Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(26): e2306318120, 2023 06 27.
Article in English | MEDLINE | ID: mdl-37307435

ABSTRACT

Histidine-rich protein II (HRPII) is secreted by Plasmodium falciparum during the blood stage of malaria infection. High plasma levels of HRPII are associated with cerebral malaria, a severe and highly fatal complication of malaria. HRPII has been shown to induce vascular leakage, the hallmark of cerebral malaria, in blood-brain barrier (BBB) and animal models. We have discovered an important mechanism for BBB disruption that is driven by unique features of HRPII. By characterizing serum from infected patients and HRPII produced by P. falciparum parasites in culture, we found that HRPII exists in large multimeric particles of 14 polypeptides that are richly laden with up to 700 hemes per particle. Heme loading of HRPII is required for efficient binding and internalization via caveolin-mediated endocytosis in hCMEC/D3 cerebral microvascular endothelial cells. Upon acidification of endolysosomes, two-thirds of the hemes are released from acid-labile binding sites and metabolized by heme oxygenase 1, generating ferric iron and reactive oxygen species. Subsequent activation of the NLRP3 inflammasome and IL-1ß secretion resulted in endothelial leakage. Inhibition of these pathways with heme sequestration, iron chelation, or anti-inflammatory drugs protected the integrity of the BBB culture model from HRPII:heme. Increased cerebral vascular permeability was seen after injection of young mice with heme-loaded HRPII (HRPII:heme) but not with heme-depleted HRPII. We propose that during severe malaria infection, HRPII:heme nanoparticles in the bloodstream deliver an overwhelming iron load to endothelial cells to cause vascular inflammation and edema. Disrupting this process is an opportunity for targeted adjunctive therapies to reduce the morbidity and mortality of cerebral malaria.


Subject(s)
Hemeproteins , Malaria, Cerebral , Malaria, Falciparum , Animals , Mice , Histidine , Endothelial Cells , Inflammation , Heme , Iron
2.
Pathogens ; 12(3)2023 Feb 24.
Article in English | MEDLINE | ID: mdl-36986296

ABSTRACT

This study analyzed fifty years of severe malaria research worldwide. Malaria is a parasitic disease that continues to have a significant impact on global health, particularly in sub-Saharan Africa. Severe malaria, a severe and often fatal form of the disease, is a major public health concern. The study used different bibliometric indicators such as the number of publications, citations, authorship, and keywords to analyze the research trends, patterns, and progress made in the field of severe malaria. The study covers the period from 1974 to 2021 and includes articles from Scopus. The results of the study indicated that there has been a steady increase in the number of publications on severe malaria over the past fifty years, with a particular increase in the last decade. The study also showed that most of the publications are from USA and Europe, while the disease occurs in Africa, South-East Asia, and the Americas. The study also identified the most frequent keywords used in the publications, and the most influential journals and authors in the field. In conclusion, this bibliometric study provides a comprehensive overview of the research trends and patterns in the field of severe malaria over the past fifty years and highlights the areas that need more attention and research efforts.

3.
Glob Pediatr ; 6: None, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38440360

ABSTRACT

Purpose: The pediatric population, especially under-five children, is highly susceptible to malaria and accounts for 76 % of global malaria deaths according to the World Malaria Report 2022. The purpose of this manuscript is to discuss the various factors involved in the susceptibility of the pediatric population to Malaria and the importance of this age group for malaria elimination. Methodology: Data on pediatric malaria epidemiology that includes prevalence, risk factors, immune factors, socioeconomic factors, control methods, etc. were extracted from published literature using PubMed and Google Scholar. This data was further correlated with malaria incidence data from the World Health Organization (WHO) and the National Center for Vector Borne Diseases Control (NCVBDC). Results: The younger age group is vulnerable to severe malaria due to an immature immune system. The risk of infection and clinical disease increases after the waning of maternal immunity. In the initial years of life, the developing brain is more susceptible to malaria infection and its after-effects. The pediatric population may act as a malaria transmission reservoir due to parasite density and asymptomatic infections. WHO recommended RTS,S/AS01 has limitations and may not be applicable in all settings to propel malaria elimination. Conclusion: The diagnosis of malaria is based on clinical suspicion and confirmed with microscopy and/or rapid diagnostic testing. The school-age pediatric population serves as a transmission reservoir in the form of asymptomatic malaria since they have acquired some immunity due to exposure in early childhood. Targeting the hidden reservoir in the pediatric population and protecting this vulnerable group will be essential for malaria elimination from the countries targeting elimination.

