Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.681
Filter
1.
Expert Rev Vaccines ; 23(1): 645-654, 2024.
Article in English | MEDLINE | ID: mdl-38888098

ABSTRACT

INTRODUCTION: Malaria continues to remain a major global health problem with nearly a quarter of a billion clinical cases and more than 600,000 deaths in 2022. There has been significant progress toward vaccine development, however, poor efficacy of approved vaccines requiring multiple immunizing doses emphasizes the need for continued efforts toward improved vaccines. Progress to date, nonetheless, has provided impetus for malaria elimination. AREAS COVERED: In this review we will focus on diverse immune mechanisms targeting gametocytes in the human host and gametocyte-mediated malaria transmission via the mosquito vector. EXPERT OPINION: To march toward the goal of malaria elimination it will be critical to target the process of malaria transmission by mosquitoes, mediated exclusively by the sexual stages, i.e. male, and female gametocytes, ingested from infected vertebrate host. Studies over several decades have established antigens in the parasite sexual stages developing in the mosquito midgut as attractive targets for the development of transmission blocking vaccines (TBVs). Immune clearance of gametocytes in the vertebrate host can synergize with TBVs and directly aid in maintaining effective transmission reducing immune potential.


Subject(s)
Malaria Vaccines , Malaria , Mosquito Vectors , Vaccine Development , Humans , Malaria Vaccines/immunology , Malaria Vaccines/administration & dosage , Animals , Malaria/prevention & control , Malaria/transmission , Malaria/immunology , Malaria/parasitology , Mosquito Vectors/parasitology , Mosquito Vectors/immunology , Plasmodium/immunology
2.
Trends Parasitol ; 40(6): 466-476, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38714463

ABSTRACT

The Plasmodium parasites that cause malaria undergo asymptomatic development in the parenchymal cells of the liver, the hepatocytes, prior to infecting erythrocytes and causing clinical disease. Traditionally, hepatocytes have been perceived as passive bystanders that allow hepatotropic pathogens such as Plasmodium to develop relatively unchallenged. However, now there is emerging evidence suggesting that hepatocytes can mount robust cell-autonomous immune responses that target Plasmodium, limiting its progression to the blood and reducing the incidence and severity of clinical malaria. Here we discuss our current understanding of hepatocyte cell-intrinsic immune responses that target Plasmodium and how these pathways impact malaria.


Subject(s)
Hepatocytes , Malaria , Plasmodium , Plasmodium/immunology , Plasmodium/physiology , Humans , Malaria/immunology , Malaria/parasitology , Hepatocytes/parasitology , Hepatocytes/immunology , Animals
4.
Rev Alerg Mex ; 71(1): 54, 2024 Feb 01.
Article in Spanish | MEDLINE | ID: mdl-38683072

ABSTRACT

OBJECTIVE: Analyze the molecular mimicry between Plasmodium spp. and autoantigens associated with GBS, identifying possible antigenic epitopes. METHODS: PSI-Blast, Praline, Emboss, Protein Data Bank, Swiss Model Server, AlphaFold 2, Ellipro and PyMol 2.3 were used to search for homologies, perform alignments, obtain protein structures, and predict epitopes. RESULTS: 17 autoantigens and seven immunological targets of the peripheral nervous system were included, identifying 72 possible epitopes associated with GBS. From the proteome of Plasmodium spp. (298 proteins), only two showed similarities close to 30% with TRIM21 and BACE1, generating seven possible epitopes. CONCLUSION: No significant homologies were observed between the proteome of GBS and Plasmodium spp. The exploration of other mechanisms such as immune-mediated capillary damage, Epitope Spreading or Bystander Activation is suggested to explain the mentioned association. These findings underscore the need to clarify the etiology of autoimmune diseases and the role of pathogens. The need for experimental studies to validate these results is emphasized.


