infections (either microscopic or submicroscopic) and uninfected controls using Luminex. Results We show that individuals with microscopic asymptomatic malaria had significantly increased levels of TNF-α and IL-6 compared to uninfected controls. Children with either microscopic or submicroscopic asymptomatic malaria exhibited higher levels of IFN-γ, IL-17A, and IL-4 compared to uninfected controls. The levels of most of the pro and anti-inflammatory cytokines were comparable between children with microscopic and submicroscopic infections. The ratio of IFN-γ/IL-10, TNF-α/IL-10, IL-6/IL-10 as well as IFN-γ/IL-4 and IL-6/IL-4 did not differ significantly between the groups. Additionally, using a principal component analysis, the cytokines measured could not distinguish amongst the three study populations. This may imply that neither microscopic nor submicroscopic asymptomatic infections were polarized toward a pro-inflammatory or anti-inflammatory response. Conclusion The data show that asymptomatic malaria infections result in increased plasma levels of both pro and anti-inflammatory cytokines relative to uninfected persons. The balance between pro- and anti-inflammatory cytokines are, however, largely maintained and this may in part, explain the lack of clinical symptoms. This is consistent with the generally accepted observation that clinical symptoms develop as a result of immunopathology involving dysregulation of inflammatory mediator balance in favor of pro-inflammatory mediators.
Collapse
Affiliation(s)
- Augustina Frimpong
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana.,Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana.,African Institute for Mathematical Sciences, Accra, Ghana
| | - Jones Amponsah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Abigail Sena Adjokatseh
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Dorothy Agyemang
- Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| | - Lutterodt Bentum-Ennin
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Ebenezer Addo Ofori
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana.,Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Eric Kyei-Baafour
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Kwadwo Akyea-Mensah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Bright Adu
- Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Gloria Ivy Mensah
- Department of Bacteriology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Linda Eva Amoah
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana.,Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana
| | - Kwadwo Asamoah Kusi
- West African Centre for Cell Biology of Infectious Pathogens (WACCBIP), University of Ghana, Accra, Ghana.,Department of Immunology, Noguchi Memorial Institute for Medical Research, College of Health Sciences, University of Ghana, Accra, Ghana.,Department of Biochemistry, Cell and Molecular Biology, College of Basic and Applied Sciences, University of Ghana, Accra, Ghana
| |
Collapse
|
20
|
RTP4 inhibits IFN-I response and enhances experimental cerebral malaria and neuropathology. Proc Natl Acad Sci U S A 2020; 117:19465-19474. [PMID: 32709745 DOI: 10.1073/pnas.2006492117] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Infection by malaria parasites triggers dynamic immune responses leading to diverse symptoms and pathologies; however, the molecular mechanisms responsible for these reactions are largely unknown. We performed Trans-species Expression Quantitative Trait Locus analysis to identify a large number of host genes that respond to malaria parasite infections. Here we functionally characterize one of the host genes called receptor transporter protein 4 (RTP4) in responses to malaria parasite and virus infections. RTP4 is induced by type I IFN (IFN-I) and binds to the TANK-binding kinase (TBK1) complex where it negatively regulates TBK1 signaling by interfering with expression and phosphorylation of both TBK1 and IFN regulatory factor 3. Rtp4 -/- mice were generated and infected with malaria parasite Plasmodiun berghei ANKA. Significantly higher levels of IFN-I response in microglia, lower parasitemia, fewer neurologic symptoms, and better survival rates were observed in Rtp4 -/- than in wild-type mice. Similarly, RTP4 deficiency significantly reduced West Nile virus titers in the brain, but not in the heart and the spleen, of infected mice, suggesting a specific role for RTP4 in brain infection and pathology. This study reveals functions of RTP4 in IFN-I response and a potential target for therapy in diseases with neuropathology.
Collapse
|
21
|
Babamale OA, Opeyemi OA, Bukky AA, Musleem AI, Kelani EO, Okhian BJ, Abu-Bakar N. Association Between Farming Activities and Plasmodium falciparum Transmission in Rural Communities in Nigeria. Malays J Med Sci 2020; 27:105-116. [PMID: 32684811 PMCID: PMC7337949 DOI: 10.21315/mjms2020.27.3.11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 05/08/2020] [Indexed: 11/09/2022] Open
Abstract
Background The connection between malaria-associated morbidities and farming activities has not been succinctly reported. This study aimed to address the connectivity between farming activities and malaria transmission. Methods The study took place in the agricultural setting of Nigeria Edu local government (9° N, 4.9° E) between March 2016 and December 2018. A pre-tested structured questionnaire was administered to obtain information on their occupation and malaria infection. Infection status was confirmed with blood film and microscopic diagnosis of Plasmodium falciparum was based on the presence of ring form or any other blood stages. Individuals who are either critically ill or lived in the community less than 3 months were excluded from the study. Results Of the 341 volunteers, 58.1% (52.9% in Shigo and 61.4% in Sista) were infected (parasitaemia density of 1243.7 parasites/μL blood). The prevalence and intensity of infection were higher among farmers (71.3%, 1922.9 parasites/μL blood, P = 0.005), particularly among rice farmers (2991.6 parasites/μL blood) compared to non-farmer participants. The occurrence and parasite density follow the same pattern for sex and age (P < 0.05). Children in the age of 6 to 10 years (AOR: 2.168, CI: 1.63–2.19) and ≥ 11 years (AOR: 3.750, CI: 2.85–3.80) groups were two-and four-fold more likely to be infected with malaria. The analysis revealed that the proximity of bush and stagnant water to the farmer (73.9%, AOR: 3.242, CI: 2.57–3.61) and non-farmer (38.1%, AOR: 1.362, CI: 1.25–1.41) habitations influence malaria transmission. Conclusion This study highlights farming activities as a risk factor for malaria infection in agro-communities. Integrated malaria control measures in agricultural communities should therefore include water and environmental management practices.
Collapse
Affiliation(s)
| | | | - Abiodun Adebayo Bukky
- Parasitology Unit, Department of Zoology, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Akinkunmi Idris Musleem
- Parasitology Unit, Department of Zoology, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Eniola Olashile Kelani
- Parasitology Unit, Department of Zoology, University of Ilorin, Ilorin, Kwara State, Nigeria
| | | | | |
Collapse
|
22
|
RAGE modulatory effects on cytokines network and histopathological conditions in malarial mice. Exp Parasitol 2020; 216:107946. [PMID: 32622941 DOI: 10.1016/j.exppara.2020.107946] [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: 02/17/2020] [Revised: 06/25/2020] [Accepted: 06/28/2020] [Indexed: 11/23/2022]
Abstract
This study was aimed at investigating the involvement of Receptor for Advanced Glycation End Products (RAGE) during malaria infection and the effects of modulating RAGE on the inflammatory cytokines release and histopathological conditions of affected organs in malarial animal model. Plasmodium berghei (P. berghei) ANKA-infected ICR mice were treated with mRAGE/pAb and rmRAGE/Fc Chimera drugs from day 1 to day 4 post infection. Survival and parasitaemia levels were monitored daily. On day 5 post infection, mice were sacrificed, blood were drawn for cytokines analysis and major organs including kidney, spleen, liver, brain and lungs were extracted for histopathological analysis. RAGE levels were increased systemically during malaria infection. Positive correlation between RAGE plasma concentration and parasitaemia development was observed. Treatment with RAGE related drugs did not improve survival of malaria-infected mice. However, significant reduction on the parasitaemia levels were recorded. On the other hand, inhibition and neutralization of RAGE production during the infection significantly increased the plasma levels of interleukin (IL-4, IL-17A, IL-10 and IL-2) and reduced interferon (IFN)-γ secretion. Histopathological analysis revealed that all treated malarial mice showed a better outcome in histological assessment of affected organs (brain, liver, spleen, lungs and kidney). RAGE is involved in malaria pathogenesis and targeting RAGE could be beneficial in malaria infected host in which RAGE inhibition or neutralization increased the release of anti-inflammatory cytokines (IL-10 and IL-4) and reduce pro-inflammatory cytokine (IFNγ) which may help alleviate tissue injury and improve histopathological conditions of affected organs during the infection.
Collapse
|
23
|
Aguilar R, Campo JJ, Chicuecue S, Cisteró P, Català A, Luis L, Ubillos I, Galatas B, Aide P, Guinovart C, Moncunill G, Dobaño C. Changing plasma cytokine, chemokine and growth factor profiles upon differing malaria transmission intensities. Malar J 2019; 18:406. [PMID: 31806027 PMCID: PMC6896751 DOI: 10.1186/s12936-019-3038-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/26/2019] [Indexed: 12/12/2022] Open
Abstract
Background Malaria epidemiological and immunological data suggest that parasite tolerance wanes in the absence of continuous exposure to the parasite, potentially enhancing pathogenesis. The expansion of control interventions and elimination campaigns raises the necessity to better understand the host factors leading to susceptibility or tolerance that are affected by rapid changes in malaria transmission intensity (MTI). Mediators of cellular immune responses are responsible for the symptoms and pathological alterations during disease and are expected to change rapidly upon malaria exposure or cessation. Methods The plasma concentrations of 30 cytokine, chemokine and growth factors in individuals of all ages from a malaria endemic area of southern Mozambique were compared between 2 years of different MTI: 2010 (lower, n = 234) and 2013 (higher, n = 143). The effect of the year on the correlations between cytokines, chemokines and growth factors and IgGs to Plasmodium falciparum (markers of exposure) was explored. The effects of age, sex, neighbourhood and parasitaemia on analyte levels and their interactions with year were also assessed. Results An inverse correlation of several cellular immune mediators with malarial antibodies in 2013, and a lack of correlation or even a positive correlation in 2010 were observed. Most cytokines, chemokines and growth factors, regardless of their immune function, had higher concentrations in 2010 compared with 2013 in P. falciparum-infected and uninfected subjects. Age and neighbourhood showed an effect on analyte concentrations. Conclusions The results show a different regulation of the cellular immune response in 2010 vs 2013 which could be related to a loss of immune-tolerance after a decline in MTI in 2010 and previous years, and a rapid re-establishment of tolerance as a consequence of more continuous exposure as MTI began increasing in 2012. Cellular immune mediators warrant further investigation as possible surrogates of MTI-associated host susceptibility or tolerance.
