Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Biomolecules ; 13(5)2023 05 09.
Article in English | MEDLINE | ID: covidwho-20237553

ABSTRACT

The triamine spermidine is a key metabolite of the polyamine pathway. It plays a crucial role in many infectious diseases caused by viral or parasitic infections. Spermidine and its metabolizing enzymes, i.e., spermidine/spermine-N1-acetyltransferase, spermine oxidase, acetyl polyamine oxidase, and deoxyhypusine synthase, fulfill common functions during infection in parasitic protozoa and viruses which are obligate, intracellular parasites. The competition for this important polyamine between the infected host cell and the pathogen determines the severity of infection in disabling human parasites and pathogenic viruses. Here, we review the impact of spermidine and its metabolites in disease development of the most important, pathogenic human viruses such as SARS-CoV-2, HIV, Ebola, and in the human parasites Plasmodium and Trypanosomes. Moreover, state-of-the-art translational approaches to manipulate spermidine metabolism in the host and the pathogen are discussed to accelerate drug development against these threatful, infectious human diseases.


Subject(s)
COVID-19 , Parasitic Diseases , Trypanosoma brucei brucei , Humans , Spermidine , Trypanosoma brucei brucei/metabolism , Plasmodium falciparum/metabolism , SARS-CoV-2/metabolism , Polyamines/metabolism
2.
EMBO Mol Med ; 15(6): e17556, 2023 06 07.
Article in English | MEDLINE | ID: covidwho-2300566

ABSTRACT

Malaria remains a huge burden on global public health. Annually there are more than 200 million cases with > 600,000 deaths worldwide, the vast majority of which occur within Sub-Saharan Africa (WHO; World Malaria Report, 2021). Malaria disease is the consequence of infection by a protozoan parasite from the genus Plasmodium with most morbidity and mortality caused by P. falciparum. With rates of infection plateauing and rebounding in some areas (in particular, as a result of the disruption caused by the COVID-19 pandemic), there have been increasing calls for new initiatives that can reduce malaria incidence towards local elimination or the hoped for goal of global eradication. In 2021, the World Health Organisation approved the first malaria vaccine RTS,S/AS01 (also called Mosquirix™), indicating it to be safe for use in young children and advocating its integration into routine immunisation programmes. Approval of this vaccine clearly represents a major landmark in global efforts towards malaria control and eradication aspirations. RTS,S modest efficacy, however, points at the need to better understand immune responses to the parasite if we hope to design next generation malaria vaccines with increased potency.


Subject(s)
COVID-19 , Malaria Vaccines , Malaria, Falciparum , Malaria , Child , Humans , Child, Preschool , Plasmodium falciparum , Pandemics , COVID-19/epidemiology , Malaria, Falciparum/prevention & control , Antibodies , Malaria/epidemiology , Protozoan Proteins/genetics
3.
Lancet Infect Dis ; 23(5): 578-588, 2023 05.
Article in English | MEDLINE | ID: covidwho-2299258