OBJETIVO: Analizar el mimetismo molecular entre Plasmodium spp. y autoantígenos asociados al SGB, identificando posibles epítopos antigénicos. MÉTODOS: Se emplearon PSI-Blast, Praline, Emboss, Protein Data Bank, Swiss Model Server, AlphaFold 2, Ellipro y PyMol 2.3 para buscar homologías, realizar alineamientos, obtener estructuras proteicas y predecir epítopos. RESULTADOS: Se incluyeron 17 autoantígenos y siete objetivos inmunológicos del sistema nervioso periférico, identificándose 72 posibles epítopos asociados al SGB. Del proteoma de Plasmodium spp. (298 proteínas), solo dos mostraron similitud cercana al 30% con TRIM21 y BACE1, generando siete posibles epítopos. CONCLUSIÓN: No se observaron homologías significativas entre el proteoma de SGB y Plasmodium spp. Se sugiere la exploración de otros mecanismos como el daño capilar inmunomediado, Epitope Spreading o Bystander Activation para explicar la asociación mencionada. Estos hallazgos subrayan la necesidad de aclarar la etiología de las enfermedades autoinmunes y el papel de los patógenos. Se enfatiza la necesidad de estudios experimentales para validar estos resultados.


Subject(s)
Guillain-Barre Syndrome , Molecular Mimicry , Molecular Mimicry/immunology , Guillain-Barre Syndrome/immunology , Humans , Plasmodium/immunology , Autoantigens/immunology , Epitopes/immunology
5.
J Immunol ; 212(9): 1467-1478, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38477614

ABSTRACT

Development of Plasmodium-specific humoral immunity is critically dependent on CD4 Th cell responses and germinal center (GC) reactions during blood-stage Plasmodium infection. IL-21, a cytokine primarily produced by CD4 T cells, is an essential regulator of affinity maturation, isotype class-switching, B cell differentiation, and maintenance of GC reactions in response to many infection and immunization models. In models of experimental malaria, mice deficient in IL-21 or its receptor IL-21R fail to develop memory B cell populations and are not protected against secondary infection. However, whether sustained IL-21 signaling in ongoing GCs is required for maintaining GC magnitude, organization, and output is unclear. In this study, we report that CD4+ Th cells maintain IL-21 expression after resolution of primary Plasmodium yoelii infection. We generated an inducible knockout mouse model that enabled cell type-specific and timed deletion of IL-21 in peripheral, mature CD4 T cells. We found that persistence of IL-21 signaling in active GCs had no impact on the magnitude of GC reactions or their capacity to produce memory B cell populations. However, the memory B cells generated in the absence of IL-21 exhibited reduced recall function upon challenge. Our data support that IL-21 prevents premature cellular dissolution within the GC and promotes stringency of selective pressures during B cell fate determination required to produce high-quality Plasmodium-specific memory B cells. These data are additionally consistent with a temporal requirement for IL-21 in fine-tuning humoral immune memory responses during experimental malaria.


Subject(s)
CD4-Positive T-Lymphocytes , Interleukins , Malaria , Plasmodium , Animals , Mice , B-Lymphocytes , CD4-Positive T-Lymphocytes/metabolism , Germinal Center/immunology , Germinal Center/metabolism , Malaria/immunology , Memory B Cells/immunology , Mice, Inbred C57BL , Plasmodium/immunology
6.
Parasitology ; 151(4): 380-389, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38361461

ABSTRACT

Naturally acquired immunity to the different types of malaria in humans occurs in areas of endemic transmission and results in asymptomatic infection of peripheral blood. The current study examined the possibility of naturally acquired immunity in Bornean orangutans, Pongo pygmaeus, exposed to endemic Plasmodium pitheci malaria. A total of 2140 peripheral blood samples were collected between January 2017 and December 2022 from a cohort of 135 orangutans housed at a natural forested Rescue and Rehabilitation Centre in West Kalimantan, Indonesia. Each individual was observed for an average of 4.3 years during the study period. Blood samples were examined by microscopy and polymerase chain reaction for the presence of plasmodial parasites. Infection rates and parasitaemia levels were measured among age groups and all 20 documented clinical malaria cases were reviewed to estimate the incidence of illness and risk ratios among age groups. A case group of all 17 individuals that had experienced clinical malaria and a control group of 34 individuals having an event of >2000 parasites µL−1 blood but with no outward or clinical sign of illness were studied. Immature orangutans had higher-grade and more frequent parasitaemia events, but mature individuals were more likely to suffer from clinical malaria than juveniles. The case orangutans having patent clinical malaria were 256 times more likely to have had no parasitaemia event in the prior year relative to asymptomatic control orangutans. The findings are consistent with rapidly acquired immunity to P. pitheci illness among orangutans that wanes without re-exposure to the pathogen.