4.
Elife ; 112022 12 14.
Article in English | MEDLINE | ID: mdl-36515978

ABSTRACT

Malaria parasites avoid immune clearance through their ability to systematically alter antigens exposed on the surface of infected red blood cells. This is accomplished by tightly regulated transcriptional control of individual members of a large, multicopy gene family called var and is the key to both the virulence and chronic nature of malaria infections. Expression of var genes is mutually exclusive and controlled epigenetically, however how large populations of parasites coordinate var gene switching to avoid premature exposure of the antigenic repertoire is unknown. Here, we provide evidence for a transcriptional network anchored by a universally conserved gene called var2csa that coordinates the switching process. We describe a structured switching bias that shifts overtime and could shape the pattern of var expression over the course of a lengthy infection. Our results provide an explanation for a previously mysterious aspect of malaria infections and shed light on how parasites possessing a relatively small repertoire of variant antigen-encoding genes can coordinate switching events to limit antigen exposure, thereby maintaining chronic infections.


Malaria causes severe illness and deaths in hundreds of thousands of people each year. Most of them are young children in Sub-Saharan Africa. The disease is transmitted when a mosquito carrying single-celled Plasmodium parasites bites a human, introducing the parasites into the bloodstream, where they enter red blood cells. When a red blood cell becomes infected, the parasite presents a protein on the cell's surface that the immune system can recognize to start fighting the infection. Immune cells then produce antibodies that flag infected cells for destruction, relieving the symptoms of the disease. To avoid being destroyed in this manner, the parasites repeatedly 'change' the protein that ends up on the surface of the red blood cells. With each change, the number of parasites rebounds, symptoms return, and the immune system must produce new antibodies. As the parasites and immune system battle it out, patients may experience repeated flare-ups of symptoms for well over a year. To change the protein that is presented on the surface of red blood cells, Plasmodium parasites switch the genes in the var gene family on and off one at a time. Each of these genes encodes a different surface protein, and the parasites may cycle through the entire var gene family during an infection. However, it remains a mystery how the millions of infecting parasites coordinate to produce the same surface protein each time. Zhang et al. show that a gene from Plasmodium parasites called var2csa is responsible for coordinating protein switching through a set pattern that allows the parasites to synchronize which protein they switch to next. Deleting the var2csa gene in malaria parasites blocks protein switching and disables this coordinated immune evasion tactic. Zhang et al.'s experiments provide new insights about protein switching in malaria parasites. Further research may help scientists characterize each step in the process and identify which steps can be targeted to treat malaria. While not a cure, treatments that disable protein switching could reduce the number of times patients relapse and relieve symptoms. More generally, the results of Zhang et al. describe a mechanism for coordinated gene expression that may be used in organisms other than Plasmodium, including humans.


Subject(s)
Malaria, Falciparum , Malaria , Parasites , Animals , Humans , Malaria, Falciparum/parasitology , Plasmodium falciparum , Protozoan Proteins/metabolism , Antigenic Variation/genetics , Antigens
5.
Front Immunol ; 13: 855795, 2022.
Article in English | MEDLINE | ID: mdl-35634341

ABSTRACT

Eryptosis is a programmed cell death-like process that occurs in red blood cells. Although the red blood cells are anucleated, there are similarities between eryptosis and apoptosis, such as increased calcium efflux, calpain activation, phosphatidylserine exposure, cell blebbing and cell shrinkage. Eryptosis occurs physiologically in red blood cells, as a consequence of the natural senescence process of these cells, but it can also be stimulated in pathological situations such as metabolic syndromes, uremic syndromes, polycythemia vera, anemias such as sickle cell anemia and thalassemia, and infectious processes including Plasmodium infection. Infection-induced eryptosis is believed to contribute to damage caused by Plasmodium, but it's still a topic of debate in the literature. In this review, we provided an overview of eryptosis mechanisms and its possible pathogenic role in malaria.


Subject(s)
Anemia, Sickle Cell , Eryptosis , Malaria , Anemia, Sickle Cell/metabolism , Apoptosis/physiology , Erythrocytes/metabolism , Humans , Malaria/metabolism
6.
Tropical Biomedicine ; : 187-204, 2021.
Article in English | WPRIM (Western Pacific) | ID: wpr-886635