ABSTRACT

BACKGROUND: Human monoclonal antibodies might offer an important new approach to reduce malaria morbidity and mortality. In the first two parts of a three-part clinical trial, the antimalarial monoclonal antibody CIS43LS conferred high protection against parasitaemia at doses of 20 mg/kg or 40 mg/kg administered intravenously followed by controlled human malaria infection. The ability of CIS43LS to confer protection at lower doses or by the subcutaneous route is unknown. We aimed to provide data on the safety and optimisation of dose and route for the human antimalaria monoclonal antibody CIS43LS. METHODS: VRC 612 Part C was the third part of a three-part, first-in-human, phase 1, adaptive trial, conducted at the University of Maryland, Baltimore Center for Vaccine Development and Global Health, Baltimore, MD, USA. We enrolled adults aged 18-50 years with no previous malaria vaccinations or infections, in a sequential, dose-escalating manner. Eligible participants received the monoclonal antibody CIS43LS in a single, open-label dose of 1 mg/kg, 5 mg/kg, or 10 mg/kg intravenously, or 5 mg/kg or 10 mg/kg subcutaneously. Participants underwent controlled human malaria infection by the bites of five mosquitoes infected with Plasmodium falciparum 3D7 strain approximately 8 weeks after their monoclonal antibody inoculation. Six additional control participants who did not receive CIS43LS underwent controlled human malaria infection simultaneously. Participants were followed-up daily on days 7-18 and day 21, with qualitative PCR used for P falciparum detection. Participants who tested positive for P falciparum were treated with atovaquone-proguanil and those who remained negative were treated at day 21. Participants were followed-up until 24 weeks after dosing. The primary outcome was safety and tolerability of CIS43LS at each dose level, assessed in the as-treated population. Secondary outcomes included protective efficacy of CIS43LS after controlled human malaria infection. This trial is now complete and is registered with ClinicalTrials.gov, NCT04206332. FINDINGS: Between Sept 1, 2021, and Oct 29, 2021, 47 people were assessed for eligibility and 31 were enrolled (one subsequently withdrew and was replaced) and assigned to receive doses of 1 mg/kg (n=7), 5 mg/kg (n=4), and 10 mg/kg (n=3) intravenously and 5 mg/kg (n=4) and 10 mg/kg (n=4) subcutaneously, or to the control group (n=8). CIS43LS administration was safe and well tolerated; no serious adverse events occurred. CIS43LS protected 18 (82%) of 22 participants who received a dose. No participants developed parasitaemia following dosing at 5 mg/kg intravenously or subcutaneously, or at 10 mg/kg intravenously or subcutaneously. All six control participants and four of seven participants dosed at 1 mg/kg intravenously developed parasitaemia after controlled human malaria infection. INTERPRETATION: CIS43LS was safe and well tolerated, and conferred protection against P falciparum at low doses and by the subcutaneous route, providing evidence that this approach might be useful to prevent malaria across several clinical use cases. FUNDING: National Institute of Allergy and Infectious Diseases, National Institutes of Health.


Subject(s)
Antimalarials , Malaria Vaccines , Malaria, Falciparum , Adult , Animals , Humans , Antibodies, Monoclonal/therapeutic use , Malaria, Falciparum/drug therapy , Malaria, Falciparum/prevention & control , Plasmodium falciparum , Malaria Vaccines/therapeutic use
4.
BMC Med ; 21(1): 137, 2023 04 07.
Article in English | MEDLINE | ID: covidwho-2303147

ABSTRACT

BACKGROUND: Whole sporozoite immunization under chemoprophylaxis (CPS regime) induces long-lasting sterile homologous protection in the controlled human malaria infection model using Plasmodium falciparum strain NF54. The relative proficiency of liver-stage parasite development may be an important factor determining immunization efficacy. Previous studies show that Plasmodium falciparum strain NF135 produces relatively high numbers of large liver-stage schizonts in vitro. Here, we evaluate this strain for use in CPS immunization regimes. METHODS: In a partially randomized, open-label study conducted at the Radboudumc, Nijmegen, the Netherlands, healthy, malaria-naïve adults were immunized by three rounds of fifteen or five NF135-infected mosquito bites under mefloquine prophylaxis (cohort A) or fifteen NF135-infected mosquito bites and presumptive treatment with artemether/lumefantrine (cohort B). Cohort A participants were exposed to a homologous challenge 19 weeks after immunization. The primary objective of the study was to evaluate the safety and tolerability of CPS immunizations with NF135. RESULTS: Relatively high liver-to-blood inocula were observed during immunization with NF135 in both cohorts. Eighteen of 30 (60%) high-dose participants and 3/10 (30%) low-dose participants experienced grade 3 adverse events 7 to 21 days following their first immunization. All cohort A participants and two participants in cohort B developed breakthrough blood-stage malaria infections during immunizations requiring rescue treatment. The resulting compromised immunizations induced modest sterile protection against homologous challenge in cohort A (5/17; 29%). CONCLUSIONS: These CPS regimes using NF135 were relatively poorly tolerated and frequently required rescue treatment, thereby compromising immunization efficiency and protective efficacy. Consequently, the full potential of NF135 sporozoites for induction of immune protection remains inconclusive. Nonetheless, the high liver-stage burden achieved by this strain highlights it as an interesting potential candidate for novel whole sporozoite immunization approaches. TRIAL REGISTRATION: The trial was registered at ClinicalTrials.gov under identifier NCT03813108.


