1
|
Sallam M, Al-Khatib AO, Al-Mahzoum KS, Abdelaziz DH, Sallam M. Current Developments in Malaria Vaccination: A Concise Review on Implementation, Challenges, and Future Directions. Clin Pharmacol 2025; 17:29-47. [PMID: 40191019 PMCID: PMC11971972 DOI: 10.2147/cpaa.s513282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Accepted: 03/25/2025] [Indexed: 04/09/2025] Open
Abstract
Introduction Malaria remains a persistent challenge in global health, disproportionately affecting populations in endemic regions (eg, sub-Saharan Africa). Despite decades of international collaborative efforts, malaria continues to claim hundreds of thousands of lives each year, with young children and pregnant women enduring the heaviest burden. This concise review aimed to provide an up-to-date assessment of malaria vaccines progress, challenges, and future directions. Methods A PubMed/MEDLINE search (2015-2024) was conducted to identify studies on malaria vaccine development, implementation barriers, efficacy, and vaccination hesitancy. Clinical trials, reviews, and global health reports were included based on relevance to the review aims. No strict inclusion criteria were applied, and selection was guided by key review themes and policy relevance. Results The introduction of pre-erythrocytic malaria vaccines (RTS,S/AS01 and R21/Matrix-M), represents an important milestone in malaria control efforts with promising results from the erythrocytic vaccine RH5.1/Matrix-M in recent clinical trials. However, the approval of these vaccines is accompanied by significant challenges such as the limited efficacy, the complexity of multi-dose regimens, and numerous barriers to widespread implementation in resource-limited settings. The review identified the complex challenges to broad malaria vaccination coverage, including logistical barriers, healthcare infrastructure effect, financial limitations, malaria vaccine hesitancy, among other obstacles in malaria-endemic regions. Promising developments in malaria vaccination, such as next-generation candidates (eg, mRNA-based vaccines), hold the potential to offer improved efficacy, longer-lasting protection, and greater scalability. There is a critical need to integrate malaria vaccination efforts with established malaria control interventions (eg, insecticide-treated bed nets, vector control strategies, and anti-malarial drugs). Conclusion Achieving sustained control of malaria morbidity and mortality will require strong global collaboration, sufficient funding, and continuous efforts to address inequities in access and delivery of malaria control measures including the malaria vaccines.
Collapse
Affiliation(s)
- Malik Sallam
- Department of Pathology, Microbiology and Forensic Medicine, School of Medicine, The University of Jordan, Amman, Jordan
- Department of Clinical Laboratories and Forensic Medicine, Jordan University Hospital, Amman, Jordan
- Department of Translational Medicine, Faculty of Medicine, Lund University, Malmö, Sweden
| | - Arwa Omar Al-Khatib
- Faculty of Pharmacy, Hourani Center for Applied Scientific Research, Al-Ahliyya Amman University, Amman, Jordan
| | | | - Doaa H Abdelaziz
- Department of Clinical Pharmacy, Faculty of Pharmacy, Al-Baha University, Al-Baha, Saudi Arabia
- Department of Clinical Pharmacy, the National Hepatology and Tropical Medicine Research Institute, Cairo, Egypt
| | - Mohammed Sallam
- Department of Pharmacy, Mediclinic Parkview Hospital, Mediclinic Middle East, Dubai, United Arab Emirates
| |
Collapse
|
2
|
Adeleke MA. Computational Development of Transmission-Blocking Vaccine Candidates Based on Fused Antigens of Pre- and Post-fertilization Gametocytes Against Plasmodium falciparum. Bioinform Biol Insights 2025; 19:11779322241306215. [PMID: 40034580 PMCID: PMC11873872 DOI: 10.1177/11779322241306215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 11/21/2024] [Indexed: 03/05/2025] Open
Abstract
Plasmodium falciparum is the most fatal species of malaria parasites in humans. Attempts at developing vaccines against the malaria parasites have not been very successful even after the approval of the RTS, S/AS01 vaccine. There is a continuous need for more effective vaccines including sexual-stage antigens that could block the transmission of malaria parasites between mosquitoes and humans. Low immunogenicity, expression, and stability are some of the challenges of transmission-blocking vaccine (TBV). This study was designed to computationally identify TBV candidates based on fused antigens by combining highly antigenic peptides from prefertilization (Pfs230, Pfs48/45) and postfertilization (Pfs25, Pfs28) gametocytes. The peptides were selected based on their antigenicity, nonallergenicity, and lack of similarity with the human proteome. Two fused antigens vaccine candidates (FAVCs) were constructed using Flagellin Salmonella enterica (FAVC-FSE) and Cholera toxin B (FAVC-CTB) as adjuvants. The constructs were evaluated for their physicochemical properties, structural stability, immunogenicity, and potential to elicit cross-protection across multiple Plasmodium species. The results yielded antigenic peptides, with antigenicity scores between 0.7589 and 1.1821. The structural analysis of FAVC-FSE and FAVC-CTB showed a Z-score of -6.70 and -4.79, a Ramachandran plot of 96.94% and 94.86% with overall quality of 94.20% and 89.85%, respectively. The FAVCs contained CD8+, CD4+, and linear B-cell epitopes with antigenicity scores between 1.2089 and 2.8623, 0.5663 and 2.4132, and 1.5196 and 2.2212, respectively. Each FAVC generated 6 conformational B-cells. High population coverage values were recorded for the FAVCs. The ability of the FAVCs to trigger immune response was evaluated through an in silico immune stimulation. The low-binding interaction energy that resulted from molecular docking and dynamics simulations showed a strong affinity of FAVCs to Toll-like receptor 5 (TLR5). The results indicate that the FAVC-FSE vaccine candidate is more promising to interrupt P falciparum transmission and provides a baseline for experimental validation.