Collapse
Affiliation(s)
- Ruth Aguilar
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Roselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Joseph J Campo
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Roselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Silvia Chicuecue
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Pau Cisteró
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Roselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Alba Català
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Roselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Leopoldina Luis
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Itziar Ubillos
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Roselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain
| | - Beatriz Galatas
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Roselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Pedro Aide
- Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Caterina Guinovart
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Roselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain.,Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Gemma Moncunill
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Roselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain.
| | - Carlota Dobaño
- ISGlobal, Hospital Clínic, Universitat de Barcelona, Carrer Roselló 153 (CEK Building), 08036, Barcelona, Catalonia, Spain.
| |
Collapse
|
24
|
Nalwoga A, Webb EL, Chihota B, Miley W, Walusimbi B, Nassuuna J, Sanya RE, Nkurunungi G, Labo N, Elliott AM, Cose S, Whitby D, Newton R. Kaposi's sarcoma-associated herpesvirus seropositivity is associated with parasite infections in Ugandan fishing communities on Lake Victoria islands. PLoS Negl Trop Dis 2019; 13:e0007776. [PMID: 31618208 PMCID: PMC6816576 DOI: 10.1371/journal.pntd.0007776] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 10/28/2019] [Accepted: 09/11/2019] [Indexed: 12/20/2022] Open
Abstract
We investigated the impact of helminths and malaria infection on Kaposi's sarcoma associated herpesvirus (KSHV) seropositivity, using samples and data collected from a cluster-randomised trial of intensive versus standard anthelminthic treatment. The trial was carried out in 2012 to 2016 among fishing communities on Lake Victoria islands in Uganda. Plasma samples from 2881 participants from two household surveys, the baseline (1310 participants) and the final (1571 participants) surveys were tested for KSHV IgG antibody responses to K8.1 and ORF73 recombinant proteins using ELISA. The baseline survey was carried out before the trial intervention while the final survey was carried out after three years of the trial intervention. Additionally, a subset sample of 372 participants from the final survey was tested for IgE, IgG and IgG4 antibody concentrations to S. mansoni adults worm antigen (SWA) and S. mansoni egg antigen (SEA) using ELISA. Infection by helminths (S. mansoni, N. americanus, T. trichiura and S. stercoralis) was diagnosed using real-time PCR, urine circulating cathodic antigen (CCA) and stool microscopy (Kato-Katz method) while malaria infection was diagnosed using microscopy. We analysed the relationship between helminth and malaria infections and KSHV seropositivity using regression modelling, allowing for survey design. At baseline, 56% of the participants were male while 48% of the participants were male in the final survey. The most prevalent helminth infection was S. mansoni (at baseline 52% and 34% in the final survey by microscopy, 86% by CCA and 50% by PCR in the final survey). KSHV seropositivity was 66% (baseline) and 56% (final survey) among those 1-12 years and >80% in those 13+ years in both surveys; malaria parasitaemia prevalence was 7% (baseline) and 4% (final survey). At baseline, individuals infected with S. mansoni (detected by microscopy) were more likely to be KSHV seropositive (aOR = 1.86 (1.16, 2.99) p = 0.012) and had higher anti-K8.1 antibody levels (acoefficient = 0.03 (0.01, 0.06) p = 0.02). In the final survey, S. mansoni (by microscopy, adjusted Odds Ratio (aOR = 1.43 (1.04-1.95), p = 0.028) and malaria parasitaemia (aOR = 3.49 (1.08-11.28), p = 0.038) were positively associated with KSHV seropositivity. Additionally, KSHV seropositive participants had higher S. mansoni-specific IgE and IgG antibody concentrations in plasma. Furthermore, HIV infected individuals on cART were less likely to be KSHV seropositive compared to HIV negative individuals (aOR = 0.46 (0.30, 0.71) p = 0.002). Schistosoma species skew the immune response towards Th2 and regulatory responses, which could impact on KSHV reactivation if co-infected with both organisms.
Collapse
Affiliation(s)
- Angela Nalwoga
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Emily L. Webb
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Belinda Chihota
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Wendell Miley
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | | | | | - Richard E. Sanya
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- College of Health Sciences, Makerere University, Kampala, Uganda
| | - Gyaviira Nkurunungi
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Nazzarena Labo
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Alison M. Elliott
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Stephen Cose
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine, London, United Kingdom
| | - Denise Whitby
- Viral Oncology Section, AIDS and Cancer Virus Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, United States of America
| | - Robert Newton
- MRC/UVRI and LSHTM Uganda Research Unit, Entebbe, Uganda
- University of York, York; United Kingdom
| |
Collapse
|
25
|
Huang B, Huang S, Chen X, Liu XB, Wu Q, Wang Y, Li X, Li K, Gao H, Cen S, Lin R, Liu Z, Jin X. Activation of Mast Cells Promote Plasmodium berghei ANKA Infection in Murine Model. Front Cell Infect Microbiol 2019; 9:322. [PMID: 31552201 PMCID: PMC6747038 DOI: 10.3389/fcimb.2019.00322] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 08/27/2019] [Indexed: 12/12/2022] Open
Abstract
Malaria, a mosquito-borne infectious disease, is a severe health problem worldwide. As reported, some anti-malarial drugs with anti-parasitic properties also block mast cells (MCs) activities. It is hypothesized that MCs activity may be correlated with the pathogenesis of malaria. Thus, the role of MCs on malarial pathogenesis and the involved physiological action and pathways need to be further investigated. This study aimed to investigate the effect of MCs activation on malaria disease severity using KunMing mice with Plasmodium berghei ANKA (PbANKA) infection treated with MCs degranulator (compound 48/80, C48/80) or MCs stabilizer (disodium cromoglycate, DSCG). PbANKA infection caused a dramatic increase in MCs density and level of MCs degranulation in cervical lymph node (CLN) and skin. Compared with infected control, C48/80 treatment had shortened survival time, increased parasitemia, exacerbated liver inflammation and CLN hyperplasia, accompanied with increase in vascular leakage and leukocyte number. The infected mice with C48/80 treatment also elevated the release of CCL2, CXCL1, and MMP-9 from MCs in CLN and skin, and TNF-α, IFN-γ, CCR2, and CXCR2 mRNA expression in CLN and liver. In contrast, the infected mice treated with DSCG showed longer survival time, lower parasitemia, improved liver inflammation and CLN hyperplasia, followed by a decline of vascular leakage and leukocyte number. Decreased MCs-derived CCL2, CXCL1, and MMP-9 from CLN and skin, mRNA expression in CLN and liver (TNF-α, IFN-γ, CCR2, and CXCR2) were also observed in infected mice with DSCG treatment. Our data indicated that MCs activation may facilitate the pathogenesis of PbANKA infection.
Collapse
Affiliation(s)
- Bo Huang
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China.,Department of Pathogen Biology and Immunology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Shiguang Huang
- School of Stomatology, Jinan University, Guangzhou, China
| | - Xiaoyan Chen
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Xiao Bo Liu
- Department of Pathogen Biology and Immunology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Qiang Wu
- Department of Pathogen Biology and Immunology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Yongfei Wang
- College of Life Science and Technology, Jinan University, Guangzhou, China
| | - Xiaobo Li
- Department of Pathogen Biology and Immunology, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| | - Kunning Li
- Lady Davis institute for Medical Research, Jewish General Hospital, McGill University, Montreal, QC, Canada.,Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Hongzhi Gao
- Department of Neurosurgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Shan Cen
- Department of Immunology, Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences, Beijing, China
| | - Rongtuan Lin
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Zhenlong Liu
- Division of Experimental Medicine, Department of Medicine, McGill University, Montreal, QC, Canada
| | - Xiaobao Jin
- Guangdong Provincial Key Laboratory of Pharmaceutical Bioactive Substances, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
26
|
The Immunological Plasmodium falciparum Malaria Characteristics of Children in Tajikistan Republic. J Trop Med 2019; 2019:5147252. [PMID: 31308850 PMCID: PMC6594323 DOI: 10.1155/2019/5147252] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/27/2019] [Accepted: 05/21/2019] [Indexed: 11/17/2022] Open
Abstract
The epidemiological situation in Tajikistan Republic deteriorated in the 1990s, when an influx of refugees from Afghanistan resulted in mass importation of Plasmodium vivax and Plasmodium falciparum malaria to Khatlon region. The National Programme of Malaria Control was successful and malaria transmission was interrupted in 2009. Background. The aim of this study was to investigate the mechanisms of immunological response in Tajik children with tropical Plasmodium falciparum malaria. Materials and Methods. We examined 124 patients with P. falciparum malaria at the age of 6 months up to 14 years that were hospitalized in Clinical Infectious Diseases Hospital in Dushanbe city and in Regional hospital of Khatlon region in the period 2000-2007. In most cases, they were school-age children (56%). The peak incidence was recorded in July-October. Verification of the diagnosis was based on clinical, epidemiological data, and the results of blood microscopy. In all patients, along with the standard, clinical, and laboratory tests, a number of indicators of the immune status were performed that include the T-immunity, the content of serum immunoglobulins of three main classes, the level of circulating immune complexes (CIC), C3 complement, and the concentration of key serum cytokines that have been studied in the dynamics of infectious process. Finding. The study of cellular and humoral immunity in patients with Plasmodium falciparum malaria is an obvious additional criterion in assessing the severity of infection. The imbalance of cytokine profile is an important pathogenic factor in the development of severe and recurrent forms of the disease, since the formation of a defective immune response to parasitic antigens contributes to adverse outcomes. Conclusions. Plasmodium falciparum malaria was characterized by depression of cellular and humoral immunity, the degree of which depended on the severity of the pathological process.