Subject(s)
Antimalarials , Insect Bites and Stings , Malaria Vaccines , Malaria , Adult , Animals , Humans , Antimalarials/therapeutic use , Artemether, Lumefantrine Drug Combination/therapeutic use , Immunization/methods , Insect Bites and Stings/drug therapy , Malaria/prevention & control , Malaria Vaccines/adverse effects , Plasmodium falciparum , Sporozoites
5.
Front Cell Infect Microbiol ; 13: 955134, 2023.
Article in English | MEDLINE | ID: covidwho-2255628

ABSTRACT

Malaria, which infected more than 240 million people and killed around six hundred thousand only in 2021, has reclaimed territory after the SARS-CoV-2 pandemic. Together with parasite resistance and a not-yet-optimal vaccine, the need for new approaches has become critical. While earlier, limited, studies have suggested that malaria parasites are affected by electromagnetic energy, the outcomes of this affectation vary and there has not been a study that looks into the mechanism of action behind these responses. In this study, through development and implementation of custom applicators for in vitro experimentation, conditions were generated in which microwave energy (MW) killed more than 90% of the parasites, not by a thermal effect but via a MW energy-induced programmed cell death that does not seem to affect mammalian cell lines. Transmission electron microscopy points to the involvement of the haemozoin-containing food vacuole, which becomes destroyed; while several other experimental approaches demonstrate the involvement of calcium signaling pathways in the resulting effects of exposure to MW. Furthermore, parasites were protected from the effects of MW by calcium channel blockers calmodulin and phosphoinositol. The findings presented here offer a molecular insight into the elusive interactions of oscillating electromagnetic fields with P. falciparum, prove that they are not related to temperature, and present an alternative technology to combat this devastating disease.


Subject(s)
COVID-19 , Malaria, Falciparum , Malaria , Parasites , Animals , Humans , Microwaves , SARS-CoV-2 , Malaria, Falciparum/parasitology , Plasmodium falciparum , Mammals
6.
Front Immunol ; 14: 1120298, 2023.
Article in English | MEDLINE | ID: covidwho-2269662

ABSTRACT

The co-occurrence and the similarities between malaria and COVID-19 diseases raise the question of whether SARS-CoV-2 is capable of infecting red blood cells and, if so, whether these cells represent a competent niche for the virus. In this study, we first tested whether CD147 functions as an alternative receptor of SARS-CoV-2 to infect host cells. Our results show that transient expression of ACE2 but not CD147 in HEK293T allows SARS-CoV-2 pseudoviruses entry and infection. Secondly, using a SARS-CoV-2 wild type virus isolate we tested whether the new coronavirus could bind and enter erythrocytes. Here, we report that 10,94% of red blood cells had SARS-CoV-2 bound to the membrane or inside the cell. Finally, we hypothesized that the presence of the malaria parasite, Plasmodium falciparum, could make erythrocytes more vulnerable to SARS-CoV-2 infection due to red blood cell membrane remodelling. However, we found a low coinfection rate (9,13%), suggesting that P. falciparum would not facilitate the entry of SARS-CoV-2 virus into malaria-infected erythrocytes. Besides, the presence of SARS-CoV-2 in a P. falciparum blood culture did not affect the survival or growth rate of the malaria parasite. Our results are significant because they do not support the role of CD147 in SARS-CoV-2 infection, and indicate, that mature erythrocytes would not be an important reservoir for the virus in our body, although they can be transiently infected.