Subject(s)
Ape Diseases , Malaria , Plasmodium , Pongo pygmaeus , Animals , Malaria/epidemiology , Malaria/immunology , Malaria/parasitology , Plasmodium/immunology , Indonesia/epidemiology , Pongo pygmaeus/parasitology , Male , Female , Ape Diseases/parasitology , Ape Diseases/epidemiology , Parasitemia/veterinary , Parasitemia/epidemiology , Parasitemia/parasitology , Incidence
7.
Curr Med Chem ; 30(39): 4450-4465, 2023.
Article in English | MEDLINE | ID: mdl-36748809

ABSTRACT

Although the burden of malaria has been successfully controlled globally, this disease remains a major public health issue. To date, neither existing drugs nor vaccines against malaria are sufficient in eliminating malaria worldwide. To achieve the eradication of malaria by 2040, effective interventions targeting all Plasmodium species are urgently needed. As the cornerstone of vaccine design, immune memory serves a significant role in the host's defense against Plasmodium infections. It has long been considered that innate immunity is non-specific and lacks immunologic memory. However, emerging evidence has suggested that innate immunity can be trained following exposure of the body to infectious agents, such as Plasmodium or its products, which, in turn, promotes the onset of a type of memory in innate immune cells. The above "trained" innate immune cells, whose phenotype is modified in response to epigenetic modifications, metabolic recombination, or cytokine secretion, exhibit differential pathophysiology after the exposure of the body to a pathogen. In addition, Plasmodium-infected red blood cells and other host cells can secrete exosomes that contain conserved parasite-specific information, such as proteins, RNA, non-coding RNA molecules, and nucleic acids. These molecules can act as stimuli for promoting the establishment of "trained" innate immunity against malaria, thereby altering the onset and progression of the parasitic disease. A deeper understanding of the role of exosomes in the development of "trained" innate immunity during Plasmodium infection could provide novel therapeutic and prevention strategies against malaria infections.


Subject(s)
Immunity, Innate , Malaria , Plasmodium , Plasmodium/immunology , Malaria/immunology , Malaria/therapy , Extracellular Vesicles/immunology , Humans , Animals , Malaria Vaccines/immunology
8.
Front Immunol ; 13: 795463, 2022.
Article in English | MEDLINE | ID: mdl-35197971

ABSTRACT

Protection from liver-stage malaria requires high numbers of CD8+ T cells to find and kill Plasmodium-infected cells. A new malaria vaccine strategy, prime-target vaccination, involves sequential viral-vectored vaccination by intramuscular and intravenous routes to target cellular immunity to the liver. Liver tissue-resident memory (TRM) CD8+ T cells have been shown to be necessary and sufficient for protection against rodent malaria by this vaccine regimen. Ultimately, to most faithfully assess immunotherapeutic responses by these local, specialised, hepatic T cells, periodic liver sampling is necessary, however this is not feasible at large scales in human trials. Here, as part of a phase I/II P. falciparum challenge study of prime-target vaccination, we performed deep immune phenotyping, single-cell RNA-sequencing and kinetics of hepatic fine needle aspirates and peripheral blood samples to study liver CD8+ TRM cells and circulating counterparts. We found that while these peripheral 'TRM-like' cells differed to TRM cells in terms of previously described characteristics, they are similar phenotypically and indistinguishable in terms of key T cell residency transcriptional signatures. By exploring the heterogeneity among liver CD8+ TRM cells at single cell resolution we found two main subpopulations that each share expression profiles with blood T cells. Lastly, our work points towards the potential for using TRM-like cells as a correlate of protection by liver-stage malaria vaccines and, in particular, those adopting a prime-target approach. A simple and reproducible correlate of protection would be particularly valuable in trials of liver-stage malaria vaccines as they progress to phase III, large-scale testing in African infants. We provide a blueprint for understanding and monitoring liver TRM cells induced by a prime-target malaria vaccine approach.


