1
|
Vinh DC. Human immunity to fungal infections. J Exp Med 2025; 222:e20241215. [PMID: 40232283 PMCID: PMC11998751 DOI: 10.1084/jem.20241215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/07/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Fungi increasingly threaten health globally. Mycoses range from life-threatening, often iatrogenic conditions, to enigmatic syndromes occurring without apparent immunosuppression. Despite some recent advances in antifungal drug development, complementary therapeutic strategies are essential for addressing these opportunistic pathogens. One promising avenue is leveraging host immunity to combat fungal infections; this necessitates deeper understanding of the molecular immunology of human fungal susceptibility to differentiate beneficial versus harmful immunopathological responses. Investigating human models of fungal diseases in natural settings, particularly through genetic immunodeficiencies and ethnographic-specific genetic vulnerabilities, reveals crucial immune pathways essential for fighting various yeasts and molds. This review highlights the diversity in intrinsic fungal susceptibility across individuals and populations, through genetic- and autoantibody-mediated processes, complementing previous principles learned from animal studies and iatrogenic contexts. Improved understanding of human immunity to fungal diseases will facilitate the development of host-directed immunotherapies and targeted public health interventions, paving the way for precision medicine in fungal disease management.
Collapse
Affiliation(s)
- Donald C. Vinh
- Department of Medicine (Division of Infectious Diseases), McGill University Health Center, Montreal, Canada
- Department of OptiLab (Division of Medical Microbiology, Division of Molecular Genetics-Immunology), McGill University Health Center, Montreal, Canada
- Department of Human Genetics, McGill University, Montreal, Canada
- Center of Reference for Genetic Research in Infection and Immunity, McGill University Health Center Research Institute, Montreal, Canada
| |
Collapse
|
2
|
Parry ERS, Pevsner R, Poulton BC, Purusothaman DK, Adam AI, Issiaka S, Ant TH, Rainey SM, Bilgo E, Diabaté A, Sinkins SP. Imaging the lifecycle of Microsporidia sp. MB in Anopheles coluzzii from western Burkina Faso reveals octosporogony. mSphere 2025:e0085124. [PMID: 40401914 DOI: 10.1128/msphere.00851-24] [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: 10/08/2024] [Accepted: 04/24/2025] [Indexed: 05/23/2025] Open
Abstract
A newly discovered microsporidian, Microsporidia sp. MB (MB), was recently found to block Plasmodium falciparum transmission in Anopheles arabiensis mosquitoes from Kenya. Here, we describe the lifecycle of the first laboratory colony of Anopheles coluzzii with MB, originating from western Burkina Faso. The lifecycle of MB was explored using fluorescent in situ hybridization and confocal microscopy, facilitated by the development of optimized protocols to produce histological sections of whole adult, larval, and embryo tissues. As in An. arabiensis, transmission appears to be predominantly vertical, with MB highly localized to the ovaries across multiple lifecycle stages. MB was sparsely distributed within the majority of developing oocytes in the gravid female. After oviposition, in the majority of embryos, MB relocated to the developing gonad at the onset of tissue differentiation, suggesting a highly specialized adaptation to host tissues. Sporogony was identified for the first time in a proportion of developing oocytes and in embryos post-oviposition. Microsporidian spore characteristics were subsequently confirmed with electron microscopy. Identification of MB sporogony in eggs suggests there are alternative horizontal routes of transmission which could play an important role in developing MB as a malaria control strategy. IMPORTANCE Malaria in West Africa, caused by Plasmodium falciparum infection and spread by anopheline mosquitoes, is responsible for hundreds of thousands of deaths annually and resulted in over 120 million cases in 2022 . The transmission-blocking effect of Microsporidia sp. MB (MB) suggests its potential as an agent for combating the spread of malaria. Understanding the routes of transmission and their effect on MB in mosquito populations is crucial for its development as a control tool. The identification of MB spores reveals the potential for another avenue of transmission beyond the vertical transmission from female to offspring. Spores could also have the potential for alternative MB dissemination methods, alongside or instead of adult mosquito releases.
Collapse
Affiliation(s)
- Ewan R S Parry
- School of Infection and Immunity, University of Glasgow and MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Roland Pevsner
- School of Infection and Immunity, University of Glasgow and MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Beth C Poulton
- School of Infection and Immunity, University of Glasgow and MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Deepak-Kumar Purusothaman
- School of Infection and Immunity, University of Glasgow and MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Abdelhakeem I Adam
- School of Infection and Immunity, University of Glasgow and MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Sare Issiaka
- Institut de Recherche en Sciences de la Santé (IRSS), BoboDioulasso, Burkina Faso
| | - Thomas H Ant
- School of Infection and Immunity, University of Glasgow and MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Stephanie M Rainey
- School of Infection and Immunity, University of Glasgow and MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| | - Etienne Bilgo
- Institut de Recherche en Sciences de la Santé (IRSS), BoboDioulasso, Burkina Faso
| | - Abdoulaye Diabaté
- Institut de Recherche en Sciences de la Santé (IRSS), BoboDioulasso, Burkina Faso
| | - Steven P Sinkins
- School of Infection and Immunity, University of Glasgow and MRC-University of Glasgow Centre for Virus Research, Glasgow, United Kingdom
| |
Collapse
|
3
|
Yanik S, Venkatesh V, Gordy JT, Alameh MG, Meza J, Li Y, Glass E, Flores-Garcia Y, Tam Y, Chaiyawong N, Sarkar D, Weissman D, Markham R, Srinivasan P. iDC-targeting PfCSP mRNA vaccine confers superior protection against Plasmodium compared to conventional mRNA. NPJ Vaccines 2025; 10:34. [PMID: 39971939 PMCID: PMC11840135 DOI: 10.1038/s41541-025-01089-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
Malaria resurgence in 2022 saw 249 million clinical cases and 608,000 deaths, mostly in children under five. The WHO-approved circumsporozoite protein (CSP)-targeting vaccines, RTS,S and R21, remain limited in availability. Strong humoral responses are crucial for sporozoite neutralization before hepatocyte infection, yet first-generation vaccines provide suboptimal protection, necessitating improved strategies. With the success of mRNA-LNP vaccines against COVID-19, there is interest in leveraging this approach to malaria. Here, we developed a novel chemokine fusion mRNA vaccine targeting immature dendritic cells (iDC) to enhance immunity against P. falciparum CSP (PfCSP). Mice immunized with MIP3α-CSP mRNA-LNP exhibited stronger CD4 + T cell responses and higher anti-NANP6 antibody titers than conventional CSP mRNA-LNP. Importantly, upon P. berghei PfCSP transgenic sporozoite challenge, MIP3α-CSP mRNA provided significantly greater protection from liver infection, strongly associated with multifunctional CD4 + T cells and anti-NANP6 titers. This study underscores iDC targeting as a promising strategy to enhance malaria vaccine efficacy.
Collapse
Affiliation(s)
- Sean Yanik
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA
| | - Varsha Venkatesh
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA
| | - James T Gordy
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | | | - Jacob Meza
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Yangchen Li
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Elizabeth Glass
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA
| | - Ying Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | - Nattawat Chaiyawong
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA
| | - Deepti Sarkar
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA
| | - Drew Weissman
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Markham
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA
| | - Prakash Srinivasan
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, USA.
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, USA.
| |
Collapse
|
4
|
Cardoso-Jaime V, Dimopoulos G. Anopheles gambiae phagocytic hemocytes promote Plasmodium falciparum infection by regulating midgut epithelial integrity. Nat Commun 2025; 16:1465. [PMID: 39920122 PMCID: PMC11805967 DOI: 10.1038/s41467-025-56313-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 01/15/2025] [Indexed: 02/09/2025] Open
Abstract
For successful transmission, the malaria parasite must traverse tissue epithelia and survive attack from the insect's innate immune system. Hemocytes play a multitude of roles in mosquitoes, including defense against invading pathogens. Here, we show that hemocytes of the major malaria vector Anopheles gambiae promote Plasmodium falciparum infection by maintaining midgut epithelial integrity by controlling cell proliferation upon blood feeding. The mosquito's hemocytes also control the midgut microbiota and immune gene expression. Our study unveils novel hemocyte functions that are exploited by the human malaria parasite to evade the mosquito's immune system.
Collapse
Affiliation(s)
- Victor Cardoso-Jaime
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - George Dimopoulos
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Malaria Research Institute, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
5
|
Goeury T, Faye N, Gerbault P, Černý V, Crubézy E, Chiaroni J, Brouk H, Brunet L, Galan M, de Groot NG, Nunes JM, Sanchez‐Mazas A. Evidence for Pathogen-Driven Selection Acting on HLA-DPB1 in Response to Plasmodium falciparum Malaria in West Africa. Ecol Evol 2025; 15:e70933. [PMID: 40008064 PMCID: PMC11850448 DOI: 10.1002/ece3.70933] [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: 01/10/2025] [Accepted: 01/16/2025] [Indexed: 02/27/2025] Open
Abstract
African populations remain underrepresented in studies of human genetic diversity, despite a growing interest in understanding how they have adapted to the diverse environments they live in. In particular, understanding the genetic basis of immune adaptation to pathogens is of paramount importance in a continent such as Africa, where the burden of infectious diseases is a major public health challenge. In this study, we investigated the molecular variation of four Human Leukocyte Antigens (HLA) class II genes (DRB1, DQA1, DQB1 and DPB1), directly involved in the immune response to parasitic infections, in more than 1000 individuals from 23 populations across North, East, Central and West Africa. By analyzing the HLA molecular diversity of these populations in relation to various geographical, cultural and environmental factors, we identified divergent genetic profiles for several (semi-)nomadic populations of the Sahel belt as a signature of their unique demography. In addition, we observed significant genetic structuring supporting both substantial geographic and linguistic differentiations within West Africa. Furthermore, neutrality tests suggest balancing selection has been shaping the diversity of these four HLA class II genes, which is consistent with molecular comparisons between HLA genes and their orthologs in chimpanzees (Patr). However, the most striking observation comes from linear modeling, demonstrating that the prevalence of Plasmodium falciparum, the primary pathogen of malaria in Africa, significantly explains a large proportion of the nucleotide diversity observed at the DPB1 gene. DPB1*01:01, a highly frequent allele in Burkinabé populations, is identified as a potential protective allele against malaria, suggesting that strong pathogen-driven positive selection at this gene has shaped HLA variation in Africa. Additionally, two low-frequency DRB1 alleles, DRB1*08:06 and DRB1*11:02, also show significant associations with P. falciparum prevalence, supporting resistance to malaria is determined by multigenic and/or multiallelic combinations rather than single allele effects.
Collapse
Affiliation(s)
- Thomas Goeury
- Laboratory of Anthropology, Genetics and Peopling History (AGP), Department of Genetics and EvolutionUniversity of GenevaGenevaSwitzerland
| | - Ndeye Faye
- Laboratory of Anthropology, Genetics and Peopling History (AGP), Department of Genetics and EvolutionUniversity of GenevaGenevaSwitzerland
| | - Pascale Gerbault
- Laboratory of Anthropology, Genetics and Peopling History (AGP), Department of Genetics and EvolutionUniversity of GenevaGenevaSwitzerland
| | - Viktor Černý
- Institute of Archaeology of the Academy of Sciences of the Czech RepublicArchaeogenetics LaboratoryCzech Academy of SciencesPragueCzech Republic
| | - Eric Crubézy
- Institut Universitaire de FranceUMR5288 CNRSUniversity of Toulouse III Paul SabatierToulouseFrance
| | | | - Hacene Brouk
- Service of Hemobiology and Blood TransfusionUniversity Hospital Center Ibn Rochd of AnnabaFaculty of MedicineBadji Mokhtar University of AnnabaAnnabaAlgeria
| | - Lydie Brunet
- Laboratory of Anthropology, Genetics and Peopling History (AGP), Department of Genetics and EvolutionUniversity of GenevaGenevaSwitzerland
- Transplantation Immunology Unit and National Reference Laboratory for Histocompatibility (UIT/LNRH)Geneva University HospitalGenevaSwitzerland
| | - Maxime Galan
- CBGP UMR 1062INRAEIRDCIRADMontpellier SupAgroUniversity of MontpellierMontpellierFrance
| | - Natasja G. de Groot
- Department of Comparative Genetics and RefinementBiomedical Primate Research Centre (BPRC)Rijswijkthe Netherlands
| | - José Manuel Nunes
- Laboratory of Anthropology, Genetics and Peopling History (AGP), Department of Genetics and EvolutionUniversity of GenevaGenevaSwitzerland
- Institute of Genetics and Genomics in Geneva (IGE3)University of GenevaGenevaSwitzerland
| | - Alicia Sanchez‐Mazas
- Laboratory of Anthropology, Genetics and Peopling History (AGP), Department of Genetics and EvolutionUniversity of GenevaGenevaSwitzerland
- Institute of Genetics and Genomics in Geneva (IGE3)University of GenevaGenevaSwitzerland
| |
Collapse
|
6
|
Tiwari R, Kumar A, Singh VK, Rajneesh, Chauhan SB, Sundar S, Nylén S, Engwerda C, Kumar R. The development and maintenance of immunity against visceral leishmaniasis. Front Immunol 2024; 15:1486407. [PMID: 39781380 PMCID: PMC11707418 DOI: 10.3389/fimmu.2024.1486407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Accepted: 11/20/2024] [Indexed: 01/12/2025] Open
Abstract
Understanding the development and maintenance of immunological memory is important for efforts to eliminate parasitic diseases like leishmaniasis. Leishmaniasis encompasses a range of pathologies, resulting from infection with protozoan parasites belonging to the subgenera Leishmania and Viannia of the genus Leishmania. A striking feature of these infections is that natural or drug-mediated cure of infection generally confers life-long protection against disease. The generation of protective T cell responses are necessary to control Leishmania infections. CD4+ T helper (Th) cells orchestrate immune responses in leishmaniasis and IFNγ+ Tbet+ CD4+ T (Th1) cells are required for the activation of phagocytes to kill captured or resident parasites, while other Th cell subset, including FoxP3+ natural regulatory T cells and Th2 cells can promote disease progression by suppressing the activities of Th1 cells. Upon resolution of a primary Leishmania infection, different subsets of CD4+ T cells, including tissue-resident memory T cells, effector memory T cells, central memory T cells, and short-lived effector T cells, help to confer resistance against reinfection. To maintain long-term protective Leishmania-specific CD4+ T cells responses, it is believed that persistent parasites or re-exposure to parasites at regular intervals is required (concomitant immunity). Despite the advances in our understanding about the immune responses during leishmaniasis, the generation of long-lasting protective immunity via vaccination has yet to be achieved. In this review, we summarize our current understanding about the formation and maintenance of immunological memory and control of leishmaniasis at the individual and population level. We will focus on Indian visceral leishmaniasis and discuss T cell responses that contribute to susceptibility to leishmaniasis, parasite persistence in populations and the environment, as well as describing advances in the development of leishmaniasis vaccines aimed at inducing protective CD4+ T cell responses.