Collapse
|
27
|
Association between Inflammatory Cytokine Levels and Thrombocytopenia during Plasmodium falciparum and P. vivax Infections in South-Western Coastal Region of India. Malar Res Treat 2019; 2019:4296523. [PMID: 31110658 PMCID: PMC6487116 DOI: 10.1155/2019/4296523] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Revised: 02/11/2019] [Accepted: 03/26/2019] [Indexed: 12/20/2022] Open
Abstract
Background Thrombocytopenia is a most commonly observed complication during malaria infections. Inflammatory cytokines such as IL-1, IL-6, and IL-10 have been documented in malaria induced thrombocytopaenia. This study was aimed to understand the possible relationship between inflammatory cytokines across varying degrees of thrombocytopenia during P. vivax, P. falciparum, and mixed infections. Methods A hospital-based cross sectional study was conducted at District Wenlock Hospital in Mangaluru, a city situated along the south-western coastal region of Arabian Sea in India. In this study, blood samples from 627 malaria patients were analyzed for infected parasite species, clinical conditions, platelet levels, and key cytokines that are produced in response to infection; samples from 176 uninfected healthy individuals were used as controls. Results The results of our study showed a high prevalence of malarial thrombocytopenia (platelets <150 ×103/μl) in this endemic settings. About 62.7% patients had mild-to-moderate levels of thrombocytopenia and 16% patients had severe thrombocytopenia (platelets <50 × 103/μl). Upon comparison of cytokines across varying degrees of thrombocytopenia, irrespective of infecting species, the levels of TNF-α and IL-10 were significantly higher during thrombocytopenia, whereas IL-6 levels were considerably lower in severe thrombocytopenia patients suffering from P. vivax or P. falciparum infections. The severe clinical complications observed in patients with malarial thrombocytopenia included severe anemia (17.5%), acute renal failure (12.7%), jaundice (27.0%), metabolic acidosis (36.5%), spontaneous bleeding (3.2%), hypoglycemia (25.4%), hyperparasitemia (4.8%), acute respiratory distress syndrome (1.6%), pulmonary edema (19.0%), and cerebral malaria (1.6%) in various combinations. Conclusion Overall, the results of our study suggest that inflammatory cytokines influence the transformation of mild forms of thrombocytopenia into severe forms during malarial infections. Further studies are needed to understand the association of inflammatory cytokine responses with severe malaria complications and thrombocytopenia.
Collapse
|
28
|
Gleeson PJ, O'Regan JA, McHale T, Tuite H, Giblin L, Reddan D. Acute interstitial nephritis with podocyte foot-process effacement complicating Plasmodium falciparum infection. Malar J 2019; 18:58. [PMID: 30823883 PMCID: PMC6397492 DOI: 10.1186/s12936-019-2674-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2017] [Accepted: 02/14/2019] [Indexed: 11/28/2022] Open
Abstract
Background Malarial acute renal failure (MARF) is a component of the severe malaria syndrome, and complicates 1–5% of malaria infections. This form of renal failure has not been well characterized by histopathology. Case presentation A 44 year-old male presented to the emergency department with a 5-day history of fever and malaise after returning from Nigeria. A blood film was positive for Plasmodium falciparum. His creatinine was 616 µmol/L coming from a normal baseline of 89 µmol/L. He had a urine protein:creatinine ratio of 346 mg/mmol (4.4 g/L). He required dialysis. A renal biopsy showed acute interstitial nephritis with podocyte foot-process effacement. He was treated with artesunate and his renal function improved. At 1 year follow-up his creatinine had plateaued at 120 µmol/L with persistent low-grade proteinuria. Conclusion Acute interstitial nephritis and podocyte foot-process effacement might be under-recognized lesions in MARF. Studying the mechanisms of MARF could give insight into the immunopathology of severe malaria.
Collapse
Affiliation(s)
- Patrick J Gleeson
- Department of Nephrology, University College Hospital, Galway, Republic of Ireland. .,Immune Receptors and Renal Immunopathology, INSERM Unit 1149, Centre de Recherche sur l'Inflammation, Université Sorbonne Paris Cité, Paris, France.
| | - John A O'Regan
- Department of Nephrology, University College Hospital, Galway, Republic of Ireland
| | - Teresa McHale
- Department of Pathology, University College Hospital, Galway, Republic of Ireland
| | - Helen Tuite
- Department of Infectious Disease, University College Hospital, Galway, Republic of Ireland
| | - Louise Giblin
- Department of Nephrology, University College Hospital, Galway, Republic of Ireland
| | - Donal Reddan
- Department of Nephrology, University College Hospital, Galway, Republic of Ireland
| |
Collapse
|
29
|
Effect of Indigofera oblongifolia on the Hepatic Oxidative Status and Expression of Inflammatory and Apoptotic Genes during Blood-Stage Murine Malaria. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:8264861. [PMID: 30838089 PMCID: PMC6374864 DOI: 10.1155/2019/8264861] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Revised: 11/27/2018] [Accepted: 12/09/2018] [Indexed: 01/24/2023]
Abstract
Malaria is a dangerous disease spread across several countries. Recent studies have focused on medicinal plants to discover alternative agents to the currently used drugs for malaria treatment. Here, we investigated the potential role of Indigofera oblongifolia leaf extract (IE) on hepatic inflammation in mice with Plasmodium chabaudi-infected erythrocytes. Female C57BL/6 mice were divided into three groups. The first group served as a control noninfected group, while the second and third groups were intraperitoneally injected with 106 erythrocytes parasitized by P. chabaudi. Mice from the third group were treated daily with a dose of 100 mg/kg of IE for 7 days. IE significantly reduced the number of leukocytes and apoptotic cells. The numbers of CD68-positive cells decreased in the livers of mice from the treatment group. Moreover, IE raised the hepatic antioxidant levels (glutathione and catalase) and reduced the levels of hepatic oxidative stress markers (malondialdehyde, nitric oxide, and reactive oxygen species). IE regulated some functions of the genes related to immune responses, including apoptotic genes (B-cell lymphoma-2, Bax, and caspase-3) and cytokine genes (interleukin-1β (IL-1β), IL-6, interferon-γ, and tumor necrosis factor-α). Therefore, IE exerts significant effects against malaria and protects the liver from injury caused by P. chabaudi via antioxidant and anti-inflammatory ways.
Collapse
|
30
|
Gowda DC, Wu X. Parasite Recognition and Signaling Mechanisms in Innate Immune Responses to Malaria. Front Immunol 2018; 9:3006. [PMID: 30619355 PMCID: PMC6305727 DOI: 10.3389/fimmu.2018.03006] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2018] [Accepted: 12/05/2018] [Indexed: 12/20/2022] Open
Abstract
Malaria caused by the Plasmodium family of parasites, especially P.falciparum and P. vivax, is a major health problem in many countries in the tropical and subtropical regions of the world. The disease presents a wide array of systemic clinical conditions and several life-threatening organ pathologies, including the dreaded cerebral malaria. Like many other infectious diseases, malaria is an inflammatory response-driven disease, and positive outcomes to infection depend on finely tuned regulation of immune responses that efficiently clear parasites and allow protective immunity to develop. Immune responses initiated by the innate immune system in response to parasites play key roles both in protective immunity development and pathogenesis. Initial pro-inflammatory responses are essential for clearing infection by promoting appropriate cell-mediated and humoral immunity. However, elevated and prolonged pro-inflammatory responses owing to inappropriate cellular programming contribute to disease conditions. A comprehensive knowledge of the molecular and cellular mechanisms that initiate immune responses and how these responses contribute to protective immunity development or pathogenesis is important for developing effective therapeutics and/or a vaccine. Historically, in efforts to develop a vaccine, immunity to malaria was extensively studied in the context of identifying protective humoral responses, targeting proteins involved in parasite invasion or clearance. The innate immune response was thought to be non-specific. However, during the past two decades, there has been a significant progress in understanding the molecular and cellular mechanisms of host-parasite interactions and the associated signaling in immune responses to malaria. Malaria infection occurs at two stages, initially in the liver through the bite of a mosquito, carrying sporozoites, and subsequently, in the blood through the invasion of red blood cells by merozoites released from the infected hepatocytes. Soon after infection, both the liver and blood stage parasites are sensed by various receptors of the host innate immune system resulting in the activation of signaling pathways and production of cytokines and chemokines. These immune responses play crucial roles in clearing parasites and regulating adaptive immunity. Here, we summarize the knowledge on molecular mechanisms that underlie the innate immune responses to malaria infection.
Collapse
Affiliation(s)
- D Channe Gowda
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| | - Xianzhu Wu
- Department of Biochemistry and Molecular Biology, The Pennsylvania State University College of Medicine, Hershey, PA, United States
| |
Collapse
|
31
|
Vallejo AF, Read RC, Arevalo-Herrera M, Herrera S, Elliott T, Polak ME. Malaria systems immunology: Plasmodium vivax induces tolerance during primary infection through dysregulation of neutrophils and dendritic cells. J Infect 2018; 77:440-447. [PMID: 30248353 PMCID: PMC6203889 DOI: 10.1016/j.jinf.2018.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2018] [Revised: 09/14/2018] [Accepted: 09/15/2018] [Indexed: 12/19/2022]
Abstract
OBJECTIVES To dissect the transcriptional networks underpinning immune cells responses during primary Plasmodium vivax infection of healthy human adults. METHODS We conducted network co-expression analysis of next-generation RNA sequencing data from whole blood from P. vivax and P. falciparum controlled human malaria infection (CHMI) of healthy naïve and malaria-exposed volunteers. Single cell transcription signatures were used to deconvolute the bulk RNA-Seq data into cell-specific signals. RESULTS Initial exposure to P. vivax induced activation of innate immunity, including efficient antigen presentation and complement activation. However, this effect was accompanied by strong immunosuppression mediated by dendritic cells via the induction of Indoleamine 2,3-Dioxygenase 1(IDO1) and Lymphocyte Activation Gene 3 (LAG3). Additionally, P. vivax induced depletion of neutrophil populations associated with down regulation of 3G-protein coupled receptors, CRXCR1, CXCR2 and CSF3R. Accordingly, in malaria-exposed volunteers the inflammatory response was attenuated, with a decreased class II antigen presentation in dendritic cells. While the immunosuppressive signalling was maintained between plasmodium species, response to P. falciparum was significantly more immunogenic. CONCLUSIONS In silico analyses suggest that primary infection with P. vivax induces potent immunosuppression mediated by dendritic cells, conditioning subsequent anti-malarial immune responses. Targeting immune evasion mechanisms could be an effective alternative for improving vaccine efficacy.