ABSTRACT

@#Malaria infection still remains as one of the most prominent parasitic diseases afflicting mankind in tropical and subtropical regions. The severity of malaria infection has often been associated to exuberant host immune inflammatory responses that could possibly lead to severe immunopathological conditions and subsequent death of host tissues. Activin A is a protein belonging to the transforming growth factor-beta (TGF-β) family that regulates multiple physiological processes and pathological-associated diseases. The biological roles of activin A have been associated with manipulation of inflammation-related processes and modulation of host immune responses. This implies that activin A protein could play a role in malaria pathogenesis since malaria infection has been closely linked to severe immune responses leading to death, However, the actual in vivo role of activin A in malaria infection remains elusive. Hence, this study was undertaken to investigate the involvement of activin A in malaria infection as well as to assess the modulating effects of activin A on the cytokine releases (TNF-α, IFN-γ and IL-10) and histopathological changes in major affected organs (kidney, liver, lung, brain and spleen) in malarial mice infected with Plasmodium berghei ANKA. Our results showed that the concentrations of plasma activin A were significantly increased in malarial mice throughout the study periods. Also. the systemic activin A level was positively correlated with malaria parasitemia. This indicates that activin A could play a role in malaria pathogenesis and malaria parasitemia development. Plasma TNF-α, IFN-γ and IL-10 cytokine levels were significantly increased in malarial mice at day-5 post infection, suggesting that these cytokines attributed to severe malaria pathogenesis. Histopathological features such as sequestration of parasitized red blood cells (pRBCs) and hemozoin formation were amongst the most common pathological conditions observed in tissues of major affected organs (kidney, liver, lung, brain and spleen) in malarial mice. Neutralization of activin A production via recombinant mouse activin RIIA Fc chimera (rmActivin RIIA Fc chimera) had significantly reduced the parasitemia levels in malarial mice. The release of TNF-α cytokine was significantly reduced as well as the sequestration of parasitized pRBCs and hemozoin formation in major affected organs in malarial mice were also alleviated following inhibition of activin A production. Overall, this preliminary study suggests that activin A could play an immune modulation role in malaria pathogenesis through modulation of TNF-α release that benefits host from severe pathological destructions provoked by intensified inflammatory responses. Further studies are warranted to elucidate the precise mechanism of immune modulation mediated by activin A and its associated immune-modulation mediators in regulating the inflammatory responses elicited during the course of malaria infection.

7.
Parasitology ; 147(1): 1-11, 2020 01.
Article in English | MEDLINE | ID: mdl-31455446

ABSTRACT

Malaria remains a major cause of mortality in African children, with no adjunctive treatments currently available to ameliorate the severe clinical forms of the disease. Rosetting, the adhesion of infected erythrocytes (IEs) to uninfected erythrocytes, is a parasite phenotype strongly associated with severe malaria, and hence is a potential therapeutic target. However, the molecular mechanisms of rosetting are complex and involve multiple distinct receptor-ligand interactions, with some similarities to the diverse pathways involved in P. falciparum erythrocyte invasion. This review summarizes the current understanding of the molecular interactions that lead to rosette formation, with a particular focus on host uninfected erythrocyte receptors including the A and B blood group trisaccharides, complement receptor one, heparan sulphate, glycophorin A and glycophorin C. There is strong evidence supporting blood group A trisaccharides as rosetting receptors, but evidence for other molecules is incomplete and requires further study. It is likely that additional host erythrocyte rosetting receptors remain to be discovered. A rosette-disrupting low anti-coagulant heparin derivative is being investigated as an adjunctive therapy for severe malaria, and further research into the receptor-ligand interactions underlying rosetting may reveal additional therapeutic approaches to reduce the unacceptably high mortality rate of severe malaria.


Subject(s)
Erythrocytes/metabolism , Malaria, Falciparum/diagnosis , Cell Adhesion/physiology , Erythrocytes/parasitology , Humans , Malaria, Falciparum/physiopathology , Plasmodium falciparum , Rosette Formation , Trisaccharides/metabolism
8.
mBio ; 9(6)2018 12 11.
Article in English | MEDLINE | ID: mdl-30538191

ABSTRACT

The most advanced vaccine against Plasmodium falciparum malaria, RTS,S/AS01, provides partial protection in infants and children living in areas of malaria endemicity. Further understanding its mechanisms of protection may allow the development of improved second-generation vaccines. The RTS,S/AS01 vaccine targets the sporozoites injected by mosquito vectors into the dermis which then travel into the blood stream to establish infection in the liver. Flores-Garcia et al. (Y. Flores-Garcia, G. Nasir, C. S. Hopp, C. Munoz, et al., mBio 9:e02194-18, 2018, https://doi.org/10.1128/mBio.02194-18) shed light on early protective responses occurring in the dermis in immunized animals. They demonstrated that immunization impairs sporozoite motility and entry into blood vessels. Furthermore, they established that challenge experiments performed using a dermal route conferred greater protection than intravenous challenge in immunized mice. Thus, the dermal challenge approach captures the additional protective mechanisms occurring in the dermis that reflect the natural physiology of infection. Those studies highlighted the fascinating biology of skin-stage sporozoites and provided additional insights into vaccine-induced protection.