Collapse
Affiliation(s)
- Matthew A. Adeleke
- Discipline of Genetics, School of Life Sciences, College of Agriculture, Engineering and Sciences, University of KwaZulu-Natal, Durban, South Africa
| |
Collapse
|
3
|
Chick JA, Abongdia NN, Shey RA, Apinjoh TO. Computational design, expression, and characterization of a Plasmodium falciparum multi-epitope, multi-stage vaccine candidate (PfCTMAG). Heliyon 2025; 11:e42014. [PMID: 39906795 PMCID: PMC11791285 DOI: 10.1016/j.heliyon.2025.e42014] [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: 09/25/2024] [Revised: 01/13/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025] Open
Abstract
Malaria, a tropical disease, claims the lives of thousands of people annually and the development of resistance to insecticides and antimalarial drugs poses a great challenge to current prevention and control strategies. Current malaria vaccines are limited in efficacy, duration of protection, and safety, due to the high antigenic diversity and complex life cycle of the Plasmodium parasite. This study sought to design and assess a more effective multi-stage, multi-epitope vaccine candidate for the control of malaria. A multi-epitope malaria vaccine candidate was designed in silico using multiple antigens from both the pre-erythrocytic and erythrocytic stages, expressed in bacteria, and its sero-reactivity to antibodies in plasma from malaria-positive (cases) and negative individuals (controls) was assessed using enzyme-linked immunosorbent assay (ELISA). Immunization experiments were equally conducted with BALB/c mice. In-silico analysis revealed that the designed antigen, PfCTMAG (Plasmodium falciparum Circumsporozoite, Thrombospondin-related adhesion protein, Merozoite surface protein 2, Apical asparagine (Asn)-rich protein and Glutamate-Rich Protein), effectively bound to Toll-like receptor 4 (TLR-4) and triggered a strong immune response. In sero-reactivity studies, malaria-positives (cases) had higher anti-PfCTMAG IgG ( p = 0.024) and IgM ( p < 0.001) levels compared to malaria negatives (controls). The mice immunized with PfCTMAG did not show adverse reactions and had higher levels of IgG antibodies (p = 0.002) compared to controls, thereby validating the safety and immunogenicity of PfCTMAG as a promising vaccine candidate.
Collapse
Affiliation(s)
- Joan A. Chick
- Department of Chemical and Biological Engineering, National Higher Polytechnic Institute, The University of Bamenda, Cameroon
| | - Nadege N. Abongdia
- Department of Chemical and Biological Engineering, National Higher Polytechnic Institute, The University of Bamenda, Cameroon
| | - Robert A. Shey
- Department of Biochemistry and Molecular Biology, Faculty of Science, University of Buea, Cameroon
| | - Tobias O. Apinjoh
- Department of Chemical and Biological Engineering, National Higher Polytechnic Institute, The University of Bamenda, Cameroon
- Department of Biochemistry and Molecular Biology, Faculty of Science, University of Buea, Cameroon
| |
Collapse
|
4
|
Quadros Gomes AR, Castro ALG, Ferreira GG, Brígido HPC, Varela ELP, Vale VV, Carneiro LA, Dolabela MF, Percario S. Impact on parasitemia, survival time and pro-inflammatory immune response in mice infected with Plasmodium berghei treated with Eleutherine plicata. Front Pharmacol 2024; 15:1484934. [PMID: 39703398 PMCID: PMC11656046 DOI: 10.3389/fphar.2024.1484934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 11/12/2024] [Indexed: 12/21/2024] Open
Abstract
In vitro studies with Plasmodium falciparum have demonstrated the antiparasitic activity of E. plicata, attributed to its naphthoquinones. This study reports on pro-inflammatory changes in mice infected with P. berghei and correlates these changes with parasitemia and survival. The ethanol extract of Eleutherine plicata (EEEp) was fractionated under reflux to obtain the dichloromethane fraction (FDMEp) and isolated compounds from E. plicata, relating these to survival time and parasitemia. Antimalarial activity was evaluated using the Peters suppressive test, with mice infected with Plasmodium berghei and treated with E. plicata, assessing parasitemia and survival over 30 days. The pro-inflammatory profile was determined by measuring interleukin-10, interferon-γ (IFN-γ), and nitric oxide levels. EEEp, FDMEp, and eleutherol showed activity on the 5th day of infection, with only FDMEp being active on the 8th day. Treatment with EEEp and FDMEp extended animal survival, reduced IFN-γ and NO levels, and increased IL-10 levels. Eleutherol significantly altered the response, with eleutherol glucuronide seemingly active by binding to lactate dehydrogenase, inhibiting hemozoin metabolism, leading to parasite death. Pro-inflammatory changes did not appear to correlate with survival and reduced parasitemia. In summary, FDMEp and eleutherol reduced parasitemia, extended survival, and modulated the inflammatory response. FDMEp and eleutherol are promising candidates for developing new antimalarial drugs.