Collapse
Affiliation(s)
- Andres F Vallejo
- Clinical and Experimental Sciences and NIHR Southampton Biomedical Research Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, LE59, MP813, SO16 6YD, Southampton, UK
| | - Robert C Read
- Clinical and Experimental Sciences and NIHR Southampton Biomedical Research Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, LE59, MP813, SO16 6YD, Southampton, UK
| | - Myriam Arevalo-Herrera
- Caucaseco Scientific Research Center, Cali, 760043, Colombia; School of Health, Universidad del Valle, Cali, 76001, Colombia
| | | | - Tim Elliott
- Centre for Cancer Immunology, Faculty of Medicine, University of Southampton, SO16 6YD Southampton, UK; Institute for Life Sciences, University of Southampton, SO17 1BJ, UK
| | - Marta E Polak
- Clinical and Experimental Sciences and NIHR Southampton Biomedical Research Centre, Faculty of Medicine, University of Southampton, Southampton General Hospital, LE59, MP813, SO16 6YD, Southampton, UK; Institute for Life Sciences, University of Southampton, SO17 1BJ, UK.
| |
Collapse
|
32
|
Xia L, Wu J, Pattaradilokrat S, Tumas K, He X, Peng YC, Huang R, Myers TG, Long CA, Wang R, Su XZ. Detection of host pathways universally inhibited after Plasmodium yoelii infection for immune intervention. Sci Rep 2018; 8:15280. [PMID: 30327482 PMCID: PMC6191451 DOI: 10.1038/s41598-018-33599-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/26/2018] [Indexed: 12/16/2022] Open
Abstract
Malaria is a disease with diverse symptoms depending on host immune status and pathogenicity of Plasmodium parasites. The continuous parasite growth within a host suggests mechanisms of immune evasion by the parasite and/or immune inhibition in response to infection. To identify pathways commonly inhibited after malaria infection, we infected C57BL/6 mice with four Plasmodium yoelii strains causing different disease phenotypes and 24 progeny of a genetic cross. mRNAs from mouse spleens day 1 and/or day 4 post infection (p.i.) were hybridized to a mouse microarray to identify activated or inhibited pathways, upstream regulators, and host genes playing an important role in malaria infection. Strong interferon responses were observed after infection with the N67 strain, whereas initial inhibition and later activation of hematopoietic pathways were found after infection with 17XNL parasite, showing unique responses to individual parasite strains. Inhibitions of pathways such as Th1 activation, dendritic cell (DC) maturation, and NFAT immune regulation were observed in mice infected with all the parasite strains day 4 p.i., suggesting universally inhibited immune pathways. As a proof of principle, treatment of N67-infected mice with antibodies against T cell receptors OX40 or CD28 to activate the inhibited pathways enhanced host survival. Controlled activation of these pathways may provide important strategies for better disease management and for developing an effective vaccine.
Collapse
Affiliation(s)
- Lu Xia
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA.,State Key Laboratory of Medical Genetics, Xiangya School of Medicine, Central South University, Changsha, Hunan, 410078, The People's Republic of China
| | - Jian Wu
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Sittiporn Pattaradilokrat
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA.,Department of Biology, Faculty of Science, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Keyla Tumas
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Xiao He
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Yu-Chih Peng
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Ruili Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Timothy G Myers
- Genomic Technologies Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Carole A Long
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA
| | - Rongfu Wang
- Center for Inflammation and Epigenetics, Houston Methodist Research Institute, Houston, TX, 77030, USA
| | - Xin-Zhuan Su
- Malaria Functional Genomics Section, Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Disease, National Institutes of Health, Bethesda, MD, 20892-8132, USA.
| |
Collapse
|
33
|
Costa AG, Ramasawmy R, Val FFA, Ibiapina HNS, Oliveira AC, Tarragô AM, Garcia NP, Heckmann MIO, Monteiro WM, Malheiro A, Lacerda MVG. Polymorphisms in TLRs influence circulating cytokines production in Plasmodium vivax malaria. Cytokine 2018; 110:374-380. [DOI: 10.1016/j.cyto.2018.04.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 03/22/2018] [Accepted: 04/07/2018] [Indexed: 02/08/2023]
|
34
|
Ourives SS, Borges QI, Dos Santos DSA, Melo ECM, de Souza RM, Damazo AS. Analysis of the lymphocyte cell population during malaria caused by Plasmodium vivax and its correlation with parasitaemia and thrombocytopaenia. Malar J 2018; 17:303. [PMID: 30126413 PMCID: PMC6102853 DOI: 10.1186/s12936-018-2443-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2018] [Accepted: 08/04/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND The mechanisms of activation and regulation of T lymphocytes and their cytokines in malaria caused by Plasmodium vivax are complex and poorly understood. Previous data suggest that T cells balance protective immune responses with immune mediated pathology in malaria. This study investigates the lymphocytic profile of patients infected with P. vivax by identifying and quantifying the specific sub-populations of Th1, Th2, Th17 and Treg cells and observing the correlation between parasitaemia and the number of platelets. METHODS A cross-sectional study was carried out in an endemic area of the state of Acre, Brazil. In order to obtain identification and quantification of lymphocyte sub-populations through flow cytometry, blood samples were collected from 50 individuals infected with P. vivax and 20 non-infected controls. To differentiate Th1 from Th2, the presence of cytokines IL-4 and TNF was examined by enzyme-linked immunosorbent assay. Utilizing the Mann-Whitney and Spearman coefficient tests, comparison and correlation analysis were rendered to test the parasitaemia and the number of platelets relationship. RESULTS The data indicate that individuals infected with P. vivax present a significant reduction in Th1, Th2 and Th17 cell sub-populations when compared to the non-infected control group. A negative correlation exists between parasitaemia and platelet counts in individuals infected with P. vivax. There is no correlation of parasitaemia or thrombocytopaenia with any sub-population of T lymphocytes analysed. Interestingly, patients with serum Th1 cytokine profile present inversely proportional parasitaemia to the increase in the number of Th1, Th2, Th17 and Treg cells while patients with serum Th2 cytokine profile present directly proportional parasitaemia to the increase in number of Th1 and Th2 cells. Regarding the number of platelets, patients with serum Th1 cytokine profile show a correlation directly proportional to the Th17 sub-population. In contrast, platelet counts are directly proportional only to Treg and activated Treg cells in patients with serum Th2 cytokine profile. CONCLUSIONS During the P. vivax infection patients with serum Th1 versus Th2 cytokine profile present different biological mechanisms for activating the immune system against parasite load.
Collapse
Affiliation(s)
- Samantha Soares Ourives
- Faculty of Medicine (FM), Federal University of Mato Grosso (UFMT), Cuiabá, Mato Grosso, 78060-900, Brazil
| | - Quessi Irias Borges
- Faculty of Medicine (FM), Federal University of Mato Grosso (UFMT), Cuiabá, Mato Grosso, 78060-900, Brazil
| | | | | | - Rodrigo Medeiros de Souza
- Centre for Health Sciences and Sport, Federal University of Acre (UFAC), Cruzeiro do Sul, AC, 69980000, Brazil
| | - Amílcar Sabino Damazo
- Faculty of Medicine (FM), Federal University of Mato Grosso (UFMT), Cuiabá, Mato Grosso, 78060-900, Brazil. .,Department of Basic Science in Health Faculty of Medicine (FM), Federal University of Mato Grosso (UFMT), Cuiabá, Mato Grosso, 78060-900, Brazil.
| |
Collapse
|
35
|
Interferon- γ and Interleukin-10 Responses during Clinical Malaria Episodes in Infants Aged 0-2 Years Prenatally Exposed to Plasmodium falciparum: Tanzanian Birth Cohort. J Trop Med 2018; 2018:6847498. [PMID: 30154871 PMCID: PMC6091450 DOI: 10.1155/2018/6847498] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Revised: 06/21/2018] [Accepted: 07/11/2018] [Indexed: 12/17/2022] Open
Abstract
Background Infants born to mothers with placental malaria are prenatally exposed to Plasmodium falciparum antigens. However, the effect of that exposure to subsequent immune responses has not been fully elucidated. This study aimed at determining the effect of prenatal exposure to P. falciparum on Interleukin-10 and Interferon-γ responses during clinical malaria episodes in the first 24 months of life. Methods This prospective cohort study involved 215 infants aged 0-2 years born to mothers with or without placental malaria. Enzyme-linked immunosorbent assay (ELISA) was used to determine levels of IL-10 and IFN-γ in infants and detect IgM in cord blood. Data were analyzed using SPSS version 20. Findings Geometric mean for IFN-γ in exposed infants was 557.9 pg/ml (95% CI: 511.6-604.1) and in unexposed infants it was 634.4 pg/ml (95% CI: 618.2-668.5) (P=0.02). Mean IL-10 was 22.4 pg/ml (95% CI: 19.4-28.4) and 15.1 pg/ml (95%CI: 12.4-17.6), respectively (P=0.01). Conclusions Prenatal exposure to P. falciparum antigens significantly affects IL-10 and IFN-γ responses during clinical malaria episodes in the first two years of life.