Subject(s)
Malaria Vaccines , Sporozoites/immunology , Animals , Child , Dermis , Humans , Infant , Malaria , Mice , Plasmodium
9.
Annu Rev Microbiol ; 72: 501-519, 2018 Sep 08.
Article in English | MEDLINE | ID: mdl-29975590

ABSTRACT

Sexual differentiation of malaria parasites from the asexual blood stage into gametocytes is an essential part of the life cycle, as gametocytes are the form that is taken up by the mosquito host. Because of the essentiality of this process for transmission to the mosquito, gametocytogenesis is an extremely attractive target for therapeutic interventions. The subject of this review is the considerable progress that has been made in recent years in elucidating the molecular mechanisms governing this important differentiation process. In particular, a number of critical transcription factors and epigenetic regulators have emerged as crucial elements in the regulation of commitment. The identification of these factors has allowed us to understand better than ever before the events occurring prior to and during commitment to sexual development and offers potential for new therapeutic interventions.


Subject(s)
Cell Differentiation , Gene Expression Regulation , Plasmodium/growth & development , Plasmodium/genetics , Gene Regulatory Networks , Transcription Factors/metabolism
10.
J Proteomics ; 127(Pt A): 103-13, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-25982387

ABSTRACT

India significantly contributes to the global malaria burden and has the largest population in the world at risk of malaria. This study aims to analyze alterations in the human serum proteome as a consequence of non-severe and severe infections by the malaria parasite Plasmodium falciparum to identify markers related to disease severity and to obtain mechanistic insights about disease pathogenesis and host immune responses. In discovery phase of the study, a comprehensive quantitative proteomic analysis was performed using gel-based (2D-DIGE) and gel-free (iTRAQ) techniques on two independent mass spectrometry platforms (ESI-Q-TOF and Q-Exactive mass spectrometry), and selected targets were validated by ELISA. Proteins showing altered serum abundance in falciparum malaria patients revealed the modulation of different physiological pathways including chemokine and cytokine signaling, IL-12 signaling and production in macrophages, complement cascades, blood coagulation, and protein ubiquitination pathways. Some muscle related and cytoskeletal proteins such as titin and galectin-3-binding protein were found to be up-regulated in severe malaria patients. Hemoglobin levels and platelet counts were also found to be drastically lower in severe malaria patients. Identified proteins including serum amyloid A, C-reactive protein, apolipoprotein E and haptoglobin, which exhibited sequential alterations in their serum abundance in different severity levels of malaria, could serve as potential predictive markers for disease severity. To the best of our information, we report here the first comprehensive analysis describing the serum proteomic alterations observed in severe P. falciparum infected patients from different malaria endemic regions of India. This article is part of a Special Issue entitled: Proteomics in India.


Subject(s)
Blood Proteins/metabolism , Malaria, Falciparum/blood , Plasmodium falciparum , Proteomics , Severity of Illness Index , Adult , Biomarkers/blood , Female , Humans , India , Malaria, Falciparum/epidemiology , Malaria, Falciparum/pathology , Male
11.
Mol Immunol ; 67(1): 71-84, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25816986

ABSTRACT

Malaria remains one of the world's deadliest diseases. Plasmodium falciparum is responsible for the most severe and lethal form of human malaria. P. falciparum's life cycle involves two obligate hosts: human and mosquito. From initial entry into these hosts, malaria parasites face the onslaught of the first line of host defence, the complement system. In this review, we discuss the complex interaction between complement and malaria infection in terms of hosts immune responses, parasite survival and pathogenesis of severe forms of malaria. We will focus on the role of complement receptor 1 and its associated polymorphisms in malaria immune complex clearance, as a mediator of parasite rosetting and as an entry receptor for P. falciparum invasion. Complement evasion strategies of P. falciparum parasites will also be highlighted. The sexual forms of the malaria parasites recruit the soluble human complement regulator Factor H to evade complement-mediated killing within the mosquito host. A novel evasion strategy is the deployment of parasite organelles to divert complement attack from infective blood stage parasites. Finally we outline the future challenge to understand the implications of these exploitation mechanisms in the interplay between successful infection of the host and pathogenesis observed in severe malaria.


Subject(s)
Complement C3b Inactivator Proteins/immunology , Life Cycle Stages/immunology , Malaria, Falciparum/immunology , Plasmodium falciparum/immunology , Receptors, Complement/immunology , Antigen-Antibody Complex/chemistry , Antigen-Antibody Complex/genetics , Complement Activation , Complement C3b Inactivator Proteins/genetics , Complement Factor H/genetics , Complement Factor H/immunology , Gene Expression , Humans , Immune Evasion , Life Cycle Stages/genetics , Malaria, Falciparum/genetics , Malaria, Falciparum/parasitology , Malaria, Falciparum/pathology , Plasmodium falciparum/genetics , Plasmodium falciparum/growth & development , Polymorphism, Genetic , Receptors, Complement/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...