Subject(s)
COVID-19 , Coinfection , Malaria, Falciparum , Humans , SARS-CoV-2 , Plasmodium falciparum , HEK293 Cells , Malaria, Falciparum/parasitology , Erythrocytes
7.
Infect Dis (Lond) ; 55(4): 299-302, 2023 04.
Article in English | MEDLINE | ID: covidwho-2232941

ABSTRACT

BACKGROUND: A rare case of coinfection of Plasmodium falciparum and SARS-CoV-2 disease in Croatia is presented in this report. METHODS: We tracked epidemiological and laboratory findings in a patient with SARS-CoV-2 and Plasmodium falciparum coinfection. A complete blood count was performed using the Sysmex XN-2000 analyser (Sysmex Corporation, Kobe, Japan), coagulation analyses were performed using the BCS XP coagulometer (Siemens Healthineers AG, Erlangen, Germany). Procalcitonin (PCT) and Interleukin-6 (IL-6) were measured by electrochemiluminescence immunoassay (ECLIA) using the Cobas e411 (Roche Diagnostics GmbH, Mannheim, Germany) analyser and high sensitivity troponin I (hsTnI) was measured using the Dimension EXL with LM analyser (Siemens Healthcare Diagnostics, Newark, USA). All other biochemistry analyses were performed using the Olympus AU680 (Beckman Coulter, Brea, California, USA) analyser. White blood cell differential analysis has been performed by examining the blood smear using the CellaVision DM1200 (CellaVision AB, Lund, Sweden) automatic analyser. RESULTS: Even though the patient's initial health condition was disturbed, as a result of the physician's comprehensive anamnesis accompanied by laboratory findings, prompt diagnosis and appropriate therapy were assured, and consequently, the patient recovered. CONCLUSION: In a pandemic, testing each febrile patient for the SARS-CoV-2 virus is of essential importance. However, the possibility of coinfection with another infectious disease agent cannot be disregarded.


Subject(s)
COVID-19 , Coinfection , Humans , Plasmodium falciparum , SARS-CoV-2 , Coinfection/diagnosis , COVID-19/diagnosis , Blood Cell Count/methods
8.
ChemMedChem ; 18(7): e202200586, 2023 04 03.
Article in English | MEDLINE | ID: covidwho-2219680

ABSTRACT

We report the structural functionalization of the terminal amino group of N1 -(7-chloroquinolin-4-yl) butane-1,4-diamine, leading to a series of 7-chloro-4-aminoquinoline derivatives, and their evaluation as potent anti-malarial and anti-viral agents. Some compounds exhibited promising anti-malarial effects against the Plasmodium falciparum 3D7 (chloroquine-sensitive) and Dd2 (chloroquine-resistant) strains. In addition, these compounds were assayed in vitro against influenza A virus (IAV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Compound 5 h, bearing an N-mesityl thiourea group, displayed pronounced anti-infectious effects against malaria, IAV, and SARS-CoV-2. These results provide new insights into drug discovery for the prevention or treatment of malaria and virus co-infection.


Subject(s)
Antimalarials , COVID-19 , Malaria , Humans , Antimalarials/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/therapeutic use , SARS-CoV-2 , Chloroquine/pharmacology , Malaria/drug therapy , Plasmodium falciparum
10.
Clin Lab ; 69(1)2023 Jan 01.
Article in English | MEDLINE | ID: covidwho-2203270

ABSTRACT

BACKGROUND: COVID-19 and malaria share some similar symptoms such as fever, difficulty in breathing, fatigue, and headaches of acute onset. With overlapping symptoms and travel history significant for COVID-19 and malaria, healthcare systems and professionals will face a great challenge in the case of COVID-19 and malaria co-infection. METHODS: Here we presented a patient with COVID-19 infection and refractory anemia of unknown reason. A diagnostic test for malaria was later performed. RESULTS: The patient was ultimately diagnosed with COVID-19 and plasmodium falciparum malaria co-infection. He recovered gradually after receiving anti-malaria treatment. CONCLUSIONS: The present case highlights the danger of focusing only on a diagnosis of COVID-19, reminding clinicians to be vigilant about the possibility of co-infections.


Subject(s)
Anemia , COVID-19 , Coinfection , Malaria, Falciparum , Malaria , Humans , Male , Anemia/diagnosis , Coinfection/diagnosis , COVID-19/complications , East Asian People , Malaria, Falciparum/complications , Malaria, Falciparum/diagnosis , Plasmodium falciparum , China
11.
BMJ Case Rep ; 15(11)2022 Nov 24.
Article in English | MEDLINE | ID: covidwho-2137561

ABSTRACT

Malaria remains one of the most prevalent infectious diseases globally. Despite targets set out by the WHO in 2015, there has been a rise in the number of cases since 2019 as an indirect effect of the COVID-19 pandemic.Cardiac complications are very rarely witnessed with severe malaria. Of the cardiac sequelae, myocarditis is one of the most frequently observed with a handful of case reports in the literature. We report a case of a man in his 50s who developed myocarditis while being managed for severe Plasmodium falciparum malaria in an intensive care unit in the UK and review the literature relevant to this case. This is the second reported case of this condition in the UK.


Subject(s)
COVID-19 , Malaria, Falciparum , Malaria , Myocarditis , Male , Humans , Plasmodium falciparum , Myocarditis/complications , Pandemics , Malaria, Falciparum/complications , Malaria, Falciparum/drug therapy , Malaria/complications
12.
Biomed Res Int ; 2022: 2545830, 2022.
Article in English | MEDLINE | ID: covidwho-2079082

ABSTRACT

The global malaria morbidity and mortality witnessed an increase from 2019 to 2020 partly due to disruptions in control programs' activities imposed by the COVID-19 pandemic. Therefore, there is still a significant burden of malaria in Cameroon which needs attention from all fronts to attain elimination goals. It is normally expected that a typical forest ecology that has undergone urbanization and subjected to high rates of ecological instabilities should also have a shift from characteristic perennial malaria transmission and a shift in the type of malaria endemicity plaguing such distorted forest ecology. In this observational comparative study, we randomly enrolled participants from rural and urban settings of a forest zone during a low malaria transmission period, which coincided with the onset of COVID-19 pandemic. An optimized structured questionnaire was employed, to collect socio-demographic data and associated risk factors. The CareStart™ Malaria HRP2 antigen test was performed on participants from both settings to determine the prevalence of community asymptomatic malaria. Of 307 participants, 188 (61.0%) were from the rural, while 119 (38.8%) from the urban community. The overall prevalence of asymptomatic malaria (27.0%) detected Plasmodium falciparum antigen in 83 participants. The urban community's prevalence was 4.2% (5 positives) while the rural community's was 41.5% (78 positives). In simple logistic regression models, rural forest community and farm around the house were statistically significant predictors of testing positive (coefficient 2.8, 95% CI 1.8-3.7, p value<0.001) and (coefficient 3.1, 95% CI 1.1-5.1, p value =0.003), respectively. In the multivariate model, the strongest predictor of testing positive was living in a rural community, with p < 0.001 and odds ratio of 10.9 (95% CI, 3.8-31.8). These results indicate that during a low transmission period, the prevalence of asymptomatic malaria differs between depleted urban and rural forested settings, suggesting a need for strategic target intervention for the control of asymptomatic malaria.