Subject(s)
Malaria Vaccines/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Genetic Vectors , Hepatocytes/immunology , Humans , Immunity, Cellular , Immunologic Memory/immunology , Liver/immunology , Malaria/immunology , Plasmodium/immunology , Sporozoites/immunology , Transcriptome , Vaccination
9.
Front Immunol ; 12: 770246, 2021.
Article in English | MEDLINE | ID: mdl-34970259

ABSTRACT

Protozoan parasite infection causes severe diseases in humans and animals, leading to tremendous economic and medical pressure. Natural immunity is the first line of defence against parasitic infection. Currently, the role of natural host immunity in combatting parasitic infection is unclear, so further research on natural host immunity against parasites will provide a theoretical basis for the prevention and treatment of related parasitic diseases. Extracellular traps (ETs) are an important natural mechanism of immunity involving resistance to pathogens. When immune cells such as neutrophils and macrophages are stimulated by external pathogens, they release a fibrous network structure, consisting mainly of DNA and protein, that can capture and kill a variety of extracellular pathogenic microorganisms. In this review, we discuss the relevant recently reported data on ET formation induced by protozoan parasite infection, including the molecular mechanisms involved, and discuss the role of ETs in the occurrence and development of parasitic diseases.


Subject(s)
Extracellular Traps/immunology , Immunity, Innate/immunology , Neutrophils/immunology , Protozoan Infections, Animal/immunology , Protozoan Infections/immunology , Signal Transduction/immunology , Animals , Extracellular Traps/parasitology , Host-Parasite Interactions/immunology , Humans , Leishmania/immunology , Leishmania/physiology , Neutrophils/parasitology , Plasmodium/immunology , Plasmodium/physiology , Protozoan Infections/parasitology , Protozoan Infections, Animal/parasitology , Toxoplasma/immunology , Toxoplasma/physiology
10.
J Immunol Res ; 2021: 7785180, 2021.
Article in English | MEDLINE | ID: mdl-34790829

ABSTRACT

Malaria is a serious and, in some unfortunate cases, fatal disease caused by a parasite of the Plasmodium genus. It predominantly occurs in tropical areas where it is transmitted through the bite of an infected Anopheles mosquito. The pathogenesis of malaria is complex and incompletely elucidated. During blood-stage infection, in response to the presence of the parasite, the host's immune system produces proinflammatory cytokines including IL-6, IL-8, IFN-γ, and TNF, cytokines which play a pivotal role in controlling the growth of the parasite and its elimination. Regulatory cytokines such as transforming growth factor- (TGF-) ß and IL-10 maintain the balance between the proinflammatory and anti-inflammatory responses. However, in many cases, cytokines have a double role. On the one hand, they contribute to parasitic clearance, and on the other, they are responsible for pathological changes encountered in malaria. Cytokine-modulating strategies may represent a promising modern approach in disease management. In this review, we discuss the host immune response in malaria, analyzing the latest studies on the roles of pro- and anti-inflammatory cytokines.


Subject(s)
Cytokines/immunology , Inflammation/immunology , Malaria/immunology , Animals , Anopheles/immunology , Anopheles/parasitology , Humans , Inflammation/parasitology , Malaria/parasitology , Plasmodium/immunology
12.
Front Immunol ; 12: 680020, 2021.
Article in English | MEDLINE | ID: mdl-34484178

ABSTRACT

Lipid-derived signaling molecules known as eicosanoids have integral roles in mediating immune and inflammatory processes across metazoans. This includes the function of prostaglandins and their cognate G protein-coupled receptors (GPCRs) to employ their immunological actions. In insects, prostaglandins have been implicated in the regulation of both cellular and humoral immune responses, yet in arthropods of medical importance, studies have been limited. Here, we describe a prostaglandin E2 receptor (AgPGE2R) in the mosquito Anopheles gambiae and demonstrate that its expression is most abundant in oenocytoid immune cell populations. Through the administration of prostaglandin E2 (PGE2) and AgPGE2R-silencing, we demonstrate that prostaglandin E2 signaling regulates a subset of prophenoloxidases (PPOs) and antimicrobial peptides (AMPs) that are strongly expressed in populations of oenocytoids. We demonstrate that PGE2 signaling via the AgPGE2R significantly limits both bacterial replication and Plasmodium oocyst survival. Additional experiments establish that PGE2 treatment increases phenoloxidase (PO) activity through the increased expression of PPO1 and PPO3, genes essential to anti-Plasmodium immune responses that promote oocyst killing. We also provide evidence that the mechanisms of PGE2 signaling are concentration-dependent, where high concentrations of PGE2 promote oenocytoid lysis, negating the protective effects of lower concentrations of PGE2 on anti-Plasmodium immunity. Taken together, our results provide new insights into the role of PGE2 signaling on immune cell function and its contributions to mosquito innate immunity that promote pathogen killing.