Collapse
|
36
|
Wu X, Gowda NM, Kawasawa YI, Gowda DC. A malaria protein factor induces IL-4 production by dendritic cells via PI3K-Akt-NF-κB signaling independent of MyD88/TRIF and promotes Th2 response. J Biol Chem 2018; 293:10425-10434. [PMID: 29666186 PMCID: PMC6036203 DOI: 10.1074/jbc.ac118.001720] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 04/06/2018] [Indexed: 11/06/2022] Open
Abstract
Dendritic cells (DC) and cytokines produced by DC play crucial roles in inducing and regulating pro-/anti-inflammatory and Th1/Th2 responses. DC are known to produce a Th1-promoting cytokine, interleukin (IL)-12, in response to malaria and other pathogenic infections, but it is thought that DC do not produce Th2-promoting cytokine, IL-4. Here, we show that a protein factor of malaria parasites induces IL-4 responses by CD11chiMHCIIhiCD3ϵ-CD49b-CD19-FcϵRI- DC via PI3K-Akt-NF-κB signaling independent of TLR-MyD88/TRIF. Malaria parasite-activated DC induced IL-4 responses by T cells both in vitro and in vivo, favoring Th2, and il-4-deficient DC were unable to induce IL-4 expression by T cells. Interestingly, lethal parasites, Plasmodium falciparum and Plasmodium berghei ANKA, induced IL-4 response primarily by CD8α- DC, whereas nonlethal Plasmodium yoelii induced IL-4 by both CD8α+ and CD8α- DC. In both P. berghei ANKA- and P. yoelii-infected mice, IL-4-expressing CD8α- DC did not express IL-12, but a distinct CD8α- DC subset expressed IL-12. In P. berghei ANKA infection, CD8α+ DC expressed IL-12 but not IL-4, whereas in P. yoelii infection, CD8α+ DC expressed IL-4 but not IL-12. These differential IL-4 and IL-12 responses by DC subsets may contribute to different Th1/Th2 development and clinical outcomes in lethal and nonlethal malaria. Our results for the first time demonstrate that a malaria protein factor induces IL-4 production by DC via PI3K-Akt-NF-κB signaling, revealing signaling and molecular mechanisms that initiate and promote Th2 development.
Collapse
Affiliation(s)
- Xianzhu Wu
- From the Department of Biochemistry and Molecular Biology and
| | - Nagaraj M Gowda
- From the Department of Biochemistry and Molecular Biology and
| | - Yuka I Kawasawa
- From the Department of Biochemistry and Molecular Biology and
- the Department of Pharmacology and the Institute for Personalized Medicine, Pennsylvania State University College of Medicine, Hershey, Pennsylvania 17033
| | - D Channe Gowda
- From the Department of Biochemistry and Molecular Biology and
| |
Collapse
|
37
|
Bwanika R, Kato CD, Welishe J, Mwandah DC. Cytokine profiles among patients co-infected with Plasmodium falciparum malaria and soil borne helminths attending Kampala International University Teaching Hospital, in Uganda. ALLERGY, ASTHMA, AND CLINICAL IMMUNOLOGY : OFFICIAL JOURNAL OF THE CANADIAN SOCIETY OF ALLERGY AND CLINICAL IMMUNOLOGY 2018; 14:10. [PMID: 29560020 PMCID: PMC5858126 DOI: 10.1186/s13223-018-0235-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 02/06/2018] [Indexed: 01/22/2023]
Abstract
BACKGROUND Malaria and helminths share the same geographical distribution in tropical Africa. Studies of the interaction of helminth and malaria co-infection in humans have been few and are mainly epidemiological, with little information on cellular immune responses. This study aimed to determine Cytokine profiles among patients co-infected with Plasmodium falciparum malaria and soil borne helminth attending Kampala International University Teaching Hospital (KIU). METHODS A case control study of 240 patients were recruited at KIU teaching hospital. Patients with Plasmodium falciparum malaria were 55 (22.9%) and those with soil-borne helminths were 63 (26.3%). The controls were 89 (37.1%), while those co-infected with Plasmodium falciparum malaria and soil-borne helminths were 33 (13.8%). Cases were defined as having a positive blood smear for P. falciparum malaria, those with helminths or co-infections of the two. Negative controls were those with a negative blood smear for P. falciparum malaria and those with no stool parasitic infections. Patients presenting with signs and symptoms of malaria or those suspected of having helminths were recruited for the study. A panel of five cytokines (IFN-γ, TNF-α, IL-6, TGF-β and IL-10) were assayed from plasma samples in patients with and without Plasmodium falciparum malaria, patients with and without helminth, and then those co-infected with the two diseases diagnosis was done using thick blood smears stained with 10% Giemsa and stool examination was done following the Kato Katz technique following standard procedures. RESULTS The prevalence of Plasmodium falciparum malaria by sex was 28 (11.7%) and 27 (11.3%) in male and female respectively. The overall prevalence of soil borne helminth was 26.3%, and among those harbouring helminths, 13.8% were co-infected with Plasmodium falciparum. Cytokine levels significantly differed across Plasmodium falciparum malaria, soil borne helminth infected patients and health controls for IFN-γ (P = 0.023), IL-10 (P = 0.008) and TGF-β (P = 0.0001). Cytokine levels significantly differed across Plasmodium falciparum malaria, soil borne helminth infected patients and patients co-infected with Plasmodium falciparum malaria and soil borne helminth for IL-10 (P = 0.004), IL-6 (P = 0.011) and TGF-β (P = 0.003). CONCLUSION An up-regulation of IFN-γ during Plasmodium falciparum malaria and an up-regulation of IL-10 and TGF-β in soil borne helminth infections was demonstrated. We demonstrate that co-infections of Plasmodium falciparum and soil borne helminth lead to an up-regulation of IL-10 and IL-6 and a down-regulation of TGF-β.Trial registration No17/10-16.
Collapse
Affiliation(s)
- Richard Bwanika
- School of Biomedical Sciences, Department of Microbiology, Kampala International University, Western Campus, Ishaka, Box 71, Bushenyi, Uganda
| | - Charles D. Kato
- School of Biomedical Sciences, Department of Microbiology, Kampala International University, Western Campus, Ishaka, Box 71, Bushenyi, Uganda
- School of Bio-security, Biotechnical & Laboratory Sciences, College of Veterinary Medicine, Animal Resources & Bio-security, Makerere University, P.O Box 7062, Kampala, Uganda
| | - Johnson Welishe
- School of Biomedical Sciences, Department of Microbiology, Kampala International University, Western Campus, Ishaka, Box 71, Bushenyi, Uganda
- School of Bio-security, Biotechnical & Laboratory Sciences, College of Veterinary Medicine, Animal Resources & Bio-security, Makerere University, P.O Box 7062, Kampala, Uganda
| | - Daniel C. Mwandah
- School of Biomedical Sciences, Department of Microbiology, Kampala International University, Western Campus, Ishaka, Box 71, Bushenyi, Uganda
| |
Collapse
|
38
|
Junaid QO, Khaw LT, Mahmud R, Ong KC, Lau YL, Borade PU, Liew JWK, Sivanandam S, Wong KT, Vythilingam I. Pathogenesis of Plasmodium berghei ANKA infection in the gerbil (Meriones unguiculatus) as an experimental model for severe malaria. ACTA ACUST UNITED AC 2017; 24:38. [PMID: 29034874 PMCID: PMC5642054 DOI: 10.1051/parasite/2017040] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 09/26/2017] [Indexed: 01/10/2023]
Abstract
Background: As the quest to eradicate malaria continues, there remains a need to gain further understanding of the disease, particularly with regard to pathogenesis. This is facilitated, apart from in vitro and clinical studies, mainly via in vivo mouse model studies. However, there are few studies that have used gerbils (Meriones unguiculatus) as animal models. Thus, this study is aimed at characterizing the effects of Plasmodium berghei ANKA (PbA) infection in gerbils, as well as the underlying pathogenesis. Methods: Gerbils, 5-7 weeks old were infected by PbA via intraperitoneal injection of 1 × 106 (0.2 mL) infected red blood cells. Parasitemia, weight gain/loss, hemoglobin concentration, red blood cell count and body temperature changes in both control and infected groups were monitored over a duration of 13 days. RNA was extracted from the brain, spleen and whole blood to assess the immune response to PbA infection. Organs including the brain, spleen, heart, liver, kidneys and lungs were removed aseptically for histopathology. Results: Gerbils were susceptible to PbA infection, showing significant decreases in the hemoglobin concentration, RBC counts, body weights and body temperature, over the course of the infection. There were no neurological signs observed. Both pro-inflammatory (IFNγ and TNF) and anti-inflammatory (IL-10) cytokines were significantly elevated. Splenomegaly and hepatomegaly were also observed. PbA parasitized RBCs were observed in the organs, using routine light microscopy and in situ hybridization. Conclusion: Gerbils may serve as a good model for severe malaria to further understand its pathogenesis.
Collapse
Affiliation(s)
- Quazim Olawale Junaid
- Department of Parasitology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia - Department of Biological Science, Faculty of Science, Federal University Kashere, Gombe State, Nigeria
| | - Loke Tim Khaw
- Department of Parasitology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia - Department of Pathology, School of Medicine, International Medical University, 57000 Kuala Lumpur, Malaysia
| | - Rohela Mahmud
- Department of Parasitology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Kien Chai Ong
- Department of Biomedical Science, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Yee Ling Lau
- Department of Parasitology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Prajakta Uttam Borade
- Department of Pathology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Jonathan Wee Kent Liew
- Department of Parasitology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Sinnadurai Sivanandam
- Department of Parasitology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Kum Thong Wong
- Department of Pathology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| | - Indra Vythilingam
- Department of Parasitology, Faculty of Medicine, University of Malaya, Lembah Pantai, 50603 Kuala Lumpur, Malaysia
| |
Collapse
|
39
|
Soni R, Sharma D, Rai P, Sharma B, Bhatt TK. Signaling Strategies of Malaria Parasite for Its Survival, Proliferation, and Infection during Erythrocytic Stage. Front Immunol 2017; 8:349. [PMID: 28400771 PMCID: PMC5368685 DOI: 10.3389/fimmu.2017.00349] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 03/10/2017] [Indexed: 12/22/2022] Open
Abstract
Irrespective of various efforts, malaria persist the most debilitating effect in terms of morbidity and mortality. Moreover, the existing drugs are also vulnerable to the emergence of drug resistance. To explore the potential targets for designing the most effective antimalarial therapies, it is required to focus on the facts of biochemical mechanism underlying the process of parasite survival and disease pathogenesis. This review is intended to bring out the existing knowledge about the functions and components of the major signaling pathways such as kinase signaling, calcium signaling, and cyclic nucleotide-based signaling, serving the various aspects of the parasitic asexual stage and highlighted the Toll-like receptors, glycosylphosphatidylinositol-mediated signaling, and molecular events in cytoadhesion, which elicit the host immune response. This discussion will facilitate a look over essential components for parasite survival and disease progression to be implemented in discovery of novel antimalarial drugs and vaccines.
Collapse
Affiliation(s)
- Rani Soni
- Department of Biotechnology, School of Life sciences, Central University of Rajasthan , Ajmer , India
| | - Drista Sharma
- Department of Biotechnology, School of Life sciences, Central University of Rajasthan , Ajmer , India
| | - Praveen Rai
- Department of Biotechnology, School of Life sciences, Central University of Rajasthan , Ajmer , India
| | - Bhaskar Sharma
- Department of Biotechnology, School of Life sciences, Central University of Rajasthan , Ajmer , India
| | - Tarun K Bhatt
- Department of Biotechnology, School of Life sciences, Central University of Rajasthan , Ajmer , India
| |
Collapse
|
40
|
Hojo-Souza NS, Pereira DB, de Souza FSH, de Oliveira Mendes TA, Cardoso MS, Tada MS, Zanini GM, Bartholomeu DC, Fujiwara RT, Bueno LL. On the cytokine/chemokine network during Plasmodium vivax malaria: new insights to understand the disease. Malar J 2017; 16:42. [PMID: 28118834 PMCID: PMC5260126 DOI: 10.1186/s12936-017-1683-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 01/05/2017] [Indexed: 11/10/2022] Open
Abstract
Background The clinical outcome of malaria depends on the delicate balance between pro-inflammatory and immunomodulatory cytokine responses triggered during infection. Despite the numerous reports on characterization of plasma levels of cytokines/chemokines, there is no consensus on the profile of these mediators during blood stage malaria. The identification of acute phase biomarkers might contribute to a better understanding of the disease, allowing the use of more effective therapeutic approaches to prevent the progression towards severe disease. In the present study, the plasma levels of cytokines and chemokines and their association with parasitaemia and number of previous malaria episodes were evaluated in Plasmodium vivax-infected patients during acute and convalescence phase, as well as in healthy donors. Methods Samples of plasma were obtained from peripheral blood samples from four different groups: P. vivax-infected, P. vivax-treated, endemic control and malaria-naïve control. The cytokine (IL-6, IL-10, IL-17, IL-27, TGF-β, IFN-γ and TNF) and chemokine (MCP-1/CCL2, IP-10/CXCL10 and RANTES/CCL5) plasma levels were measured by CBA or ELISA. The network analysis was performed using Spearman correlation coefficient. Results Plasmodium vivax infection induced a pro-inflammatory response driven by IL-6 and IL-17 associated with an immunomodulatory profile mediated by IL-10 and TGF-β. In addition, a reduction was observed of IFN-γ plasma levels in P. vivax group. A lower level of IL-27 was observed in endemic control group in comparison to malaria-naïve control group. No significant results were found for IL-12p40 and TNF. It was also observed that P. vivax infection promoted higher levels of MCP-1/CCL2 and IP-10/CXCL10 and lower levels of RANTES/CCL5. The plasma level of IL-10 was elevated in patients with high parasitaemia and with more than five previous malaria episodes. Furthermore, association profile between cytokine and chemokine levels were observed by correlation network analysis indicating signature patterns associated with different parasitaemia levels. Conclusions The P. vivax infection triggers a balanced immune response mediated by IL-6 and MCP-1/CCL2, which is modulated by IL-10. In addition, the results indicated that IL-10 plasma levels are influenced by parasitaemia and number of previous malaria episodes.
Collapse
Affiliation(s)
- Natália Satchiko Hojo-Souza
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Fernanda Sumika Hojo de Souza
- Departamento de Ciência da Computação, Universidade Federal de São João del-Rei, São João del-Rei, Minas Gerais, Brazil
| | | | - Mariana Santos Cardoso
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | | | - Graziela Maria Zanini
- Instituto de Pesquisa Clínica Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniella Castanheira Bartholomeu
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Ricardo Toshio Fujiwara
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil
| | - Lilian Lacerda Bueno
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, Minas Gerais, Brazil.
| |
Collapse
|
41
|
Chakarov N, Pauli M, Krüger O. Immune responses link parasite genetic diversity, prevalence and plumage morphs in common buzzards. Evol Ecol 2016. [DOI: 10.1007/s10682-016-9871-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
42
|
Fotoran WL, Colhone MC, Ciancaglini P, Stabeli RG, Wunderlich G. Merozoite-Protein Loaded Liposomes Protect against Challenge in Two Murine Models of Plasmodium Infection. ACS Biomater Sci Eng 2016; 2:2276-2286. [DOI: 10.1021/acsbiomaterials.6b00492] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Wesley L. Fotoran
- Department
of Parasitology, Institute for Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Marcelle C. Colhone
- Department
of Chemistry, FFCLRP-USP, University of São Paulo, Ribeirão
Preto, Brazil
| | - Pietro Ciancaglini
- Department
of Chemistry, FFCLRP-USP, University of São Paulo, Ribeirão
Preto, Brazil
| | - Rodrigo G. Stabeli
- Centro de Estudos
de Biomoléculas Aplicadas a Saúde, Fiocruz−Fundação
Oswaldo Cruz, Ministério da Saúde, e Departamento de
Medicina da Universidade Federal de Rondônia (UNIR), Porto Velho, Rondônia, Brazil
| | - Gerhard Wunderlich
- Department
of Parasitology, Institute for Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
43
|
Zander RA, Guthmiller JJ, Graham AC, Pope RL, Burke BE, Carr DJ, Butler NS. Type I Interferons Induce T Regulatory 1 Responses and Restrict Humoral Immunity during Experimental Malaria. PLoS Pathog 2016; 12:e1005945. [PMID: 27732671 PMCID: PMC5061386 DOI: 10.1371/journal.ppat.1005945] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 09/21/2016] [Indexed: 12/21/2022] Open
Abstract
CD4 T cell-dependent antibody responses are essential for limiting Plasmodium parasite replication and the severity of malaria; however, the factors that regulate humoral immunity during highly inflammatory, Th1-biased systemic infections are poorly understood. Using genetic and biochemical approaches, we show that Plasmodium infection-induced type I interferons limit T follicular helper accumulation and constrain anti-malarial humoral immunity. Mechanistically we show that CD4 T cell-intrinsic type I interferon signaling induces T-bet and Blimp-1 expression, thereby promoting T regulatory 1 responses. We further show that the secreted effector cytokines of T regulatory 1 cells, IL-10 and IFN-γ, collaborate to restrict T follicular helper accumulation, limit parasite-specific antibody responses, and diminish parasite control. This circuit of interferon-mediated Blimp-1 induction is also operational during chronic virus infection and can occur independently of IL-2 signaling. Thus, type I interferon-mediated induction of Blimp-1 and subsequent expansion of T regulatory 1 cells represent generalizable features of systemic, inflammatory Th1-biased viral and parasitic infections that are associated with suppression of humoral immunity. Humoral immunity is essential for host resistance to pathogens that trigger highly inflammatory immune responses, including Plasmodium parasites, the causative agents of malaria. Long-lived, secreted antibody responses depend on a specialized subset of CD4 T cells called T follicular helper (Tfh) cells. However, anti-Plasmodium humoral immunity is often short-lived, non-sterilizing, and immunity rapidly wanes, leaving individuals susceptible to repeated bouts of malaria. Here we explored the relationship between inflammatory type I interferons, the regulation of pathogen-specific CD4 T cell responses, and humoral immunity using models of experimental malaria and systemic virus infection. We identified that type I interferons promote the formation and accumulation of pathogen-specific CD4 T regulatory 1 cells that co-express interferon-gamma and interleukin-10. Moreover, we show that the combined activity of interferon-gamma and interleukin-10 limits the magnitude of infection-induced Tfh responses, the secretion of parasite-specific secreted antibody, and parasite control. Our study provides new insight into the regulation of T regulatory 1 responses and humoral immunity during inflammatory immune reactions against systemic infections.