Collapse
Affiliation(s)
- Rahul Tiwari
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Awnish Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Vishal Kumar Singh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Rajneesh
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shashi Bhushan Chauhan
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Shyam Sundar
- Department of Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Susanne Nylén
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Engwerda
- Infection and Inflammation Division, QIMR Berghofer Medical Research Institute, Brisbane, QLD, Australia
| | - Rajiv Kumar
- Centre of Experimental Medicine and Surgery, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| |
Collapse
|
7
|
Thellier M, Gemegah AAJ, Tantaoui I. Global Fight against Malaria: Goals and Achievements 1900-2022. J Clin Med 2024; 13:5680. [PMID: 39407740 PMCID: PMC11477079 DOI: 10.3390/jcm13195680] [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: 08/29/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
This article examines the historical and ongoing efforts to fight malaria, a parasitic disease caused by Plasmodium species and transmitted by Anopheles mosquitoes. Despite over a century of control efforts, malaria remains a major global health issue. In 2022, there were an estimated 249 million cases across 85 countries, leading to approximately 600,000 deaths. In the recently published Global Technical Strategy for Malaria 2016-2030, the World Health Organization (WHO) has prioritized malaria eradication. The main goals are to reduce malaria incidence and mortality by 90% by 2030 compared to 2015 levels. However, as of 2022, progress has been limited, with only a 2% reduction in incidence and a 6% reduction in mortality. This review traces the historical context of malaria, highlighting its ancient origins and the pivotal scientific discoveries in the late 19th century that paved the way for modern control measures. The Global Malaria Eradication Programme launched by the WHO in 1955 initially showed promise, largely due to the insecticide DDT, but ultimately failed to achieve its goals mainly due to logistical problems, vector resistance to DDT, and inadequate funding. Despite significant advances in the early 21st century, including the Roll Back Malaria initiative and increased international funding, malaria eradication remains a distant goal. Persistent challenges, such as weak healthcare systems, parasite and vector resistance to drugs and insecticides, and inadequate funding, continue to hamper global efforts. Therefore, this article underscores the need for a deeper understanding of malaria's history and recent evolution to inform future strategies for eradication.
Collapse
Affiliation(s)
- Marc Thellier
- AP-HP, Centre National de Référence du Paludisme, Hôpital Pitié–Salpêtrière, 75013 Paris, France; (A.A.J.G.); (I.T.)
- Sorbonne Université, Institut National de la Santé et de la Recherche Médicale (INSERM), IPLESP Institut Pierre–Louis d’Épidémiologie et de Santé Publique, 75013 Paris, France
| | | | - Ilhame Tantaoui
- AP-HP, Centre National de Référence du Paludisme, Hôpital Pitié–Salpêtrière, 75013 Paris, France; (A.A.J.G.); (I.T.)
| |
Collapse
|
8
|
Bi B, Wu L, Liu Y, Zhou XN, Shen T, Cao L, White M, Yang GJ. Intervention portfolios analysis of Plasmodium vivax control in central China. Malar J 2024; 23:242. [PMID: 39138510 PMCID: PMC11321059 DOI: 10.1186/s12936-024-05063-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND The effects of a diverse spectrum of malaria interventions were evaluated through a deterministic Plasmodium vivax transmission model. This approach aimed to provide theoretical evidence of the performance of these interventions once implemented for achieving malaria elimination. METHODS An integrated intervention portfolio, including mass drug administration, insecticide treatment, and untreated bed nets, was analyzed through modeling. Additionally, data-driven calibration was implemented to infer coverages that effectively reproduced historical malaria patterns in China from 1971 to 1983. RESULTS MDA utilizing primaquine emerged as the most effective single intervention, achieving a 70% reduction in malaria incidence when implemented at full coverage. Furthermore, a strategic combination of MDA with primaquine, chloroquine, untreated bed nets, and seasonal insecticide treatments effectively eradicated malaria, attaining elimination at a coverage level of 70%. It was conclusively demonstrated that an integrated approach combining MDA and vector control measures is essential for the successful elimination of malaria. CONCLUSION High coverage of mass drug administration with primaquine and chloroquine before transmission was the key driver of the malaria decline in China from 1971 to 1983. The best-fit intervention coverage combinations derived from calibration are provided as a reference for malaria control in other countries.
Collapse
Affiliation(s)
- Bo Bi
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, School of Public Health, Hainan Medical University, Haikou, People's Republic of China
| | - Logan Wu
- Department of Population Health and Immunity, Walter and Eliza Hall Institute, Melbourne, Australia
- Department of Medical Biology, University of Melbourne, Melbourne, Australia
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ying Liu
- Henan Center for Disease Control and Prevention, Zhengzhou, Henan, People's Republic of China
| | - Xiao-Nong Zhou
- National Institute of Parasitic Diseases at Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tianren Shen
- Infection Medicine, Deanery of Biomedical Sciences, College of Medicine and Veterinary Medicine, The University of Edinburgh, Edinburgh, Scotland, UK
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, China
| | - Li Cao
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, School of Public Health, Hainan Medical University, Haikou, People's Republic of China
| | - Michael White
- Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
| | - Guo-Jing Yang
- NHC Key Laboratory of Tropical Disease Control, School of Tropical Medicine, School of Public Health, Hainan Medical University, Haikou, People's Republic of China.
| |
Collapse
|
9
|
Pande V, Bahal M, Dua J, Gupta A. Ronald Ross: Pioneer of Malaria Research and Nobel Laureate. Cureus 2024; 16:e65993. [PMID: 39221334 PMCID: PMC11366399 DOI: 10.7759/cureus.65993] [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: 06/25/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Sir Ronald Ross, a British medical doctor and researcher, is renowned for his pioneering work in malaria research. His discovery of the malaria parasite's lifecycle within mosquitoes revolutionized the understanding and control of malaria, transitioning the field from the miasma theory to vector-based strategies. This literature review aims to explore the comprehensive contributions of Ronald Ross to malaria research and their enduring impact on public health.
Collapse
Affiliation(s)
- Vineeta Pande
- Pediatrics, Dr. D.Y. Patil Medical College, Hospital and Research Center, Pune, IND
| | - Mridu Bahal
- Pediatrics, Dr. D.Y. Patil Medical College, Hospital and Research Center, Pune, IND
| | - Jasleen Dua
- Pediatrics, Dr. D.Y. Patil Medical College, Hospital and Research Center, Pune, IND
| | - Aryan Gupta
- Pediatric Neurology, Dr. D.Y. Patil Medical College, Hospital and Research Center, Pune, IND
| |
Collapse
|
10
|
Gozalo AS, Robinson CK, Holdridge J, Franco Mahecha OL, Elkins WR. Overview of Plasmodium spp. and Animal Models in Malaria Research. Comp Med 2024; 74:205-230. [PMID: 38902006 PMCID: PMC11373680 DOI: 10.30802/aalas-cm-24-000019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/08/2024] [Accepted: 05/13/2024] [Indexed: 06/22/2024]
Abstract
Malaria is a parasitic disease caused by protozoan species of the genus Plasmodium and transmitted by female mosquitos of the genus Anopheles and other Culicidae. Most of the parasites of the genus Plasmodium are highly species specific with more than 200 species described affecting different species of mammals, birds, and reptiles. Plasmodium species strictly affecting humans are P. falciparum, P. vivax, P. ovale, and P. malariae. More recently, P. knowlesi and other nonhuman primate plasmodia were found to naturally infect humans. Currently, malaria occurs mostly in poor tropical and subtropical areas of the world, and in many of these countries it is the leading cause of illness and death. For more than 100 y, animal models, have played a major role in our understanding of malaria biology. Avian Plasmodium species were the first to be used as models to study human malaria. Malaria parasite biology and immunity were first studied using mainly P. gallinaceum and P. relictum. Rodent malarias, particularly P. berghei and P. yoelii, have been used extensively as models to study malaria in mammals. Several species of Plasmodium from nonhuman primates have been used as surrogate models to study human malaria immunology, pathogenesis, candidate vaccines, and treatments. Plasmodium cynomolgi, P. simiovale, and P. fieldi are important models for studying malaria produced by P. vivax and P. ovale, while P. coatneyi is used as a model for study- ing severe malaria. Other nonhuman primate malarias used in research are P. fragile, P. inui, P. knowlesi, P. simium, and P. brasilianum. Very few nonhuman primate species can develop an infection with human malarias. Macaques in general are resistant to infection with P. falciparum, P. vivax, P. malariae, and P. ovale. Only apes and a few species of New World monkeys can support infection with human malarias. Herein we review the most common, and some less common, avian, reptile, and mammal plasmodia species used as models to study human malaria.
Collapse
Affiliation(s)
- Alfonso S Gozalo
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Christen K Robinson
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Julie Holdridge
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Olga L Franco Mahecha
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - William R Elkins
- Comparative Medicine Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
11
|
Yanik S, Venkatesh V, Gordy JT, Gabriel-Alameh M, Meza J, Li Y, Glass E, Flores-Garcia Y, Tam Y, Chaiyawong N, Sarkar D, Weissman D, Markham R, Srinivasan P. Immature dendritic cell-targeting mRNA vaccine expressing PfCSP enhances protective immune responses against Plasmodium liver infection. RESEARCH SQUARE 2024:rs.3.rs-4656309. [PMID: 39041038 PMCID: PMC11261966 DOI: 10.21203/rs.3.rs-4656309/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2024]
Abstract
Resurgence in malaria has been noted in 2022 with 249 million clinical cases resulting in 608,000 deaths, mostly in children under five. Two vaccines, RTS, S, and more recently R21, targeting the circumsporozoite protein (CSP) are recommended by the WHO but are not yet widely available. Strong humoral responses to neutralize sporozoites before they can infect the hepatocytes are important for vaccine-mediated protection. Suboptimal protection conferred by these first-generation vaccines highlight the need for approaches to improve vaccine-induced immune responses. With the recent success of mRNA-LNP vaccines against COVID-19, there is growing interest in leveraging this approach to enhance malaria vaccines. Here, we present the development of a novel chemokine fusion mRNA vaccine aimed at boosting immune responses to PfCSP by targeting the immunogen to immature dendritic cells (iDC). Vaccination of mice with mRNA encoding full-length CSP fused to macrophage inflammatory protein 3 alpha (MIP3α) encapsulated within lipid nanoparticles (LNP) elicited robust CD4+ T cell responses and enhanced antibody titers against NANP repeat epitopes compared to a conventional CSP mRNA-LNP vaccine. Importantly, the CSP-MIP3α fusion vaccine provided significantly greater protection against liver infection upon challenge with P. berghei PfCSP transgenic sporozoites. This enhanced protection was associated with multifunctional CD4+ T cells levels and anti-NANP repeat titers. This study highlights the potential to augment immune responses to PfCSP through iDC targeting and bolster protection against malaria liver infection.
Collapse
Affiliation(s)
- Sean Yanik
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Varsha Venkatesh
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - James T Gordy
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
| | | | - Jacob Meza
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
| | - Yangchen Li
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
| | - Elizabeth Glass
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
| | - Yevel Flores-Garcia
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Ying Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | - Nattawat Chaiyawong
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Deepti Sarkar
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| | - Drew Weissman
- Penn Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA 19104
| | - Richard Markham
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
| | - Prakash Srinivasan
- Department of Molecular Microbiology and Immunology, Johns Hopkins School of Public Health, Baltimore, MD, 21205, USA
- The Johns Hopkins Malaria Research Institute, Baltimore, MD, 21205, USA
| |
Collapse
|
12
|
Habibzadeh F. The effect on the equilibrium sickle cell allele frequency of the probable protection conferred by malaria and sickle cell gene against other infectious diseases. Sci Rep 2024; 14:15399. [PMID: 38965406 PMCID: PMC11224252 DOI: 10.1038/s41598-024-66289-2] [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: 03/20/2024] [Accepted: 07/01/2024] [Indexed: 07/06/2024] Open
Abstract
If a mutated gene with heterozygous advantage against malaria, e.g., hemoglobin S (HbS) gene, is introduced in a small tribe, the gene (allele) frequency (fgene) increases until it reaches a steady state value (feq) where the total mortality from malaria and sickle cell disease is a minimum. This is a classic example of balanced-polymorphism named malaria hypothesis. In a previous in silico study, assuming realistic initial conditions, it has been shown that the feq is around 14%, far less than the fgene observed in certain parts of Africa, 24%. It seems that the malaria hypothesis, per se, could not explain such a high fgene, unless it is assumed that malaria and HbS gene can provide protection against other diseases. Using Monte-Carlo simulation, the current study was conducted to examine the effect on feq of five scenarios was examined. The studied scenarios consisted of different combinations of mortality of other diseases and the possible amounts of protections conferred by malaria and HbS gene against the diseases. Taking into account other diseases causing mortality in the population makes the fgene rate of change steeper over generations. feq is an increasing function of the amount of protection conferred by HbS gene against other diseases. The effect of protection provided by malaria against other diseases on feq, is however, variable-depending on the amount of protection conferred by HbS gene against other diseases, it may increase or decrease feq. If malaria and HbS gene provide protections of 1.5-fold and threefold against other diseases, respectively, the feq is around 24%, the amount reported in certain tribes of Africa. Under certain scenarios, the feq attained is even higher.
Collapse
|
13
|
Hernández-Zambrano LJ, Alfonso-González H, Buitrago SP, Castro-Cavadía CJ, Garzón-Ospina D. Exploring the genetic diversity pattern of PvEBP/DBP2: A promising candidate for an effective Plasmodium vivax vaccine. Acta Trop 2024; 255:107231. [PMID: 38685340 DOI: 10.1016/j.actatropica.2024.107231] [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: 02/22/2024] [Revised: 04/25/2024] [Accepted: 04/26/2024] [Indexed: 05/02/2024]
Abstract
Malaria remains a public health challenge. Since many control strategies have proven ineffective in eradicating this disease, new strategies are required, among which the design of a multivalent vaccine stands out. However, the effectiveness of this strategy has been hindered, among other reasons, by the genetic diversity observed in parasite antigens. In Plasmodium vivax, the Erythrocyte Binding Protein (PvEBP, also known as DBP2) is an alternate ligand to Duffy Binding Protein (DBP); given its structural resemblance to DBP, EBP/DBP2 is proposed as a promising antigen for inclusion in vaccine design. However, the extent of genetic diversity within the locus encoding this protein has not been comprehensively assessed. Thus, this study aimed to characterize the genetic diversity of the locus encoding the P. vivax EBP/DBP2 protein and to determine the evolutionary mechanisms modulating this diversity. Several intrapopulation genetic variation parameters were estimated using 36 gene sequences of PvEBP/DBP2 from Colombian P. vivax clinical isolates and 186 sequences available in databases. The study then evaluated the worldwide genetic structure and the evolutionary forces that may influence the observed patterns of genetic variation. It was found that the PvEBP/DBP2 gene exhibits one of the lowest levels of genetic diversity compared to other vaccine-candidate antigens. Four major haplotypes were shared worldwide. Analysis of the protein's 3D structure and epitope prediction identified five regions with potential antigenic properties. The results suggest that the PvEBP/DBP2 protein possesses ideal characteristics to be considered when designing a multivalent effective antimalarial vaccine against P. vivax.