Subject(s)
Anopheles/immunology , Anopheles/microbiology , Anopheles/parasitology , Dinoprostone/metabolism , Oocysts/immunology , Plasmodium/immunology , Signal Transduction , Animals , Anopheles/classification , Hemocytes/metabolism , Host-Pathogen Interactions/immunology , Immunity, Innate , Microbial Viability , Mosquito Vectors/immunology , Mosquito Vectors/microbiology , Mosquito Vectors/parasitology , Phylogeny , Plasmodium/growth & development , Pore Forming Cytotoxic Proteins/metabolism , Receptors, Prostaglandin E/genetics , Receptors, Prostaglandin E/metabolism
13.
Front Immunol ; 12: 729086, 2021.
Article in English | MEDLINE | ID: mdl-34512663

ABSTRACT

A successful malaria transmission blocking vaccine (TBV) requires the induction of a high antibody titer that leads to abrogation of parasite traversal of the mosquito midgut following ingestion of an infectious bloodmeal, thereby blocking the cascade of secondary human infections. Previously, we developed an optimized construct UF6b that elicits an antigen-specific antibody response to a neutralizing epitope of Anopheline alanyl aminopeptidase N (AnAPN1), an evolutionarily conserved pan-malaria mosquito midgut-based TBV target, as well as established a size-controlled lymph node targeting biodegradable nanoparticle delivery system that leads to efficient and durable antigen-specific antibody responses using the model antigen ovalbumin. Herein, we demonstrate that co-delivery of UF6b with the adjuvant CpG oligodeoxynucleotide immunostimulatory sequence (ODN ISS) 1018 using this biodegradable nanoparticle vaccine delivery system generates an AnAPN1-specific immune response that blocks parasite transmission in a standard membrane feeding assay. Importantly, this platform allows for antigen dose-sparing, wherein lower antigen payloads elicit higher-quality antibodies, therefore less antigen-specific IgG is needed for potent transmission-reducing activity. By targeting lymph nodes directly, the resulting immunopotentiation of AnAPN1 suggests that the de facto assumption that high antibody titers are needed for a TBV to be successful needs to be re-examined. This nanovaccine formulation is stable at -20°C storage for at least 3 months, an important consideration for vaccine transport and distribution in regions with poor healthcare infrastructure. Together, these data support further development of this nanovaccine platform for malaria TBVs.


Subject(s)
Adjuvants, Immunologic/pharmacology , Anopheles/immunology , Lymph Nodes/drug effects , Malaria Vaccines/pharmacology , Malaria/prevention & control , Nanoparticles , Oligodeoxyribonucleotides/pharmacology , Plasmodium/immunology , Vaccine Development , Animals , Anopheles/parasitology , Antibodies, Neutralizing/blood , Antibodies, Protozoan/blood , CD13 Antigens/antagonists & inhibitors , CD13 Antigens/immunology , CD13 Antigens/metabolism , Drug Compounding , Epitopes , Female , Host-Parasite Interactions , Immunoglobulin G/blood , Lymph Nodes/immunology , Lymph Nodes/parasitology , Malaria/immunology , Malaria/parasitology , Malaria/transmission , Malaria Vaccines/immunology , Mice , Nanomedicine , Plasmodium/pathogenicity , Vaccination
14.
Cell Rep ; 36(8): 109586, 2021 08 24.
Article in English | MEDLINE | ID: mdl-34433049