Collapse
Affiliation(s)
- Ryan A. Zander
- Departments of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Jenna J. Guthmiller
- Departments of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Amy C. Graham
- Departments of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Rosemary L. Pope
- Departments of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Bradly E. Burke
- Departments of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Daniel J.J. Carr
- Departments of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Department of Ophthalmology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Graduate Program in Biosciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
| | - Noah S. Butler
- Departments of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- Graduate Program in Biosciences, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, United States of America
- * E-mail:
| |
Collapse
|
44
|
Increased CD40 Expression Enhances Early STING-Mediated Type I Interferon Response and Host Survival in a Rodent Malaria Model. PLoS Pathog 2016; 12:e1005930. [PMID: 27716849 PMCID: PMC5055354 DOI: 10.1371/journal.ppat.1005930] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 09/12/2016] [Indexed: 01/01/2023] Open
Abstract
Both type I interferon (IFN-I) and CD40 play a significant role in various infectious diseases, including malaria and autoimmune disorders. CD40 is mostly known to function in adaptive immunity, but previous observations of elevated CD40 levels early after malaria infection of mice led us to investigate its roles in innate IFN-I responses and disease control. Using a Plasmodium yoelii nigeriensis N67 and C57BL/6 mouse model, we showed that infected CD40-/- mice had reduced STING and serum IFN-β levels day-2 post infection, higher day-4 parasitemia, and earlier deaths. CD40 could greatly enhance STING-stimulated luciferase signals driven by the IFN-β promoter in vitro, which was mediated by increased STING protein levels. The ability of CD40 to influence STING expression was confirmed in CD40-/- mice after malaria infection. Substitutions at CD40 TRAF binding domains significantly decreased the IFN-β signals and STING protein level, which was likely mediated by changes in STING ubiquitination and degradation. Increased levels of CD40, STING, and ISRE driven luciferase signal in RAW Lucia were observed after phagocytosis of N67-infected red blood cells (iRBCs), stimulation with parasite DNA/RNA, or with selected TLR ligands [LPS, poly(I:C), and Pam3CSK4]. The results suggest stimulation of CD40 expression by parasite materials through TLR signaling pathways, which was further confirmed in bone marrow derived dendritic cells/macrophages (BMDCs/BMDMs) and splenic DCs from CD40-/-, TLR3-/- TLR4-/-, TRIF-/-, and MyD88-/- mice after iRBC stimulation or parasite infection. Our data connect several signaling pathways consisting of phagocytosis of iRBCs, recognition of parasite DNA/RNA (possibly GPI) by TLRs, elevated levels of CD40 and STING proteins, increased IFN-I production, and longer host survival time. This study reveals previously unrecognized CD40 function in innate IFN-I responses and protective pathways in infections with malaria strains that induce a strong IFN-I response, which may provide important information for better understanding and management of malaria.
Collapse
|
45
|
Fontana MF, Baccarella A, Craft JF, Boyle MJ, McIntyre TI, Wood MD, Thorn KS, Anidi C, Bayat A, Chung MR, Hamburger R, Kim CY, Pearman E, Pham J, Tang JJ, Boon L, Kamya MR, Dorsey G, Feeney ME, Kim CC. A Novel Model of Asymptomatic Plasmodium Parasitemia That Recapitulates Elements of the Human Immune Response to Chronic Infection. PLoS One 2016; 11:e0162132. [PMID: 27583554 PMCID: PMC5008831 DOI: 10.1371/journal.pone.0162132] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Accepted: 08/17/2016] [Indexed: 11/18/2022] Open
Abstract
In humans, immunity to Plasmodium sp. generally takes the form of protection from symptomatic malaria (i.e., 'clinical immunity') rather than infection ('sterilizing immunity'). In contrast, mice infected with Plasmodium develop sterilizing immunity, hindering progress in understanding the mechanistic basis of clinical immunity. Here we present a novel model in which mice persistently infected with P. chabaudi exhibit limited clinical symptoms despite sustaining patent parasite burdens for many months. Characterization of immune responses in persistently infected mice revealed development of CD4+ T cell exhaustion, increased production of IL-10, and expansion of B cells with an atypical surface phenotype. Additionally, persistently infected mice displayed a dramatic increase in circulating nonclassical monocytes, a phenomenon that we also observed in humans with both chronic Plasmodium exposure and asymptomatic infection. Following pharmacological clearance of infection, previously persistently infected mice could not control a secondary challenge, indicating that persistent infection disrupts the sterilizing immunity that typically develops in mouse models of acute infection. This study establishes an animal model of asymptomatic, persistent Plasmodium infection that recapitulates several central aspects of the immune response in chronically exposed humans. As such, it provides a novel tool for dissection of immune responses that may prevent development of sterilizing immunity and limit pathology during infection.
Collapse
Affiliation(s)
- Mary F. Fontana
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Alyssa Baccarella
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Joshua F. Craft
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Michelle J. Boyle
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
- The Burnet Institute, Center for Biomedical Research, Melbourne, Australia
| | - Tara I. McIntyre
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Matthew D. Wood
- Department of Pathology, Division of Neuropathology, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Kurt S. Thorn
- Department of Biochemistry and Biophysics, University of California San Francisco, San Francisco, California, 94158, United States of America
| | - Chioma Anidi
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Aqieda Bayat
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Me Ree Chung
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Rebecca Hamburger
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Chris Y. Kim
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Emily Pearman
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Jennifer Pham
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Jia J. Tang
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Louis Boon
- EPIRUS Biopharmaceuticals, Utrecht, Netherlands BV
| | - Moses R. Kamya
- School of Medicine, Makerere University College of Health Sciences, Kampala, Uganda
| | - Grant Dorsey
- Division of Infectious Diseases, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Margaret E. Feeney
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
- Division of Pediatric Infectious Diseases and Global Health, Department of Pediatrics, University of California San Francisco, San Francisco, California, 94143, United States of America
| | - Charles C. Kim
- Division of Experimental Medicine, Department of Medicine, University of California San Francisco, San Francisco, California, 94143, United States of America
- * E-mail:
| |
Collapse
|
46
|
Chaves YO, da Costa AG, Pereira MLM, de Lacerda MVG, Coelho-Dos-Reis JG, Martins-Filho OA, Teixeira-Carvalho A, Malheiro A, Monteiro WM, Orlandi PP, Marinho CRF, Nogueira PA. Immune response pattern in recurrent Plasmodium vivax malaria. Malar J 2016; 15:445. [PMID: 27581163 PMCID: PMC5007810 DOI: 10.1186/s12936-016-1501-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Accepted: 08/23/2016] [Indexed: 01/01/2023] Open
Abstract
Background Plasmodium vivax is the causative agent of human malaria of large geographic distribution, with 35 million cases annually. In Brazil, it is the most prevalent species, being responsible by around 70 % of the malaria cases. Methods A cross-sectional study was performed in Manaus (Amazonas, Brazil), including 36 adult patients with primary malaria, 19 with recurrent malaria, and 20 endemic controls. The ex vivo phenotypic features of circulating leukocyte subsets (CD4+ T-cells, CD8+ T-cells, NK, NKT, B, B1 and Treg cells) as well as the plasmatic cytokine profile (IL-2, IL-4, IL-6, IL-10, TNF and IFN-γ) were assessed, aiming at establishing patterns of immune response characteristic of primary malaria vs recurrent malaria as compared to endemic controls. Results The proportion of subjects with high levels of WBC was reduced in malaria patients as compared to the endemic control. Monocytes were diminished particularly in patients with primary malaria. The proportion of subjects with high levels of all lymphocyte subsets was decreased in all malaria groups, regardless their clinical status. Decreased proportion of subjects with high levels of CD4+ and CD8+ T-cells was found especially in the group of patients with recurrent malaria. Data analysis indicated significant increase in the proportion of the subjects with high plasmatic cytokine levels in both malaria groups, characterizing a typical cytokine storm. Recurrent malaria patients displayed the highest plasmatic IL-10 levels, that correlated directly with the CD4+/CD8+ T-cells ratio and the number of malaria episodes. Conclusion The findings confirm that the infection by the P. vivax causes a decrease in peripheral blood lymphocyte subsets, which is intensified in the cases of “recurrent malaria”. The unbalanced CD4+/CD8+ T-cells ratio, as well as increased IL-10 levels were correlated with the number of recurrent malaria episodes. These results suggest that the gradual remodelling of the immune response is dependent on the repeated exposure to the parasite, which involves a strict control of the immune response mediated by the CD4+/CD8+ T-cell unbalance and exacerbated IL-10 secretion. Electronic supplementary material The online version of this article (doi:10.1186/s12936-016-1501-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Yury Oliveira Chaves
- Instituto Leônidas e Maria Deane, Fundação Oswaldo Cruz (FIOCRUZ), Manaus, AM, Brazil
| | - Allyson Guimarães da Costa
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil.,Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, AM, Brazil.,Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil
| | - Marcelo Luís Monteiro Pereira
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Marcus Vinícius Guimarães de Lacerda
- Instituto Leônidas e Maria Deane, Fundação Oswaldo Cruz (FIOCRUZ), Manaus, AM, Brazil.,Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil.,Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, AM, Brazil
| | - Jordana Grazziela Coelho-Dos-Reis
- Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, MG, Brazil
| | - Olindo Assis Martins-Filho
- Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, MG, Brazil
| | - Andréa Teixeira-Carvalho
- Grupo Integrado de Pesquisas em Biomarcadores de Diagnóstico e Monitoração, Centro de Pesquisas René Rachou, Fundação Oswaldo Cruz (FIOCRUZ), Belo Horizonte, MG, Brazil
| | - Adriana Malheiro
- Departamento de Ensino e Pesquisa, Fundação Hospitalar de Hematologia e Hemoterapia do Amazonas (HEMOAM), Manaus, AM, Brazil.,Programa de Pós-Graduação em Imunologia Básica e Aplicada, Universidade Federal do Amazonas (UFAM), Manaus, AM, Brazil
| | - Wuelton Marcelo Monteiro
- Programa de Pós-Graduação em Medicina Tropical, Universidade do Estado do Amazonas (UEA), Manaus, AM, Brazil.,Fundação de Medicina Tropical Dr. Heitor Vieira Dourado (FMT-HVD), Manaus, AM, Brazil
| | | | - Claudio Romero Farias Marinho
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade de São Paulo (USP), São Paulo, SP, Brazil
| | - Paulo Afonso Nogueira
- Instituto Leônidas e Maria Deane, Fundação Oswaldo Cruz (FIOCRUZ), Manaus, AM, Brazil.
| |
Collapse
|
47
|
Sharma D, Soni R, Patel S, Joshi D, Bhatt TK. In-silico studies on DegP protein of Plasmodium falciparum in search of anti-malarials. J Mol Model 2016; 22:201. [PMID: 27491850 DOI: 10.1007/s00894-016-3064-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 07/03/2016] [Indexed: 10/21/2022]
Abstract
Despite encouraging progress over the past decade, malaria caused by the Plasmodium parasite continues to pose an enormous disease burden and is one of the major global health problems. The extreme challenge in malaria management is the resistance of parasites to traditional monochemotherapies like chloroquine and sulfadoxine-pyrimethamine. No vaccine is yet in sight, and the foregoing effective drugs are also losing ground against the disease due to the resistivity of parasites. New antimalarials with novel mechanisms of action are needed to circumvent existing or emerging drug resistance. DegP protein, secretory in nature has been shown to be involved in regulation of thermo-oxidative stress generated during asexual life cycle of Plasmodium, probably required for survival of parasite in host. Considering the significance of protein, in this study, we have generated a three-dimensional structure of PfDegP followed by validation of the modeled structure using several tools like RAMPAGE, ERRAT, and others. We also performed an in-silico screening of small molecule database against PfDegP using Glide. Furthermore, molecular dynamics simulation of protein and protein-ligand complex was carried out using GROMACS. This study substantiated potential drug-like molecules and provides the scope for development of novel antimalarial drugs.