Collapse
Affiliation(s)
- Antônio Rafael Quadros Gomes
- Postgraduate Program in Pharmaceutical Innovation, Federal University of Pará, Belém, Brazil
- Postgraduate Program in Biodiversity and Biotechnology, Federal University of Pará, Belém, Brazil
| | - Ana Laura Gadelha Castro
- Postgraduate Program in Pharmaceutical Innovation, Federal University of Pará, Belém, Brazil
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Pará, Belém, Brazil
| | | | | | - Everton Luiz Pompeu Varela
- Postgraduate Program in Biodiversity and Biotechnology, Federal University of Pará, Belém, Brazil
- Oxidative Stress Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| | - Valdicley Vieira Vale
- Postgraduate Program in Pharmaceutical Innovation, Federal University of Pará, Belém, Brazil
| | | | - Maria Fâni Dolabela
- Postgraduate Program in Pharmaceutical Innovation, Federal University of Pará, Belém, Brazil
- Postgraduate Program in Biodiversity and Biotechnology, Federal University of Pará, Belém, Brazil
- Postgraduate Program in Pharmaceutical Sciences, Federal University of Pará, Belém, Brazil
| | - Sandro Percario
- Postgraduate Program in Biodiversity and Biotechnology, Federal University of Pará, Belém, Brazil
- Oxidative Stress Laboratory, Institute of Biological Sciences, Federal University of Pará, Belém, Brazil
| |
Collapse
|
5
|
Asare KK, Kwapong SS, Tey P, Sackey V, Nuvor SV, Amoah LE. Plasmodium Falciparum and mosquito vector IgG patterns across suspected malaria cases in Ghana. BMC Infect Dis 2024; 24:1374. [PMID: 39623362 PMCID: PMC11613542 DOI: 10.1186/s12879-024-10248-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
INTRODUCTION Malaria, a widespread tropical disease, remains a significant global health issue, resulting in numerous deaths each year. In Ghana, malaria is a leading cause of illness, contributing to a large proportion of hospital outpatient visits. The study assessed the pattern of malaria and vector IgG antibody levels among suspected malaria patients seeking healthcare at selected health facilities across Ghana. METHODS Samples from a total of 823 participants aged 1 to 85 years with clinical malaria from the ten regions of Ghana were recruited into the study. Archived plasma obtained from each participant was used to assess antibody responses against MSP1 (19 k), MSP2 (FC27 & 3D7), MSP3, gSG6-P1, and GLURP-RO using ELISA. The data were categorized according to study site, age group, gender, and diagnostic tests. Data were analyzed using Kruskal-Wallis's statistics. The statistical significance was assessed at 0.05. RESULTS The mean ± standard error of the mean (S.E) of MSP3 IgG concentration for the different age groups were 16, 847 ± 3, 031 ng/mL for 0-4 years, 18, 973 ± 4,357 ng/mL for 5-10 years, 25,961 ± 5,436 ng/mL for 11-15 years and 76, 244 ± 8, 209 ng/mL for ≥ 16 years. A significant (Kruskal-Wallis statistic = 122.6, p < 0.0001) increase in P. falciparum MSP 3 (p < 0.0001) and gSG6-P1(p < 0.0001) IgG concentration was observed with increasing age categories. There were significant differences in antibody responses against MSP2 (FC27) IgG (Kruskal-Wallis statistic = 29.63, p = 0.0005), MSP3 IgG (Kruskal-Wallis statistic = 32.53, p = 0.0002), GLURP-RO IgG (Kruskal-Wallis statistic = 52.8, p < 0.0001) and gSG6-P1 IgG (Kruskal-Wallis statistic = 152.8, p < 0.0001) across the study regions. CONCLUSION The study reveals that IgG against merozoite surface proteins MSP3, GLURP-RO, and gSG6-P1 but not MSP1 and MSP2 antibodies increase with age. The mean IgG antibody concentrations varied in the selected regions of Ghana. A longitudinal study where confounding factors are controlled for is recommended to provide insights into the development of immunity and antibody efficacy, and to enhance the effectiveness of malaria prevention efforts in Ghana. This will help improve the overall understanding of malaria transmission.