Subject(s)
COVID-19 , Malaria, Falciparum , Malaria , Humans , Rural Population , Plasmodium falciparum , COVID-19/epidemiology , Pandemics , Malaria/epidemiology , Forests , Prevalence , Malaria, Falciparum/epidemiology
13.
Adv Sci (Weinh) ; 9(28): e2105396, 2022 10.
Article in English | MEDLINE | ID: covidwho-2047424

ABSTRACT

In many malaria-endemic regions, current detection tools are inadequate in diagnostic accuracy and accessibility. To meet the need for direct, phenotypic, and automated malaria parasite detection in field settings, a portable platform to process, image, and analyze whole blood to detect Plasmodium falciparum parasites, is developed. The liberated parasites from lysed red blood cells suspended in a magnetic field are accurately detected using this cellphone-interfaced, battery-operated imaging platform. A validation study is conducted at Ugandan clinics, processing 45 malaria-negative and 36 malaria-positive clinical samples without external infrastructure. Texture and morphology features are extracted from the sample images, and a random forest classifier is trained to assess infection status, achieving 100% sensitivity and 91% specificity against gold-standard measurements (microscopy and polymerase chain reaction), and limit of detection of 31 parasites per µL. This rapid and user-friendly platform enables portable parasite detection and can support malaria diagnostics, surveillance, and research in resource-constrained environments.


Subject(s)
Malaria, Falciparum , Malaria , Parasites , Animals , Erythrocytes , Malaria/diagnosis , Malaria/parasitology , Malaria, Falciparum/diagnosis , Malaria, Falciparum/epidemiology , Malaria, Falciparum/parasitology , Plasmodium falciparum
14.
Malar J ; 21(1): 255, 2022 Sep 06.
Article in English | MEDLINE | ID: covidwho-2038759

ABSTRACT

BACKGROUND: Circulating myeloid-derived-suppressor-cells (MDSC) with immunosuppressive function are increased in human experimental Plasmodium falciparum infection, but have not been studied in clinical malaria. METHODS: Using flow-cytometry, circulating polymorphonuclear-MDSC were evaluated in cryopreserved samples from patients with uncomplicated Plasmodium vivax (n = 8) and uncomplicated (n = 4) and severe (n = 16) falciparum malaria from Papua, Indonesia. RESULTS: The absolute number of circulating polymorphonuclear-MDSC were significantly elevated in severe falciparum malaria patients compared to controls (n = 10). Polymorphonuclear-MDSC levels in uncomplicated vivax malaria were also elevated to levels comparable to that seen in severe falciparum malaria. CONCLUSION: Control of expansion of immunosuppressive MDSC may be important for development of effective immune responses in falciparum and vivax malaria.


Subject(s)
Malaria, Falciparum , Malaria, Vivax , Malaria , Myeloid-Derived Suppressor Cells , Humans , Indonesia , Malaria/complications , Plasmodium falciparum , Plasmodium vivax
15.
Molecules ; 27(17)2022 Aug 31.
Article in English | MEDLINE | ID: covidwho-2033062

ABSTRACT

Malaria is one of the most important infectious diseases worldwide. The causative of the most severe forms of malaria, Plasmodium falciparum, has developed resistances against all the available antimalarial drugs. In the present study, the phytochemical investigation of the green seaweed Halimeda macroloba has afforded two new compounds 1-2, along with 4 known ones 3-6. The structures of the compounds had been confirmed using 1& 2D-NMR and HRESIMS analyses. Extensive machine-learning-supported virtual-screening suggested cytochrome-C enzyme as a potential target for compound 2. Docking, absolute-binding-free-energy (ΔGbinding) and molecular-dynamics-simulation (MDS) of compound 2 revealed the strong binding interaction of this compound with cytochrome-C. In vitro testing for crude extract and isolated compounds revealed the potential in vitro inhibitory activity of both extract and compound 2 against P. falciparum. The crude extract was able to inhibit the parasite growth with an IC50 value of 1.8 ± 0.35 µg/mL. Compound 2 also showed good inhibitory activity with an IC50 value of 3.2 ± 0.23 µg/mL. Meanwhile, compound 6 showed moderate inhibitory activity with an IC50 value of 19.3 ± 0.51 µg/mL. Accordingly, the scaffold of compound 2 can be considered as a good lead compound for the future development of new antimalarial agents.


Subject(s)
Antimalarials , Malaria, Falciparum , Malaria , Seaweed , Antimalarials/chemistry , Cytochromes , Humans , Malaria/drug therapy , Malaria, Falciparum/drug therapy , Plant Extracts/chemistry , Plasmodium falciparum
17.
PLoS One ; 17(9): e0270863, 2022.
Article in English | MEDLINE | ID: covidwho-2021856

ABSTRACT

Plasmodium falciparum, a protozoan parasite and causative agent of human malaria, has one of the most A/T-biased genomes sequenced to date. This may give the genome and the transcriptome unusual structural features. Recent progress in sequencing techniques has made it possible to study the secondary structures of RNA molecules at the transcriptomic level. Thus, in this study we produced the in vivo RNA structurome of a protozoan parasite with a highly A/U-biased transcriptome. We showed that it is possible to probe the secondary structures of P. falciparum RNA molecules in vivo using two different chemical probes, and obtained structures for more than half of all transcripts in the transcriptome. These showed greater stability (lower free energy) than the same structures modelled in silico, and structural features appeared to influence translation efficiency and RNA decay. Finally, we compared the P. falciparum RNA structurome with the predicted RNA structurome of an A/U-balanced species, P. knowlesi, finding a bias towards lower overall transcript stability and more hairpins and multi-stem loops in P. falciparum. This unusual protozoan RNA structurome will provide a basis for similar studies in other protozoans and also in other unusual genomes.


Subject(s)
Malaria, Falciparum , Malaria , Parasites , Animals , Genome, Protozoan , Humans , Malaria/genetics , Malaria, Falciparum/parasitology , Parasites/genetics , Plasmodium falciparum/genetics , Protozoan Proteins/genetics , RNA , RNA, Protozoan/genetics , Transcriptome
18.
Virulence ; 13(1): 634-653, 2022 12.
Article in English | MEDLINE | ID: covidwho-2008470

ABSTRACT

Globally, malaria is a public health concern, with severe malaria (SM) contributing a major share of the disease burden in malaria endemic countries. In this context, identification and validation of SM biomarkers are essential in clinical practice. Some biomarkers (C-reactive protein, angiopoietin 2, angiopoietin-2/1 ratio, platelet count, histidine-rich protein 2) have yielded interesting results in the prognosis of Plasmodium falciparum severe malaria, but for severe P. vivax and P. knowlesi malaria, similar evidence is missing. The validation of these biomarkers is hindered by several factors such as low sample size, paucity of evidence-evaluating studies, suboptimal values of sensitivity/specificity, poor clinical practicality of measurement methods, mixed Plasmodium infections, and good clinical value of the biomarkers for concurrent infections (pneumonia and current COVID-19 pandemic). Most of these biomarkers are non-specific to pathogens as they are related to host response and hence should be regarded as prognostic/predictive biomarkers that complement but do not replace pathogen biomarkers for clinical evaluation of SM patients. This review highlights the importance of research on diagnostic/predictive/therapeutic biomarkers, neglected malaria species, and clinical practicality of measurement methods in future studies. Finally, the importance of omics technologies for faster identification/validation of SM biomarkers is also included.


Subject(s)
COVID-19 , Malaria, Falciparum , Malaria , Biomarkers , Humans , Pandemics , Plasmodium falciparum , Plasmodium vivax
19.
Anal Chim Acta ; 1225: 340246, 2022 Sep 08.
Article in English | MEDLINE | ID: covidwho-1982438

ABSTRACT

Protein-based diagnostics are the standard of care for screening and diagnosing a broad range of diseases and medical conditions. The current gold standard method for quantifying proteins in clinical specimens is the enzyme-linked immunosorbent assay (ELISA), which offers high analytical sensitivity, can process many samples at once, and is widely available in many diagnostic laboratories worldwide. However, running an ELISA is cumbersome, requiring multiple liquid handling and washing steps, and time-intensive (∼2 - 4 h per test). Here, we demonstrate a unique magneto-ELISA that utilizes dually labeled magnetic nanoparticles (DMPs) coated with horseradish peroxidase (HRP) and an HRP-conjugated detection antibody, enabling rapid immunomagnetic enrichment and signal amplification. For proof of concept, this assay was used to detect Plasmodium falciparum histidine-rich protein 2 (PfHRP2), a malaria parasite biomarker, which exhibited a lower limit of detection of 2 pg mL-1 (33 fM) in human serum. Measurements of PfHRP2 in clinical blood samples from individuals with and without P. falciparum infection revealed that this magneto-ELISA offers a superior diagnostic accuracy compared to a commercial PfHRP2 ELISA kit. We also demonstrate the versatility of this platform by adapting it for the detection of SARS-CoV-2 nucleocapsid protein, which could be detected at concentrations as low as 8 pg mL-1 (174 fM) in human serum. In addition to its high analytical performance, this assay can be completed in 30 min, requires no specialized equipment, and is compatible with standard microplate readers and ELISA protocols, allowing it to integrate readily into current clinical practice.


Subject(s)
COVID-19 , Malaria, Falciparum , Nanoparticles , Enzyme-Linked Immunosorbent Assay/methods , Humans , Malaria, Falciparum/diagnosis , Malaria, Falciparum/parasitology , Plasmodium falciparum , SARS-CoV-2
20.
Front Immunol ; 13: 809264, 2022.
Article in English | MEDLINE | ID: covidwho-1979036

ABSTRACT

Memory B cells (MBCs) and plasma antibodies against Plasmodium falciparum (Pf) merozoite antigens are important components of the protective immune response against malaria. To gain understanding of how responses against Pf develop in these two arms of the humoral immune system, we evaluated MBC and antibody responses against the most abundant merozoite antigen, full-length Pf merozoite surface protein 1 (PfMSP1FL), in individuals from a region in Uganda with high Pf transmission. Our results showed that PfMSP1FL-specific B cells in adults with immunological protection against malaria were predominantly IgG+ classical MBCs, while children with incomplete protection mainly harbored IgM+ PfMSP1FL-specific classical MBCs. In contrast, anti-PfMSP1FL plasma IgM reactivity was minimal in both children and adults. Instead, both groups showed high plasma IgG reactivity against PfMSP1FL, with broadening of the response against non-3D7 strains in adults. The B cell receptors encoded by PfMSP1FL-specific IgG+ MBCs carried high levels of amino acid substitutions and recognized relatively conserved epitopes on the highly variable PfMSP1 protein. Proteomics analysis of PfMSP119-specific IgG in plasma of an adult revealed a limited repertoire of anti-MSP1 antibodies, most of which were IgG1 or IgG3. Similar to B cell receptors of PfMSP1FL-specific MBCs, anti-PfMSP119 IgGs had high levels of amino acid substitutions and their sequences were predominantly found in classical MBCs, not atypical MBCs. Collectively, these results showed evolution of the PfMSP1-specific humoral immune response with cumulative Pf exposure, with a shift from IgM+ to IgG+ B cell memory, diversification of B cells from germline, and stronger recognition of PfMSP1 variants by the plasma IgG repertoire.


Subject(s)
Malaria , Merozoite Surface Protein 1 , Adult , Animals , Antibodies, Protozoan , Antibody Formation , Child , Humans , Immunoglobulin G , Immunoglobulin M/metabolism , Memory B Cells , Merozoites , Plasmodium falciparum , Receptors, Antigen, B-Cell/metabolism , Uganda
SELECTION OF CITATIONS
SEARCH DETAIL