ABSTRACT

During acute malaria, most individuals mount robust inflammatory responses that limit parasite burden. However, long-lived sterilizing anti-malarial memory responses are not efficiently induced, even following repeated Plasmodium exposures. Using multiple Plasmodium species, genetically modified parasites, and combinations of host genetic and pharmacologic approaches, we find that the deposition of the malarial pigment hemozoin directly limits the abundance and capacity of conventional type 1 dendritic cells to prime helper T cell responses. Hemozoin-induced dendritic cell dysfunction results in aberrant Plasmodium-specific CD4 T follicular helper cell differentiation, which constrains memory B cell and long-lived plasma cell formation. Mechanistically, we identify that dendritic cell-intrinsic NLRP3 inflammasome activation reduces conventional type 1 dendritic cell abundance, phagocytosis, and T cell priming functions in vivo. These data identify biological consequences of hemozoin deposition during malaria and highlight the capacity of the malarial pigment to program immune evasion during the earliest events following an initial Plasmodium exposure.


Subject(s)
Hemeproteins/pharmacology , Inflammasomes/drug effects , Lymphocyte Activation/immunology , Malaria/drug therapy , Animals , Antimalarials/pharmacology , Dendritic Cells/immunology , Inflammasomes/metabolism , Malaria/immunology , Memory B Cells/drug effects , Memory B Cells/immunology , Mice, Inbred C57BL , Phagocytosis/physiology , Plasmodium/immunology , T-Lymphocytes, Helper-Inducer/immunology
15.
Immunobiology ; 226(5): 152091, 2021 09.
Article in English | MEDLINE | ID: mdl-34303920

ABSTRACT

The spike protein of coronavirus is key target for drug development and other pharmacological interventions. In current study, we performed an integrative approach to predict antigenic sites in SARS-CoV-2 spike receptor binding domain and found nine potential antigenic sites. The predicted antigenic sites were then assessed for possible molecular similarity with other known antigens in different organisms. Out of nine sites, seven sites showed molecular similarity with 54 antigenic determinants found in twelve pathogenic bacterial species (Mycobacterium tuberculosis, Mycobacterium leprae, Bacillus anthracis, Borrelia burgdorferi, Clostridium perfringens, Clostridium tetani, Helicobacter Pylori, Listeria monocytogenes, Staphylococcus aureus, Streptococcus pyogenes, Vibrio cholera and Yersinia pestis), two malarial parasites (Plasmodium falciparum and Plasmodium knowlesi) and influenza virus A. Most of the bacterial antigens that displayed molecular similarity with antigenic sites in SARS-CoV-2 RBD (receptor binding domain) were toxins and virulent factors. Antigens from Mycobacterium that showed similarity were mainly involved in modulating host cell immune response and ensuring persistence and survival of pathogen in host cells. Presence of a large number of antigenic determinants, similar to those in highly pathogenic microorganisms, not merely accounts for complex etiology of the disease but also provides an explanation for observed pathophysiological complications, such as deregulated immune response, unleashed or dysregulated cytokine secretion (cytokine storm), multiple organ failure etc., that are more evident in aged and immune-compromised patients. Over-representation of antigenic determinants from Plasmodium and Mycobacterium in all antigenic sites suggests that anti-malarial and anti-TB drugs can prove to be clinical beneficial for COVID-19 treatment. Besides this, anti-leprosy, anti-lyme, anti-plague, anti-anthrax drugs/vaccine etc. are also expected to be beneficial in COVID-19 treatment. Moreover, individuals previously immunized/vaccinated or had previous history of malaria, tuberculosis or other disease caused by fifteen microorganisms are expected to display a considerable degree of resistance against SARS-CoV-2 infection. Out of the seven antigenic sites predicted in SARS-CoV-2, a part of two antigenic sites were also predicted as potent T-cell epitopes (KVGGNYNYL444-452 and SVLYNSASF366-374) against MHC class I and three (KRISNCVADYSVLYN356-370, DLCFTNVYADSFVI389-402, and YRVVVLSFELLHA508-520) against MHC class II. All epitopes possessed significantly lower predicted IC50 value which is a prerequisite for a preferred vaccine candidate for COVID-19.