Collapse
Affiliation(s)
- Drista Sharma
- Department of Biotechnology, Central University of Rajasthan, NH-8, Bandarsindri, Rajasthan, India, 305801
| | - Rani Soni
- Department of Biotechnology, Central University of Rajasthan, NH-8, Bandarsindri, Rajasthan, India, 305801
| | - Sachin Patel
- Department of Biotechnology, Central University of Rajasthan, NH-8, Bandarsindri, Rajasthan, India, 305801
| | - Deepti Joshi
- Department of Biotechnology, Central University of Rajasthan, NH-8, Bandarsindri, Rajasthan, India, 305801
| | - Tarun Kumar Bhatt
- Department of Biotechnology, Central University of Rajasthan, NH-8, Bandarsindri, Rajasthan, India, 305801.
| |
Collapse
|
48
|
Goddard A, Leisewitz AL, Kjelgaard-Hansen M, Kristensen AT, Schoeman JP. Excessive Pro-Inflammatory Serum Cytokine Concentrations in Virulent Canine Babesiosis. PLoS One 2016; 11:e0150113. [PMID: 26953797 PMCID: PMC4783066 DOI: 10.1371/journal.pone.0150113] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Accepted: 02/09/2016] [Indexed: 12/21/2022] Open
Abstract
Babesia rossi infection causes a severe inflammatory response in the dog, which is the result of the balance between pro- and anti-inflammatory cytokine secretion. The aim of this study was to determine whether changes in cytokine concentrations were present in dogs with babesiosis and whether it was associated with disease outcome. Ninety-seven dogs naturally infected with B. rossi were studied and fifteen healthy dogs were included as controls. Diagnosis of babesiosis was confirmed by polymerase chain reaction and reverse line blot. Blood samples were collected from the jugular vein at admission, prior to any treatment. Cytokine concentrations were assessed using a canine-specific multiplex assay on an automated analyser. Serum concentrations of interleukin (IL)-2, IL-6, IL-8, IL-10, IL-18, granulocyte-macrophage colony stimulating factor (GM-CSF) and monocyte chemotactic protein-1 (MCP-1) were measured. Twelve of the Babesia-infected dogs died (12%) and 85 survived (88%). Babesia-infected dogs were also divided into those that presented within 48 hours from displaying clinical signs, and those that presented more than 48 hours after displaying clinical signs. Cytokine concentrations were compared between the different groups using the Mann-Whitney U test. IL-10 and MCP-1 concentrations were significantly elevated for the Babesia-infected dogs compared to the healthy controls. In contrast, the IL-8 concentration was significantly decreased in the Babesia-infected dogs compared to the controls. Concentrations of IL-6 and MCP-1 were significantly increased in the non-survivors compared to the survivors. Concentrations for IL-2, IL-6, IL-18 and GM-CSF were significantly higher in those cases that presented during the more acute stage of the disease. These findings suggest that a mixed cytokine response is present in dogs with babesiosis caused by B. rossi, and that an excessive pro-inflammatory response may result in a poor outcome.
Collapse
Affiliation(s)
- Amelia Goddard
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
- * E-mail:
| | - Andrew L. Leisewitz
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - Mads Kjelgaard-Hansen
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Annemarie T. Kristensen
- Department of Veterinary Clinical and Animal Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Johan P. Schoeman
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
49
|
Ribeiro BDP, Cassiano GC, de Souza RM, Cysne DN, Grisotto MAG, de Azevedo dos Santos APS, Marinho CRF, Machado RLD, Nascimento FRF. Polymorphisms in Plasmodium vivax Circumsporozoite Protein (CSP) Influence Parasite Burden and Cytokine Balance in a Pre-Amazon Endemic Area from Brazil. PLoS Negl Trop Dis 2016; 10:e0004479. [PMID: 26943639 PMCID: PMC4778932 DOI: 10.1371/journal.pntd.0004479] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Accepted: 02/01/2016] [Indexed: 12/17/2022] Open
Abstract
Mechanisms involved in severe P. vivax malaria remain unclear. Parasite polymorphisms, parasite load and host cytokine profile may influence the course of infection. In this study, we investigated the influence of circumsporozoite protein (CSP) polymorphisms on parasite load and cytokine profile in patients with vivax malaria. A cross-sectional study was carried out in three cities: São Luís, Cedral and Buriticupu, Maranhão state, Brazil, areas of high prevalence of P. vivax. Interleukin (IL)-2, IL-4, IL-10, IL-6, IL-17, tumor necrosis factor alpha (TNF-α, interferon gamma (IFN-γ and transforming growth factor beta (TGF-β were quantified in blood plasma of patients and in supernatants from peripheral blood mononuclear cell (PBMC) cultures. Furthermore, the levels of cytokines and parasite load were correlated with VK210, VK247 and P. vivax-like CSP variants. Patients infected with P. vivax showed increased IL-10 and IL-6 levels, which correlated with the parasite load, however, in multiple comparisons, only IL-10 kept this association. A regulatory cytokine profile prevailed in plasma, while an inflammatory profile prevailed in PBMC culture supernatants and these patterns were related to CSP polymorphisms. VK247 infected patients showed higher parasitaemia and IL-6 concentrations, which were not associated to IL-10 anti-inflammatory effect. By contrast, in VK210 patients, these two cytokines showed a strong positive correlation and the parasite load was lower. Patients with the VK210 variant showed a regulatory cytokine profile in plasma, while those infected with the VK247 variant have a predominantly inflammatory cytokine profile and higher parasite loads, which altogether may result in more complications in infection. In conclusion, we propose that CSP polymorphisms is associated to the increase of non-regulated inflammatory immune responses, which in turn may be associated with the outcome of infection. Recent evidences have associated P. vivax infections with clinical complications, previously only attributed to P. falciparum malaria. The interaction between host and parasite may contribute to severity of the disease, however, the specific contribution of each factor remains unclear. Previous studies have shown that polymorphisms in Plasmodium vivax CSP may interfere in systemic reactions, response to drug treatments, leading to different symptoms as well as humoral responses. In this study, we investigate whether these polymorphisms could influence the parasite load and cytokine profile, which altogether may influence the malaria outcome. In this sense, studies have shown that subjects with high parasitic loads, responding with production of pro-inflammatory cytokines develop a more severe disease. The present data indicate that VK247 variant are associated with significant higher parasite loads and pro-inflammatory cytokine profile compared to VK210 variant. In this regard, this study demonstrates that P. vivax CSP polymorphisms have systemic effects in the host immune response, and the investigation of immunogenicity of parasite proteins may provide evidences for a better understanding of this infection.
Collapse
Affiliation(s)
- Bruno de Paulo Ribeiro
- Programa de Pós- graduação em Ciências da Saúde, Universidade Federal do Maranhão (UFMA), São Luís, Maranhão, Brazil
- Laboratório de Imunofisiologia, Universidade Federal do Maranhão (UFMA), São Luís, Maranhão, Brazil
| | - Gustavo Capatti Cassiano
- Centro de Investigação de Microrganismos, Faculdade de Medicina de São José do Rio Preto (FAMERP), São José do Rio Preto, São Paulo, Brazil
| | - Rodrigo Medeiros de Souza
- Departamento de Parasitologia, Universidade de São Paulo (ICB/USP), São Paulo, São Paulo, Brazil
- Centro Multidisciplinar, Campus Floresta (Universidade Federal do Acre), Cruzeiro do Sul, Acre, Brazil
| | - Dalila Nunes Cysne
- Laboratório de Imunofisiologia, Universidade Federal do Maranhão (UFMA), São Luís, Maranhão, Brazil
| | | | | | | | - Ricardo Luiz Dantas Machado
- Laboratório de Pesquisa Básica em Malária (Instituto Evandro Chagas / Secretaria de Vigilância em Saúde / Ministério da Saúde—IEC/SVS/MS), Belém, Pará, Brazil
| | | |
Collapse
|
50
|
Galatas B, Bassat Q, Mayor A. Malaria Parasites in the Asymptomatic: Looking for the Hay in the Haystack. Trends Parasitol 2015; 32:296-308. [PMID: 26708404 DOI: 10.1016/j.pt.2015.11.015] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2015] [Revised: 11/21/2015] [Accepted: 11/24/2015] [Indexed: 12/11/2022]
Abstract
With malaria elimination back on the international agenda, programs face the challenge of targeting all Plasmodium infections, not only symptomatic cases. As asymptomatic individuals are unlikely to seek treatment, they are missed by passive surveillance while remaining infectious to mosquitoes, thus acting as silent reservoirs of transmission. To estimate the risk of asymptomatic infections in various phases of malaria elimination, we need a deeper understanding of the underlying mechanisms favoring carriage over disease, which may involve both pathogen and host factors. Here we review our current knowledge on the determinants leading to Plasmodium falciparum symptomless infections. Understanding the host-pathogen interactions that are most likely to affect transitions between malaria disease states could guide the development of tools to tackle asymptomatic carriers in elimination settings.
Collapse
Affiliation(s)
- Beatriz Galatas
- ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Quique Bassat
- ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique
| | - Alfredo Mayor
- ISGlobal, Barcelona Centre for International Health Research (CRESIB), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain; Centro de Investigação em Saúde de Manhiça (CISM), Maputo, Mozambique.
| |
Collapse
|