Collapse
Affiliation(s)
- Kwame Kumi Asare
- Biomedical and Clinical Research Centre, College of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
- Department of Biomedical Science, School of Allied Health Sciences, University of Cape Coast, Cape Coast, Ghana
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Sebastian Shine Kwapong
- Department of Microbiology and Immunology, School of Medical Sciences, University of Cape Coast, Cape Coast, Ghana
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Prosper Tey
- Department of Microbiology and Immunology, School of Medical Sciences, University of Cape Coast, Cape Coast, Ghana
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana
| | - Vincent Sackey
- Department of Microbiology and Immunology, School of Medical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Samuel Victor Nuvor
- Department of Microbiology and Immunology, School of Medical Sciences, University of Cape Coast, Cape Coast, Ghana
| | - Linda Eva Amoah
- Department of Immunology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra, Ghana.
| |
Collapse
|
6
|
Yadav N, Kalata AC, Reynolds RA, Raappana A, Sather DN, Murphy SC. Identifying Plasmodium P36 and P52 antigens for co-administration with circumsporozoite protein to enhance vaccine efficacy. RESEARCH SQUARE 2024:rs.3.rs-4909396. [PMID: 39399676 PMCID: PMC11469399 DOI: 10.21203/rs.3.rs-4909396/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2024]
Abstract
Vaccines targeting the complex pre-erythrocytic stage of Plasmodium parasites may benefit from inclusion of multiple antigens. However, discerning protective effects can be difficult because newer candidates may not be as protective as leading antigens like the circumsporozoite protein (CSP) in the conventional pre-clinical mouse model. We developed a modified mouse model challenge strategy that maximizes the contribution of T cells induced by novel candidate antigens at the sporozoite challenge time point and used this approach to test Plasmodium P36 and P52 vaccine candidates alone and in concert with non-protective doses of CSP. Co-administration of P36 and/or P52 with CSP achieved 80-100% sterile protection in mice, compared to only 7-30% protection for each individual antigen. P36 and P52 vaccination induced murine CD4+ and CD8+ T cell responses, but not antibody responses. This study adds P36 and P52 as promising vaccine antigens that may enhance protection achieved by CSP vaccination.
Collapse
Affiliation(s)
- Naveen Yadav
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
| | - Anya C. Kalata
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
| | - Rebekah A. Reynolds
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
| | - Andrew Raappana
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - D. Noah Sather
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| | - Sean C. Murphy
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, United States of America
- Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, United States of America
- Department of Microbiology, University of Washington, Seattle, WA, United States of America
- Seattle Children’s Research Institute, Seattle, WA, United States of America
| |
Collapse
|
7
|
Obeagu EI, Obeagu GU. Adapting to the shifting landscape: Implications of climate change for malaria control: A review. Medicine (Baltimore) 2024; 103:e39010. [PMID: 39029063 PMCID: PMC11398779 DOI: 10.1097/md.0000000000039010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/21/2024] Open
Abstract
Malaria, a global public health challenge, continues to affect millions of lives, particularly in regions where its transmission is endemic. The interplay between climate change and malaria dynamics has emerged as a critical concern, reshaping the landscape of this vector-borne disease. This review publication, titled "Adapting to the shifting landscape: Implications of climate change for malaria control," explores the multifaceted relationship between climate change and the control of malaria. The paper begins by dissecting the influence of climate change on malaria dynamics, including alterations in temperature, precipitation, and other climatic factors that impact the habitat and life cycle of malaria vectors. It delves into the evolving ecology and behavior of malaria vectors in response to changing climatic conditions, emphasizing the importance of understanding these adaptations. As a response to this shifting landscape, the review discusses adaptive strategies for malaria control, ranging from vector control measures to the utilization of climate data in early warning systems. Community engagement and education are highlighted as essential components of these strategies, recognizing the vital role of local communities in effective malaria control efforts. The paper also identifies future directions and research needs, underscoring the importance of staying ahead of the evolving climate-malaria relationship. This review underscores the urgency of adapting to the changing landscape of malaria transmission driven by climate change. It emphasizes the significance of proactively addressing climate-related challenges to enhance malaria control and protect the health and well-being of vulnerable populations.
Collapse
|
8
|
Jackeline Pérez-Vega M, Manuel Corral-Ruiz G, Galán-Salinas A, Silva-García R, Mancilla-Herrera I, Barrios-Payán J, Fabila-Castillo L, Hernández-Pando R, Enid Sánchez-Torres L. Acute lung injury is prevented by monocyte locomotion inhibitory factor in an experimental severe malaria mouse model. Immunobiology 2024; 229:152823. [PMID: 38861873 DOI: 10.1016/j.imbio.2024.152823] [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: 08/29/2023] [Revised: 06/01/2024] [Accepted: 06/05/2024] [Indexed: 06/13/2024]
Abstract
Acute lung injury caused by severe malaria (SM) is triggered by a dysregulated immune response towards the infection with Plasmodium parasites. Postmortem analysis of human lungs shows diffuse alveolar damage (DAD), the presence of CD8 lymphocytes, neutrophils, and increased expression of Intercellular Adhesion Molecule 1 (ICAM-1). P. berghei ANKA (PbA) infection in C57BL/6 mice reproduces many SM features, including acute lung injury characterized by DAD, CD8+ T lymphocytes and neutrophils in the lung parenchyma, and tissular expression of proinflammatory cytokines and adhesion molecules, such as IFNγ, TNFα, ICAM, and VCAM. Since this is related to a dysregulated immune response, immunomodulatory agents are proposed to reduce the complications of SM. The monocyte locomotion inhibitory factor (MLIF) is an immunomodulatory pentapeptide isolated from axenic cultures of Entamoeba hystolitica. Thus, we evaluated if the MLIF intraperitoneal (i.p.) treatment prevented SM-induced acute lung injury. The peptide prevented SM without a parasiticidal effect, indicating that its protective effect was related to modifications in the immune response. Furthermore, peripheral CD8+ leukocytes and neutrophil proportions were higher in infected treated mice. However, the treatment prevented DAD, CD8+ cell infiltration into the pulmonary tissue and downregulated IFNγ. Moreover, VCAM-1 expression was abrogated. These results indicate that the MLIF treatment downregulated adhesion molecule expression, impeding cell migration and proinflammatory cytokine tissular production, preventing acute lung injury induced by SM. Our findings represent a potential novel strategy to avoid this complication in various events where a dysregulated immune response triggers lung injury.
Collapse
Affiliation(s)
- Martha Jackeline Pérez-Vega
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico; Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Gerardo Manuel Corral-Ruiz
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico; Posgrado en Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Adrian Galán-Salinas
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico
| | - Raúl Silva-García
- Unidad de Investigación Médica en Inmunología, Hospital de Pediatría, CMN-Siglo XXI, IMSS, Ciudad de México, Mexico
| | - Ismael Mancilla-Herrera
- Departamento de Infectología e Inmunología, Instituto Nacional de Perinatología, Ciudad de México, Mexico
| | - Jorge Barrios-Payán
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico
| | | | - Rogelio Hernández-Pando
- Sección de Patología Experimental, Departamento de Patología, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Ciudad de México, Mexico.
| | - Luvia Enid Sánchez-Torres
- Departamento de Inmunología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Ciudad de México, Mexico.
| |
Collapse
|
9
|
McNitt SA, Dick JK, Hernandez Castaneda M, Sangala JA, Pierson M, Macchietto M, Burrack KS, Crompton PD, Seydel KB, Hamilton SE, Hart GT. Phenotype and function of IL-10 producing NK cells in individuals with malaria experience. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.11.593687. [PMID: 38798324 PMCID: PMC11118352 DOI: 10.1101/2024.05.11.593687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Plasmodium falciparum infection can trigger high levels of inflammation that lead to fever and sometimes severe disease. People living in malaria-endemic areas gradually develop resistance to symptomatic malaria and control both parasite numbers and the inflammatory response. We previously found that adaptive natural killer (NK) cells correlate with reduced parasite load and protection from symptoms. We also previously found that murine NK cell production of IL-10 can protect mice from experimental cerebral malaria. Human NK cells can also secrete IL-10, but it was unknown what NK cell subsets produce IL-10 and if this is affected by malaria experience. We hypothesize that NK cell immunoregulation may lower inflammation and reduce fever induction. Here, we show that NK cells from subjects with malaria experience make significantly more IL-10 than subjects with no malaria experience. We then determined the proportions of NK cells that are cytotoxic and produce interferon gamma and/or IL-10 and identified a signature of adaptive and checkpoint molecules on IL-10-producing NK cells. Lastly, we find that co-culture with primary monocytes, Plasmodium -infected RBCs, and antibody induces IL-10 production by NK cells. These data suggest that NK cells may contribute to protection from malaria symptoms via IL-10 production.
Collapse
|
10
|
López-Guzmán C, García AM, Ramirez JD, Aliaga TT, Fernández-Moya A, Kemmerling U, Vásquez AM. Plasmodium falciparum alters the trophoblastic barrier and stroma villi organization of human placental villi explants. Malar J 2024; 23:130. [PMID: 38693572 PMCID: PMC11064279 DOI: 10.1186/s12936-024-04960-9] [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: 01/10/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024] Open
Abstract
BACKGROUND The sequestration of Plasmodium falciparum infected erythrocytes in the placenta, and the resulting inflammatory response affects maternal and child health. Despite existing information, little is known about the direct impact of P. falciparum on the placental barrier formed by trophoblast and villous stroma. This study aimed to assess placental tissue damage caused by P. falciparum in human placental explants (HPEs). METHODS HPEs from chorionic villi obtained of human term placentas (n = 9) from normal pregnancies were exposed to P. falciparum-infected erythrocytes (IE) for 24 h. HPEs were embedded in paraffin blocks and used to study tissue damage through histopathological and histochemical analysis and apoptosis using TUNEL staining. Culture supernatants were collected to measure cytokine and angiogenic factors and to determine LDH activity as a marker of cytotoxicity. A subset of archived human term placenta paraffin-embedded blocks from pregnant women with malaria were used to confirm ex vivo findings. RESULTS Plasmodium falciparum-IE significantly damages the trophoblast layer and the villous stroma of the chorionic villi. The increased LDH activity and pathological findings such as syncytial knots, fibrin deposits, infarction, trophoblast detachment, and collagen disorganization supported these findings. The specific damage to the trophoblast and the thickening of the subjacent basal lamina were more pronounced in the ex vivo infection. In contrast, apoptosis was higher in the in vivo infection. This disparity could be attributed to the duration of exposure to the infection, which significantly varied between individuals naturally exposed over time and the 24-h exposure in the ex vivo HPE model. CONCLUSION Exposure to P. falciparum-IE induces a detachment of the syncytiotrophoblast, disorganization of the stroma villi, and an increase in apoptosis, alterations that may be associated with adverse results such as intrauterine growth restriction and low birth weight.