Subject(s)
Antigens, Viral/immunology , Epitopes, T-Lymphocyte/immunology , Peptides/immunology , SARS-CoV-2/immunology , Spike Glycoprotein, Coronavirus/immunology , Bacteria/immunology , Binding Sites , COVID-19/prevention & control , COVID-19 Vaccines , Influenza A virus/immunology , Plasmodium/immunology , Protein Domains
16.
Sci Rep ; 11(1): 13248, 2021 06 24.
Article in English | MEDLINE | ID: mdl-34168264

ABSTRACT

Multiplex assays for malaria antigen detection can gather data from large sample sets, but considerations for the consistency and quality assurance (QA) of mass testing lack evaluation. We present a QA framework for a study occurring November 2019 to March 2020 involving 504 assay plates detecting four Plasmodium antigens: pan-Plasmodium aldolase and lactate dehydrogenase (LDH), histidine-rich protein 2 (HRP2), P. vivax LDH (PvLDH). Controls on each plate included buffer blank, antigen negative blood, and 4-point positive dilution curve. The blank and negative blood provided consistently low signal for all targets except for pAldolase, which showed variability. Positive curve signals decreased throughout the 5-month study duration but retained a coefficient of variation (CV) of < 5%, with the exception of HRP2 in month 5 (CV of 11%). Regression fittings for inter-plate control signals provided mean and standard deviations (SDs), and of 504 assay plates, 6 (1.2%) violated the acceptable deviation limits and were repeated. For the 40,272 human blood samples assayed in this study, of 161,088 potential data points (each sample × 4 antigens), 160,641 (99.7%) successfully passed quality checks. The QA framework presented here can be utilized to ensure quality of laboratory antigen detection for large sample sets.


Subject(s)
Antigens, Protozoan/immunology , Malaria/immunology , Plasmodium/immunology , Adolescent , Antigens, Protozoan/blood , Child , Fructose-Bisphosphate Aldolase/immunology , Humans , L-Lactate Dehydrogenase/immunology , Nigeria , Protozoan Proteins/immunology , Quality Control , Serologic Tests/methods
17.
Front Immunol ; 12: 683404, 2021.
Article in English | MEDLINE | ID: mdl-34168652

ABSTRACT

Antibody immunity against malaria is effective but non-sterile. In addition to antibody-mediated inhibition, neutralisation or opsonisation of malaria parasites, antibody-mediated complement activation is also important in defense against infection. Antibodies form immune complexes with parasite-derived antigens that can activate the classical complement pathway. The complement system provides efficient surveillance for infection, and its activation leads to parasite lysis or parasite opsonisation for phagocytosis. The induction of complement-fixing antibodies contributes significantly to the development of protective immunity against clinical malaria. These complement-fixing antibodies can form immune complexes that are recognised by complement receptors on innate cells of the immune system. The efficient clearance of immune complexes is accompanied by complement receptor internalisation, abrogating the detrimental consequences of excess complement activation. Here, we review the mechanisms of activation of complement by alternative, classical, and lectin pathways in human malaria at different stages of the Plasmodium life cycle with special emphasis on how complement-fixing antibodies contribute to protective immunity. We briefly touch upon the action of anaphylatoxins, the assembly of membrane attack complex, and the possible reasons underlying the resistance of infected erythrocytes towards antibody-mediated complement lysis, relevant to their prolonged survival in the blood of the human host. We make suggestions for further research on effector functions of antibody-mediated complement activation that would guide future researchers in deploying complement-fixing antibodies in preventive or therapeutic strategies against malaria.


Subject(s)
Antibodies, Protozoan/immunology , Antibody-Dependent Cell Cytotoxicity , Complement Activation/immunology , Complement System Proteins/immunology , Host-Parasite Interactions/immunology , Malaria/immunology , Plasmodium/immunology , Antigen-Antibody Complex/immunology , Complement Pathway, Classical/immunology , Erythrocytes/immunology , Erythrocytes/parasitology , Humans , Life Cycle Stages , Malaria/parasitology , Plasmodium/growth & development
18.
Front Immunol ; 12: 661241, 2021.
Article in English | MEDLINE | ID: mdl-34122419