Collapse
Affiliation(s)
- Laura J Hernández-Zambrano
- Grupo de Estudios en Genética y Biología Molecular (GEBIMOL), School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia - UPTC, Tunja, Boyacá, Colombia; Population Genetics And Molecular Evolution (PGAME), Fundación Scient, Tunja, Boyacá, Colombia
| | - Heliairis Alfonso-González
- Grupo de Estudios en Genética y Biología Molecular (GEBIMOL), School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia - UPTC, Tunja, Boyacá, Colombia; Population Genetics And Molecular Evolution (PGAME), Fundación Scient, Tunja, Boyacá, Colombia
| | - Sindy P Buitrago
- Grupo de Estudios en Genética y Biología Molecular (GEBIMOL), School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia - UPTC, Tunja, Boyacá, Colombia; Population Genetics And Molecular Evolution (PGAME), Fundación Scient, Tunja, Boyacá, Colombia
| | - Carlos J Castro-Cavadía
- Grupo de Investigaciones Microbiológicas y Biomédicas de Córdoba (GIMBIC), School of Health Sciences, Universidad de Córdoba, Montería, Córdoba, Colombia
| | - Diego Garzón-Ospina
- Grupo de Estudios en Genética y Biología Molecular (GEBIMOL), School of Biological Sciences, Universidad Pedagógica y Tecnológica de Colombia - UPTC, Tunja, Boyacá, Colombia; Population Genetics And Molecular Evolution (PGAME), Fundación Scient, Tunja, Boyacá, Colombia.
| |
Collapse
|
14
|
Michel M, Skourtanioti E, Pierini F, Guevara EK, Mötsch A, Kocher A, Barquera R, Bianco RA, Carlhoff S, Coppola Bove L, Freilich S, Giffin K, Hermes T, Hiß A, Knolle F, Nelson EA, Neumann GU, Papac L, Penske S, Rohrlach AB, Salem N, Semerau L, Villalba-Mouco V, Abadie I, Aldenderfer M, Beckett JF, Brown M, Campus FGR, Chenghwa T, Cruz Berrocal M, Damašek L, Duffett Carlson KS, Durand R, Ernée M, Fântăneanu C, Frenzel H, García Atiénzar G, Guillén S, Hsieh E, Karwowski M, Kelvin D, Kelvin N, Khokhlov A, Kinaston RL, Korolev A, Krettek KL, Küßner M, Lai L, Look C, Majander K, Mandl K, Mazzarello V, McCormick M, de Miguel Ibáñez P, Murphy R, Németh RE, Nordqvist K, Novotny F, Obenaus M, Olmo-Enciso L, Onkamo P, Orschiedt J, Patrushev V, Peltola S, Romero A, Rubino S, Sajantila A, Salazar-García DC, Serrano E, Shaydullaev S, Sias E, Šlaus M, Stančo L, Swanston T, Teschler-Nicola M, Valentin F, Van de Vijver K, Varney TL, Vigil-Escalera Guirado A, Waters CK, Weiss-Krejci E, Winter E, Lamnidis TC, Prüfer K, Nägele K, Spyrou M, Schiffels S, Stockhammer PW, Haak W, Posth C, Warinner C, Bos KI, Herbig A, Krause J. Ancient Plasmodium genomes shed light on the history of human malaria. Nature 2024; 631:125-133. [PMID: 38867050 PMCID: PMC11222158 DOI: 10.1038/s41586-024-07546-2] [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: 09/10/2023] [Accepted: 05/09/2024] [Indexed: 06/14/2024]
Abstract
Malaria-causing protozoa of the genus Plasmodium have exerted one of the strongest selective pressures on the human genome, and resistance alleles provide biomolecular footprints that outline the historical reach of these species1. Nevertheless, debate persists over when and how malaria parasites emerged as human pathogens and spread around the globe1,2. To address these questions, we generated high-coverage ancient mitochondrial and nuclear genome-wide data from P. falciparum, P. vivax and P. malariae from 16 countries spanning around 5,500 years of human history. We identified P. vivax and P. falciparum across geographically disparate regions of Eurasia from as early as the fourth and first millennia BCE, respectively; for P. vivax, this evidence pre-dates textual references by several millennia3. Genomic analysis supports distinct disease histories for P. falciparum and P. vivax in the Americas: similarities between now-eliminated European and peri-contact South American strains indicate that European colonizers were the source of American P. vivax, whereas the trans-Atlantic slave trade probably introduced P. falciparum into the Americas. Our data underscore the role of cross-cultural contacts in the dissemination of malaria, laying the biomolecular foundation for future palaeo-epidemiological research into the impact of Plasmodium parasites on human history. Finally, our unexpected discovery of P. falciparum in the high-altitude Himalayas provides a rare case study in which individual mobility can be inferred from infection status, adding to our knowledge of cross-cultural connectivity in the region nearly three millennia ago.
Collapse
MESH Headings
- Female
- Humans
- Male
- Altitude
- Americas/epidemiology
- Asia/epidemiology
- Biological Evolution
- Disease Resistance/genetics
- DNA, Ancient/analysis
- Europe/epidemiology
- Genome, Mitochondrial/genetics
- Genome, Protozoan/genetics
- History, Ancient
- Malaria/parasitology
- Malaria/history
- Malaria/transmission
- Malaria/epidemiology
- Malaria, Falciparum/epidemiology
- Malaria, Falciparum/history
- Malaria, Falciparum/parasitology
- Malaria, Falciparum/transmission
- Malaria, Vivax/epidemiology
- Malaria, Vivax/history
- Malaria, Vivax/parasitology
- Malaria, Vivax/transmission
- Plasmodium/genetics
- Plasmodium/classification
- Plasmodium falciparum/genetics
- Plasmodium falciparum/isolation & purification
- Plasmodium malariae/genetics
- Plasmodium malariae/isolation & purification
- Plasmodium vivax/genetics
- Plasmodium vivax/isolation & purification
Collapse
Affiliation(s)
- Megan Michel
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany.
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, USA.
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean, .
| | - Eirini Skourtanioti
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean
| | - Federica Pierini
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Evelyn K Guevara
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Department of Forensic Medicine, University of Helsinki, Helsinki, Finland
| | - Angela Mötsch
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean
| | - Arthur Kocher
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Transmission, Infection, Diversification and Evolution Group, Max Planck Institute of Geoanthropology, Jena, Germany
| | - Rodrigo Barquera
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Raffaela A Bianco
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean
| | - Selina Carlhoff
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Lorenza Coppola Bove
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean
- Department of Legal Medicine, Toxicology and Physical Anthropology, University of Granada, Granada, Spain
| | - Suzanne Freilich
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Department of Evolutionary Anthropology, University of Vienna, Vienna, Austria
| | - Karen Giffin
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Department of Environmental Sciences, University of Basel, Basel, Switzerland
| | - Taylor Hermes
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Department of Anthropology, University of Arkansas, Fayetteville, AR, USA
| | - Alina Hiß
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Florian Knolle
- Department of Medical Engineering and Biotechnology, University of Applied Sciences Jena, Jena, Germany
| | - Elizabeth A Nelson
- Microbial Palaeogenomics Unit, Department of Genomes and Genetics, Institut Pasteur, Paris, France
| | - Gunnar U Neumann
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean
| | - Luka Papac
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Sandra Penske
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Adam B Rohrlach
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- School of Computer and Mathematical Sciences, University of Adelaide, Adelaide, Australia
- Adelaide Data Science Centre, University of Adelaide, Adelaide, Australia
| | - Nada Salem
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean
| | - Lena Semerau
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Vanessa Villalba-Mouco
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean
- Instituto Universitario de Investigación en Ciencias Ambientales de Aragón, IUCA-Aragosaurus, Universitity of Zaragoza, Zaragoza, Spain
| | - Isabelle Abadie
- Inrap - Institut national de recherches archéologiques préventives, Paris, France
- Centre Michel de Boüard, Centre de recherches archéologiques et historiques anciennes et médiévales, Université de Caen Normandie, Caen, France
| | - Mark Aldenderfer
- Department of Anthropology and Heritage Studies, University of California, Merced, Merced, CA, USA
| | | | - Matthew Brown
- Sociology and Anthropology Department, Farmingdale State College, Farmingdale, NY, USA
| | - Franco G R Campus
- Department of History, Human Sciences, and Education, University of Sassari, Sassari, Italy
| | - Tsang Chenghwa
- Institute of Anthropology, National Tsing Hua University, Hsinchu, Taiwan
| | - María Cruz Berrocal
- Institute of Heritage Sciences (INCIPIT), Spanish National Research Council (CSIC), Santiago de Compostela, Spain
| | - Ladislav Damašek
- Institute of Classical Archaeology, Faculty of Arts, Charles University, Prague, Czech Republic
| | | | - Raphaël Durand
- Service d'archéologie préventive Bourges plus, Bourges, France
- UMR 5199 PACEA, Université de Bordeaux, Pessac Cedex, France
| | - Michal Ernée
- Department of Prehistoric Archaeology, Institute of Archaeology of the Czech Academy of Sciences, Prague, Czech Republic
| | | | - Hannah Frenzel
- Anatomy Institute, University of Leipzig, Leipzig, Germany
| | - Gabriel García Atiénzar
- Instituto Universitario de Investigación en Arqueología y Patrimonio Histórico, Universidad de Alicante, San Vicente del Raspeig (Alicante), Spain
| | | | - Ellen Hsieh
- Institute of Anthropology, National Tsing Hua University, Hsinchu, Taiwan
| | - Maciej Karwowski
- Institut für Urgeschichte und Historische Archäologie, University of Vienna, Vienna, Austria
| | - David Kelvin
- Department of Microbiology and Immunology, Dalhousie University, Halifax, Nova Scotia, Canada
| | - Nikki Kelvin
- Division of Ancient Pathogens, BioForge Canada Limited, Halifax, Nove Scotia, Canada
| | - Alexander Khokhlov
- Samara State University of Social Sciences and Education, Samara, Russia
| | - Rebecca L Kinaston
- BioArch South, Waitati, New Zealand
- Griffith Centre for Social and Cultural Studies, Griffith University, Nathan, Queensland, Australia
| | - Arkadii Korolev
- Samara State University of Social Sciences and Education, Samara, Russia
| | - Kim-Louise Krettek
- Senckenberg Centre for Human Evolution and Palaeoenvironment, University of Tübingen, Tübingen, Germany
| | - Mario Küßner
- Thuringian State Office for Heritage Management and Archaeology, Weimar, Germany
| | - Luca Lai
- Department of Anthropology, University of North Carolina at Charlotte, Charlotte, NC, USA
| | - Cory Look
- Sociology and Anthropology Department, Farmingdale State College, Farmingdale, NY, USA
| | - Kerttu Majander
- Department of Environmental Science, Integrative Prehistory and Archaeological Science, University of Basel, Basel, Switzerland
| | - Kirsten Mandl
- Department of Evolutionary Anthropology, University of Vienna, Vienna, Austria
| | | | - Michael McCormick
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean
- Initiative for the Science of the Human Past at Harvard, Department of History, Harvard University, Cambridge, MA, USA
| | - Patxuka de Miguel Ibáñez
- Instituto Universitario de Investigación en Arqueología y Patrimonio Histórico, Universidad de Alicante, San Vicente del Raspeig (Alicante), Spain
- Servicio de Obstetricia, Hospital Virgen de los Lirios-Fisabio, Alcoi, Spain
- Sección de Antropología, Sociedad de Ciencias Aranzadi, Donostia - San Sebastián, Spain
| | - Reg Murphy
- University of Nebraska-Lincoln, Lincoln, NE, USA
| | | | - Kerkko Nordqvist
- Helsinki Collegium for Advanced Studies, University of Helsinki, Helsinki, Finland
| | - Friederike Novotny
- Department of Anthropology, Natural History Museum Vienna, Vienna, Austria
| | - Martin Obenaus
- Silva Nortica Archäologische Dienstleistungen, Thunau am Kamp, Austria
| | - Lauro Olmo-Enciso
- Department of History, University of Alcalá, Alcalá de Henares, Spain
| | - Päivi Onkamo
- Department of Biology, University of Turku, Turku, Finland
| | - Jörg Orschiedt
- Landesamt für Denkmalpflege und Archäologie Sachsen-Anhalt, Halle, Germany
- Institut für Prähistorische Archäologie, Freie Universität Berlin, Berlin, Germany
| | - Valerii Patrushev
- Centre of Archaeological and Ethnographical Investigation, Mari State University, Yoshkar-Ola, Russia
| | - Sanni Peltola
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Faculty of Biological and Environmental Sciences, University of Helsinki, Helsinki, Finland
| | - Alejandro Romero
- Instituto Universitario de Investigación en Arqueología y Patrimonio Histórico, Universidad de Alicante, San Vicente del Raspeig (Alicante), Spain
- Departamento de Biotecnología, Universidad de Alicante, San Vicente del Raspeig, Spain
| | - Salvatore Rubino
- Department of Biomedical Sciences, University of Sassari, Sassari, Italy
| | - Antti Sajantila
- Department of Forensic Medicine, University of Helsinki, Helsinki, Finland
- Forensic Medicine Unit, Finnish Institute for Health and Welfare, Helsinki, Finland
| | - Domingo C Salazar-García
- Departament de Prehistòria, Arqueologia i Història Antiga, Universitat de València, Valencia, Spain
- Department of Geological Sciences, University of Cape Town, Cape Town, South Africa
| | - Elena Serrano
- Instituto Internacional de Investigaciones Prehistóricas, Universidad de Cantabria, Santander, Spain
- TAR Arqueología, Madrid, Spain
| | | | - Emanuela Sias
- Centro Studi sulla Civiltà del Mare, Stintino, Italy
| | - Mario Šlaus
- Anthropological Center, Croatian Academy of Sciences and Arts, Zagreb, Croatia
| | - Ladislav Stančo
- Institute of Classical Archaeology, Faculty of Arts, Charles University, Prague, Czech Republic
| | - Treena Swanston
- Department of Anthropology, Economics and Political Science, MacEwan University, Edmonton, Alberta, Canada
| | - Maria Teschler-Nicola
- Department of Evolutionary Anthropology, University of Vienna, Vienna, Austria
- Department of Anthropology, Natural History Museum Vienna, Vienna, Austria
| | | | - Katrien Van de Vijver
- Royal Belgian Institute of Natural Sciences, Brussels, Belgium
- Center for Archaeological Sciences, University of Leuven, Leuven, Belgium
- Dienst Archeologie - Stad Mechelen, Mechelen, Belgium
| | - Tamara L Varney
- Department of Anthropology, Lakehead University, Thunder Bay, Ontario, Canada
| | | | - Christopher K Waters
- Heritage Department, National Parks of Antigua and Barbuda, St. Paul's Parish, Antigua and Barbuda
| | - Estella Weiss-Krejci
- Austrian Archaeological Institute, Austrian Academy of Sciences, Vienna, Austria
- Institut für Ur- und Frühgeschichte, Heidelberg University, Heidelberg, Germany
- Department of Social and Cultural Anthropology, University of Vienna, Vienna, Austria
| | - Eduard Winter
- Department of Anthropology, Natural History Museum Vienna, Vienna, Austria
| | - Thiseas C Lamnidis
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Kay Prüfer
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Kathrin Nägele
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Maria Spyrou
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Archaeo- and Palaeogenetics, Institute for Archaeological Sciences, Department of Geosciences, University of Tübingen, Tübingen, Germany
| | - Stephan Schiffels
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Philipp W Stockhammer
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean
- Institute for Pre- and Protohistoric Archaeology and Archaeology of the Roman Provinces, Ludwig Maximilian University, Munich, Germany
| | - Wolfgang Haak
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Cosimo Posth
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Senckenberg Centre for Human Evolution and Palaeoenvironment, University of Tübingen, Tübingen, Germany
- Archaeo- and Palaeogenetics, Institute for Archaeological Sciences, Department of Geosciences, University of Tübingen, Tübingen, Germany
| | - Christina Warinner
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean
- Department of Anthropology, Harvard University, Cambridge, MA, USA
| | - Kirsten I Bos
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany
| | - Alexander Herbig
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany.
| | - Johannes Krause
- Department of Archaeogenetics, Max Planck Institute for Evolutionary Anthropology, Leipzig, Germany.
- Max Planck-Harvard Research Center for the Archaeoscience of the Ancient Mediterranean, .
| |
Collapse
|
15
|
Kombate G, Kone I, Douti B, Soubeiga KAM, Grobbee DE, van der Sande MAB. Malaria risk mapping among children under five in Togo. Sci Rep 2024; 14:8213. [PMID: 38589576 PMCID: PMC11001891 DOI: 10.1038/s41598-024-58287-1] [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: 01/02/2024] [Accepted: 03/27/2024] [Indexed: 04/10/2024] Open
Abstract
Malaria is a major health threat in sub-Sahara Africa, especially for children under five. However, there is considerable heterogeneity between areas in malaria risk reported, associated with environmental and climatic. We used data from Togo to explore spatial patterns of malaria incidence. Geospatial covariate datasets, including climatic and environmental variables from the 2017 Malaria Indicator Survey in Togo, were used for this study. The association between malaria incidence and ecological predictors was assessed using three regression techniques, namely the Ordinary Least Squares (OLS), spatial lag model (SLM), and spatial error model (SEM). A total of 171 clusters were included in the survey and provided data on environmental and climate variables. Spatial autocorrelation showed that the distribution of malaria incidence was not random and revealed significant spatial clustering. Mean temperature, precipitation, aridity and proximity to water bodies showed a significant and direct association with malaria incidence rate in the SLM model, which best fitted the data according to AIC. Five malaria incidence hotspots were identified. Malaria incidence is spatially clustered in Togo associated with climatic and environmental factors. The results can contribute to the development of specific malaria control plans taking geographical variation into consideration and targeting transmission hotspots.
Collapse
Affiliation(s)
- Gountante Kombate
- Ministry of Health and Public Hygiene, Lomé, Togo.
- Interdisciplinary Research Laboratory in Social Health Sciences University Joseph Ki-Zerbo, Ouagadougou, Burkina Faso.
| | - Issouf Kone
- African School of Economics (ASE), Cotonou, Benin
| | - Bili Douti
- Ministry of Health and Public Hygiene, Lomé, Togo
| | - Kamba André-Marie Soubeiga
- Interdisciplinary Research Laboratory in Social and University Joseph Ki-Zerbo, Ouagadougou, Burkina Faso
| | - Diederick E Grobbee
- Global Public Health, Julius Centre, University Medical Centre Utrecht, Utrecht, The Netherlands
| | - Marianne A B van der Sande
- Global Public Health, Julius Centre, University Medical Centre Utrecht, Utrecht, The Netherlands
- Department of Public Health, Institute of Tropical Medicine, Antwerp, Belgium
| |
Collapse
|
16
|
Habibzadeh F. On the feasibility of malaria hypothesis. Sci Rep 2024; 14:5800. [PMID: 38461305 PMCID: PMC10924941 DOI: 10.1038/s41598-024-56515-2] [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: 09/27/2023] [Accepted: 03/07/2024] [Indexed: 03/11/2024] Open
Abstract
In 1954, Allison proposed that hemoglobin S (HbS) gene causes protection against fatal malaria. This would explain the high HbS gene frequency observed in certain regions hyperendemic for malaria, so-called "malaria hypothesis". This in silico study was conducted to examine the feasibility of the hypothesis under more realistic initial conditions, where a mutant gene with heterozygous advantage against malaria (e.g., HbS) was introduced in a group of Neolithic hunter-gatherers who decided to start agriculture nearby water where malaria killed a proportion of population. The tribe population size, number of children born to each woman in each generation, mortality from malaria and sickle cell disease, the protection factor provided by the gene carriers against malaria, the probability of mating between the members of the parent and offspring populations, population growth, and increased fertility in women heterozygous for HbS, were also considered. For effectively confer protection against malaria within the shortest possible period, the mutation needs to be happened in a small population. For a large population, the process would take around 100 generations (~ 2500 years) or more to provide an effective protection. Even then, the probability that the new gene could survive and propagate to future generations is about 35%. Conventional population genetics equations with differential or difference equations, give totally incorrect estimates of the gene frequency in small populations; discrete mathematics should be used, instead. After introduction of the advantageous mutation, the gene frequency increased until a steady state value. This value is far less than the gene frequency reported in certain tribes of Africa. It seems that the malaria hypothesis, per se, could not explain such a high observed gene frequency, unless HbS is associated with lower mortality from other causes too.
Collapse
|
17
|
Omorou R, Delabie B, Lavoignat A, Chaker V, Bonnot G, Traore K, Bienvenu AL, Picot S. Nanoparticle tracking analysis of natural hemozoin from Plasmodium parasites. Acta Trop 2024; 250:107105. [PMID: 38135133 DOI: 10.1016/j.actatropica.2023.107105] [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: 07/25/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/24/2023]
Abstract
BACKGROUND Hemozoin is a byproduct of hemoglobin digestion crucial for parasite survival. It forms crystals that can be of interest as drug targets or biomarkers of malaria infection. However, hemozoin has long been considered as an amorphous crystal of simple morphology. Studying the consequences of biomineralization of this crystal during the parasite growth may provide more comprehensive evidence of its role during malaria. OBJECTIVES This study aimed to investigate the interest of nanoparticles tracker analysis for measuring the concentration and size of hemozoin particles produced from different parasite sources and conditions. METHODS Hemozoin was extracted from several clones of Plasmodium falciparum both asexual and sexual parasites. Hemozoin was also extracted from blood samples of malaria patients and from saliva of asymptomatic malaria carriers. Nanoparticles tracking analysis (NTA) was performed to assess the size and concentration of hemozoin. RESULTS NTA data showed variation in hemozoin concentration, size, and crystal clusters between parasite clones, species, and stages. Among parasite clones, hemozoin concentration ranged from 131 to 2663 particles/infected red blood cell (iRBC) and size ranged from 149.6 ± 6.3 nm to 234.8 ± 40.1 nm. The mean size was lower for Plasmodium vivax (176 ± 79.2 nm) than for Plasmodium falciparum (254.8 ± 74.0 nm). Sexual NF54 parasites showed a 7.5-fold higher concentration of hemozoin particles (28.7 particles/iRBC) compared to asexual parasites (3.8 particles/iRBC). In addition, the mean hemozoin size also increased by approximately 60 % for sexual parasites. Compared to in vitro cultures of parasites, blood samples showed low hemozoin concentrations. CONCLUSIONS This study highlights the potential of NTA as a useful method for analyzing hemozoin, demonstrating its ability to provide detailed information on hemozoin characterization. However, further research is needed to adapt the NTA for hemozoin analysis.
Collapse
Affiliation(s)
- Roukayatou Omorou
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE, University Lyon1, University Lyon, Villeurbanne 69100, France.
| | - Blanche Delabie
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE, University Lyon1, University Lyon, Villeurbanne 69100, France
| | - Adeline Lavoignat
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE, University Lyon1, University Lyon, Villeurbanne 69100, France
| | - Victorien Chaker
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE, University Lyon1, University Lyon, Villeurbanne 69100, France
| | - Guillaume Bonnot
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE, University Lyon1, University Lyon, Villeurbanne 69100, France
| | - Karim Traore
- Malaria Research and Training Center, University of Sciences, Techniques and Technologies, Bamako, Mali
| | - Anne-Lise Bienvenu
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE, University Lyon1, University Lyon, Villeurbanne 69100, France; Service Pharmacie, Groupement Hospitalier Nord, Hospices Civils de Lyon, Lyon 69004, France
| | - Stephane Picot
- Malaria Research Unit, UMR 5246 CNRS-INSA-CPE, University Lyon1, University Lyon, Villeurbanne 69100, France; Institute of Parasitology and Medical Mycology, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon 69004, France
| |
Collapse
|
18
|
Platon L, Zhang Q, Cao J, Ménard D. Malaria in the horn of Africa: The ongoing battle against drug resistance. Clin Transl Med 2023; 13:e1482. [PMID: 37983939 PMCID: PMC10659765 DOI: 10.1002/ctm2.1482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 11/06/2023] [Indexed: 11/22/2023] Open
Affiliation(s)
- Lucien Platon
- Institute of Parasitology and Tropical DiseasesUR7292 Dynamics of Host‐Pathogen InteractionsUniversité de StrasbourgStrasbourgFrance
| | - Qingfeng Zhang
- Laboratory of Molecular ParasitologyKey Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of EducationTongji HospitalClinical Center for Brain and Spinal Cord ResearchSchool of MedicineTongji UniversityShanghaiChina
| | - Jun Cao
- National Health Commission Key Laboratory of Parasitic Disease Control and PreventionJiangsu Provincial Key Laboratory on Parasite and Vector Control TechnologyJiangsu Institute of Parasitic DiseasesWuxiChina
- Center for Global HealthSchool of Public HealthNanjing Medical UniversityNanjingChina
| | - Didier Ménard
- Institute of Parasitology and Tropical DiseasesUR7292 Dynamics of Host‐Pathogen InteractionsUniversité de StrasbourgStrasbourgFrance
- Laboratory of Parasitology and Medical MycologyCentre Hospitalier Universitaire StrasbourgStrasbourgFrance
- Malaria Parasite Biology and Vaccines UnitInstitut PasteurUniversité Paris CitéParisFrance
| |
Collapse
|
19
|
Powrel J, Koirala RP, Adhikari NP. Structural stability of R-state conformation of carbonmonoxyl sickle and normal hemoglobin dimer. J Biomol Struct Dyn 2023; 41:9957-9966. [PMID: 36473709 DOI: 10.1080/07391102.2022.2150890] [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: 06/01/2022] [Accepted: 11/12/2022] [Indexed: 12/12/2022]
Abstract
A mutation at the sixth residue, glutamic acid to valine, in beta chain of hemoglobin distorts the entire shape of hemoglobin into a sickle shape. The investigation of the binding mechanisms of different chains of hemoglobin under the mutated condition can give an understanding of the molecular distortion. In this work, we have studied the binding mechanism between two chains in the dimer structure of the R-state conformation of carbonmonoxyl sickle hemoglobin and is compared with that of normal hemoglobin by using molecular dynamics simulations. The binding strength between α-chain (PROA) and β-chain (PROB) in hemoglobin dimer has been analyzed by estimating hydrogen bonds, salt bridges, hydrophobic interactions and non-bonded interactions (electrostatics and van der Waals). The quantitative estimation of aforementioned interactions depicts that the structural stability of normal hemoglobin dimer is found to be greater than that of sickle one. The outcomes of such interactions are also supported by the estimated free energy between the chains in R-state conformation of the dimers. The difference of binding free energy, calculated by utilizing the umbrella sampling technique, is found to be ≈ (0.67 ± 0.06) kcal/mol.Communicated by Ramaswamy H. Sarma.
Collapse
Affiliation(s)
- Jhulan Powrel
- Department of Physics, Butwal Multiple Campus, Butwal, Nepal
- Central Department of Physics, Tribhuvan University, Kathmandu, Nepal
| | | | | |
Collapse
|
20
|
Siqueira-Neto JL, Wicht KJ, Chibale K, Burrows JN, Fidock DA, Winzeler EA. Antimalarial drug discovery: progress and approaches. Nat Rev Drug Discov 2023; 22:807-826. [PMID: 37652975 PMCID: PMC10543600 DOI: 10.1038/s41573-023-00772-9] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2023] [Indexed: 09/02/2023]
Abstract
Recent antimalarial drug discovery has been a race to produce new medicines that overcome emerging drug resistance, whilst considering safety and improving dosing convenience. Discovery efforts have yielded a variety of new molecules, many with novel modes of action, and the most advanced are in late-stage clinical development. These discoveries have led to a deeper understanding of how antimalarial drugs act, the identification of a new generation of drug targets, and multiple structure-based chemistry initiatives. The limited pool of funding means it is vital to prioritize new drug candidates. They should exhibit high potency, a low propensity for resistance, a pharmacokinetic profile that favours infrequent dosing, low cost, preclinical results that demonstrate safety and tolerability in women and infants, and preferably the ability to block Plasmodium transmission to Anopheles mosquito vectors. In this Review, we describe the approaches that have been successful, progress in preclinical and clinical development, and existing challenges. We illustrate how antimalarial drug discovery can serve as a model for drug discovery in diseases of poverty.
Collapse
Affiliation(s)
| | - Kathryn J Wicht
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
| | - Kelly Chibale
- Holistic Drug Discovery and Development (H3D) Centre, University of Cape Town, Rondebosch, South Africa
- South African Medical Research Council Drug Discovery and Development Research Unit, Department of Chemistry and Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Rondebosch, South Africa
| | | | - David A Fidock
- Department of Microbiology and Immunology and Center for Malaria Therapeutics and Antimicrobial Resistance, Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, NY, USA
| | | |
Collapse
|
21
|
Fitri LE, Pawestri AR, Winaris N, Endharti AT, Khotimah ARH, Abidah HY, Huwae JTR. Antimalarial Drug Resistance: A Brief History of Its Spread in Indonesia. Drug Des Devel Ther 2023; 17:1995-2010. [PMID: 37431492 PMCID: PMC10329833 DOI: 10.2147/dddt.s403672] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 04/25/2023] [Indexed: 07/12/2023] Open
Abstract
Malaria remains to be a national and global challenge and priority, as stated in the strategic plan of the Indonesian Ministry of Health and Sustainable Development Goals. In Indonesia, it is targeted that malaria elimination can be achieved by 2030. Unfortunately, the development and spread of antimalarial resistance inflicts a significant risk to the national malaria control programs which can lead to increased malaria morbidity and mortality. In Indonesia, resistance to widely used antimalarial drugs has been reported in two human species, Plasmodium falciparum and Plasmodium vivax. With the exception of artemisinin, resistance has surfaced towards all classes of antimalarial drugs. Initially, chloroquine, sulfadoxine-pyrimethamine, and primaquine were the most widely used antimalarial drugs. Regrettably, improper use has supported the robust spread of their resistance. Chloroquine resistance was first reported in 1974, while sulfadoxine-pyrimethamine emerged in 1979. Twenty years later, most provinces had declared treatment failures of both drugs. Molecular epidemiology suggested that variations in pfmdr1 and pfcrt genes were associated with chloroquine resistance, while dhfr and dhps genes were correlated with sulfadoxine-pyrimethamine resistance. Additionally, G453W, V454C and E455K of pfk13 genes appeared to be early warning sign to artemisinin resistance. Here, we reported mechanisms of antimalarial drugs and their development of resistance. This insight could provide awareness toward designing future treatment guidelines and control programs in Indonesia.
Collapse
Affiliation(s)
- Loeki Enggar Fitri
- Department of Parasitology Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- AIDS, Toxoplasma, Opportunistic Disease and Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Aulia Rahmi Pawestri
- Department of Parasitology Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- AIDS, Toxoplasma, Opportunistic Disease and Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Nuning Winaris
- Department of Parasitology Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- AIDS, Toxoplasma, Opportunistic Disease and Malaria Research Group, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Agustina Tri Endharti
- Department of Parasitology Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| | - Alif Raudhah Husnul Khotimah
- Master Program in Biomedical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Medical Doctor Profession Education, Faculty of Medical and Health Science, Maulana Malik Ibrahim State Islamic University, Malang, Indonesia
| | - Hafshah Yasmina Abidah
- Master Program in Biomedical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Medical Doctor Profession Education, Faculty of Medical and Health Science, Maulana Malik Ibrahim State Islamic University, Malang, Indonesia
| | - John Thomas Rayhan Huwae
- Master Program in Biomedical Science, Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
- Medical Doctor Profession Study Program Faculty of Medicine, Universitas Brawijaya, Malang, Indonesia
| |
Collapse
|
22
|
Bauer IL. The oral repellent - science fiction or common sense? Insects, vector-borne diseases, failing strategies, and a bold proposition. Trop Dis Travel Med Vaccines 2023; 9:7. [PMID: 37381000 DOI: 10.1186/s40794-023-00195-9] [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: 12/21/2022] [Accepted: 06/09/2023] [Indexed: 06/30/2023] Open
Abstract
Over the last decades, unimaginable amounts of money have gone into research and development of vector control measures, repellents, treatment, and vaccines for vector borne diseases. Technological progress and scientific breakthroughs allowed for ever more sophisticated and futuristic strategies. Yet, each year, millions of people still die or suffer from potentially serious consequences of malaria or dengue to more recent infections, such as zika or chikungunya, or of debilitating consequences of neglected tropical diseases. This does not seem value for money. In addition, all current vector control strategies and personal protection methods have shortcomings, some serious, that are either destructive to non-target species or unsatisfactory in their effectiveness. On the other hand, the rapid decline in insect populations and their predators reflects decades-long aggressive and indiscriminate vector control. This major disruption of biodiversity has an impact on human life not anticipated by the well-meaning killing of invertebrates. The objective of this paper is to re-examine current control methods, their effectiveness, their impact on biodiversity, human and animal health, and to call for scientific courage in the pursuit of fresh ideas. This paper brings together topics that are usually presented in isolation, thereby missing important links that offer potential solutions to long-standing problems in global health. First, it serves as a reminder of the importance of insects to human life and discusses the few that play a role in transmitting disease. Next, it examines critically the many currently employed vector control strategies and personal protection methods. Finally, based on new insights into insect chemo-sensation and attractants, this perspective makes a case for revisiting a previously abandoned idea, the oral repellent, and its use via currently successful methods of mass-application. The call is out for focused research to provide a powerful tool for public health, tropical medicine, and travel medicine.
Collapse
Affiliation(s)
- Irmgard L Bauer
- College of Healthcare Sciences, Academy - Tropical Health and Medicine, James Cook University, Townsville, QLD, 4811, Australia.
| |
Collapse
|
23
|
Oxendine Harp K, Bashi A, Botchway F, Addo-Gyan D, Tetteh-Tsifoanya M, Lamptey A, Djameh G, Iqbal SA, Lekpor C, Banerjee S, Wilson MD, Dei-Adomakoh Y, Adjei AA, Stiles JK, Driss A. Sickle Cell Hemoglobin Genotypes Affect Malaria Parasite Growth and Correlate with Exosomal miR-451a and let-7i-5p Levels. Int J Mol Sci 2023; 24:7546. [PMID: 37108709 PMCID: PMC10141851 DOI: 10.3390/ijms24087546] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 04/04/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Malaria affects a significant portion of the global population, with 247 million cases in 2021, primarily in Africa. However, certain hemoglobinopathies, such as sickle cell trait (SCT), have been linked to lower mortality rates in malaria patients. Hemoglobin (Hb) mutations, including HbS and HbC, can cause sickle cell disease (SCD) when both alleles are inherited (HbSS and HbSC). In SCT, one allele is inherited and paired with a normal allele (HbAS, HbAC). The high prevalence of these alleles in Africa may be attributed to their protective effect against malaria. Biomarkers are crucial for SCD and malaria diagnosis and prognosis. Studies indicate that miRNAs, specifically miR-451a and let-7i-5p, are differentially expressed in HbSS and HbAS compared to controls. Our research examined the levels of exosomal miR-451a and let-7i-5p in red blood cells (RBCs) and infected red blood cells (iRBCs) from multiple sickle Hb genotypes and their impact on parasite growth. We assessed exosomal miR-451a and let-7i-5p levels in vitro in RBC and iRBC supernatants. Exosomal miRNAs exhibited distinct expression patterns in iRBCs from individuals with different sickle Hb genotypes. Additionally, we discovered a correlation between let-7i-5p levels and trophozoite count. Exosomal miR-451a and let-7i-5p could modulate SCD and malaria severity and serve as potential biomarkers for malaria vaccines and therapies.
Collapse
Affiliation(s)
- Keri Oxendine Harp
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (K.O.H.); (A.B.); (S.A.I.); (S.B.)
| | - Alaijah Bashi
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (K.O.H.); (A.B.); (S.A.I.); (S.B.)
| | - Felix Botchway
- Department of Pathology, Korle-Bu Teaching Hospital, University of Ghana Medical School, Accra P.O. Box 4236, Ghana; (F.B.); (C.L.); (A.A.A.)
| | - Daniel Addo-Gyan
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra P.O. Box LG581, Ghana; (D.A.-G.); (M.T.-T.); (A.L.); (G.D.); (M.D.W.)
| | - Mark Tetteh-Tsifoanya
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra P.O. Box LG581, Ghana; (D.A.-G.); (M.T.-T.); (A.L.); (G.D.); (M.D.W.)
| | - Amanda Lamptey
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra P.O. Box LG581, Ghana; (D.A.-G.); (M.T.-T.); (A.L.); (G.D.); (M.D.W.)
| | - Georgina Djameh
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra P.O. Box LG581, Ghana; (D.A.-G.); (M.T.-T.); (A.L.); (G.D.); (M.D.W.)
| | - Shareen A. Iqbal
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (K.O.H.); (A.B.); (S.A.I.); (S.B.)
| | - Cecilia Lekpor
- Department of Pathology, Korle-Bu Teaching Hospital, University of Ghana Medical School, Accra P.O. Box 4236, Ghana; (F.B.); (C.L.); (A.A.A.)
| | - Saswati Banerjee
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (K.O.H.); (A.B.); (S.A.I.); (S.B.)
| | - Michael D. Wilson
- Department of Parasitology, Noguchi Memorial Institute for Medical Research, University of Ghana, Accra P.O. Box LG581, Ghana; (D.A.-G.); (M.T.-T.); (A.L.); (G.D.); (M.D.W.)
| | - Yvonne Dei-Adomakoh
- Department of Haematology, Korle-Bu Teaching Hospital, Accra P.O. Box 77, Ghana;
| | - Andrew A. Adjei
- Department of Pathology, Korle-Bu Teaching Hospital, University of Ghana Medical School, Accra P.O. Box 4236, Ghana; (F.B.); (C.L.); (A.A.A.)
| | - Jonathan K. Stiles
- Department of Microbiology, Biochemistry and Immunology, Morehouse School of Medicine, Atlanta, GA 30310, USA;
| | - Adel Driss
- Department of Physiology, Morehouse School of Medicine, Atlanta, GA 30310, USA; (K.O.H.); (A.B.); (S.A.I.); (S.B.)
| |
Collapse
|
24
|
Maharaj R, Ward A, Didier B, Seocharan I, Firas N, Balawanth R, Lucero D, Morris N, Shandukani M, Raswiswi E, Malatjie G, Mabunda E, Moonasar D. The effect of the COVID-19 lockdown on malaria transmission in South Africa. Malar J 2023; 22:107. [PMID: 36964548 PMCID: PMC10038361 DOI: 10.1186/s12936-023-04542-1] [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: 11/19/2022] [Accepted: 03/22/2023] [Indexed: 03/26/2023] Open
Abstract
BACKGROUND For a country such as South Africa which is targeting malaria elimination, mobile and migrant populations pose a substantial risk to importation of malaria parasites. It has been hypothesized that halting cross-border movement of mobile and migrant populations will decrease the importation of malaria, however this option is not a politically, operationally, and financially viable prospect. It has social impacts as well, since families live on either side of the border and preventing travel will challenge family ties. Due to the COVID-19 pandemic and closure of ports of entry (land and air) for non-essential travel into South Africa, a unique opportunity arose to test the hypothesis. METHODOLOGY An interrupted time series analysis was done to assess whether the post-lockdown trends (April-December 2020) in monthly reported imported and local cases differed from the pre-lockdown trends (January 2015-March 2020). The analysis was conducted separately for KwaZulu-Natal, Mpumalanga, and Limpopo provinces. RESULTS On average, imported cases were lower in the post-intervention period in all three provinces, and local cases were lower in Mpumalanga and Limpopo, though no results were statistically significant. CONCLUSION Since population movement continued after the travel restrictions were lifted, border screening with testing and treating should be considered for reducing parasite movement. Another option is reducing malaria cases at the source in neighbouring countries by implementing proven, effective vector and parasite control strategies and through a downstream effect reduce malaria entering South Africa.
Collapse
Affiliation(s)
| | | | | | | | - Nina Firas
- Clinton Health Access Initiative, Mbabane, Eswatini
| | | | | | | | | | - Eric Raswiswi
- KwaZulu-Natal Provincial Department of Health, Jozini, South Africa
| | - Gillian Malatjie
- Mpumalanga Provincial Department of Health, Nelspruit, South Africa
| | - Erik Mabunda
- Limpopo Provincial Department of Health, Polokwane, South Africa
| | | |
Collapse
|
25
|
Dwivedi SR, Mishra LC, Mishra G. An Analytical Approach to Progression in Malaria Therapeutics. NATURAL PRODUCT BASED DRUG DISCOVERY AGAINST HUMAN PARASITES 2023:471-490. [DOI: 10.1007/978-981-19-9605-4_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
26
|
Scully EJ, Liu W, Li Y, Ndjango JBN, Peeters M, Kamenya S, Pusey AE, Lonsdorf EV, Sanz CM, Morgan DB, Piel AK, Stewart FA, Gonder MK, Simmons N, Asiimwe C, Zuberbühler K, Koops K, Chapman CA, Chancellor R, Rundus A, Huffman MA, Wolfe ND, Duraisingh MT, Hahn BH, Wrangham RW. The ecology and epidemiology of malaria parasitism in wild chimpanzee reservoirs. Commun Biol 2022; 5:1020. [PMID: 36167977 PMCID: PMC9515101 DOI: 10.1038/s42003-022-03962-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 09/01/2022] [Indexed: 11/09/2022] Open
Abstract
Chimpanzees (Pan troglodytes) harbor rich assemblages of malaria parasites, including three species closely related to P. falciparum (sub-genus Laverania), the most malignant human malaria parasite. Here, we characterize the ecology and epidemiology of malaria infection in wild chimpanzee reservoirs. We used molecular assays to screen chimpanzee fecal samples, collected longitudinally and cross-sectionally from wild populations, for malaria parasite mitochondrial DNA. We found that chimpanzee malaria parasitism has an early age of onset and varies seasonally in prevalence. A subset of samples revealed Hepatocystis mitochondrial DNA, with phylogenetic analyses suggesting that Hepatocystis appears to cross species barriers more easily than Laverania. Longitudinal and cross-sectional sampling independently support the hypothesis that mean ambient temperature drives spatiotemporal variation in chimpanzee Laverania infection. Infection probability peaked at ~24.5 °C, consistent with the empirical transmission optimum of P. falciparum in humans. Forest cover was also positively correlated with spatial variation in Laverania prevalence, consistent with the observation that forest-dwelling Anophelines are the primary vectors. Extrapolating these relationships across equatorial Africa, we map spatiotemporal variation in the suitability of chimpanzee habitat for Laverania transmission, offering a hypothetical baseline indicator of human exposure risk.
Collapse
Affiliation(s)
- Erik J Scully
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA
- Department of Immunology & Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA
| | - Weimin Liu
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Yingying Li
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jean-Bosco N Ndjango
- Department of Ecology and Management of Plant and Animal Resources, Faculty of Sciences, University of Kisangani, BP 2012, Kisangani, Democratic Republic of the Congo
| | - Martine Peeters
- Recherche Translationnelle Appliquée au VIH et aux Maladies Infectieuses, Institut de Recherche pour le Développement, University of Montpellier, INSERM, 34090, Montpellier, France
| | - Shadrack Kamenya
- Gombe Stream Research Centre, The Jane Goodall Institute, Tanzania, Kigoma, Tanzania
| | - Anne E Pusey
- Department of Evolutionary Anthropology, Duke University, Durham, NC, 27708, USA
| | - Elizabeth V Lonsdorf
- Department of Psychology, Franklin and Marshall College, Lancaster, PA, 17604, USA
| | - Crickette M Sanz
- Department of Anthropology, Washington University in St. Louis, St Louis, MO, 63130, USA
- Congo Program, Wildlife Conservation Society, BP 14537, Brazzaville, Republic of the Congo
| | - David B Morgan
- Lester E. Fisher Center for the Study and Conservation of Apes, Lincoln Park Zoo, Chicago, IL, 60614, USA
| | - Alex K Piel
- Department of Anthropology, University College London, 14 Taviton St, Bloomsbury, WC1H OBW, London, UK
| | - Fiona A Stewart
- Department of Anthropology, University College London, 14 Taviton St, Bloomsbury, WC1H OBW, London, UK
- School of Biological and Environmental Sciences, Liverpool John Moores University, Liverpool, L3 3AF, UK
| | - Mary K Gonder
- Department of Biology, Drexel University, Philadelphia, PA, 19104, USA
| | - Nicole Simmons
- Zoology Department, Makerere University, P.O. Box 7062, Kampala, Uganda
| | | | - Klaus Zuberbühler
- School of Psychology and Neuroscience, University of St Andrews, St Andrews, UK
- Department of Comparative Cognition, Institute of Biology, University of Neuchâtel, Neuchâtel, Switzerland
| | - Kathelijne Koops
- Department of Ape Behaviour & Ecology Group, University of Zurich, Zurich, Switzerland
| | - Colin A Chapman
- Department of Anthropology, Center for the Advanced Study of Human Paleobiology, George Washington University, Washington, DC, USA
- School of Life Sciences, University of KwaZulu-Natal, Scottsville, Pietermaritzburg, South Africa
| | - Rebecca Chancellor
- Department of Anthropology & Sociology, West Chester University, West Chester, PA, USA
- Department of Psychology, West Chester University, West Chester, PA, USA
| | - Aaron Rundus
- Department of Psychology, West Chester University, West Chester, PA, USA
| | - Michael A Huffman
- Center for International Collaboration and Advanced Studies in Primatology, Primate Research Institute, Kyoto University, Inuyama, Aichi, Japan
| | | | - Manoj T Duraisingh
- Department of Immunology & Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, 02115, USA.
| | - Beatrice H Hahn
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Richard W Wrangham
- Department of Human Evolutionary Biology, Harvard University, Cambridge, MA, 02138, USA.
| |
Collapse
|
27
|
Cravo P. On the contribution of the rodent model Plasmodium chabaudi for understanding the genetics of drug resistance in malaria. Parasitol Int 2022; 91:102623. [PMID: 35803536 DOI: 10.1016/j.parint.2022.102623] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 06/22/2022] [Accepted: 06/30/2022] [Indexed: 10/17/2022]
Abstract
Malaria is a devastating disease that still claims over half a million lives every year, mostly in sub-Saharan Africa. One of the main barriers to malaria control is the evolution and propagation of drug-resistant mutant parasites. Knowing the genes and respective mutations responsible for drug resistance facilitates the design of drugs with novel modes of action and allows predicting and monitoring drug resistance in natural parasite populations in real-time. The best way to identify these mutations is to experimentally evolve resistance to the drug in question and then comparing the genomes of the drug-resistant mutants to that of the sensitive progenitor parasites. This simple evolutive concept was the starting point for the development of a paradigm over the years, based on the use of the rodent malaria parasite Plasmodium chabaudi to unravel the genetics of drug resistance in malaria. It involves the use of a cloned parasite isolate (P. chabaudi AS) whose genome is well characterized, to artificially select resistance to given drugs through serial passages in mice under slowly increasing drug pressure. The end resulting parasites are cloned and the genetic mutations are then discovered through Linkage Group Selection, a technique conceived by Prof. Richard Carter and his group, and/or Whole Genome Sequencing. The precise role of these mutations can then be interrogated in malaria parasites of humans through allelic replacement experiments and/or genotype-phenotype association studies in natural parasite populations. Using this paradigm, all the mutations underlying resistance to the most important antimalarial drugs were identified, most of which were pioneering and later shown to also play a role in drug resistance in natural infections of human malaria parasites. This supports the use of P. chabaudi a fast-track predictive model to identify candidate genetic markers of resistance to present and future antimalarial drugs and improving our understanding of the biology of resistance.
Collapse
Affiliation(s)
- Pedro Cravo
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical, Universidade Nova de Lisboa, Rua da Junqueira, n° 100, 1349-008 Lisboa, Portugal.
| |
Collapse
|
28
|
Yee DA, Dean Bermond C, Reyes-Torres LJ, Fijman NS, Scavo NA, Nelsen J, Yee SH. Robust network stability of mosquitoes and human pathogens of medical importance. Parasit Vectors 2022; 15:216. [PMID: 35725618 PMCID: PMC9208160 DOI: 10.1186/s13071-022-05333-4] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 05/23/2022] [Indexed: 11/22/2022] Open
Abstract
Background The exact number of mosquito species relevant to human health is unknown, posing challenges in understanding the scope and breadth of vector–pathogen relationships, and how resilient mosquito vector–pathogen networks are to targeted eradication of vectors. Methods We performed an extensive literature survey to determine the associations between mosquito species and their associated pathogens of human medical importance. For each vector–pathogen association, we then determined the strength of the associations (i.e., natural infection, lab infection, lab dissemination, lab transmission, known vector). A network analysis was used to identify relationships among all pathogens and vectors. Finally, we examined how elimination of either random or targeted species affected the extinction of pathogens. Results We found that 88 of 3578 mosquito species (2.5%) are known vectors for 78 human disease-causing pathogens; however, an additional 243 species (6.8%) were identified as potential or likely vectors, bringing the total of all mosquitos implicated in human disease to 331 (9.3%). Network analysis revealed that known vectors and pathogens were compartmentalized, with the removal of six vectors being enough to break the network (i.e., cause a pathogen to have no vector). However, the presence of potential or likely vectors greatly increased redundancies in the network, requiring more than 41 vectors to be eliminated before breaking the network. Conclusion Although < 10% of mosquitoes are involved in transmitting pathogens that cause human disease, our findings point to inherent robustness in global mosquito vector–pathogen networks. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s13071-022-05333-4.
Collapse
Affiliation(s)
- Donald A Yee
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, 118 College Drive, Hattiesburg, MS, 39406, USA.
| | - Catherine Dean Bermond
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, 118 College Drive, Hattiesburg, MS, 39406, USA
| | - Limarie J Reyes-Torres
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, 118 College Drive, Hattiesburg, MS, 39406, USA
| | - Nicole S Fijman
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, 118 College Drive, Hattiesburg, MS, 39406, USA
| | - Nicole A Scavo
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, 118 College Drive, Hattiesburg, MS, 39406, USA
| | - Joseph Nelsen
- School of Biological, Environmental, and Earth Sciences, University of Southern Mississippi, 118 College Drive, Hattiesburg, MS, 39406, USA
| | - Susan H Yee
- Gulf Ecosystem Measurement and Modeling Division, U.S. Environmental Protection Agency, 1 Sabine Island Drive, Gulf Breeze, FL, 32561, USA
| |
Collapse
|
29
|
Nosten F, Richard-Lenoble D, Danis M. A Brief History of Malaria. Presse Med 2022; 51:104130. [PMID: 35667599 DOI: 10.1016/j.lpm.2022.104130] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 05/31/2022] [Indexed: 11/29/2022] Open
Abstract
For millions of years, invertebrates and malaria parasites have coexisted and to date, malaria remains the most important human parasitic disease. This co-evolution had profound impacts on the movements of early hominids and on the genome of modern humans. Over the past two centuries, progress has been made with the discovery of the parasite, its transmission, and medicines, paving the way to the control of the disease and its elimination in some countries. However, the Plasmodium parasite is a formidable foe capable of developing resistance to drugs, and the mosquito vector has adapted to insecticides, foiling all attempts to eradicate the disease. Over recent years the economic and social costs of malaria have been recognized and more funds have been mobilized than ever before, however further efforts are needed. National programs, international institutions and researchers will need to do better if the preventable deaths of hundreds of thousands of mostly African children are to be averted.
Collapse
Affiliation(s)
- François Nosten
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine Research building, University of Oxford Old Road campus, Oxford, UK; Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand.
| | - Dominique Richard-Lenoble
- Professeur émérite des Universités - CHR de Tours, Parasitologie-Médecine tropicale. Membre de l'Académie Nationale de Médecine
| | - Martin Danis
- Professeur émérite, Parasitologie, Médecine Tropicale, Centre Hospitalier Universitaire Pitié-Salpêtrière, Sorbonne Université Paris Médecine, Académie Nationale de Médecine
| |
Collapse
|
30
|
Marzal A, Magallanes S, Garcia-Longoria L. Stimuli Followed by Avian Malaria Vectors in Host-Seeking Behaviour. BIOLOGY 2022; 11:726. [PMID: 35625454 PMCID: PMC9138572 DOI: 10.3390/biology11050726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 03/30/2022] [Accepted: 04/29/2022] [Indexed: 06/15/2023]
Abstract
Vector-borne infectious diseases (e.g., malaria, dengue fever, and yellow fever) result from a parasite transmitted to humans and other animals by blood-feeding arthropods. They are major contributors to the global disease burden, as they account for nearly a fifth of all infectious diseases worldwide. The interaction between vectors and their hosts plays a key role driving vector-borne disease transmission. Therefore, identifying factors governing host selection by blood-feeding insects is essential to understand the transmission dynamics of vector-borne diseases. Here, we review published information on the physical and chemical stimuli (acoustic, visual, olfactory, moisture and thermal cues) used by mosquitoes and other haemosporidian vectors to detect their vertebrate hosts. We mainly focus on studies on avian malaria and related haemosporidian parasites since this animal model has historically provided important advances in our understanding on ecological and evolutionary process ruling vector-borne disease dynamics and transmission. We also present relevant studies analysing the capacity of feather and skin symbiotic bacteria in the production of volatile compounds with vector attractant properties. Furthermore, we review the role of uropygial secretions and symbiotic bacteria in bird-insect vector interactions. In addition, we present investigations examining the alterations induced by haemosporidian parasites on their arthropod vector and vertebrate host to enhance parasite transmission. Finally, we propose future lines of research for designing successful vector control strategies and for infectious disease management.
Collapse
Affiliation(s)
- Alfonso Marzal
- Department of Anatomy, Cellular Biology and Zoology, University of Extremadura, Avenida de Elvas s/n, 06006 Badajoz, Spain;
- Grupo de Investigación y Sostenibilidad Ambiental, Universidad Nacional Federico Villarreal, Lima 15007, Peru
| | - Sergio Magallanes
- Department of Wetland Ecology, Biological Station (EBD-CSIC), Avda, Américo Vespucio 26, 41092 Sevilla, Spain;
| | - Luz Garcia-Longoria
- Department of Anatomy, Cellular Biology and Zoology, University of Extremadura, Avenida de Elvas s/n, 06006 Badajoz, Spain;
| |
Collapse
|
31
|
Simwela NV, Waters AP. Current status of experimental models for the study of malaria. Parasitology 2022; 149:1-22. [PMID: 35357277 PMCID: PMC9378029 DOI: 10.1017/s0031182021002134] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/07/2021] [Accepted: 12/08/2021] [Indexed: 01/09/2023]
Abstract
Infection by malaria parasites (Plasmodium spp.) remains one of the leading causes of morbidity and mortality, especially in tropical regions of the world. Despite the availability of malaria control tools such as integrated vector management and effective therapeutics, these measures have been continuously undermined by the emergence of vector resistance to insecticides or parasite resistance to frontline antimalarial drugs. Whilst the recent pilot implementation of the RTS,S malaria vaccine is indeed a remarkable feat, highly effective vaccines against malaria remain elusive. The barriers to effective vaccines result from the complexity of both the malaria parasite lifecycle and the parasite as an organism itself with consequent major gaps in our understanding of their biology. Historically and due to the practical and ethical difficulties of working with human malaria infections, research into malaria parasite biology has been extensively facilitated by animal models. Animals have been used to study disease pathogenesis, host immune responses and their (dys)regulation and further disease processes such as transmission. Moreover, animal models remain at the forefront of pre-clinical evaluations of antimalarial drugs (drug efficacy, mode of action, mode of resistance) and vaccines. In this review, we discuss commonly used animal models of malaria, the parasite species used and their advantages and limitations which hinder their extrapolation to actual human disease. We also place into this context the most recent developments such as organoid technologies and humanized mice.
Collapse
Affiliation(s)
- Nelson V. Simwela
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| | - Andrew P. Waters
- Institute of Infection, Immunity & Inflammation, Wellcome Centre for Integrative Parasitology, University of Glasgow, Glasgow, UK
| |
Collapse
|
32
|
Greener M. Parasites and mental health. PROGRESS IN NEUROLOGY AND PSYCHIATRY 2022. [DOI: 10.1002/pnp.733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
|
33
|
Melamed D, Nov Y, Malik A, Yakass MB, Bolotin E, Shemer R, Hiadzi EK, Skorecki KL, Livnat A. De novo mutation rates at the single-mutation resolution in a human HBB gene-region associated with adaptation and genetic disease. Genome Res 2022; 32:488-498. [PMID: 35031571 PMCID: PMC8896469 DOI: 10.1101/gr.276103.121] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 01/10/2022] [Indexed: 11/25/2022]
Abstract
Although it is known that the mutation rate varies across the genome, previous estimates were based on averaging across various numbers of positions. Here, we describe a method to measure the origination rates of target mutations at target base positions and apply it to a 6-bp region in the human hemoglobin subunit beta (HBB) gene and to the identical, paralogous hemoglobin subunit delta (HBD) region in sperm cells from both African and European donors. The HBB region of interest (ROI) includes the site of the hemoglobin S (HbS) mutation, which protects against malaria, is common in Africa, and has served as a classic example of adaptation by random mutation and natural selection. We found a significant correspondence between de novo mutation rates and past observations of alleles in carriers, showing that mutation rates vary substantially in a mutation-specific manner that contributes to the site frequency spectrum. We also found that the overall point mutation rate is significantly higher in Africans than in Europeans in the HBB region studied. Finally, the rate of the 20A→T mutation, called the “HbS mutation” when it appears in HBB, is significantly higher than expected from the genome-wide average for this mutation type. Nine instances were observed in the African HBB ROI, where it is of adaptive significance, representing at least three independent originations; no instances were observed elsewhere. Further studies will be needed to examine mutation rates at the single-mutation resolution across these and other loci and organisms and to uncover the molecular mechanisms responsible.
Collapse
|
34
|
Kim YH, Ahn HJ, Kim D, Hong SJ, Kim TS, Nam HW. Recent Spatial and Temporal Trends of Malaria in Korea. THE KOREAN JOURNAL OF PARASITOLOGY 2021; 59:585-593. [PMID: 34974665 PMCID: PMC8721308 DOI: 10.3347/kjp.2021.59.6.585] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 11/30/2021] [Indexed: 12/03/2022]
Abstract
This study was done to provide an analytical overview on the latest malaria infection clusters by evaluating temporal trends during 2010–2019 in Korea. Incheon was the most likely cluster (MLC) for all cases of malaria during the total period. MLCs for P. falciparum, vivax, malariae, ovale, and clinically diagnosed malaria without parasitological confirmation were Jeollanam-do, Incheon, Gangwon-do, Gyeongsangnam-do, and Jeollabuk-do, respectively. Malaria was decreasing in most significant clusters, but Gwangju showed an increase for all cases of malaria, P. vivax and clinically diagnosed cases. Malaria overall, P. falciparum and P. vivax seem to be under control thanks to aggressive health measures. This study might provide a sound scientific basis for future control measures against malaria in Korea.
Collapse
Affiliation(s)
- Yeong Hoon Kim
- Department of Ophthalmology, College of Medicine, Catholic University of Korea, Seoul 06591,
Korea
| | - Hye-Jin Ahn
- Department of Parasitology, College of Medicine, Catholic University of Korea, Seoul 06591,
Korea
| | - Dongjae Kim
- Department of Biomedicine Health Science, College of Medicine, The Catholic University of Korea, Seoul 06591,
Korea
| | - Sung-Jong Hong
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012,
Korea
| | - Tong-Soo Kim
- Convergence Research Center for Insect Vectors, Incheon National University, Incheon 22012,
Korea
| | - Ho-Woo Nam
- Department of Parasitology, College of Medicine, Catholic University of Korea, Seoul 06591,
Korea
- Corresponding author ()
| |
Collapse
|
35
|
Arora G, Sajid A, Chuang YM, Dong Y, Gupta A, Gambardella K, DePonte K, Almeras L, Dimopolous G, Fikrig E. Immunomodulation by Mosquito Salivary Protein AgSAP Contributes to Early Host Infection by Plasmodium. mBio 2021; 12:e0309121. [PMID: 34903042 PMCID: PMC8669493 DOI: 10.1128/mbio.03091-21] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 10/29/2021] [Indexed: 11/20/2022] Open
Abstract
Malaria is caused when Plasmodium sporozoites are injected along with saliva by an anopheline mosquito into the dermis of a vertebrate host. Arthropod saliva has pleiotropic effects that can influence local host responses, pathogen transmission, and exacerbation of the disease. A mass spectrometry screen identified mosquito salivary proteins that are associated with Plasmodium sporozoites during saliva secretions. In this study, we demonstrate that one of these salivary antigens, Anopheles gambiae sporozoite-associated protein (AgSAP), interacts directly with Plasmodium falciparum and Plasmodium berghei sporozoites. AgSAP binds to heparan sulfate and inhibits local inflammatory responses in the skin. The silencing of AgSAP in mosquitoes reduces their ability to effectively transmit sporozoites to mice. Moreover, immunization with AgSAP decreases the Plasmodium burden in mice that are bitten by Plasmodium-infected mosquitoes. These data suggest that AgSAP facilitates early Plasmodium infection in the vertebrate host and serves as a target for the prevention of malaria. IMPORTANCE Malaria is a vector-borne disease caused by Plasmodium sporozoites. When an anopheline mosquito bites its host, it releases Plasmodium sporozoites as well as saliva components. Mosquito proteins have the potential to serve as antigens to prevent or influence malaria without directly targeting the pathogen. This may help set a new paradigm for vaccine development. In this study, we have elucidated the role of a novel salivary antigen, named Anopheles gambiae sporozoite-associated protein (AgSAP). The results presented here show that AgSAP interacts with Plasmodium falciparum and Plasmodium berghei sporozoites and modulates local inflammatory responses in the skin. Furthermore, our results show that AgSAP is a novel mosquito salivary antigen that influences the early stages of Plasmodium infection in the vertebrate host. Individuals living in countries where malaria is endemic generate antibodies against AgSAP, which indicates that AgSAP can serve as a biomarker for disease prevalence and epidemiological analysis.
Collapse
Affiliation(s)
- Gunjan Arora
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Andaleeb Sajid
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yu-Min Chuang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Yuemei Dong
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Akash Gupta
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kristen Gambardella
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Kathleen DePonte
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Lionel Almeras
- Unité de Parasitologie et Entomologie, Département de Microbiologie et Maladies Infectieuses, Institut de Recherche Biomédicale des Armées, Marseille, France
- Aix Marseille Université, IRD, AP-HM, SSA, UMR Vecteurs-Infections Tropicales et Méditerranéennes (VITROME), IHU-Méditerranée Infection, Marseille, France
| | - George Dimopolous
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
36
|
Sternberg RJ. Adaptive Intelligence: Intelligence Is Not a Personal Trait but Rather a Person × Task × Situation Interaction. J Intell 2021; 9:jintelligence9040058. [PMID: 34940380 PMCID: PMC8704265 DOI: 10.3390/jintelligence9040058] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/03/2021] [Accepted: 11/21/2021] [Indexed: 01/10/2023] Open
Abstract
This article explores the advantages of viewing intelligence not as a fixed trait residing within an individual, but rather as a person × task × situation interaction. The emphasis in the article is on the role of persons solving tasks embedded in situations involving learning, intellectual abilities, and competencies. The article opens with a consideration of the role of situations in intelligent behavior. The article then discusses how intelligence is more similar to creativity and wisdom, in terms of the role of situations, than many psychologists have realized. Then the article reviews the role of situations in identity-based and irrational thinking and in conspiratorial thinking and cults. Next the article discusses the demonstrated importance of situations in assessment, but also notes the difficulties in sampling situations. Finally, the article draws conclusions, in particular, that, given our lack of situation-based tests, we need to be more modest in our interpretations results from conventional tests of intelligence.
Collapse
Affiliation(s)
- Robert J Sternberg
- Department of Psychology, College of Human Ecology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
37
|
Doolan DL. Malaria research in Australia: looking through the lens of the past towards the future. Int J Parasitol 2021; 51:1255-1263. [PMID: 34780720 DOI: 10.1016/j.ijpara.2021.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 11/05/2021] [Accepted: 11/05/2021] [Indexed: 10/19/2022]
Abstract
Malaria remains a global health priority, with substantial resources devoted to control and intervention since the causative parasite was first identified in 1880. Major advances have been made in discovery and translational research activities aimed at prevention, treatment and control. Laboratory-based, clinical, and field-based studies have complemented public health approaches. Australian scientists have played important roles, developing and applying innovative approaches, novel research tools and cutting-edge technologies in animal and human models of disease, as well as in disease-endemic settings. This article will provide an insight into 50 years of Australian efforts to discover mechanisms and targets of immunity and pathogenesis; develop new diagnostics, drugs, vaccines, and therapeutics; and assess new public health interventions and control measures in malaria-endemic settings.
Collapse
Affiliation(s)
- Denise L Doolan
- Centre for Molecular Therapeutics, Australian Institute of Tropical Health & Medicine, James Cook University, Cairns QLD 4878, Australia.
| |
Collapse
|
38
|
Alano P, Culleton R, Doerig C, Miller L. Professor Richard Carter (1945-2021). Trends Parasitol 2021. [PMID: 34742625 DOI: 10.1016/j.pt.2021.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The malaria research community lost a pioneer when Professor Richard Carter passed away at the age of 76 on 4 September 2021. Richard was an exceptionally brilliant malariologist, always inquisitive and gifted with an unorthodox way of thinking.
Collapse
Affiliation(s)
| | | | | | - Louis Miller
- National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
39
|
Golding TM, Mbaba M, Smith GS. Modular synthesis of antimalarial quinoline-based PGM metallarectangles. Dalton Trans 2021; 50:15274-15286. [PMID: 34633398 DOI: 10.1039/d1dt02842a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
A new ditopic, quinoline-based ligand L (7-chloro-4-(pyridin-4-yl)quinoline) was synthesized via a Suzuki cross-coupling reaction. The ligand was utilized to synthesize the corresponding half-sandwich iridium(III) and ruthenium(II) binuclear complexes (1c and 1d) and the subsequent metallarectangles (2c, 2d, 3c, and 3d), via [2 + 2] coordination-driven self-assembly. Single-crystal X-ray diffraction confirmed the proposed molecular structure of the binuclear complex [{IrCl2(Cp*)}2(μ-L)] (1c) and DFT calculations were used to predict the optimized geometry of the rectangular nature of [{Ir(μ-Cl)(Cp*)}4(μ-L)2](CF3SO3)4 (2c). All of the metallarectangles were isolated as their triflate salts and characterized using various spectroscopic (1H, 13C{1H}, DOSY NMR, and IR spectroscopy) and analytical techniques (ESI-MS). The synthesized compounds were screened against the NF54 chloroquine-sensitive (CQS) and K1 chloroquine-resistant (CQR) strains of Plasmodium falciparum. Incorporation of the ubiquitous quinoline core and metal complexation significantly enhanced the in vitro biological activity, with an increase in the nuclearity correlating with an increase in the resultant antiplasmodial activity. This was observed across both parasitic strains, alluding to the potential of supramolecular metallarectangles to act as antiplasmodial agents. Inhibition of haemozoin formation was considered a potential mechanism of action and selected metallarectangles exhibit β-haematin inhibition activity with near comparable activity to chloroquine.
Collapse
Affiliation(s)
- Taryn M Golding
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, South Africa.
| | - Mziyanda Mbaba
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, South Africa.
| | - Gregory S Smith
- Department of Chemistry, University of Cape Town, Rondebosch, Cape Town, South Africa.
| |
Collapse
|
40
|
Fatunla OAT, Olatunya OS, Ogundare EO, Fatunla TO, Oluwayemi IO, Oluwadiya KS, Oyelami OA. Towards malaria control in Nigeria: implications of the malaria parasite rate and spleen rate among children living in a rural community in southwest Nigeria. Trans R Soc Trop Med Hyg 2021; 115:1330-1338. [PMID: 34460920 DOI: 10.1093/trstmh/trab131] [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: 04/20/2021] [Revised: 07/14/2021] [Accepted: 08/11/2021] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND The World Health Organization prioritizes malaria surveillance for accurate tracking of progress of intervention programmes. The malaria parasite rate (PR) and spleen rate (SR) are economical surveillance tools. There has been a global decline in the burden of malaria over the last decade, but most African countries, like Nigeria, have a slow rate of decline. There is a need for adequate malaria surveillance to guide malaria control strategies and policymaking. METHODS A community-based cross-sectional study was conducted among 363 children ages 1-15 y in rural southwest Nigeria. The participants' PR was determined by microscopy and the SR was determined by palpation and ultrasonography. The associations between PR and SR and other covariates were assessed. RESULTS The PR was 26.7% and the SR was 12.9%. There was no significant association between PR or SR across age groups, but low social class was significantly associated with PR (55 [33.5%], p=0.004) and SR (29 [17.3%], p=0.013). The odds of having splenomegaly doubled with malaria parasitaemia (odds ratio 2.03 [95% confidence interval 1.06 to 3.88). CONCLUSIONS The PR and SR suggest that the study area is meso-endemic. The PR in the study area was almost equal across age groups; our findings suggest there may be a need for policy review to plan malaria intervention programmes and include older children, not just children <5 y of age. Routine malaria surveillance using simple tools such as the PR and SR are necessary for reviewing malaria control programmes in the community.
Collapse
Affiliation(s)
- Odunayo A T Fatunla
- Department of Paediatrics, Ekiti State University Teaching Hospital, Ado-Ekiti, Ekiti State, Nigeria
| | - Oladele S Olatunya
- Department of Paediatrics, Ekiti State University Teaching Hospital, Ado-Ekiti, Ekiti State, Nigeria.,Department of Paediatrics, College of Medicine, Ekiti State University, Ado-Ekiti, Ekiti State, Nigeria
| | - Ezra O Ogundare
- Department of Paediatrics, Ekiti State University Teaching Hospital, Ado-Ekiti, Ekiti State, Nigeria.,Department of Paediatrics, College of Medicine, Ekiti State University, Ado-Ekiti, Ekiti State, Nigeria
| | - Tolulope O Fatunla
- Department of Family Medicine, Ekiti State University Teaching Hospital, Ado-Ekiti, Ekiti State, Nigeria
| | - Isaac O Oluwayemi
- Department of Paediatrics, Ekiti State University Teaching Hospital, Ado-Ekiti, Ekiti State, Nigeria.,Department of Paediatrics, College of Medicine, Ekiti State University, Ado-Ekiti, Ekiti State, Nigeria
| | - Kehinde S Oluwadiya
- Department of Surgery, College of Medicine, Ekiti State University, Ado-Ekiti, Ekiti State, Nigeria
| | - Oyeku A Oyelami
- Department of Paediatrics, Ekiti State University Teaching Hospital, Ado-Ekiti, Ekiti State, Nigeria.,Department of Paediatrics, College of Medicine, Ekiti State University, Ado-Ekiti, Ekiti State, Nigeria
| |
Collapse
|
41
|
Ainembabazi P, Castelnuovo B, Okoboi S, Arinaitwe WJ, Parkes-Ratanshi R, Byakika-Kibwika P. A situation analysis of competences of research ethics committee members regarding review of research protocols with complex and emerging study designs in Uganda. BMC Med Ethics 2021; 22:132. [PMID: 34565355 PMCID: PMC8474911 DOI: 10.1186/s12910-021-00692-6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 09/08/2021] [Indexed: 11/23/2022] Open
Abstract
Background Over the past two decades, Uganda has experienced a significant increase in clinical research driven by both academia and industry. This has been combined with a broader spectrum of research proposals, with respect to methodologies and types of intervention that need evaluation by Research Ethics Committees (RECs) with associated increased requirement for expertise. We assessed the competencies of REC members regarding review of research protocols with complex and emerging research study designs. The aim was to guide development of a training curriculum to improve the quality of scientific and ethical review. Methods This was a cross-sectional study design, with quantitative data collection methods. Research Ethics Committee members completed a structured pre-coded questionnaire on current competence with complex and emerging study design. REC members were asked to outline a list of additional topics for which they needed training. Data from coded questions were entered into Epidata Version 3.1 and then exported to STATA Version14.1 for analysis. Descriptive analysis was performed and findings are presented using percentages and frequencies. Results We enrolled 55 REC members from 6 RECs who have a total of 97 members. The majority of whom were males (56.4%, n = 31/55). The level of competence for review of selected study design was lowest for Controlled Human Infection Model (10.9%, n = 6) and reverse pharmacology design (10.9%, n = 6), and highest for cluster randomized study design (52.7%, n = 29) and implementation science research (52.7%, n = 29). Conclusion Competence for review of research protocols with complex and emerging study design was low among participating REC members. We recommend prioritising training of REC members on complex and emerging study designs to enhance quality of research protocol review. Supplementary Information The online version contains supplementary material available at 10.1186/s12910-021-00692-6.
Collapse
Affiliation(s)
- Provia Ainembabazi
- Infectious Diseases Institute, Makerere University, College of Health Sciences, P.O BOX 22418, Kampala, Uganda
| | - Barbara Castelnuovo
- Infectious Diseases Institute, Makerere University, College of Health Sciences, P.O BOX 22418, Kampala, Uganda
| | - Stephen Okoboi
- Infectious Diseases Institute, Makerere University, College of Health Sciences, P.O BOX 22418, Kampala, Uganda
| | - Walter Joseph Arinaitwe
- Infectious Diseases Institute, Makerere University, College of Health Sciences, P.O BOX 22418, Kampala, Uganda
| | - Rosalind Parkes-Ratanshi
- Infectious Diseases Institute, Makerere University, College of Health Sciences, P.O BOX 22418, Kampala, Uganda.,Clinical School, University of Cambridge, Cambridge, UK
| | - Pauline Byakika-Kibwika
- Infectious Diseases Institute, Makerere University, College of Health Sciences, P.O BOX 22418, Kampala, Uganda. .,Department of Medicine, Makerere University College of Health Sciences, P.O Box 7072, Kampala, Uganda.
| |
Collapse
|
42
|
Maurya R, Kanakan A, Vasudevan JS, Chattopadhyay P, Pandey R. Infection outcome needs two to tango: human host and the pathogen. Brief Funct Genomics 2021; 21:90-102. [PMID: 34402498 PMCID: PMC8385967 DOI: 10.1093/bfgp/elab037] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/15/2021] [Accepted: 07/21/2021] [Indexed: 12/15/2022] Open
Abstract
Infectious diseases are potential drivers for human evolution, through a complex, continuous and dynamic interaction between the host and the pathogen/s. It is this dynamic interaction that contributes toward the clinical outcome of a pathogenic disease. These are modulated by contributions from the human genetic variants, transcriptional response (including noncoding RNA) and the pathogen’s genome architecture. Modern genomic tools and techniques have been crucial for the detection and genomic characterization of pathogens with respect to the emerging infectious diseases. Aided by next-generation sequencing (NGS), risk stratification of host population/s allows for the identification of susceptible subgroups and better disease management. Nevertheless, many challenges to a general understanding of host–pathogen interactions remain. In this review, we elucidate how a better understanding of the human host-pathogen interplay can substantially enhance, and in turn benefit from, current and future applications of multi-omics based approaches in infectious and rare diseases. This includes the RNA-level response, which modulates the disease severity and outcome. The need to understand the role of human genetic variants in disease severity and clinical outcome has been further highlighted during the Coronavirus disease 2019 (COVID-19) pandemic. This would enhance and contribute toward our future pandemic preparedness.
Collapse
Affiliation(s)
- Ranjeet Maurya
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi-110007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Akshay Kanakan
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi-110007, India
| | - Janani Srinivasa Vasudevan
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi-110007, India
| | - Partha Chattopadhyay
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi-110007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| | - Rajesh Pandey
- INtegrative GENomics of HOst-PathogEn (INGEN-HOPE) laboratory, CSIR-Institute of Genomics and Integrative Biology (CSIR-IGIB), Mall Road, Delhi-110007, India.,Academy of Scientific and Innovative Research (AcSIR), Ghaziabad-201002, India
| |
Collapse
|
43
|
Abstract
African apes harbor at least twelve Plasmodium species, some of which have been a source of human infection. It is now well established that Plasmodium falciparum emerged following the transmission of a gorilla parasite, perhaps within the last 10,000 years, while Plasmodium vivax emerged earlier from a parasite lineage that infected humans and apes in Africa before the Duffy-negative mutation eliminated the parasite from humans there. Compared to their ape relatives, both human parasites have greatly reduced genetic diversity and an excess of nonsynonymous mutations, consistent with severe genetic bottlenecks followed by rapid population expansion. A putative new Plasmodium species widespread in chimpanzees, gorillas, and bonobos places the origin of Plasmodium malariae in Africa. Here, we review what is known about the origins and evolutionary history of all human-infective Plasmodium species, the time and circumstances of their emergence, and the diversity, host specificity, and zoonotic potential of their ape counterparts.
Collapse
Affiliation(s)
- Paul M Sharp
- Institute of Evolutionary Biology and Centre for Immunity, Infection and Evolution, University of Edinburgh, EH9 3FL, United Kingdom
| | - Lindsey J Plenderleith
- Institute of Evolutionary Biology and Centre for Immunity, Infection and Evolution, University of Edinburgh, EH9 3FL, United Kingdom
| | - Beatrice H Hahn
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA;
| |
Collapse
|
44
|
Watson JA, Ndila CM, Uyoga S, Macharia A, Nyutu G, Mohammed S, Ngetsa C, Mturi N, Peshu N, Tsofa B, Rockett K, Leopold S, Kingston H, George EC, Maitland K, Day NPJ, Dondorp AM, Bejon P, Williams TN, Holmes CC, White NJ. Improving statistical power in severe malaria genetic association studies by augmenting phenotypic precision. eLife 2021; 10:e69698. [PMID: 34225842 PMCID: PMC8315799 DOI: 10.7554/elife.69698] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Accepted: 06/22/2021] [Indexed: 12/14/2022] Open
Abstract
Severe falciparum malaria has substantially affected human evolution. Genetic association studies of patients with clinically defined severe malaria and matched population controls have helped characterise human genetic susceptibility to severe malaria, but phenotypic imprecision compromises discovered associations. In areas of high malaria transmission, the diagnosis of severe malaria in young children and, in particular, the distinction from bacterial sepsis are imprecise. We developed a probabilistic diagnostic model of severe malaria using platelet and white count data. Under this model, we re-analysed clinical and genetic data from 2220 Kenyan children with clinically defined severe malaria and 3940 population controls, adjusting for phenotype mis-labelling. Our model, validated by the distribution of sickle trait, estimated that approximately one-third of cases did not have severe malaria. We propose a data-tilting approach for case-control studies with phenotype mis-labelling and show that this reduces false discovery rates and improves statistical power in genome-wide association studies.
Collapse
Affiliation(s)
- James A Watson
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol UniversityBangkokThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Carolyne M Ndila
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol UniversityBangkokThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Sophie Uyoga
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-CoastKilifiKenya
| | - Alexander Macharia
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-CoastKilifiKenya
| | - Gideon Nyutu
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-CoastKilifiKenya
| | - Shebe Mohammed
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-CoastKilifiKenya
| | - Caroline Ngetsa
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-CoastKilifiKenya
| | - Neema Mturi
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-CoastKilifiKenya
| | - Norbert Peshu
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-CoastKilifiKenya
| | - Benjamin Tsofa
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-CoastKilifiKenya
| | - Kirk Rockett
- The Wellcome Sanger InstituteCambridgeUnited Kingdom
- Wellcome Trust Centre for Human Genetics, University of OxfordOxfordUnited Kingdom
| | - Stije Leopold
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol UniversityBangkokThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Hugh Kingston
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol UniversityBangkokThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Elizabeth C George
- Medical Research Council Clinical Trials Unit, University College LondonLondonUnited Kingdom
| | - Kathryn Maitland
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-CoastKilifiKenya
- Institute of Global Health Innovation, Imperial College, LondonLondonUnited Kingdom
| | - Nicholas PJ Day
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol UniversityBangkokThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Arjen M Dondorp
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol UniversityBangkokThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| | - Philip Bejon
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-CoastKilifiKenya
| | - Thomas N Williams
- KEMRI-Wellcome Trust Research Programme, Centre for Geographic Medicine Research-CoastKilifiKenya
- Institute of Global Health Innovation, Imperial College, LondonLondonUnited Kingdom
| | - Chris C Holmes
- Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
- Department of Statistics, University of OxfordOxfordUnited Kingdom
| | - Nicholas J White
- Mahidol Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol UniversityBangkokThailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of Medicine, University of OxfordOxfordUnited Kingdom
| |
Collapse
|
45
|
de Villiers KA, Egan TJ. Heme Detoxification in the Malaria Parasite: A Target for Antimalarial Drug Development. Acc Chem Res 2021; 54:2649-2659. [PMID: 33982570 DOI: 10.1021/acs.accounts.1c00154] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Over the last century, malaria deaths have decreased by more than 85%. Nonetheless, there were 405 000 deaths in 2018, mostly resulting from Plasmodium falciparum infection. In the 21st century, much of the advance has arisen from the deployment of insecticide-treated bed nets and artemisinin combination therapy. However, over the past few decades parasites with a delayed artemisinin clearance phenotype have appeared in Southeast Asia, threatening further gains. The effort to find new drugs is thus urgent. A prominent process in blood stage malaria parasites, which we contend remains a viable drug target, is hemozoin formation. This crystalline material consisting of heme can be readily seen when parasites are viewed microscopically. The process of its formation in the parasite, however, is still not fully understood.In early work, we recognized hemozoin formation as a biomineralization process. We have subsequently investigated the kinetics of synthetic hemozoin (β-hematin) crystallization catalyzed at lipid-aqueous interfaces under biomimetic conditions. This led us to the use of neutral detergent-based high-throughput screening (HTS) for inhibitors of β-hematin formation. A good hit rate against malaria parasites was obtained. Simultaneously, we developed a pyridine-based assay which proved successful in measuring the concentrations of hematin not converted to β-hematin.The pyridine assay was adapted to determine the effects of chloroquine and other clinical antimalarials on hemozoin formation in the cell. This permitted the determination of the dose-dependent amounts of exchangeable heme and hemozoin in P. falciparum for the first time. These studies have shown that hemozoin inhibitors cause a dose-dependent increase in exchangeable heme, correlated with decreased parasite survival. Electron spectroscopic imaging (ESI) showed a relocation of heme iron into the parasite cytoplasm, while electron microscopy provided evidence of the disruption of hemozoin crystals. This cellular assay was subsequently extended to top-ranked hits from a wide range of scaffolds found by HTS. Intriguingly, the amounts of exchangeable heme at the parasite growth IC50 values of these scaffolds showed substantial variation. The amount of exchangeable heme was found to be correlated with the amount of inhibitor accumulated in the parasitized red blood cell. This suggests that heme-inhibitor complexes, rather than free heme, lead to parasite death. This was supported by ESI using a Br-containing compound which showed the colocalization of Fe and Br as well as by confocal Raman microscopy which confirmed the presence of a complex in the parasite. Current evidence indicates that inhibitors block hemozoin formation by surface adsorption. Indeed, we have successfully introduced molecular docking with hemozoin to find new inhibitors. It follows that the resulting increase in free heme leads to the formation of the parasiticidal heme-inhibitor complex. We have reported crystal structures of heme-drug complexes for several aryl methanol antimalarials in nonaqueous media. These form coordination complexes but most other inhibitors interact noncovalently, and the determination of their structures remains a major challenge.It is our view that key future developments will include improved assays to measure cellular heme levels, better in silico approaches for predicting β-hematin inhibition, and a concerted effort to determine the structure and properties of heme-inhibitor complexes.
Collapse
Affiliation(s)
- Katherine A. de Villiers
- Department of Chemistry and Polymer Science, Stellenbosch University, Private Bag, Matieland 7600, South Africa
| | - Timothy J. Egan
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch 7701, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Observatory, Cape Town 7945, South Africa
| |
Collapse
|
46
|
Li J, Chu XH, Wang XY, Feng BM, Yu ZX. Aging affects artemisinin synthesis in Artemisia annua. Sci Rep 2021; 11:11297. [PMID: 34050229 PMCID: PMC8163859 DOI: 10.1038/s41598-021-90807-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 05/18/2021] [Indexed: 11/29/2022] Open
Abstract
Artemisinin (ART) is the most effective component in malaria treatment, however, the extremely low content restricts its clinical application. Therefore, it is urgent to increase the yield of ART. ART gradually accumulates with aging, small RNA (sRNA) and transcriptome analysis were applied on the leaves of 2-week-old (2 w) and 3-month-old (3 m) A. annua respectively. Among all the annotated sRNAs, 125 were upregulated and 128 downregulated in the 3 m sample compared to the 2 w one. Whereas 2183 genes were upregulated and 2156 downregulated. Notably, the level of miR156 and several annotated miRNAs gradually decreased while SPLs increased. In addition, the genes on ART biosynthesis pathway were significantly upregulated including ADS, CYP71AV1, ADH1, DBR2 and ALDH1, and so were the positive transcription factors like AaERF1, AaORA and AaWRKY1 indicating that age influences the ART biosynthesis by activating the expression of the synthesizing genes as well as positive transcription factors. This study contributes to reveal the regulatory effects of age on ART biosynthesis both in sRNA and transcription levels.
Collapse
Affiliation(s)
- Jiao Li
- College of Life Sciences and Biotechnology, Dalian University, 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, Liaoning, China
| | - Xiao-Hui Chu
- College of Life Sciences and Biotechnology, Dalian University, 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, Liaoning, China
| | - Xiao-Yu Wang
- College of Life Sciences and Biotechnology, Dalian University, 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, Liaoning, China
| | - Bao-Min Feng
- College of Life Sciences and Biotechnology, Dalian University, 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, Liaoning, China
| | - Zong-Xia Yu
- College of Life Sciences and Biotechnology, Dalian University, 10 Xuefu Street, Dalian Economic and Technological Development Zone, Dalian, 116622, Liaoning, China.
| |
Collapse
|
47
|
Petrou A, Fesatidou M, Geronikaki A. Thiazole Ring-A Biologically Active Scaffold. Molecules 2021; 26:3166. [PMID: 34070661 PMCID: PMC8198555 DOI: 10.3390/molecules26113166] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/15/2021] [Accepted: 05/20/2021] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Thiazole is a good pharmacophore nucleus due to its various pharmaceutical applications. Its derivatives have a wide range of biological activities such as antioxidant, analgesic, and antimicrobial including antibacterial, antifungal, antimalarial, anticancer, antiallergic, antihypertensive, anti-inflammatory, and antipsychotic. Indeed, the thiazole scaffold is contained in more than 18 FDA-approved drugs as well as in numerous experimental drugs. OBJECTIVE To summarize recent literature on the biological activities of thiazole ring-containing compounds Methods: A literature survey regarding the topics from the year 2015 up to now was carried out. Older publications were not included, since they were previously analyzed in available peer reviews. RESULTS Nearly 124 research articles were found, critically analyzed, and arranged regarding the synthesis and biological activities of thiazoles derivatives in the last 5 years.
Collapse
Affiliation(s)
| | | | - Athina Geronikaki
- School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.P.); (M.F.)
| |
Collapse
|
48
|
Tennessen JA, Duraisingh MT. Three Signatures of Adaptive Polymorphism Exemplified by Malaria-Associated Genes. Mol Biol Evol 2021; 38:1356-1371. [PMID: 33185667 PMCID: PMC8042748 DOI: 10.1093/molbev/msaa294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Malaria has been one of the strongest selective pressures on our species. Many of the best-characterized cases of adaptive evolution in humans are in genes tied to malaria resistance. However, the complex evolutionary patterns at these genes are poorly captured by standard scans for nonneutral evolution. Here, we present three new statistical tests for selection based on population genetic patterns that are observed more than once among key malaria resistance loci. We assess these tests using forward-time evolutionary simulations and apply them to global whole-genome sequencing data from humans, and thus we show that they are effective at distinguishing selection from neutrality. Each test captures a distinct evolutionary pattern, here called Divergent Haplotypes, Repeated Shifts, and Arrested Sweeps, associated with a particular period of human prehistory. We clarify the selective signatures at known malaria-relevant genes and identify additional genes showing similar adaptive evolutionary patterns. Among our top outliers, we see a particular enrichment for genes involved in erythropoiesis and for genes previously associated with malaria resistance, consistent with a major role for malaria in shaping these patterns of genetic diversity. Polymorphisms at these genes are likely to impact resistance to malaria infection and contribute to ongoing host-parasite coevolutionary dynamics.
Collapse
|
49
|
Athni TS, Shocket MS, Couper LI, Nova N, Caldwell IR, Caldwell JM, Childress JN, Childs ML, De Leo GA, Kirk DG, MacDonald AJ, Olivarius K, Pickel DG, Roberts SO, Winokur OC, Young HS, Cheng J, Grant EA, Kurzner PM, Kyaw S, Lin BJ, López RC, Massihpour DS, Olsen EC, Roache M, Ruiz A, Schultz EA, Shafat M, Spencer RL, Bharti N, Mordecai EA. The influence of vector-borne disease on human history: socio-ecological mechanisms. Ecol Lett 2021; 24:829-846. [PMID: 33501751 PMCID: PMC7969392 DOI: 10.1111/ele.13675] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/08/2020] [Accepted: 12/09/2020] [Indexed: 01/14/2023]
Abstract
Vector-borne diseases (VBDs) are embedded within complex socio-ecological systems. While research has traditionally focused on the direct effects of VBDs on human morbidity and mortality, it is increasingly clear that their impacts are much more pervasive. VBDs are dynamically linked to feedbacks between environmental conditions, vector ecology, disease burden, and societal responses that drive transmission. As a result, VBDs have had profound influence on human history. Mechanisms include: (1) killing or debilitating large numbers of people, with demographic and population-level impacts; (2) differentially affecting populations based on prior history of disease exposure, immunity, and resistance; (3) being weaponised to promote or justify hierarchies of power, colonialism, racism, classism and sexism; (4) catalysing changes in ideas, institutions, infrastructure, technologies and social practices in efforts to control disease outbreaks; and (5) changing human relationships with the land and environment. We use historical and archaeological evidence interpreted through an ecological lens to illustrate how VBDs have shaped society and culture, focusing on case studies from four pertinent VBDs: plague, malaria, yellow fever and trypanosomiasis. By comparing across diseases, time periods and geographies, we highlight the enormous scope and variety of mechanisms by which VBDs have influenced human history.
Collapse
Affiliation(s)
- Tejas S. Athni
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Marta S. Shocket
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Ecology and Evolutionary Biology, University of California, Los Angeles, CA, USA
| | - Lisa I. Couper
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Nicole Nova
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Iain R. Caldwell
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, Queensland, Australia
| | - Jamie M. Caldwell
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Biology, University of Hawaii at Manoa, Honolulu, HI, USA
| | - Jasmine N. Childress
- Department of Ecology, Evolution, and Marine Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Marissa L. Childs
- Emmett Interdisciplinary Program in Environment and Resources, Stanford University, Stanford, CA, USA
| | - Giulio A. De Leo
- Hopkins Marine Station of Stanford University, Pacific Grove, CA, USA
- Woods Institute for the Environment, Stanford University, Stanford, CA, USA
| | - Devin G. Kirk
- Department of Biology, Stanford University, Stanford, CA, USA
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Andrew J. MacDonald
- Bren School of Environmental Science and Management, University of California, Santa Barbara, CA, USA
- Earth Research Institute, University of California, Santa Barbara, CA, USA
| | | | - David G. Pickel
- Department of Classics, Stanford University, Stanford, CA, USA
| | | | - Olivia C. Winokur
- Department of Pathology, Microbiology, and Immunology, School of Veterinary Medicine, University of California, Davis, CA, USA
| | - Hillary S. Young
- Department of Ecology, Evolution, and Marine Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Julian Cheng
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | | | - Saw Kyaw
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Bradford J. Lin
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | | | - Erica C. Olsen
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Maggie Roache
- Department of Biology, Stanford University, Stanford, CA, USA
| | - Angie Ruiz
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Muskan Shafat
- Department of Biology, Stanford University, Stanford, CA, USA
| | | | - Nita Bharti
- Department of Biology, Center for Infectious Disease Dynamics, Penn State University, University Park, PA, USA
| | | |
Collapse
|
50
|
Song Y, Shan L, Gbyli R, Liu W, Strowig T, Patel A, Fu X, Wang X, Xu ML, Gao Y, Qin A, Bruscia EM, Tebaldi T, Biancon G, Mamillapalli P, Urbonas D, Eynon E, Gonzalez DG, Chen J, Krause DS, Alderman J, Halene S, Flavell RA. Combined liver-cytokine humanization comes to the rescue of circulating human red blood cells. Science 2021; 371:1019-1025. [PMID: 33674488 DOI: 10.1126/science.abe2485] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Accepted: 02/01/2021] [Indexed: 12/13/2022]
Abstract
In vivo models that recapitulate human erythropoiesis with persistence of circulating red blood cells (RBCs) have remained elusive. We report an immunodeficient murine model in which combined human liver and cytokine humanization confer enhanced human erythropoiesis and RBC survival in the circulation. We deleted the fumarylacetoacetate hydrolase (Fah) gene in MISTRG mice expressing several human cytokines in place of their murine counterparts. Liver humanization by intrasplenic injection of human hepatocytes (huHep) eliminated murine complement C3 and reduced murine Kupffer cell density. Engraftment of human sickle cell disease (SCD)-derived hematopoietic stem cells in huHepMISTRGFah -/- mice resulted in vaso-occlusion that replicated acute SCD pathology. Combined liver-cytokine-humanized mice will facilitate the study of diseases afflicting RBCs, including bone marrow failure, hemoglobinopathies, and malaria, and also preclinical testing of therapies.
Collapse
Affiliation(s)
- Yuanbin Song
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.,Department of Hematologic Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Liang Shan
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA. .,Department of Medicine, Pathology and Immunology, Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Rana Gbyli
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - Wei Liu
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - Till Strowig
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA.,Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Amisha Patel
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - Xiaoying Fu
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.,Department of Laboratory Medicine, Shenzhen Children's Hospital, Shenzhen, People's Republic of China
| | - Xiaman Wang
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.,Department of Hematology, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China
| | - Mina L Xu
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Yimeng Gao
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - Ashley Qin
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - Emanuela M Bruscia
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT, USA
| | - Toma Tebaldi
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.,Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Giulia Biancon
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - Padmavathi Mamillapalli
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA.,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - David Urbonas
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Elizabeth Eynon
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - David G Gonzalez
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA
| | - Jie Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Diane S Krause
- Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA.,Department of Pathology, Yale University School of Medicine, New Haven, CT, USA.,Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Jonathan Alderman
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, and Yale Center for RNA Science and Medicine, Yale University School of Medicine, New Haven, CT, USA. .,Yale Stem Cell Center, Yale University School of Medicine, New Haven, CT, USA
| | - Richard A Flavell
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA. .,Howard Hughes Medical Institute, Yale University, New Haven, CT, USA
| |
Collapse
|