Collapse
Affiliation(s)
- Carolina López-Guzmán
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Calle 62 #52-59 Torre 1, Laboratorio 610, Medellín, Colombia
| | - Ana María García
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Calle 62 #52-59 Torre 1, Laboratorio 610, Medellín, Colombia
| | - Juan Diego Ramirez
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Calle 62 #52-59 Torre 1, Laboratorio 610, Medellín, Colombia
| | - Trinidad Torres Aliaga
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Alejandro Fernández-Moya
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Ulrike Kemmerling
- Instituto de Ciencias Biomédicas, Facultad de Medicina, Universidad de Chile, Independencia 1027, Santiago, Chile
| | - Ana María Vásquez
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Calle 62 #52-59 Torre 1, Laboratorio 610, Medellín, Colombia.
- Escuela de Microbiología, Universidad de Antioquia, Calle 67 # 53-108, Bloque 5, Oficina 5-135, Medellín, Colombia.
| |
Collapse
|
11
|
Abosalif KOA, Abdalla AE, Junaid K, Eltayeb LB, Ejaz H. The interleukin-10 family: Major regulators of the immune response against Plasmodium falciparum infections. Saudi J Biol Sci 2023; 30:103805. [PMID: 37727525 PMCID: PMC10506046 DOI: 10.1016/j.sjbs.2023.103805] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2023] [Revised: 08/16/2023] [Accepted: 09/01/2023] [Indexed: 09/21/2023] Open
Abstract
Malaria caused by the Plasmodium falciparum strain is more severe because of this protozoan's ability to disrupt the physiology of host cells during the blood stages of development by initiating the production of the interleukin-10 (IL-10) family of cytokines. P. falciparum feeds on hemoglobin and causes host cells to adhere to the walls of blood vessels by remodeling their composition. IL-10 is produced by CD4+ T cells that inhibits antigen-presenting cells' activity to prevent inflammation. This cytokine and its family members are crucial in promoting malarial infection by inhibiting the host's protective immune response, thus initiating Plasmodium parasitemia. IL-10 is also responsible for preventing severe pathology during Plasmodium infection and initiates several signaling pathways to alter the physiology of host cells during malarial infection. This review summarizes the critical aspects of P. falciparum infection, including its role in signaling pathways for cytokine exudation, its effect on microRNA, the human immune response in malaria, and the role played by the liver hormone hepcidin. Moreover, future aspects of vaccine development and therapeutic strategies to combat P. falciparum infections are also discussed in detail.
Collapse
Affiliation(s)
- Khalid Omer Abdalla Abosalif
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Abualgasim Elgaili Abdalla
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| | - Kashaf Junaid
- School of Biological and Behavioural Sciences, Queen Mary University of London, London E1 4NS, UK
| | - Lienda Bashier Eltayeb
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, Prince Sattam Bin AbdulAziz University- Al-Kharj, 11942 Riyadh, Saudi Arabia
| | - Hasan Ejaz
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, Sakaka 72388, Saudi Arabia
| |
Collapse
|
12
|
Tsoumani ME, Voyiatzaki C, Efstathiou A. Malaria Vaccines: From the Past towards the mRNA Vaccine Era. Vaccines (Basel) 2023; 11:1452. [PMID: 37766129 PMCID: PMC10536368 DOI: 10.3390/vaccines11091452] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 08/29/2023] [Accepted: 09/01/2023] [Indexed: 09/29/2023] Open
Abstract
Plasmodium spp. is the etiological agent of malaria, a life-threatening parasitic disease transmitted by infected mosquitoes. Malaria remains a major global health challenge, particularly in endemic regions. Over the years, various vaccine candidates targeting different stages of Plasmodium parasite life-cycle have been explored, including subunit vaccines, vectored vaccines, and whole organism vaccines with Mosquirix, a vaccine based on a recombinant protein, as the only currently approved vaccine for Plasmodium falciparum malaria. Despite the aforementioned notable progress, challenges such as antigenic diversity, limited efficacy, resistant parasites escaping protective immunity and the need for multiple doses have hindered the development of a highly efficacious malaria vaccine. The recent success of mRNA-based vaccines against SARS-CoV-2 has sparked renewed interest in mRNA vaccine platforms. The unique mRNA vaccine features, including their potential for rapid development, scalability, and flexibility in antigen design, make them a promising avenue for malaria vaccine development. This review provides an overview of the malaria vaccines' evolution from the past towards the mRNA vaccine era and highlights their advantages in overcoming the limitations of previous malaria vaccine candidates.
Collapse
Affiliation(s)
- Maria E. Tsoumani
- Department of Biomedical Sciences, University of West Attica, 12243 Aigaleo, Greece; (M.E.T.); (C.V.)
| | - Chrysa Voyiatzaki
- Department of Biomedical Sciences, University of West Attica, 12243 Aigaleo, Greece; (M.E.T.); (C.V.)
| | - Antonia Efstathiou
- Department of Biomedical Sciences, University of West Attica, 12243 Aigaleo, Greece; (M.E.T.); (C.V.)
- Immunology of Infection Group, Department of Microbiology, Hellenic Pasteur Institute, 11521 Athens, Greece
| |
Collapse
|
13
|
Ezema CA, Okagu IU, Ezeorba TPC. Escaping the enemy's bullets: an update on how malaria parasites evade host immune response. Parasitol Res 2023:10.1007/s00436-023-07868-6. [PMID: 37219610 DOI: 10.1007/s00436-023-07868-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2023] [Accepted: 05/08/2023] [Indexed: 05/24/2023]
Abstract
Malaria continues to cause untold hardship to inhabitants of malaria-endemic regions, causing significant morbidity and mortality that severely impact global health and the economy. Considering the complex life cycle of malaria parasites (MPs) and malaria biology, continued research efforts are ongoing to improve our understanding of the pathogenesis of the diseases. Female Anopheles mosquito injects MPs into its hosts during a blood meal, and MPs invade the host skin and the hepatocytes without causing any serious symptoms. Symptomatic infections occur only during the erythrocytic stage. In most cases, the host's innate immunity (for malaria-naïve individuals) and adaptive immunity (for pre-exposed individuals) mount severe attacks and destroy most MPs. It is increasingly understood that MPs have developed several mechanisms to escape from the host's immune destruction. This review presents recent knowledge on how the host's immune system destroys invading MPs as well as MPs survival or host immune evasion mechanisms. On the invasion of host cells, MPs release molecules that bind to cell surface receptors to reprogram the host in a way to lose the capacity to destroy them. MPs also hide from the host immune cells by inducing the clustering of both infected and uninfected erythrocytes (rosettes), as well as inducing endothelial activation. We hope this review will inspire more research to provide a complete understanding of malaria biology and promote interventions to eradicate the notorious disease.
Collapse
Affiliation(s)
- Chinonso Anthony Ezema
- Department of Microbiology, Faculty of Biological Sciences, University of Nigeria, Enugu State, 410001, Nigeria
- Division of Soft Matter, Hokkaido University, Sapporo, 060-0810, Japan
| | - Innocent Uzochukwu Okagu
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Enugu State, 410001, Nigeria
| | - Timothy Prince Chidike Ezeorba
- Department of Biochemistry, Faculty of Biological Sciences, University of Nigeria, Enugu State, 410001, Nigeria.
- Department of Genetics and Biotechnology, Faculty of Biological Sciences, University of Nigeria, Enugu State, 410001, Nigeria.
- Department of Molecular Biotechnology, School of Biosciences, University of Birmingham Edgbaston, Birmingham, B15 2TT, UK.
| |
Collapse
|
14
|
Bhattacharjee S, Ghosh D, Saha R, Sarkar R, Kumar S, Khokhar M, Pandey RK. Mechanism of Immune Evasion in Mosquito-Borne Diseases. Pathogens 2023; 12:635. [PMID: 37242305 PMCID: PMC10222277 DOI: 10.3390/pathogens12050635] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Revised: 04/20/2023] [Accepted: 04/21/2023] [Indexed: 05/28/2023] Open
Abstract
In recent decades, mosquito-borne illnesses have emerged as a major health burden in many tropical regions. These diseases, such as malaria, dengue fever, chikungunya, yellow fever, Zika virus infection, Rift Valley fever, Japanese encephalitis, and West Nile virus infection, are transmitted through the bite of infected mosquitoes. These pathogens have been shown to interfere with the host's immune system through adaptive and innate immune mechanisms, as well as the human circulatory system. Crucial immune checkpoints such as antigen presentation, T cell activation, differentiation, and proinflammatory response play a vital role in the host cell's response to pathogenic infection. Furthermore, these immune evasions have the potential to stimulate the human immune system, resulting in other associated non-communicable diseases. This review aims to advance our understanding of mosquito-borne diseases and the immune evasion mechanisms by associated pathogens. Moreover, it highlights the adverse outcomes of mosquito-borne disease.
Collapse
Affiliation(s)
| | - Debanjan Ghosh
- Department of Biotechnology, Pondicherry University, Puducherry 605014, India
| | - Rounak Saha
- Department of Biochemistry and Molecular Biology, Pondicherry University, Puducherry 605014, India
| | - Rima Sarkar
- DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Saurav Kumar
- DBT Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram 695014, India
| | - Manoj Khokhar
- Department of Biochemistry, AIIMS, Jodhpur 342005, India
| | - Rajan Kumar Pandey
- Department of Medical Biochemistry and Biophysics, Karolinska Institute, 171 77 Solna, Sweden
| |
Collapse
|
15
|
Deshmukh R. Exploring the potential of antimalarial nanocarriers as a novel therapeutic approach. J Mol Graph Model 2023; 122:108497. [PMID: 37149980 DOI: 10.1016/j.jmgm.2023.108497] [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: 12/29/2022] [Revised: 03/31/2023] [Accepted: 04/17/2023] [Indexed: 05/09/2023]
Abstract
Malaria is a life-threatening parasitic disease that affects millions of people worldwide, especially in developing countries. Despite advances in conventional therapies, drug resistance in malaria parasites has become a significant concern. Hence, there is a need for a new therapeutic approach. To combat the disease effectively means eliminating vectors and discovering potent treatments. The nanotechnology research efforts in nanomedicine show promise by exploring the potential use of nanomaterials that can surmount these limitations occurring with antimalarial drugs, which include multidrug resistance or lack of specificity when targeting parasites directly. Utilizing nanomaterials would possess unique advantages over conventional chemotherapy systems by increasing the efficacy levels while reducing side effects significantly by delivering medications precisely within the diseased area. It also provides cheap yet safe measures against Malaria infections worldwide-ultimately improving treatment efficiency holistically without reinventing new methods therapeutically. This review is an effort to provide an overview of the various stages of malaria parasites, pathogenesis, and conventional therapies, as well as the treatment gap existing with available formulations. It explores different types of nanocarriers, such as liposomes, ethosomal cataplasm, solid lipid nanoparticles, nanostructured lipid carriers, polymeric nanocarriers, and metallic nanoparticles, which are frequently employed to boost the efficiency of antimalarial drugs to overcome the challenges and develop effective and safe therapies. The study also highlights the improved pharmacokinetics, enhanced drug bioavailability, and reduced toxicity associated with nanocarriers, making them a promising therapeutic approach for treating malaria.
Collapse
Affiliation(s)
- Rohitas Deshmukh
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, 281406, India.
| |
Collapse
|
16
|
Shaikh MS, Islam F, Gargote PP, Gaikwad RR, Dhupe KC, Khan SL, Siddiqui FA, Tapadiya GG, Ali SS, Dey A, Emran TB. Potential Epha2 Receptor Blockers Involved in Cerebral Malaria from Taraxacum officinale, Tinospora cordifolia, Rosmarinus officinalis and Ocimum basilicum: A Computational Approach. Pathogens 2022; 11:1296. [PMID: 36365047 PMCID: PMC9693644 DOI: 10.3390/pathogens11111296] [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] [Received: 09/22/2022] [Revised: 10/27/2022] [Accepted: 11/02/2022] [Indexed: 09/01/2023] Open
Abstract
Cerebral malaria (CM) is a severe manifestation of parasite infection caused by Plasmodium species. In 2018, there were approximately 228 million malaria cases worldwide, resulting in about 405,000 deaths. Survivors of CM may live with lifelong post-CM consequences apart from an increased risk of childhood neurodisability. EphA2 receptors have been linked to several neurological disorders and have a vital role in the CM-associated breakdown of the blood-brain barrier. Molecular docking (MD) studies of phytochemicals from Taraxacum officinale, Tinospora cordifolia, Rosmarinus officinalis, Ocimum basilicum, and the native ligand ephrin-A were conducted to identify the potential blockers of the EphA2 receptor. The software program Autodock Vina 1.1.2 in PyRx-Virtual Screening Tool and BIOVIA Discovery Studio visualizer was used for this MD study. The present work showed that blocking the EphA2 receptor by these phytochemicals prevents endothelial cell apoptosis by averting ephrin-A ligand-expressing CD8+ T cell bioadhesion. These phytochemicals showed excellent docking scores and binding affinity, demonstrating hydrogen bond, electrostatic, Pi-sigma, and pi alkyl hydrophobic binding interactions when compared with native ligands at the EphA2 receptor. The comparative MD study using two PDB IDs showed that isocolumbin, carnosol, luteolin, and taraxasterol have better binding affinities (viz. -9.3, -9.0, -9.5, and -9.2 kcal/mol, respectively). Ocimum basilicum phytochemicals showed a lower docking score but more binding interactions than native ligands at the EphA2 receptor for both PDB IDs. This suggests that these phytochemicals may serve as potential drug candidates in the management of CM. We consider that the present MD study provides leads in drug development by targeting the EphA2 receptor in managing CM. The approach is innovative because a role for EphA2 receptors in CM has never been highlighted.
Collapse
Affiliation(s)
- Mohd Sayeed Shaikh
- Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad 431136, India
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka 1341, Bangladesh
| | - Parag P. Gargote
- Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad 431136, India
| | - Rutuja R. Gaikwad
- Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad 431136, India
| | - Kalpana C. Dhupe
- Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad 431136, India
| | - Sharuk L. Khan
- Department of Pharmaceutical Chemistry, N.B.S. Institute of Pharmacy, Ausa 413520, India
| | - Falak A. Siddiqui
- Department of Pharmaceutical Chemistry, N.B.S. Institute of Pharmacy, Ausa 413520, India
| | - Ganesh G. Tapadiya
- Shreeyash Institute of Pharmaceutical Education and Research, Aurangabad 431136, India
| | | | - Abhijit Dey
- Department of Life Sciences, Presidency University, 86/1 College Street, Kolkata 700073, India
| | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong 4381, Bangladesh
| |
Collapse
|