ABSTRACT

As a relatively successful pathogen, several parasites can establish long-term infection in host. This "harmonious symbiosis" status relies on the "precise" manipulation of host immunity and metabolism, however, the underlying mechanism is still largely elusive. Immunometabolism is an emerging crossed subject in recent years. It mainly discusses the regulatory mechanism of metabolic changes on reprogramming the key transcriptional and post-transcriptional events related to immune cell activation and effect, which provides a novel insight for understanding how parasites regulate the infection and immunity in hosts. The present study reviewed the current research progress on metabolic reprogramming mechanism exploited by parasites to modulate the function in various immune cells, highlighting the future exploitation of key metabolites or metabolic events to clarify the underlying mechanism of anti-parasite immunity and design novel intervention strategies against parasitic infection.


Subject(s)
Dendritic Cells/immunology , Lymphocytes/immunology , Macrophages/immunology , Parasitic Diseases/immunology , Plasmodium/immunology , Schistosoma/immunology , Trypanosoma/immunology , Animals , Dendritic Cells/metabolism , Dendritic Cells/parasitology , Host-Parasite Interactions/immunology , Humans , Lymphocytes/metabolism , Lymphocytes/parasitology , Macrophages/metabolism , Macrophages/parasitology , Parasitic Diseases/metabolism , Parasitic Diseases/parasitology , Plasmodium/physiology , Schistosoma/physiology , Trypanosoma/physiology
19.
Am J Trop Med Hyg ; 105(2): 477-479, 2021 06 14.
Article in English | MEDLINE | ID: mdl-34125702

ABSTRACT

The protective effect of semi-immunity to alleviate clinical complications of malaria remains incompletely understood. This ecological study quantified the proportion of unfavorable clinical outcomes among patient populations with imported malaria as a function of the reported proportion of absent semi-immunity in a patient population. Group-level proportions were extracted from published studies on imported malaria. Linear regression analyses demonstrate a consistent positive trend between the average proportion of absent semi-immunity in patient populations of imported malaria and the proportion of unfavorable clinical outcomes therein. Regression equations provide a group-level estimate of attributable fractions of clinical complications resulting from absent semi-immunity to malaria.


Subject(s)
Malaria , Plasmodium/immunology , Antimalarials/therapeutic use , Chemoprevention , Communicable Diseases, Imported/epidemiology , Communicable Diseases, Imported/immunology , Humans , Immunity , Malaria/drug therapy , Malaria/epidemiology , Malaria/immunology , Medication Adherence , Mortality , Population Surveillance , Prevalence , Travel , Treatment Outcome
20.
Nat Metab ; 3(7): 1001-1016, 2021 07.
Article in English | MEDLINE | ID: mdl-34113019

ABSTRACT

Host responses to infection with the malaria parasite Plasmodium falciparum vary among individuals for reasons that are poorly understood. Here we reveal metabolic perturbations as a consequence of malaria infection in children and identify an immunosuppressive role of endogenous steroid production in the context of P. falciparum infection. We perform metabolomics on matched samples from children from two ethnic groups in West Africa, before and after infection with seasonal malaria. Analysing 306 global metabolomes, we identify 92 parasitaemia-associated metabolites with impact on the host adaptive immune response. Integrative metabolomic and transcriptomic analyses, and causal mediation and moderation analyses, reveal an infection-driven immunosuppressive role of parasitaemia-associated pregnenolone steroids on lymphocyte function and the expression of key immunoregulatory lymphocyte genes in the Gouin ethnic group. In children from the less malaria-susceptible Fulani ethnic group, we observe opposing responses following infection, consistent with the immunosuppressive role of endogenous steroids in malaria. These findings advance our understanding of P. falciparum pathogenesis in humans and identify potential new targets for antimalarial therapeutic interventions.


Subject(s)
Adaptive Immunity , Host-Parasite Interactions , Malaria/immunology , Malaria/metabolism , Metabolome , Plasmodium/immunology , Host-Parasite Interactions/immunology , Humans , Immunomodulation , Lymphocytes/immunology , Lymphocytes/metabolism , Malaria/parasitology , Malaria, Falciparum/immunology , Malaria, Falciparum/metabolism , Malaria, Falciparum/parasitology , Parasitemia , Plasmodium falciparum/immunology , Steroids/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL